WO2018153969A1 - Combination of atr kinase inhibitors with radium-223 salt - Google Patents

Combination of atr kinase inhibitors with radium-223 salt Download PDF

Info

Publication number
WO2018153969A1
WO2018153969A1 PCT/EP2018/054362 EP2018054362W WO2018153969A1 WO 2018153969 A1 WO2018153969 A1 WO 2018153969A1 EP 2018054362 W EP2018054362 W EP 2018054362W WO 2018153969 A1 WO2018153969 A1 WO 2018153969A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrazol
naphthyridine
methylmorpholin
morpholin
naphthyridin
Prior art date
Application number
PCT/EP2018/054362
Other languages
French (fr)
Inventor
Antje Margret WENGNER
Arne Scholz
Original Assignee
Bayer Aktiengesellschaft
Bayer Pharma Aktiengesellschaft
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 Bayer Aktiengesellschaft, Bayer Pharma Aktiengesellschaft filed Critical Bayer Aktiengesellschaft
Publication of WO2018153969A1 publication Critical patent/WO2018153969A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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 covers combinations of at least two components, component A and component B, comprising component A being an ATR kinase inhibitor, particularly Compound A, and component B being radium-223, particularly a pharmaceutically acceptable salt of radium-223.
  • component A being an ATR kinase inhibitor, particularly Compound A
  • component B being radium-223, particularly a pharmaceutically acceptable salt of radium-223.
  • Another aspect of the present invention covers the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a hyper - proliferative disease, particurlarly for the treatment of prostate cancer and/or bone metastases thereof.
  • Cancer is the second most prevalent cause of death in the United States, causing 450,000 deaths per year. While substantial progress has been made in identifying some of the likely environmental and hereditary causes of cancer, there is a need for additional therapeutic modalities that target cancer and related diseases. In particular there is a need for therapeutic methods for treating diseases associated with dysregulated growth / proliferation.
  • Cancer is a complex disease arising after a selection process for cells with acquired functional capabilities like enhanced survival / resistance towards apoptosis and a limitless proliferative potential. Thus, it is preferred to develop drugs for cancer therapy addressing distinct features of established tumors.
  • DDR DNA damage response
  • Proteins that directly recognize aberrant DNA structures such as the MRE11-Rad50-Nbsl complex recognizing DNA double strand breaks by binding to double-stranded DNA ends, or RPA (replication protein A) binding to single stranded DNA, recruit and activate the most upstream kinases of the DDR pathway, ATM (ataxia-telangiectasia mutated), ATR (ATM-and Rad3-related, UniProtKB/Swiss-Prot Q13535), and DNA-PKcs (DNA-dependent protein kinase).
  • ATM is primarily activated by DNA double strand breaks
  • DNA-PKcs is mainly involved in non-homologous end joining process of DNA repair
  • ATR responds to a broad spectrum of DNA damage, including double- strand breaks and lesions derived from interference with DNA replication.
  • Major components of downstream signaling of ATM include Chk2 and p53, whereas ATR signaling involves Chkl and cdc25.
  • Knockout of the ATR gene in mice is embryonically lethal and ATR knockout cells develop chromosome breaks and undergo apoptosis [E.J. Brown, D. Baltimore: ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 14, 397-402, 2000].
  • ATM is not essential for cell survival although ATM knockout cells are hypersensitive to ionizing radiation and agents which cause DNA double-strand breaks.
  • ATR which forms a complex with ATRIP (ATR-interacting protein, UniProtKB/Swiss-Prot Q8WXE1) is mainly activated by long stretches of single-stranded DNA which are generated by the continuing DNA unwinding activity of helicases upon stalled replication.
  • This replication stress with stalled replication forks may be induced by ultraviolet light, certain chemotherapeutic drugs, hydroxyurea, or aberrant oncogenic signaling resulting in increased replication initiation or origin firing.
  • Activation of ATR results in inhibition of the cell cycle in S or G2 phase via the Chkl-cdc25 pathway and in suppression of late origin firing.
  • the cell gains time to resolve the replication stress and, eventually, to restart replication after the source of stress has been removed.
  • As the ATR pathway ensures cell survival after replication stress it potentially contributes to resistance to chemotherapy. Thus inhibition of ATR kinase activity could be useful for cancer treatment.
  • oncogene-driven tumor cells e.g. Ras mutation/upregulation, Myc upregulation, CyclinE overexpression
  • ATR suppression in Ras oncogene driven cells was reported to result in substantial tumor cell killing [O. Gilad, BY Nabet, et al.: Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. Cancer Res. 70, 9693-9702, 2010].
  • a healthy normal cell which has ATM and ATR pathways in parallel, arrests in Gl phase of the cell cycle upon induced DNA damage even in presence of an ATR inhibitor.
  • a tumor cell which most often deficient in ATM and/or p53 signaling relies on the ATR pathway and undergoes cell death in presence of an ATR inhibitor. This suggests that ATR inhibitors may be used for the treatment of tumors with deficient ATM signaling and/or p53 function.
  • inhibitors of ATR kinase represent valuable compounds that should complement therapeutic options not only as single agents but also in combination with other drugs.
  • Radium-223 dichloride (Ra 223 Ci2) (Xofigo®) uses alpha radiation from radium-223 decay to kill cancer cells. It targets to bone tissue by virtue of its chemical similarity to calcium. It has an effect over a range of 2-10 cells and causes less damage to surrounding healthy tissues compared to current radiation therapy based on beta or gamma radiation. Significant increase in median overall survival was demonstrated in Phase III clinical trials and radium-223 dichloride (Xofigo) was approved as a treatment for castration-resistant prostate cancer (CRPC) patients with symptomatic bone metastases.
  • CRPC castration-resistant prostate cancer
  • a preferred suitable pharmaceutically acceptable salt of radium-223 is the dichloride (Ra 223 Cl2).
  • Radium-223 dichloride is a novel, targeted alpha-emitter that selectively binds to areas of increased bone turnover in bone metastases and emits high-energy alpha-particles of extremely short ( ⁇ 100 ⁇ ) range (Bruland O. S. et al., High-linear energy transfer irradiation targeted to skeletal metastases by the alpha-emitter 223 Ra: adjuvant or alternative to conventional modalities?, Clin. Cancer Res. 2006; 12: 6250s-7s). It is the first targeted alpha-emitter approved for the treatment of prostate cancer with bone metastases.
  • radium-223 is bound into newly formed bone stroma, especially within the microenvironment of osteoblastic or sclerotic metastases (Henriksen G. et al., Significant antitumor effect from bone-seeking, alpha-particle-emitting (223)Ra demonstrated in an experimental skeletal metastases model, Cancer Res. 2002; 62: 3120-3125; Henriksen G. et al., Targeting of osseous sites with alpha-emitting 223Ra: comparison with the beta-emitter 89Sr in mice, J. Nucl. Med 2003; 44: 252-59).
  • the high-energy alpha-particle radiation induces mainly double-strand DNA breaks resulting in a potent and highly localized cytotoxic effect in the target areas containing metastatic cancer cells
  • Lewington V.J. Bone- seeking radionuclides for therapy, J. Nucl. Med 2005; 46 (suppl 1): 38S-47S; Liepe K., Alpharadin, a 223 Ra-based alpha-particle-emitting pharmaceutical for the treatment of bone metastases in patients with cancer, Curr. Opin. Investig. Drugs 2009; 10: 1346-58; McDevitt M.R. et al., Radioimmunotherapy with alpha- emitting nuclides, Eur. J. Nucl. Med.
  • the short path length of the alpha-particles also means that toxicity to adjacent healthy tissue and particularly the bone marrow may be reduced (Kerr C, (223)Ra targets skeletal metastases and spares normal tissue, Lancet Oncol. 2002; 3: 453; Li Y., Russell P.J., Allen B.J., Targeted alpha-therapy for control of micrometastatic prostate cancer, Expert Rev. Anticancer Ther. 2004; 4: 459-68).
  • Radium-223 has demonstrated a favorable safety profile with minimal myelotoxicity in phase 1 and 2 studies of patients with bone metastases (Nilsson S. et al., First clinical experience with alpha-emitting radium-223 in the treatment of skeletal metastases, Clin. Cancer Res. 2005; 11 : 4451-59; Nilsson S. et al., Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II study, Lancet Oncol. 2007; 8: 587-94).
  • radium-223 reduces pain, improves disease-related biomarkers (e.g., bone alkaline phosphatase [ALP] and prostate-specific antigen [PSA]), and have suggested a survival benefit in patients with CRPC and bone metastases (Parker C. et al., A randomized, double-blind, dose-finding, multicenter, phase 2 study of radium chloride (Ra-223) in patients with bone metastases and castration-resistant prostate cancer, Eur Urol. 2013 Feb; 63(2): 189-97; Nilsson S.
  • ALP bone alkaline phosphatase
  • PSA prostate-specific antigen
  • the ALSYMPCA (ALpharadin in SYMptomatic Prostate CAncer patients) trial provides proof of principle for the role of targeted alpha-emitters in oncology.
  • radium-223 significantly prolonged overall survival with a 30.5% reduction in risk of death compared with placebo in patients with CRPC (Castration Resistant Prostate Cancer) and bone metastases.
  • Median survival with radium-223 was longer than placebo by 2.8 months.
  • All main secondary efficacy endpoints were statistically significant and favored treatment with radium-223, including the clinically defined endpoint of time to first skeletal-related event, which was significantly prolonged in patients receiving radium-223 (C. Parker et al., Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer , The New England Journal of Medicine 369(3):213-23).
  • radium-223 inhibits osseous prostate cancer growth by dual targeting of cancer cells and bone microenvironment in mouse models.
  • a substantial percentage of cancer patients is affected by skeletal metastases.
  • As many as 85% of patients with advanced lung, prostate and breast carcinoma develop bony metastases (Garret R., Semin. Oncol. 72, 3433 -3435 (1993) Bone destruction in cancer; Nielsen, OS, Munro AJ, Tannock IF. J Clin Oncol 9, 509-5 24 (1991)).
  • Established treatments such as hormone therapy, chemotherapy and external radiotherapy often causes temporary responses, but ultimately most bone cancer patients experience relapses (Kanis JA.
  • 223 Ra is used as an a-emitting radiopharmaceutical for targeting of calcified tissues, e.g. bone surfaces and osseous tumor lesions. It can be suitable as a bone seeking radiopharmaceutical. It thus may be used for prophylactic cancer treatment by delivering a focused dose to bone surfaces in patients with a high probability of having undetected micro metastases at bone surfaces. Another example of its potential use would be in the treatment of painful osseous sites.
  • the alkaline-earth radionuclide radium-223 is useful for the targeting of calcified tissues, e.g., bone and a pharmaceutical acceptable solution comprising 223 Ra.
  • the alkaline-earth radionuclide radium-223 is suitable for the use of the nuclide as a cationic species and/or associated to a chelator or another form of a carrier molecule with affinity for calcified tissues. Thus may be combined with a chelator that can be subsequently conjugated to a molecule with affinity for calcified tissues.
  • the diseases where the radioisotopes could be used includes, but are not limited to skeletal metastases of prostate-, breast-, kidney- and lung cancer as well as primary bone cancer and also multiple myeloma.
  • the present invention provides combinations of at least two components, component A and component B, comprising component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223, particularly a pharmaceutically acceptable inorganic salt of the alkaline-earth radionuclide radium-223.
  • the present invention concerns combinations of at least two components A and B, comprising component A being an inhibitor of ATR kinase, particularly Compound A, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
  • kits comprising:
  • component A one or more ATR kinase inhibitors as described herein, particularly
  • component B a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof; and, optionally, in which kit optionally either or both of said components A and B in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially.
  • the components may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
  • the present invention concerns the combinations as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra.
  • the present invention covers the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra.
  • the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra, using an effective amount of the combinations as described herein.
  • inhibitor of ATR kinase or the term “ATR kinase inhibitor” as used herein means any compound that inhibits ATR kinase. Examples of such compounds are described infra ("COMPONENT A OF THE COMBINATION").
  • halogen atom halo- or Hal-
  • fluorine chlorine, bromine or iodine atom.
  • Ci-C6-alkyl is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, 4, 5, or 6 carbon atoms, e.g. a methyl, ethyl, propyl, butyl, pentyl, hexyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl, iso-pentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neo-pentyl, 1,1-dimethylpropyl, 4- methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3- dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3
  • said group has 1, 2, 3 or 4 carbon atoms ("Ci-C t-alkyl”), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert- butyl group, more particularly 1, 2 or 3 carbon atoms (“Ci-C 3 -alkyl”), e.g. a methyl, ethyl, n- propyl or iso-propyl group.
  • Si-C t-alkyl 1, 2, 3 or 4 carbon atoms
  • Ci-C6-haloalkyl is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C6-alkyl” is defined supra, and in which one or more hydrogen atoms is replaced by a halogen atom, in identically or differently, i.e. one halogen atom being independent from another. Particularly, said halogen atom is F.
  • Said C 1 -C6- haloalkyl group is, for example, -CF3, -CHF2, -CH2F, -CF2CF3 or -CH2CF3.
  • Ci-C t-hydroxyaikyl is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C t-alkyl” is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a hydroxymethyl, 1- hydroxyethyl, 2-hydroxyethyl, 1,2-dihydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2,3- dihydroxypropyl, l,3-dihydroxypropan-2-yl, 3-hydroxy-2-methyl-propyl, 2-hydroxy-2-methyl- propyl, l-hydroxy-2-methyl-propyl group.
  • a hydroxymethyl 1- hydroxyethyl, 2-hydroxyethyl, 1,2-dihydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2,3- dihydroxypropyl, l,3-dihydroxypropan-2-yl, 3-hydroxy-2-methyl-propyl
  • Ci-C6-alkoxy is to be understood as meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-alkyl, in which the term “alkyl” is defined supra, e.g. a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy, sec-butoxy, pentoxy, iso-pentoxy, or n-hexoxy group, or an isomer thereof.
  • said "Ci-C6-alkoxy” can contain 1, 2, 3, 4 or 5 carbon atoms, (a “Ci-Cs-alkoxy”), preferably 1, 2, 3 or 4 carbon atoms ("d-C ⁇ alkoxy").
  • Ci-C6-haloalkoxy is to be understood as meaning a linear or branched, saturated, monovalent Ci-C6-alkoxy group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a halogen atom.
  • said halogen atom is F.
  • Said Ci-C6-haloalkoxy group is, for example, -OCF3, -OCHF2, -OCH2F, -OCF2CF3, or
  • C 2 -C6-alkenyl is to be understood as meaning a linear or branched, monovalent hydrocarbon group, which contains one or more double bonds, and which has 2, 3, 4, 5 or 6 carbon atoms or 2, 3 or 4 carbon atoms ("C 2 -C4-alkenyl), particularly 2 or 3 carbon atoms (“C 2 - C3-alkenyl”), it being understood that in the case in which said alkenyl group contains more than one double bond, then said double bonds may be isolated from, or conjugated with, each other.
  • Said alkenyl group is, for example, a vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, homoallyl, (E)-but-2-enyl, (Z)-but-2-enyl, (E)-but-l-enyl, (Z)-but- l-enyl, pent-4-enyl, (E)-pent- 3-enyl, (Z)-pent-3-enyl, (E)-pent-2-enyl, (Z)-pent-2-enyl, (E)-pent-l-enyl, (Z)-pent- l-enyl, hex- 5-enyl, (E)-hex-4-enyl, (Z)-hex-4-enyl, (E)-hex-3-enyl, (Z)-hex-3-enyl, (E)-hex-2-enyl, (Z)-hex- 2-enyl
  • C3-Cio-cycloalkyl is to be understood as meaning a saturated, monovalent, mono-, or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms ("C3-C10- cycloalkyl").
  • Said C3-Cio-cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic hydrocarbon ring, e.g. a perhydropentalenylene or decalin ring.
  • said ring contains 3, 4, 5 or 6 carbon atoms ("C3-C6-cycloalkyl”), preferably cyclopropyl.
  • said 3- to 10-membered heterocycloalkyl can contain 2, 3, 4, or 5 carbon atoms, and one or more of the above-mentioned heteroatom-containing groups (a "3- to 6-membered heterocycloalkyl"), more particularly said heterocycloalkyl can contain 4 or 5 carbon atoms, and one or more of the above-mentioned heteroatom-containing groups (a "5- to 6-membered heterocycloalkyl").
  • said heterocycloalkyl can be a 4-membered ring, such as an azetidinyl, oxetanyl, or a 5-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl, or a 7-membered ring, such as a diazepanyl ring, for example.
  • said heterocycloalkyl can be benzo fused.
  • the 3- to 6-membered heterocycloalkyl is a tetrahydrofuranyl, tetrahydropyranyl or piperazinyl
  • Said heterocycloalkyl can be bicyclic, such as, without being limited thereto, a 5,5-membered ring, e.g. a hexahydrocyclopenta[c]pyrrol-2(lH)-yl ring, or a 5,6-membered bicyclic ring, e.g. a hexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl ring.
  • said nitrogen atom-containing ring can be partially unsaturated, i.e. it can contain one or more double bonds, such as, without being limited thereto, a 2,5-dihydro-lH- pyrrolyl, 4H-[l,3,4]thiadiazinyl, 4,5-dihydrooxazolyl, or 4H-[l,4]thiazinyl ring, for example, or, it may be benzo-fused, such as, without being limited thereto, a dihydroisoquinolinyl ring, for example.
  • heterocycloalkenyl may contain one or more double bonds, e.g. 4H-pyranyl, 2H-pyranyl, 3,6- dihydro-2H-pyran-4-yl, 3,6-dihydro-2H-thiopyran-4-yl, l,2,3,6-tetrahydropyridin-4-yl, 3H- diazirinyl, 2,5-dihydro-lH-pyrrolyl, [l,3]dioxolyl, 4H-[l,3,4]thiadiazinyl, 2,5-dihydrofuranyl, 2,3-dihydrofuranyl, 2,5-dihydrothiophenyl, 2,3-dihydrothiophenyl, 4,5-dihydrooxazolyl, 4H- [l,4]thiazinyl or 5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl group or it may be be be
  • heteroaryl is understood as meaning a monovalent, monocyclic- , bicyclic- or tricyclic aromatic ring system having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms (a "5- to 14- membered heteroaryl” group), 5 or 6 or 9 or 10 ring atoms (a "5- to 10-membered heteroaryl” group) or particularly 5 or 6 ring atoms ("5- to 6-membered heteroaryl” group), and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur, and in addition in each case can be benzocondensed.
  • heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, thia-4H-pyrazolyl etc., and benzo derivatives thereof, such as, for example, benzofuranyl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl, etc.; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc., and benzo derivatives thereof, such as, for example, quinolinyl, quinazolinyl, isoquinolinyl, etc.;
  • the heteroarylic or heteroarylenic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof.
  • the term pyridinyl or pyridinylene includes pyridin-2-yl, pyridin-2-ylene, pyridin-3-yl, pyridin-3-ylene, pyridin-4-yl and pyridin-4-ylene; or the term thienyl or thienylene includes thien-2-yl, thien-2-ylene, thien-3-yl and thien-3-ylene.
  • Ci-Ce as used throughout this text, e.g. in the context of the definition of "C1-C6- alkyl", “Ci-C6-haloalkyl", “Ci-C6-alkoxy” or “Ci-C6-haloalkoxy” is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 6, i.e. 1, 2, 3, 4, 5, or 6 carbon atoms. It is to be understood further that said term “Ci-Ce” is to be interpreted as any sub-range comprised therein, e.g.
  • C1-C6 C2-C5 , C3-C4 , C1-C2 , C1-C3 , C1-C4 , C1-C5; particularly C1-C2, C1-C3, C1-C4, C1-C5, C1-C6; more particularly C1-C4; in the case of "Ci-C6-haloalkyl” or "C1-C6- haloalkoxy” even more particularly C1-C2.
  • C 2 -C6 as used throughout this text, e.g.
  • C2-C6-alkenyl and “C2-C6-alkynyl”
  • C2-C6-alkynyl is to be understood as meaning an alkenyl group or an alkynyl group having a finite number of carbon atoms of 2 to 6, i.e. 2, 3, 4, 5, or 6 carbon atoms.
  • C 2 -C6 is to be interpreted as any subrange comprised therein, e.g. C2-C6, C3-C5, C3-C4, C2-C3, C2-C4, C2-C5; particularly C2-C3.
  • C3-C6 as used throughout this text, e.g. in the context of the definition of "C3-C6-cycloalkyl”, is to be understood as meaning a cycloalkyl group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term “C3-C6” is to be interpreted as any sub-range comprised therein, e.g. C3-C6, C4-C5, C3-C5, C3-C4, C 4 -C 6 , C 5 -C 6 ; particularly C 3 -C 6 .
  • C 2 -C4-alkenyl is to be understood as meaning a alkenyl group having a finite number of carbon atoms of 2 to 4, i.e. 2, 3 or 4 carbon atoms. It is to be understood further that said term “C 2 -C4" is to be interpreted as any sub-range comprised therein, e.g. C2-C4, C2-C3, C3-C4.
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.
  • stable compound' or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • the term "one or more”, e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning “one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two".
  • the invention also includes all suitable isotopic variations of the compound of component A, particularly of Compound A.
  • An isotopic variation of the compound of component A is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature.
  • isotopes that can be incorporated into the compound of component A include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), U C, 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F,
  • isotopic variations of the compound of component A are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon- 14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances.
  • isotopic variations of the compound of component A can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.
  • the compounds of component A may contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired.
  • Asymmetric carbon atoms may be present in the (R) or (S) configuration, resulting in racemic mixtures in the case of a single asymmetric centre, and diastereomeric mixtures in the case of multiple asymmetric centres.
  • asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
  • the compounds of component A may contain sulphur atoms which are asymmetric, such as an as mmetric sulphoxide or sulphoximine group, of structure:
  • Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention.
  • Preferred compounds of component A are those which produce the more desirable biological activity, particularly preferred is Compound A.
  • Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of component A are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers.
  • appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid.
  • Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation.
  • the optically active bases or acids are then liberated from the separated diastereomeric salts.
  • a different process for separation of optical isomers involves the use of chiral chromatography (e.g., chiral HPLC columns), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers.
  • Suitable chiral HPLC columns are manufactured by Daicel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable.
  • Enzymatic separations, with or without derivatisation are also useful.
  • the optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
  • the present invention includes all possible stereoisomers of the compounds of component A as single stereoisomers, or as any mixture of said stereoisomers, e.g. R- or S- isomers, or E- or Z- isomers, in any ratio.
  • Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of component A may be achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example.
  • the compounds of component A, particularly of Compound A may exist as tautomers.
  • any compound of component A which contains a pyrazole moiety as a heteroaryl group for example can exist as a 1H tautomer, or a 2H tautomer, or even a mixture in any amount of the two tautomers, or a triazole moiety for example can exist as a 1H tautomer, a 2H tautomer, or a 4H tautomer, or even a mixture in any amount of said 1H, 2H and 4H tautomers, namely:
  • the present combination includes all possible tautomers of the compounds of component A, particularly the 1 H-tautomer or the 2H-tautomer of the pyrazol-5-yl group in 8-position of the naphthyridine core of Compound A, as single tautomers, or as any mixture of said tautomers, in any ratio.
  • the compounds of component A can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised.
  • the present combination includes all such possible N-oxides of component A.
  • the present combination also covers useful forms of the compounds as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates.
  • the compounds of the present combination can exist as a hydrate, or as a solvate, wherein the compounds of the present combination contain polar solvents, in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds.
  • the amount of polar solvents, in particular water may exist in a stoichiometric or non- stoichiometric ratio. In the case of stoichiometric solvates, e.g.
  • a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, terra-, penta- etc. solvates or hydrates, respectively, are possible.
  • the present combination includes all such hydrates or solvates.
  • the compounds of the present combination can exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or can exist in the form of a salt.
  • Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, customarily used in pharmacy.
  • the present invention includes all possible salts of the components of the present combination as single salts, or as any mixture of said salts, in any ratio.
  • the present invention includes all possible crystalline forms, or polymorphs, of the compounds of components of the present combination, either as single polymorphs, or as a mixture of more than one polymorphs, in any ratio.
  • radicals in the compounds of the present combination When radicals in the compounds of the present combination are substituted, the radicals may be mono- or polysubstituted, unless specified otherwise. In the context of the present invention, all radicals which occur more than once are defined independently of one another. Substitution by one, two or three identical or different substituents is preferred.
  • treatment includes inhibition, retardation, checking, alleviating, attenuating, restricting, reducing, suppressing, repelling or healing of a disease or the development, the course or the progression of such states and/or the symptoms of such states.
  • disease includes but is not limited a condition, a disorder, an injury or a health problem.
  • therapy is understood here to be synonymous with the term “treatment”.
  • prevention means the avoidance or reduction of the risk of contracting, experiencing, suffering from or having a disease or a development or advancement of such states and/or the symptoms of such states.
  • the treatment or prevention of a disease may be partial or complete.
  • Component A can be selected from inhibitors of ATR kinase specifically or generically disclosed in the following publications: J. Med. Chem. 2013, 56, 2125-2138; Exp. Rev. Mol. Med. 16, elO, 2014; WO2010054398 Al; WO2010071837A1; WO2010073034A1; WO2011143399A1; WO2011143419A1; WO2011143422A1; WO2011143423 A2; WO2011143425 A2; WO2011143426A1; WO2011154737A1; WO2011163527 Al; WO2012138938A1 ; WO2012178123A1 ; WO2012178124A1; WO2012178125A1 WO2013049719A1 ; WO2013049720A1 ; WO2013049722A1; WO2013049859 A 1 WO2013071085A1 ; WO2013071088A1 ; WO2013071090A1
  • said component A is selected from VX-803, VX-970, AZD-6738 and a compound of general formula (I)
  • each Ci-C6-alkyl, Ci-C6-alkoxy, 3- to 10-membered heterocycloalkoxy, C 2 -C6- alkenyl, C 3 -C6-cycloalkyl, 3- to 10-membered heterocycloalkyl, phenyl or heteroaryl is optionally substituted, one or more times, independently from each other, with halogen, OH, -NR 7 R 8 , Ci-C6-alkyl optionally substituted one or more times with hydroxyl or phenyl, Ci-C6-haloalkyl, Ci-C6-alkoxy, C 3 -C6-cycloalkyl, 3- to 6- membered heterocycloalkyl, phenyl, -(CO)OR 7 , -(CO)NR 7 R 8 , -NR 7 (CO)R 10 , -NR 8 (CO)OR 7 , -NR 8 (CO) NR 7 R 8 , -(S0 2 )R 9
  • each 4- to 10-membered heterocycloalkenyl is optionally substituted, one or more times, independently from each other, with Ci-C t-alkyl;
  • R 3 , R 4 represent, independently from each other, hydrogen or methyl
  • R 7 , R 8 represent, independently from each other, hydrogen, Ci-C6-alkyl, C3-C6-cycloalkyl or phenyl, which phenyl is optionally substituted, one or more times, with halogen; or R 7 and R 8 together represent a 4-, 5-, 6- or 7-membered cyclic amine group, which is optionally substituted, one or more times, independently from each other, with a substituent selected from Ci-C6-alkyl, Ci-C6-haloalkyl, said 4-, 5-, 6- or 7-membered cyclic amine group optionally containing one further heteroatom selected from the group consisting of O, N and S;
  • R 9 represents Ci-C t-alkyl or phenyl, wherein each Ci-C/t-alkyl or phenyl is optionally
  • R 10 represents Ci-C/t-alkyl
  • R 11 represents hydrogen, Ci-C 4 -alkyl, -(CO)OR 7 , -(CO)NR 7 R 8 or CN;
  • R 12 represents hydrogen or Ci-C/t-alkyl
  • R 13 represents halogen, OH, -NR 7 R 8 , CN, N0 2 , G-Ce-alkyl, G-Ce-haloalkyl, G-Ce-alkoxy,
  • Ci-Ce-haloalkoxy C 2 -C 6 -alkenyl, C 3 -C 6 -cycloalkyl, -(CO)OR 7 or -(CO)NR 7 R 8 ;
  • VX-803 means 2-amino-6-fluoro-N-[5-fluoro-4- (4- ⁇ [4-(oxetan-3-yl)piperazin- l-yl]carbonyl Jpiperidin- l-yl)pyridin-3-yl]pyrazolo[ 1 ,5- a]pyrimidine-3-carboxamide. It has the following structure:
  • VX-970 means 3-(3 [(methylamino)methyl]phenyl ⁇ - 1 ,2-oxazol-5-yl)-5-[4-(propan-2-ylsulfonyl)phenyl]pyrazin-2 amine. It has the following structure:
  • AZD-6738 means 4- ⁇ 4-[(3R)-3- methylmorpholin-4-yl] -6- [ 1 -(S-methylsulfonimidoyl)cyclopropyl]pyrimidin-2-yl ⁇ - 1 H- pyrrolo[2,3-b]pyridine. It has the following structure:
  • said component A is selected from VX-803, VX-97 compound of general formula (lb)
  • said component A is a compound of general formula (lb)
  • each 4- to 6-membered heterocycloalkenyl is optionally substituted, one or more times, with methyl;
  • R 4 represents hydrogen or methyl
  • R 7 , R 8 represent, independently from each other, hydrogen or Ci-C/t-alkyl
  • R 9 represents Ci-C/t-alkyl
  • R 10 represents Ci-C t-alkyl
  • R 11 represents hydrogen, methyl, -(CO)OR 7 ;
  • component A is 2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl- lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine ("Compound A”), a tautomer, an N- oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
  • said component A is Compound A of structure
  • pharmaceutically acceptable salt of component A, particularly of Compound A, refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention.
  • Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid.
  • Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts.
  • an appropriate base e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts.
  • acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.
  • alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
  • Representative salts of a component A of this invention include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art.
  • acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-
  • Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, or butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl sulfate, or diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, or butyl chlorides,
  • Said component A may be in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially with component B and optionally component C as further described infra.
  • the components A and B and optionally C may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
  • the specific compounds of the lists as disclosed above are preferred as being component A of the combination, most preferred is "Compound A" used in the Experimental Section. It is to be understood that the present invention relates also to any combination of the embodiments of component A described above.
  • Component B is a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium- 223.
  • a pharmaceutically acceptable salt of radium-223 can be, for example, an acid addition salt with an inorganic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, bisulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2 (4 hydroxybenzoyl) benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3 hydroxy 2 naphthoic, nicotinic, pamoic, pectinic, persulfuric, 3 phenylpropionic, picric, pival
  • a preferred pharmaceutically acceptable salt of radium-223 is the dichloride ( 223 RaCl2).
  • Methods for preparation of a pharmaceutically acceptable solution comprising radium-223 are disclosed e.g. in WO 2000/40275(A2), WO 2011/134671(A1), and WO 2011/134672(A1).
  • compositions comprising radium-223 show a unique mechanism of action as a targeted radiopharmaceutical. They represent a new generation of alpha emitting therapeutic pharmaceuticals based on the natural bone-seeking nuclide radium-223.
  • an aqueous solution of radium-223 chloride ( 223 R 3 Cl 2 ) for intravenous injection, sterile and free from bacterial endotoxins is used.
  • the solution is isotonic, containing a sodium citrate buffered saline to physiological pH.
  • a preferred dosage regimen for radium-223 chloride injection is 50 kBq per kg body weight given at 4 week intervals, as a course consisting of 6 injections. Single radium-223 doses up to 250 kBq per kg body weight were evaluated in a phase I clinical trial. The observed adverse reactions at this dose were diarrhea and reversible myelosuppression (including one case (1/5) of grade 3 neutropenia).
  • the aqueous radium-223 dichloride solution may be supplied in a single-dose 10 ml vial which contains a fill volume of 6 ml.
  • This product has a radioactivity concentration of radium-223 of 1,000 kBq/mL (0.03 mCi/mL), corresponding to 0.53 ng/mL of radium at reference date.
  • the active moiety is the alpha particle emitting nuclide radium-223 (half-life is 11.4 days), present as a divalent cation ( 223 Ra 2+ ).
  • the fraction of energy emitted from radium- 223 and its daughters as alpha-particles is 95.3%
  • the fraction emitted as beta-particles is 3.6%
  • the fraction emitted as gamma-radiation is 1.1%.
  • the combined energy from the emitted radiation from complete decay of radium-223 and its daughter nuclides is 28.2 MeV.
  • Radium-223 is to be administered intravenously by qualified personnel as a slow bolus injection.
  • An intravenous access line should be used for administration of Radium-223.
  • the line must be flushed with isotonic saline before and after injection of Radium-223.
  • Radium-223 selectively targets areas of increased bone turnover, as in bone metastases, and concentrates by forming a complex with hydroxyapatite.
  • Alpha emission contributes about 93% of the total radiation absorbed dose.
  • the high linear energy alpha particle radiation induces double-strand DNA breaks, resulting in a potent and localized cytotoxic effect in the target areas containing metastatic cancer cells.
  • the short path length (less than 100 micrometers) of the alpha particles minimizes the effect on adjacent healthy tissue such as the bone marrow.
  • the present invention provides combinations of at least two components, preferably two components, comprising component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium- 223.
  • the present invention covers a combination of any component A mentioned herein with any component B mentioned herein, optionally with any component C mentioned herein.
  • the present invention concerns combinations of at least two components A and B, preferably of two components, comprising component A being an inhibitor of ATR kinase, particularly Compound A, and component B being pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
  • the present invention provides combinations of at least two components A and B, preferably of two components, comprising component A being an inhibitor of ATR kinase, particularly Compound A, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable inorganic salt of the alkaline- earth radionuclide radium-223.
  • the present invention provides combinations of at least two components, preferably of two components, comprising component A as described supra and component B as described supra, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
  • a combination of the present invention comprises Compound A or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, preferably the dichloride salt of radium-223.
  • the combination of the present invention comprises Compound A or a pharmaceutically acceptable salt thereof and the dichloride salt of radium-223.
  • kits comprising:
  • component A one or more, preferably one, ATR kinase inhibitor(s) as described supra, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof;
  • component B a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof as described supra.
  • either or both of said components A and B in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially.
  • the components A and B may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
  • Preferably component A is administered by the oral route and component B is administered by the intravenous route.
  • kits comprising:
  • component A one or more, preferably one, ATR kinase inhibitor(s) as described supra, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof;
  • component B a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof as described supra; and, optionally, component C: one or more, preferably one, further pharmaceutical agent(s),
  • components A, B and C in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially.
  • the components A and B, optionally C may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
  • component C being at least one pharmaceutical agent includes the effective compound itself as well as its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers as well as any pharmaceutical composition comprising such effective compound or its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers.
  • a list of such pharmaceutical agents of component C is being provided further below.
  • the combinations of component A and component B of this invention can be administered as the sole pharmaceutical agent or in combination with one or more further pharmaceutical agents C where the resulting combination of components A, B and C causes no unacceptable adverse effects.
  • the combinations of components A and B of this invention can be combined with component C, i.e.
  • one or more further pharmaceutical agents such as known anti-angiogenesis, anti-hyper-proliferative, antiinflammatory, analgesic, immunoregulatory, diuretic, antiarrhytmic, anti-hypercholsterolemia, anti-dyslipidemia, anti-diabetic or antiviral agents, and the like, as well as with admixtures and combinations thereof.
  • Optional pharmaceutical agents which can be added as component C to the combination of components A and B can be one or more pharmaceutical agents such as 1311-chTNT, abarelix, abiraterone, aclarubicin, ado-trastuzumab emtansine, afatinib, aflibercept, aldesleukin, alectinib, alemtuzumab, alendronic acid, alitretinoin, altretamine, amifostine, aminoglutethimide, hexyl aminolevulinate, amrubicin, amsacrine, anastrozole, ancestim, anethole dithiolethione, anetumab ravtansine, angiotensin II, antithrombin III, aprepitant, arcitumomab, arglabin, arsenic trioxide, asparaginase, axitinib, azacitidine, basilixima
  • Preferred optional pharmaceutical agents which may be added as component C to the combination of components A and B is/ are one or more agents selected from enzalutamide, bicalutamide, flutamide, nilutamide, and/or abiraterone.
  • cytotoxic and/or cytostatic agents as component C in combination with a combination of components A and B of the present invention may serve to:
  • the present invention covers a pharmaceutical composition comprising a combination of the present invention as described supra together with one or more pharmaceutically acceptable excipients.
  • the present invention covers a pharmaceutical composition
  • a pharmaceutical composition comprising a combination of at least two components, component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 together with one or more pharmaceutically acceptable excipients.
  • the present invention covers a pharmaceutical composition
  • a pharmaceutical composition comprising a combination of at least two components, component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223, optionally with any component C mentioned herein, together with one or more pharmaceutically acceptable excipients.
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof
  • component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223, optionally with any component C mentioned herein, together with one or more pharmaceutically acceptable excipients
  • components A and B, and optionally component C are present in separate formulations. In another embodiment the components A and B, and optionally component C, are present in a joint formulation.
  • compositions are non-toxic, preferably they are non-toxic and inert.
  • Pharmaceutically acceptable excipients include, inter alia,
  • ⁇ fillers and excipients for example cellulose, microcrystalline cellulose, such as, for example, Avicel®, lactose, mannitol, starch, calcium phosphate such as, for example, Di-Cafos®),
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • bases for suppositories for example polyethylene glycols, cacao butter, hard fat
  • solvents for example water, ethanol, Isopropanol, glycerol, propylene glycol, medium chain-length triglycerides fatty oils, liquid polyethylene glycols, paraffins
  • surfactants for example sodium dodecyle sulphate, lecithin, phospholipids, fatty alcohols such as, for example, Lanette®, sorbitan fatty acid esters such as, for example, Span®, polyoxyethylene sorbitan fatty acid esters such as, for example, Tween®, polyoxyethylene fatty acid glycerides such as, for example, Cremophor®, polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers such as, for example, Pluronic®),
  • surfactants for example sodium dodecyle sulphate, lecithin, phospholipids, fatty alcohols such as, for example, Lanette®, sorbitan fatty acid esters such as, for example, Span®, polyoxyethylene sorbitan fatty acid esters such as, for example, Tween®, polyoxyethylene fatty acid glycerides such as, for example, Crem
  • buffers and also acids and bases for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • acids and bases for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • isotonicity agents for example glucose, sodium chloride
  • adsorbents for example highly-disperse silicas
  • viscosity-increasing agents for example polyvinylpyrrolidon, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids such as, for example, Carbopol®, alginates, gelatine),
  • binders for example polyvinylpyrrolidon, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids such as, for example, Carbopol®, alginates, gelatine
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate such as, for example, Explotab®, cross- linked polyvinylpyrrolidon, croscarmellose-sodium such as, for example, AcDiSol®
  • modified starch carboxymethylcellulose-sodium, sodium starch glycolate such as, for example, Explotab®, cross- linked polyvinylpyrrolidon, croscarmellose-sodium such as, for example, AcDiSol®
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate such as, for example, Explotab®, cross- linked polyvinylpyrrolidon, croscarmellose-sodium such as, for example, AcDiSol®
  • lubricants for example magnesium stearate, stearic acid, talc, highly-disperse silicas such as, for example, Aerosil®
  • coating materials for example sugar, shellac
  • film formers for films or diffusion membranes which dissolve rapidly or in a modified manner for example polyvinylpyrrolidones such as, for example, Kollidon®, polyvinyl alcohol, hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit®),
  • capsule materials for example gelatine, hydroxypropylmethylcellulose
  • synthetic polymers for example polylactides, polyglycolides, polyacrylates, polymethacrylates such as, for example, Eudragit®, polyvinylpyrrolidones such as, for example, Kollidon®, polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers
  • synthetic polymers for example polylactides, polyglycolides, polyacrylates, polymethacrylates such as, for example, Eudragit®, polyvinylpyrrolidones such as, for example, Kollidon®, polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers
  • plasticizers for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate
  • stabilisers for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate
  • preservatives for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate
  • colourants for example inorganic pigments such as, for example, iron oxides, titanium dioxide
  • flavourings sweeteners, flavour- and/or odour-masking agents.
  • the components A, B and C may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
  • Component A is administered intravenously, intraperitoneally, preferably it is administered orally.
  • Component B preferably is administered by the intravenous route.
  • Component C is administered intravenously, intraperitoneally, preferably it is administered orally.
  • compositions of this invention varies by the route of administration.
  • Components of this invention can be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • binders such as acacia, corn starch or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet
  • Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives.
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.
  • Components of this invention can also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils.
  • Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Oily suspensions can be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol.
  • the suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.
  • Syrups and elixirs can be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.
  • sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.
  • Components of this invention can also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in preferably a pharmaceutically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-l,l-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfact
  • Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta- aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example dimethyl dial
  • compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • compositions of the present invention can be in the form of sterile injectable aqueous suspensions.
  • suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • Components of the invention can also be administered in the form of suppositories for rectal administration of the drug.
  • These components can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are, for example, cocoa butter and polyethylene glycol.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference).
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.
  • the present invention concerns the use of the combination of the present invention as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention concerns the kit as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention concerns the pharmaceutical composition as described supra for the treatment or prophylaxis of a disease, preferably a hyper- proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention covers the use of such combinations as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention covers the use of such kit as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention covers the use of such pharmaceutical composition as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
  • the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, using an effective amount of the combination as described supra.
  • the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, using an effective amount of the kit or pharmaceutical composition as described supra.
  • the present invention concerns a method of treating a disease in a patient, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, comprising a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223,_wherein components A and B are administered simultaneously, concurrently, separately or sequentially.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223,_wherein components A and B are administered concurrently.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 2 hours to 96 hours prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 6 hours to 84 hours prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 22 hours to 26 hours prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 42 hours to 54 hours prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 44 hours to 52 hours prior to component A.
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • the present invention concerns a method of treating a disease in a patient, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, comprising
  • component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
  • component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, and optionally
  • compositions of the present invention thus can be used for the treatment or prophylaxis of hyper-proliferative diseases, including diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, such as, for example, haematological tumors and/or metastases therof, solid tumors, and/or metastases thereof, e.g.
  • leukemias multiple myeloma thereof and myelodysplastic syndrome, malignant lymphomas, breast tumors including and bone metastases thereof, tumors of the thorax including non-small cell and small cell lung tumors and bone metastases thereof, gastrointestinal tumors, endocrine tumors, mammary and other gynaecological tumors and bone metastases thereof, urological tumors including renal, bladder and prostate tumors, skin tumors, and sarcomas, and/or metastases thereof.
  • inappropriate within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as preferably meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.
  • the present invention covers the treatment of lung carcinoma, in particular small- cell lung cancer, colorectal cancer, bladder cancer, lymphomas, in particular diffuse large B-cell lymphoma (DLBC) and mantle cell lymphoma (MCL), prostate cancer, in particular castration- resistant prostate cancer, gliomas, and ovarian cancer.
  • lung carcinoma in particular small- cell lung cancer, colorectal cancer, bladder cancer, lymphomas, in particular diffuse large B-cell lymphoma (DLBC) and mantle cell lymphoma (MCL), prostate cancer, in particular castration- resistant prostate cancer, gliomas, and ovarian cancer.
  • lung carcinoma in particular small- cell lung cancer, colorectal cancer, bladder cancer, lymphomas, in particular diffuse large B-cell lymphoma (DLBC) and mantle cell lymphoma (MCL)
  • DLBC diffuse large B-cell lymphoma
  • MCL mantle cell lymphoma
  • prostate cancer in particular castration- resistant prostate cancer, gliomas
  • the invention covers combinations, kits or pharmaceutical compositions as described supra comprising component A or a pharmaceutically acceptable salt thereof and component B being a pharmaceutically acceptable salt of the alkaline earth radionuclide radium- 223 for use in the treatment of cancer indications particularly for such cancer type which form metastases in bone.
  • cancer types are e.g. breast, prostate, lung, multiple myeloma, kidney or thyroid cancer.
  • Another aspect of the invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of a hyper-proliferative disease and/or metastases thereof.
  • the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein components A and B are administered simultaneously, concurrently, separately or sequentially.
  • the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein components A and B are administered concurrently.
  • the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein component B is administered prior to component A.
  • the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein component B is administered 2 hours to 96 hours, or 6 hours to 84 hours, or 12 hours to 72 hours, or 24 hours to 48 hours, or 18 hours to 30 hours, or 20 hours to 28 hours, or 22 hours to 26 hours, or 42 hours to 54 hours, or 44 hours to 52 hours, or 46 hours to 50 hours, or 24 hours, or 48 hours prior to component A.
  • component B is administered 2 hours to 96 hours, or 6 hours to 84 hours, or 12 hours to 72 hours, or 24 hours to 48 hours, or 18 hours to 30 hours, or 20 hours to 28 hours, or 22 hours to 26 hours, or 42 hours to 54 hours, or 44 hours to 52 hours, or 46 hours to 50 hours, or 24 hours, or 48 hours prior to component A.
  • Another embodiment covers the use of a combination, kit or pharmaceutical composition according to the present invention for the preparation of a medicament for the treatment or prophylaxis of breast cancer, prostate cancer, multiple myeloma, non- small cell lung cancer and/or metastases thereof, especially wherein the metastases are bone metastases.
  • the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer, in a subject, comprising administering to said subject a therapeutically effective amount of a combination according to the present invention.
  • the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer, in a subject, comprising administering to said subject a therapeutically effective amount of a combination according to the present invention.
  • a cancer particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer
  • the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer and/or metastases thereof in a subject, comprising administering to said subject a therapeutically effective amount of a combination accoring to the present invention.
  • a cancer particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer and/or metastases thereof in a subject, comprising administering to said subject a therapeutically effective amount of a combination accoring to the present invention.
  • prostate cancer means any histology type of prostate cancer including but not limited to acinar adenocarcinoma, ductal adenocarcinoma, transitional cell (or urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers, particularly acinar adenocarcinoma, stage MO castration-resistant prostate cancer (MO CRPC) or stage Ml castration-resistant prostate cancer (Ml CRPC), preferably Ml castration-resistant prostate cancer (Ml CRPC).
  • MO CRPC stage MO castration-resistant prostate cancer
  • Ml CRPC stage Ml castration-resistant prostate cancer
  • MO and “Ml” (including Mia, Mlb, Mlc) are used in accordance with the "TNM staging system” for prostate cancer developed by the American Joint Committee on Cancer as further described in "TNM CLASSIFICATION OF MALIGNANT TUMORS", 7th edition Edited by James D. Brierley, Mary K. Gospodarowicz, Christian Wittekind, Published by UICC 2011. According to the TNM classification the term “MO” means that there are no distant metastases and that the cancer has not spread to other parts of the body. “Ml” means that there are distant metastases and that cancer has spread to distant parts of the body.
  • CRPC castration-resistant prostate cancer
  • stage Ml CRPC preferably stage Mlb castration-resistant prostate cancer (Mlb CRPC) or stage Mlc castration-resistant prostate cancer (Mlc CRPC).
  • Mlb CRPC stage Mlb castration-resistant prostate cancer
  • Mlc CRPC stage Mlc castration-resistant prostate cancer
  • Mlb CRPC as used herein means that the castration-resistant prostate cancer has spread to the bones.
  • Mlc CRPC as used herein means that the castration-resistant prostate cancer has spread to other organs such as lungs, liver, or brain (with or without spread to the bones).
  • Preferred uses of the combinations of the invention are the treatment of prostate cancer, especially castration-resistant prostate cancer (CRPC), preferably of CRPC with symptomatic bone metastases and no known visceral metastatic disease.
  • CRPC castration-resistant prostate cancer
  • One preferred embodiment is the use of the combinations, the kits or the pharmaceutical compositions of the invention for the treatment of prostate cancer, especially of castration- resistant prostate cancer (CRPC) with symptomatic bone metastases and no known visceral metastatic disease.
  • CRPC castration- resistant prostate cancer
  • kits or the pharmaceutical compositions of the invention concern the treatment of castration resistant prostate cancer (MO CRPC) or of Ml castration-resistant prostate cancer (Ml CRPC) in a subject, wherein the subject is chemotherapy-naive.
  • MO CRPC castration resistant prostate cancer
  • Ml CRPC Ml castration-resistant prostate cancer
  • chemotherapy-naive means that the subject, prior to the treatment with the combinations, the kits or the pharmaceutical compositions of the present invention has not received a chemotherapy.
  • kits or the pharmaceutical compositions of the present invention concern the treatment of castration resistant prostate cancer (MO CRPC) or of Ml castration-resistant prostate cancer (Ml CRPC) in a subject, wherein the subject has received a chemotherapy prior to the treatment with the combinations, the kits or the pharmaceutical compositions of the invention.
  • MO CRPC castration resistant prostate cancer
  • Ml CRPC Ml castration-resistant prostate cancer
  • chemotherapeutic agents are rather non-specific agents such as for example alkylating agents, anthracyclines, taxanes, epothilones, histone deacetylase inhibitors, inhibitors of topoisomerase I, inhibitors of topoisomerase II, nucleotide analogues, platinum-based agents, vinca alkaloids, etc..
  • Another embodiment of the present invention concerns the use of the combinations of the invention for the treatment of breast cancer, in particular breast cancer with bone metastases (Amol Takalkar et al., Exp Hematol Oncol. 2014; 3: 23. "Radium-223 dichloride bone-targeted alpha particle therapy for hormone-refractory breast cancer metastatic to bone”; Suominen et al., J Natl Cancer Inst. 2013 Jun 19;105(12):908-16. "Survival benefit with radium-223 dichloride in a mouse model of breast cancer bone metastasis”).
  • the present invention concerns the use of the combinations of the present invention for the treatment of multiple myeloma.
  • kits or pharmaceutical compositions of the present invention might be utilized to inhibit, block, reduce, decrease, etc., cell proliferation and/or cell division, and/or produce apoptosis.
  • This invention includes a method comprising administering to a mammal in need thereof, including a human, an amount of a component A and an amount of component B of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof, which is effective to treat the disease.
  • Hyper-proliferative diseases include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), as well as malignant neoplasia.
  • malignant neoplasia treatable with the compounds according to the present invention include solid and hematological tumors.
  • Solid tumors can be exemplified by tumors of the breast, bladder, bone, brain, central and peripheral nervous system, colon, anum, endocrine glands (e.g. thyroid and adrenal cortex), esophagus, endometrium, germ cells, head and neck, kidney, liver, lung, larynx and hypopharynx, mesothelioma, ovary, pancreas, prostate, rectum, renal, small intestine, soft tissue, testis, stomach, skin, ureter, vagina and vulva.
  • Malignant neoplasias include inherited cancers exemplified by Retinoblastoma and Wilms tumor.
  • malignant neoplasias include primary tumors in said organs and corresponding secondary tumors in distant organs ("tumor metastases").
  • Hematological tumors can be exemplified by aggressive and indolent forms of leukemia and lymphoma, namely non- Hodgkins disease, chronic and acute myeloid leukemia (CML / AML), acute lymphoblastic leukemia (ALL), Hodgkins disease, multiple myeloma and T-cell lymphoma.
  • myelodysplastic syndrome plasma cell neoplasia, paraneoplastic syndromes, and cancers of unknown primary site as well as AIDS related malignancies.
  • breast cancer examples include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ, particularly with bone metastases.
  • cancers of the respiratory tract include, but are not limited to small-cell and non- small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
  • brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
  • Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer.
  • Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
  • Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell.
  • Lymphomas include, but are not limited to AIDS -related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin' s disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia. These diseases have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
  • Combinations of the present invention might also be used for treating diseases associated with excessive and/or abnormal angiogenesis.
  • Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism.
  • a number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity [Aiello et al. New Engl. J. Med. 1994, 331, 1480 ; Peer et al. Lab. Invest. 1995, 72, 638], age-related macular degeneration [AMD ; see, Lopez et al. Invest. Opththalmol. Vis. Sci.
  • neovascular glaucoma neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc.
  • RA rheumatoid arthritis
  • restenosis in-stent restenosis
  • vascular graft restenosis etc.
  • the increased blood supply associated with cancerous and neoplastic tissue encourages growth, leading to rapid tumor enlargement and metastases.
  • the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastases and the consequence spread of the cancer.
  • combinations of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis diseases, e.g., by inhibiting and/or reducing blood vessel formation ; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredients to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular component and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredients to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 50 mg/kg body weight per day.
  • Clinically useful dosing schedules of a compound will range from one to three times a day dosing to once every four weeks dosing.
  • "drug holidays" in which a patient is not dosed with a drug for a certain period of time may be beneficial to the overall balance between pharmacological effect and tolerability.
  • a unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day.
  • the average daily dosage for administration by injection will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
  • the average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
  • a preferred dosage regimen for radium-223 injection is 50 kBq per kg body weight given at 4 week intervals, as a course consisting of 6 injections.
  • Single radium-223 doses up to 250 kBq per kg body weight were evaluated in a phase I clinical trial.
  • the observed adverse reactions at this dose were diarrhea and reversible myelosuppression (including one case (1/5) of grade 3 neutropenia).
  • the aqueous radium-223 dichloride solution may be supplied in a single-dose 10 ml vial which contains a fill volume of 6 ml.
  • This product has a radioactivity concentration of radium-223 of 1,000 kBq/mL (0.03 mCi/mL), corresponding to 0.53 ng/mL of radium at reference date.
  • Radium-223 is to be administered intravenously by qualified personnel as a slow bolus injection.
  • An intravenous access line should be used for administration of radium-223.
  • the line must be flushed with isotonic saline before and after injection of radium-223.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compounds employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • Compound A is an example of component A.
  • Compound A is described in Example 111 of International Patent Application WO2016020320 (Al).
  • Compound A is 2-[(3R)-3-methylmorpholin-4-yl]-4-(l- methyl-lH-pyrazol-5-yl -8-(lH-pyrazol-5-yl)-l,7-naphthyridine, of structure:
  • Component B
  • component B is radium-223 dichloride, the synthesis of which is disclosed in WO2000/040275, which is incorporated herein in its entirety by reference.
  • LNCaP is an androgen-receptor (AR) positive CRPC cell line with several mutations/deletions in DNA damage or mismatch repair genes (APCR2714C, ARIDlAfs, ATG5fs, ATMA1119V/K1572N, ATRXEE2264- 2265E, BRCA2fs, CHEK2T430N, ERCC3A740T;R391W, ERCC5L1023I,
  • AR androgen-receptor
  • LNCaP cells secrete prostate specific antigen (PSA) and are known to form osteoblastic and mixed lesions when inoculated to bone marrow cavity. Therefore this model can be effectively used for testing the effects of cancer drug candidates on the growth of prostate cancer cells in bone.
  • PSA prostate specific antigen
  • LNCaP-luc a luciferase transfected LNCaP cell line
  • BLI Bioluminescent Imaging
  • mice were 5-8 weeks of age at the beginning of the study. On day 0 mice were given intratibial inoculation of LNCaP-luc human prostate cancer cells. The development of osteoblastic lesions were monitored by X-ray imaging before dosing was started and in 3 weeks intervals thereafter. The lesion area in hind limbs was determined from the images with MetaMorph image analysis software (Molecular Devices LLC, Sunnyvale, CA, USA) by drawing the outlines of radio- opaque and -lucent areas. Tumor burden was quantified by imaging the bioluminescence emitted by the LNCaP-luc cells. The obtained data were analyzed using Living Image® software (PerkinElmer, Waltham, MA, USA) version 4.2.
  • Serum Bone Turnover Markers (PINP and ICTP) for the Early Detection of Bone Metastases in Patients With Prostate Cancer: A Longitudinal Approach; Windy Dean-Colomb et al.; Breast Cancer Res Treat. 2013 January ; 137(2). Elevated serum PINP predicts development of bone metastasis and survival in early-stage breast cancer).
  • Statistical analysis was effected using single-factor ANOVA and comparison with the control group by means of pair-by-pair comparative analysis (Dunnett's test) or student's t-test.
  • Ra-223 300 kBq/kg, 5 ml/kg, iv, q4w
  • Compound A dose 2 50 mg/kg 2 days on/ 5 days off po qd+ Vehicle for Ra-223, 28 mmol/L sodium citrate, 5 ml/kg, iv, q4w
  • Combo Ra-223 + Compound A dose 1 Ra-223 (300 kBq/kg, 5 ml/kg, iv, q4W, + Compound A (20 mg/kg 2 days on/ 5 days off po qd, started on week 4 after tumor cell inoculation) 6) Combo Ra-223 + Compound A dose 2: Ra-223 (300 kBq/kg, 5 ml/kg, iv, q4W, started on week 4 after tumor cell inoculation) + Compound A (50 mg/kg 2 days on/ 5 days off po qd, started on week 4 after tumor cell inoculation)
  • T/C ratio of the PSA level of treatment versus control at the end of the study.
  • T/C ratio of the PINP level of treatment versus control at the end of the study.
  • Ra-223 150 kBq/kg, 5 ml/kg, iv, q4w
  • Total bone lesion area was significantly reduced in combination therapy compared to vehicle control when Compound A was applied in weekly cycles for 2 days on and 5 days off starting concurrently with Ra-223 or 24h after Ra-223 (see Figure 3 B).
  • Tumor burden determined by BLI signal in bone was significantly reduced by all tested combination treatment schedules in comparison to vehicle control (see Figure 3 C).
  • Treatment were well tolerated. No treatment related critical body weight loss (> 10%) or toxicities/death were recorded (data not shown).
  • T/C ratio of the PSA level of treatment versus control at the end of the study
  • T/C ratio of the total bone lesion area of treatment versus control at the end of the study
  • T/C ratio of the BLI signal of treatment versus control at the end of the study.
  • Ra-223 150 kBq/kg, 5 ml/kg, iv, q4w
  • Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (10 mg/kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223)
  • Combo Ra-223 (150 kBq kg, 5 ml/kg, iv, q4w) + Compound A (20 mg kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223)
  • T/C ratio of the PSA level of treatment versus control at the end of the study.
  • T/C ratio of the PINP level of treatment versus control at the end of the study.
  • T/C ratio of the total bone lesion area of treatment versus control at the end of the study
  • T/C ratio of the BLI signal of treatment versus control at the end of the study.

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Inorganic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention covers combinations of at least two components, component A and component B, component A being an ATR kinase inhibitor, and component B being radium-223, particularly a pharmaceutically acceptable salt of radium-223. Another aspect of the present invention covers the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a hyper-proliferative disease, particurlarly for the treatment of prostate cancer as well as bone metastases thereof.

Description

COMBINATION of ATR KINASE INHIBITORS WITH RADIUM-223 SALT
The present invention covers combinations of at least two components, component A and component B, comprising component A being an ATR kinase inhibitor, particularly Compound A, and component B being radium-223, particularly a pharmaceutically acceptable salt of radium-223. Another aspect of the present invention covers the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a hyper - proliferative disease, particurlarly for the treatment of prostate cancer and/or bone metastases thereof.
BACKGROUND
Cancer is the second most prevalent cause of death in the United States, causing 450,000 deaths per year. While substantial progress has been made in identifying some of the likely environmental and hereditary causes of cancer, there is a need for additional therapeutic modalities that target cancer and related diseases. In particular there is a need for therapeutic methods for treating diseases associated with dysregulated growth / proliferation.
Cancer is a complex disease arising after a selection process for cells with acquired functional capabilities like enhanced survival / resistance towards apoptosis and a limitless proliferative potential. Thus, it is preferred to develop drugs for cancer therapy addressing distinct features of established tumors.
The integrity of the genome of eukaryotic cells is secured by complex signaling pathways, referred to as the DNA damage response (DDR), and multiple DNA repair mechanisms. Upon recognizing DNA damage activation of the DDR pathways results in cell cycle arrest, suppression of general translation, induction of DNA repair, and, finally, in cell survival or cell death. Proteins that directly recognize aberrant DNA structures, such as the MRE11-Rad50-Nbsl complex recognizing DNA double strand breaks by binding to double-stranded DNA ends, or RPA (replication protein A) binding to single stranded DNA, recruit and activate the most upstream kinases of the DDR pathway, ATM (ataxia-telangiectasia mutated), ATR (ATM-and Rad3-related, UniProtKB/Swiss-Prot Q13535), and DNA-PKcs (DNA-dependent protein kinase). Whereas ATM is primarily activated by DNA double strand breaks, and DNA-PKcs is mainly involved in non-homologous end joining process of DNA repair, ATR responds to a broad spectrum of DNA damage, including double- strand breaks and lesions derived from interference with DNA replication. Major components of downstream signaling of ATM include Chk2 and p53, whereas ATR signaling involves Chkl and cdc25. Knockout of the ATR gene in mice is embryonically lethal and ATR knockout cells develop chromosome breaks and undergo apoptosis [E.J. Brown, D. Baltimore: ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 14, 397-402, 2000]. In contrast, ATM is not essential for cell survival although ATM knockout cells are hypersensitive to ionizing radiation and agents which cause DNA double-strand breaks.
ATR, which forms a complex with ATRIP (ATR-interacting protein, UniProtKB/Swiss-Prot Q8WXE1) is mainly activated by long stretches of single-stranded DNA which are generated by the continuing DNA unwinding activity of helicases upon stalled replication. This replication stress with stalled replication forks may be induced by ultraviolet light, certain chemotherapeutic drugs, hydroxyurea, or aberrant oncogenic signaling resulting in increased replication initiation or origin firing. Activation of ATR results in inhibition of the cell cycle in S or G2 phase via the Chkl-cdc25 pathway and in suppression of late origin firing. The cell gains time to resolve the replication stress and, eventually, to restart replication after the source of stress has been removed. As the ATR pathway ensures cell survival after replication stress it potentially contributes to resistance to chemotherapy. Thus inhibition of ATR kinase activity could be useful for cancer treatment.
In oncogene-driven tumor cells (e.g. Ras mutation/upregulation, Myc upregulation, CyclinE overexpression) increased replication stress has been observed as compared to healthy normal cells. ATR suppression in Ras oncogene driven cells was reported to result in substantial tumor cell killing [O. Gilad, BY Nabet, et al.: Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. Cancer Res. 70, 9693-9702, 2010].
Although ATM and ATR are principally activated by different types of DNA damage their signaling includes some cross-talk thus that they can, at least partially, substitute for each others function. This finding suggests some tumor-cell selectivity of pharmaceutical inhibition of ATR.
A healthy normal cell, which has ATM and ATR pathways in parallel, arrests in Gl phase of the cell cycle upon induced DNA damage even in presence of an ATR inhibitor. In contrast, a tumor cell which most often deficient in ATM and/or p53 signaling relies on the ATR pathway and undergoes cell death in presence of an ATR inhibitor. This suggests that ATR inhibitors may be used for the treatment of tumors with deficient ATM signaling and/or p53 function.
Details of DDR signaling and the functional role of ATM and ATR were recently reviewed in:
E. Fokas, R. Prevo et al.: Targeting ATR in DNA damage response and cancer therapeutics.
Cancer Treatment Rev 40, 109-117, 2014. J.M. Wagner & S.H. Kaufmann: Prospects for the use of ATR inhibitors to treat cancer. Pharmaceuticals 3, 1311-1334, 2010. D. Woods & J.J. Tuchi:
Chemotherapy induced DNA damage response. Cancer Biol. Thera. 14, 379-389, 2013. A. Marechal & L. Zou: DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb. Perspect. Biol. 5, a012716, 2013. M.K. Zeman & K.A. Cimprich: Causes and consequences of replication stress. Nat. Cell Biol. 16, 2-9, 2014. S. Llona-Minguez, A. Hoglund et al.: Chemical strategies for development of ATR inhibitors. Exp. Rev. Mol. Med. 16, elO, 2014.
Thus inhibitors of ATR kinase represent valuable compounds that should complement therapeutic options not only as single agents but also in combination with other drugs.
Radium-223 dichloride (Ra223Ci2) (Xofigo®) uses alpha radiation from radium-223 decay to kill cancer cells. It targets to bone tissue by virtue of its chemical similarity to calcium. It has an effect over a range of 2-10 cells and causes less damage to surrounding healthy tissues compared to current radiation therapy based on beta or gamma radiation. Significant increase in median overall survival was demonstrated in Phase III clinical trials and radium-223 dichloride (Xofigo) was approved as a treatment for castration-resistant prostate cancer (CRPC) patients with symptomatic bone metastases.
A preferred suitable pharmaceutically acceptable salt of radium-223 is the dichloride (Ra223Cl2). Radium-223 dichloride is a novel, targeted alpha-emitter that selectively binds to areas of increased bone turnover in bone metastases and emits high-energy alpha-particles of extremely short (<100 μιη) range (Bruland O. S. et al., High-linear energy transfer irradiation targeted to skeletal metastases by the alpha-emitter 223Ra: adjuvant or alternative to conventional modalities?, Clin. Cancer Res. 2006; 12: 6250s-7s). It is the first targeted alpha-emitter approved for the treatment of prostate cancer with bone metastases.
As a bone-seeking calcium mimetic, radium-223 is bound into newly formed bone stroma, especially within the microenvironment of osteoblastic or sclerotic metastases (Henriksen G. et al., Significant antitumor effect from bone-seeking, alpha-particle-emitting (223)Ra demonstrated in an experimental skeletal metastases model, Cancer Res. 2002; 62: 3120-3125; Henriksen G. et al., Targeting of osseous sites with alpha-emitting 223Ra: comparison with the beta-emitter 89Sr in mice, J. Nucl. Med 2003; 44: 252-59). The high-energy alpha-particle radiation induces mainly double-strand DNA breaks resulting in a potent and highly localized cytotoxic effect in the target areas containing metastatic cancer cells (Lewington V.J., Bone- seeking radionuclides for therapy, J. Nucl. Med 2005; 46 (suppl 1): 38S-47S; Liepe K., Alpharadin, a 223Ra-based alpha-particle-emitting pharmaceutical for the treatment of bone metastases in patients with cancer, Curr. Opin. Investig. Drugs 2009; 10: 1346-58; McDevitt M.R. et al., Radioimmunotherapy with alpha- emitting nuclides, Eur. J. Nucl. Med. 1998; 25: 1341-51). The short path length of the alpha-particles also means that toxicity to adjacent healthy tissue and particularly the bone marrow may be reduced (Kerr C, (223)Ra targets skeletal metastases and spares normal tissue, Lancet Oncol. 2002; 3: 453; Li Y., Russell P.J., Allen B.J., Targeted alpha-therapy for control of micrometastatic prostate cancer, Expert Rev. Anticancer Ther. 2004; 4: 459-68).
Radium-223 has demonstrated a favorable safety profile with minimal myelotoxicity in phase 1 and 2 studies of patients with bone metastases (Nilsson S. et al., First clinical experience with alpha-emitting radium-223 in the treatment of skeletal metastases, Clin. Cancer Res. 2005; 11 : 4451-59; Nilsson S. et al., Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II study, Lancet Oncol. 2007; 8: 587-94).
Phase 2 studies have shown that radium-223 reduces pain, improves disease-related biomarkers (e.g., bone alkaline phosphatase [ALP] and prostate-specific antigen [PSA]), and have suggested a survival benefit in patients with CRPC and bone metastases (Parker C. et al., A randomized, double-blind, dose-finding, multicenter, phase 2 study of radium chloride (Ra-223) in patients with bone metastases and castration-resistant prostate cancer, Eur Urol. 2013 Feb; 63(2): 189-97; Nilsson S. et al., A randomized, dose -response, multicenter phase II study of radium-223 chloride for the palliation of painful bone metastases in patients with castration-resistant prostate cancer, Eur. J. Cancer 2012; 48: 678-86).
The ALSYMPCA (ALpharadin in SYMptomatic Prostate CAncer patients) trial provides proof of principle for the role of targeted alpha-emitters in oncology. In this trial, radium-223 significantly prolonged overall survival with a 30.5% reduction in risk of death compared with placebo in patients with CRPC (Castration Resistant Prostate Cancer) and bone metastases. Median survival with radium-223 was longer than placebo by 2.8 months. All main secondary efficacy endpoints were statistically significant and favored treatment with radium-223, including the clinically defined endpoint of time to first skeletal-related event, which was significantly prolonged in patients receiving radium-223 (C. Parker et al., Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer , The New England Journal of Medicine 369(3):213-23).
According to Suominen et al. (Clin Cancer Res. 2017 Aug l;23(15):4335-4346) radium-223 inhibits osseous prostate cancer growth by dual targeting of cancer cells and bone microenvironment in mouse models. A substantial percentage of cancer patients is affected by skeletal metastases. As many as 85% of patients with advanced lung, prostate and breast carcinoma develop bony metastases (Garret R., Semin. Oncol. 72, 3433 -3435 (1993) Bone destruction in cancer; Nielsen, OS, Munro AJ, Tannock IF. J Clin Oncol 9, 509-5 24 (1991)). Established treatments such as hormone therapy, chemotherapy and external radiotherapy often causes temporary responses, but ultimately most bone cancer patients experience relapses (Kanis JA. Bone 17,101s-105s (1995), Bone and cancer. Pathophysiology and treatment of metastases). There is thus a strong need for new therapies to relieve pain and slow down tumor progression. 223Ra is used as an a-emitting radiopharmaceutical for targeting of calcified tissues, e.g. bone surfaces and osseous tumor lesions. It can be suitable as a bone seeking radiopharmaceutical. It thus may be used for prophylactic cancer treatment by delivering a focused dose to bone surfaces in patients with a high probability of having undetected micro metastases at bone surfaces. Another example of its potential use would be in the treatment of painful osseous sites.
The alkaline-earth radionuclide radium-223 is useful for the targeting of calcified tissues, e.g., bone and a pharmaceutical acceptable solution comprising 223Ra.
The alkaline-earth radionuclide radium-223 is suitable for the use of the nuclide as a cationic species and/or associated to a chelator or another form of a carrier molecule with affinity for calcified tissues. Thus may be combined with a chelator that can be subsequently conjugated to a molecule with affinity for calcified tissues. The effect of the radioisotope to generated by providing a cascade of a-particles on bone surfaces and/or in calcified tumors for the palliation of pain caused by various diseases and/or for the prophylactic use against possible minimal disease to the skeleton, and/or also for the therapeutic treatment of established cancer to the bone. The diseases where the radioisotopes could be used includes, but are not limited to skeletal metastases of prostate-, breast-, kidney- and lung cancer as well as primary bone cancer and also multiple myeloma.
However, the state of the art does not disclose the combinations of the present invention comprising an inhibitor of ATR kinase or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223. SUMMARY OF THE INVENTION
Surprisingly a synergistic effect in a prostate tumor model was observed when administering a ATR kinase inhibitor in combination with a suitable pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
Therefore, in accordance with a first aspect, the present invention provides combinations of at least two components, component A and component B, comprising component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223, particularly a pharmaceutically acceptable inorganic salt of the alkaline-earth radionuclide radium-223.
In accordance with a further aspect, the present invention concerns combinations of at least two components A and B, comprising component A being an inhibitor of ATR kinase, particularly Compound A, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
The combinations comprising at least two components A and B, as decribed and defined herein, are also referred to as "combinations of the present invention".
Further, the present invention covers a kit comprising:
component A: one or more ATR kinase inhibitors as described herein, particularly
Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof;
component B: a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof; and, optionally, in which kit optionally either or both of said components A and B in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially. The components may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
In accordance with another aspect, the present invention concerns the combinations as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra. In accordance with another aspect, the present invention covers the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra. In accordance with another aspect, the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra, using an effective amount of the combinations as described herein.
DETAILED DESCRIPTION OF THE INVENTION
The terms as mentioned in the present text have the following meanings:
The term "inhibitor of ATR kinase" or the term "ATR kinase inhibitor" as used herein means any compound that inhibits ATR kinase. Examples of such compounds are described infra ("COMPONENT A OF THE COMBINATION").
The term "halogen atom", "halo-" or "Hal-" is to be understood as meaning a fluorine, chlorine, bromine or iodine atom.
The term "Ci-C6-alkyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, 4, 5, or 6 carbon atoms, e.g. a methyl, ethyl, propyl, butyl, pentyl, hexyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl, iso-pentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neo-pentyl, 1,1-dimethylpropyl, 4- methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3- dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbutyl, or 1,2-dimethylbutyl group, or an isomer thereof. Particularly, said group has 1, 2, 3 or 4 carbon atoms ("Ci-C t-alkyl"), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert- butyl group, more particularly 1, 2 or 3 carbon atoms ("Ci-C3-alkyl"), e.g. a methyl, ethyl, n- propyl or iso-propyl group.
The term "Ci-C6-haloalkyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C6-alkyl" is defined supra, and in which one or more hydrogen atoms is replaced by a halogen atom, in identically or differently, i.e. one halogen atom being independent from another. Particularly, said halogen atom is F. Said C1-C6- haloalkyl group is, for example, -CF3, -CHF2, -CH2F, -CF2CF3 or -CH2CF3. The term "Ci-C t-hydroxyaikyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C t-alkyl" is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a hydroxymethyl, 1- hydroxyethyl, 2-hydroxyethyl, 1,2-dihydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2,3- dihydroxypropyl, l,3-dihydroxypropan-2-yl, 3-hydroxy-2-methyl-propyl, 2-hydroxy-2-methyl- propyl, l-hydroxy-2-methyl-propyl group.
The term "Ci-C6-alkoxy" is to be understood as meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-alkyl, in which the term "alkyl" is defined supra, e.g. a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy, sec-butoxy, pentoxy, iso-pentoxy, or n-hexoxy group, or an isomer thereof. Particularly, said "Ci-C6-alkoxy" can contain 1, 2, 3, 4 or 5 carbon atoms, (a "Ci-Cs-alkoxy"), preferably 1, 2, 3 or 4 carbon atoms ("d-C^alkoxy"). The term "Ci-C6-haloalkoxy" is to be understood as meaning a linear or branched, saturated, monovalent Ci-C6-alkoxy group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a halogen atom. Particularly, said halogen atom is F. Said Ci-C6-haloalkoxy group is, for example, -OCF3, -OCHF2, -OCH2F, -OCF2CF3, or
The term "C2-C6-alkenyl" is to be understood as meaning a linear or branched, monovalent hydrocarbon group, which contains one or more double bonds, and which has 2, 3, 4, 5 or 6 carbon atoms or 2, 3 or 4 carbon atoms ("C2-C4-alkenyl), particularly 2 or 3 carbon atoms ("C2- C3-alkenyl"), it being understood that in the case in which said alkenyl group contains more than one double bond, then said double bonds may be isolated from, or conjugated with, each other. Said alkenyl group is, for example, a vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, homoallyl, (E)-but-2-enyl, (Z)-but-2-enyl, (E)-but-l-enyl, (Z)-but- l-enyl, pent-4-enyl, (E)-pent- 3-enyl, (Z)-pent-3-enyl, (E)-pent-2-enyl, (Z)-pent-2-enyl, (E)-pent-l-enyl, (Z)-pent- l-enyl, hex- 5-enyl, (E)-hex-4-enyl, (Z)-hex-4-enyl, (E)-hex-3-enyl, (Z)-hex-3-enyl, (E)-hex-2-enyl, (Z)-hex- 2-enyl, (E)-hex-l-enyl, (Z)-hex-l-enyl, isopropenyl, 2-methylprop-2-enyl, l-methylprop-2-enyl,
2- methylprop-l-enyl, (E)-l-methylprop-l-enyl, (Z)-l-methylprop-l-enyl, 3-methylbut-3-enyl, 2- methylbut-3-enyl, l-methylbut-3-enyl, 3-methylbut-2-enyl, (E)-2-methylbut-2-enyl, (Z)-2- methylbut-2-enyl, (E)-l-methylbut-2-enyl, (Z)-l-methylbut-2-enyl, (E)-3-methylbut-l-enyl, (Z)-
3- methylbut-l-enyl, (E)-2-methylbut-l-enyl, (Z)-2-methylbut- l-enyl, (E)- l-methylbut-l-enyl, (Z)-l-methylbut-l-enyl, l,l-dimethylprop-2-enyl, 1-ethylprop-l-enyl, 1-propylvinyl, 1- isopropyl vinyl, 4-methylpent-4-enyl, 3-methylpent-4-enyl, 2-methylpent-4-enyl, 1-methylpent- 4-enyl, 4-methylpent-3-eriyl, (E)-3-methylpent-3-enyl, (Z)-3-methylpent-3-enyl, (E)-2- methylpent-3-enyl, (Z)-2-methylpent-3-enyl, (E)-l-methylpent-3-enyl, (Z)-l-methylpent-3-enyl, (E)-4-methylpent-2-enyl, (Z)-4-methylpent-2-enyl, (E)-3-methylpent-2-enyl, (Z)-3-mefhylpent- 2-enyl, (E)-2-methylpent-2-enyl, (Z)-2-methylpent-2-enyl, (E)-l-methylpent-2-enyl, (Z)-l- methylpent-2-enyl, (E)-4-methylpent-l-enyl, (Z)-4-methylpent-l-enyl, (E)-3-methylpent-l-eriyl, (Z)-3-methylpent-l-enyl, (E)-2-methylpent-l-enyl, (Z)-2-methylpent-l-enyl, (E)-l-mefhylpent-
1- enyl, (Z)-l-methylpent-l-enyl, 3-ethylbut-3-enyl, 2-ethylbut-3-enyl, l-ethylbut-3-enyl, (E)-3- ethylbut-2-enyl, (Z)-3-ethylbut-2-enyl, (E)-2-ethylbut-2-enyl, (Z)-2-ethylbut-2-enyl, (E)-l- ethylbut-2-enyl, (Z)-l-ethylbut-2-enyl, (E)-3-ethylbut-l-enyl, (Z)-3-ethylbut-l-enyl, 2-efhylbut- 1-enyl, (E)-l-ethylbut-l-enyl, (Z)-l-ethylbut-l-enyl, 2-propylprop-2-enyl, l-propylprop-2-enyl,
2- isopropylprop-2-enyl, l-isopropylprop-2-enyl, (E)-2-propylprop- 1-enyl, (Z)-2-propylprop-l- enyl, (E)-l-propylprop- 1-enyl, (Z)-l-propylprop-l-enyl, (E)-2-isopropylprop- 1-enyl, (Z)-2- isopropylprop- 1-enyl, (E)-l-isopropylprop-l-enyl, (Z)-l-isopropylprop- 1-enyl, (E)-3,3- dimethylprop- 1-enyl, (Z)-3,3-dimethylprop- 1-enyl, l-(l,l-dimethylethyl)ethenyl, buta-1,3- dienyl, penta-l,4-dienyl, hexa-l,5-dienyl, or methylhexadienyl group. Particularly, said group is vinyl or allyl.
The term "C3-Cio-cycloalkyl" is to be understood as meaning a saturated, monovalent, mono-, or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms ("C3-C10- cycloalkyl"). Said C3-Cio-cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic hydrocarbon ring, e.g. a perhydropentalenylene or decalin ring. Particularly, said ring contains 3, 4, 5 or 6 carbon atoms ("C3-C6-cycloalkyl"), preferably cyclopropyl.
The term "3- to 10-membered heterocycloalkyl" is to be understood as meaning a saturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), S(=0)2, NRa, in which Ra represents a hydrogen atom, or a Ci-C6-alkyl or Ci-C6-haloalkyl group ; it being possible for said heterocycloalkyl group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom.
Particularly, said 3- to 10-membered heterocycloalkyl can contain 2, 3, 4, or 5 carbon atoms, and one or more of the above-mentioned heteroatom-containing groups (a "3- to 6-membered heterocycloalkyl"), more particularly said heterocycloalkyl can contain 4 or 5 carbon atoms, and one or more of the above-mentioned heteroatom-containing groups (a "5- to 6-membered heterocycloalkyl"). Particularly, without being limited thereto, said heterocycloalkyl can be a 4-membered ring, such as an azetidinyl, oxetanyl, or a 5-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl, or a 7-membered ring, such as a diazepanyl ring, for example. Optionally, said heterocycloalkyl can be benzo fused. Preferably, the 3- to 6-membered heterocycloalkyl is a tetrahydrofuranyl, tetrahydropyranyl or piperazinyl.
Said heterocycloalkyl can be bicyclic, such as, without being limited thereto, a 5,5-membered ring, e.g. a hexahydrocyclopenta[c]pyrrol-2(lH)-yl ring, or a 5,6-membered bicyclic ring, e.g. a hexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl ring.
As mentioned supra, said nitrogen atom-containing ring can be partially unsaturated, i.e. it can contain one or more double bonds, such as, without being limited thereto, a 2,5-dihydro-lH- pyrrolyl, 4H-[l,3,4]thiadiazinyl, 4,5-dihydrooxazolyl, or 4H-[l,4]thiazinyl ring, for example, or, it may be benzo-fused, such as, without being limited thereto, a dihydroisoquinolinyl ring, for example.
The term "3- to 10-membered heterocycloalkoxy" of formula -O-heterocycloalkyl, in which the term "heterocycloalkyl" is defined supra, is to be understood as meaning a saturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), S(=0)2, NRa, in which Ra represents a hydrogen atom, a Ci-C6-alkyl or Ci-C6-haloalkyl group and which is connected to the rest of the molecule via an oxygen atom, e.g. a pyrrolidineoxy, tetrahydrofuraneoxy or tetrahydropyranoxy. The term "4- to 10-membered heterocycloalkenyl" is to be understood as meaning an unsaturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), S(=0)2, NRa, in which Ra represents a hydrogen atom, or a Ci-C6-alkyl or Ci-C6-haloalkyl group; it being possible for said heterocycloalkenyl group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Examples of said heterocycloalkenyl may contain one or more double bonds, e.g. 4H-pyranyl, 2H-pyranyl, 3,6- dihydro-2H-pyran-4-yl, 3,6-dihydro-2H-thiopyran-4-yl, l,2,3,6-tetrahydropyridin-4-yl, 3H- diazirinyl, 2,5-dihydro-lH-pyrrolyl, [l,3]dioxolyl, 4H-[l,3,4]thiadiazinyl, 2,5-dihydrofuranyl, 2,3-dihydrofuranyl, 2,5-dihydrothiophenyl, 2,3-dihydrothiophenyl, 4,5-dihydrooxazolyl, 4H- [l,4]thiazinyl or 5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl group or it may be benzo fused.
The term "heteroaryl" is understood as meaning a monovalent, monocyclic- , bicyclic- or tricyclic aromatic ring system having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms (a "5- to 14- membered heteroaryl" group), 5 or 6 or 9 or 10 ring atoms (a "5- to 10-membered heteroaryl" group) or particularly 5 or 6 ring atoms ("5- to 6-membered heteroaryl" group), and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur, and in addition in each case can be benzocondensed. Particularly, heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, thia-4H-pyrazolyl etc., and benzo derivatives thereof, such as, for example, benzofuranyl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl, etc.; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc., and benzo derivatives thereof, such as, for example, quinolinyl, quinazolinyl, isoquinolinyl, etc.; or azocinyl, indolizinyl, purinyl, etc., and benzo derivatives thereof; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthpyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, xanthenyl, oxepinyl or lH-pyrrolo[2,3-b]pyridin-4-yl, etc..
In general, and unless otherwise mentioned, the heteroarylic or heteroarylenic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof. Thus, for some illustrative non-restricting example, the term pyridinyl or pyridinylene includes pyridin-2-yl, pyridin-2-ylene, pyridin-3-yl, pyridin-3-ylene, pyridin-4-yl and pyridin-4-ylene; or the term thienyl or thienylene includes thien-2-yl, thien-2-ylene, thien-3-yl and thien-3-ylene.
The term "Ci-Ce", as used throughout this text, e.g. in the context of the definition of "C1-C6- alkyl", "Ci-C6-haloalkyl", "Ci-C6-alkoxy" or "Ci-C6-haloalkoxy" is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 6, i.e. 1, 2, 3, 4, 5, or 6 carbon atoms. It is to be understood further that said term "Ci-Ce" is to be interpreted as any sub-range comprised therein, e.g. C1-C6 , C2-C5 , C3-C4 , C1-C2 , C1-C3 , C1-C4 , C1-C5; particularly C1-C2, C1-C3, C1-C4, C1-C5, C1-C6; more particularly C1-C4; in the case of "Ci-C6-haloalkyl" or "C1-C6- haloalkoxy" even more particularly C1-C2. Similarly, as used herein, the term "C2-C6", as used throughout this text, e.g. in the context of the definitions of "C2-C6-alkenyl" and "C2-C6-alkynyl", is to be understood as meaning an alkenyl group or an alkynyl group having a finite number of carbon atoms of 2 to 6, i.e. 2, 3, 4, 5, or 6 carbon atoms. It is to be understood further that said term "C2-C6" is to be interpreted as any subrange comprised therein, e.g. C2-C6, C3-C5, C3-C4, C2-C3, C2-C4, C2-C5; particularly C2-C3.
Further, as used herein, the term "C3-C6", as used throughout this text, e.g. in the context of the definition of "C3-C6-cycloalkyl", is to be understood as meaning a cycloalkyl group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term "C3-C6" is to be interpreted as any sub-range comprised therein, e.g. C3-C6, C4-C5, C3-C5, C3-C4, C4-C6, C5-C6; particularly C3-C6.
Further, as used herein, the term "C2-C4", as used throughout this text, e.g. in the context of "C2- C4-alkenyl" is to be understood as meaning a alkenyl group having a finite number of carbon atoms of 2 to 4, i.e. 2, 3 or 4 carbon atoms. It is to be understood further that said term "C2-C4" is to be interpreted as any sub-range comprised therein, e.g. C2-C4, C2-C3, C3-C4.
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties. Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.
By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
As used herein, the term "one or more", e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning "one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two".
The invention also includes all suitable isotopic variations of the compound of component A, particularly of Compound A. An isotopic variation of the compound of component A is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into the compound of component A include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), UC, 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F,
CI, Br, I, I, I and I, respectively. Certain isotopic variations of the compound of component A, for example, those in which one or more radioactive isotopes such as 3H or 14C are incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon- 14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of the compound of component A can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.
Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, polymorph, isomer, hydrate, solvate or the like.
The compounds of component A may contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be present in the (R) or (S) configuration, resulting in racemic mixtures in the case of a single asymmetric centre, and diastereomeric mixtures in the case of multiple asymmetric centres. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
The compounds of component A may contain sulphur atoms which are asymmetric, such as an as mmetric sulphoxide or sulphoximine group, of structure:
Figure imgf000014_0001
, for example, in which * indicates atoms to which the rest of the molecule can be bound.
Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention. Preferred compounds of component A are those which produce the more desirable biological activity, particularly preferred is Compound A. Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of component A are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers. Examples of appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation. The optically active bases or acids are then liberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chiral chromatography (e.g., chiral HPLC columns), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers. Suitable chiral HPLC columns are manufactured by Daicel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable. Enzymatic separations, with or without derivatisation, are also useful. The optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
In order to limit different types of isomers from each other reference is made to IUPAC Rules Section E (Pure Appl Chem 45, 11-30, 1976).
The present invention includes all possible stereoisomers of the compounds of component A as single stereoisomers, or as any mixture of said stereoisomers, e.g. R- or S- isomers, or E- or Z- isomers, in any ratio. Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of component A may be achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example. Further, the compounds of component A, particularly of Compound A, may exist as tautomers. For example, any compound of component A which contains a pyrazole moiety as a heteroaryl group for example can exist as a 1H tautomer, or a 2H tautomer, or even a mixture in any amount of the two tautomers, or a triazole moiety for example can exist as a 1H tautomer, a 2H tautomer, or a 4H tautomer, or even a mixture in any amount of said 1H, 2H and 4H tautomers, namely:
Figure imgf000016_0001
1 H-tautomer 2H-tautomer
Figure imgf000016_0002
1 H-tautomer 2H-tautomer 4H-tautomer
The present combination includes all possible tautomers of the compounds of component A, particularly the 1 H-tautomer or the 2H-tautomer of the pyrazol-5-yl group in 8-position of the naphthyridine core of Compound A, as single tautomers, or as any mixture of said tautomers, in any ratio.
Further, the compounds of component A, particularly Compound A, can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised. The present combination includes all such possible N-oxides of component A.
The present combination also covers useful forms of the compounds as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates. The compounds of the present combination can exist as a hydrate, or as a solvate, wherein the compounds of the present combination contain polar solvents, in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds. The amount of polar solvents, in particular water, may exist in a stoichiometric or non- stoichiometric ratio. In the case of stoichiometric solvates, e.g. a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, terra-, penta- etc. solvates or hydrates, respectively, are possible. The present combination includes all such hydrates or solvates.
Further, the compounds of the present combination can exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or can exist in the form of a salt. Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, customarily used in pharmacy.
The present invention includes all possible salts of the components of the present combination as single salts, or as any mixture of said salts, in any ratio.
Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of components of the present combination, either as single polymorphs, or as a mixture of more than one polymorphs, in any ratio.
When radicals in the compounds of the present combination are substituted, the radicals may be mono- or polysubstituted, unless specified otherwise. In the context of the present invention, all radicals which occur more than once are defined independently of one another. Substitution by one, two or three identical or different substituents is preferred.
In the context of the present invention, the term "treatment" or "treating" includes inhibition, retardation, checking, alleviating, attenuating, restricting, reducing, suppressing, repelling or healing of a disease or the development, the course or the progression of such states and/or the symptoms of such states.
The term "disease" includes but is not limited a condition, a disorder, an injury or a health problem. The term "therapy" is understood here to be synonymous with the term "treatment".
The terms "prevention", "prophylaxis" or "preclusion" are used synonymously in the context of the present invention and refer to the avoidance or reduction of the risk of contracting, experiencing, suffering from or having a disease or a development or advancement of such states and/or the symptoms of such states.
The treatment or prevention of a disease may be partial or complete.
COMPONENT A OF THE COMBINATION
Component A can be selected from inhibitors of ATR kinase specifically or generically disclosed in the following publications: J. Med. Chem. 2013, 56, 2125-2138; Exp. Rev. Mol. Med. 16, elO, 2014; WO2010054398 Al; WO2010071837A1; WO2010073034A1; WO2011143399A1; WO2011143419A1; WO2011143422A1; WO2011143423 A2; WO2011143425 A2; WO2011143426A1; WO2011154737A1; WO2011163527 Al; WO2012138938A1 ; WO2012178123A1 ; WO2012178124A1; WO2012178125A1 WO2013049719A1 ; WO2013049720A1 ; WO2013049722A1; WO2013049859 A 1 WO2013071085A1 ; WO2013071088A1 ; WO2013071090A1; WO2013071093A1 WO2013071094A1 ; WO2013152298 Al ; WO2014062604A1; WO2014089379A1 WO2014143240; WO 2014143241; WO 2014143242; ACS Med. Chem. Lett. 2015. 6, 37-41; ACS Med. Chem. Lett. 2015. 6, 42-46, WO 2015085132, WO 2015187451. The ATR kinase inhibitors mentioned in the prior art have been disclosed for the treatment or prophylaxis of different diseases, especially cancer. In another embodiment of the invention, said component A is selected from VX-803, VX-970, AZD-6738 and a compound of general formula (I)
Figure imgf000018_0001
(I)
in which:
R1 represents a group selected from:
Figure imgf000018_0002
wherein * indicates the point of attachment of said group with the rest of the molecule; represents hydrogen, halogen, -NR7R8, CN, Ci-C6-alkyl, Ci-C6-alkoxy, 3- to 10- membered heterocycloalkoxy, C2-C6-alkenyl, C3-C6-cycloalkyl, 3- to 10-membered heterocycloalkyl, 4- to 10-membered heterocycloalkenyl, phenyl, heteroaryl, -(CO)OR7, -(CO)NR7R8, -(S02)R9, -(SO)R9, -SR9, -(S02)NR7R8, -NR7(S02)R9, -((SO)=NRu)R10, -N=(SO)R9R10, -SiR10RuR12, -(PO)(OR7)2, -(PO)(OR7)R10 or -(PO)(R10)2,
wherein each Ci-C6-alkyl, Ci-C6-alkoxy, 3- to 10-membered heterocycloalkoxy, C2-C6- alkenyl, C3-C6-cycloalkyl, 3- to 10-membered heterocycloalkyl, phenyl or heteroaryl is optionally substituted, one or more times, independently from each other, with halogen, OH, -NR7R8, Ci-C6-alkyl optionally substituted one or more times with hydroxyl or phenyl, Ci-C6-haloalkyl, Ci-C6-alkoxy, C3-C6-cycloalkyl, 3- to 6- membered heterocycloalkyl, phenyl, -(CO)OR7, -(CO)NR7R8, -NR7(CO)R10, -NR8(CO)OR7, -NR8(CO) NR7R8, -(S02)R9, -(SO)R9, -SR9, -(S02)NR7R8,
-NR7(S02)R9, -((SO)=NRu)R10, -N=(SO)R9R10, -(PO)(OR7)2, -(PO)(OR7)R10, -(PO)(R10)2 or with a heteroaryl group which is optionally substituted, one or more times, with Ci-C/t-alkyl;
wherein each 4- to 10-membered heterocycloalkenyl is optionally substituted, one or more times, independently from each other, with Ci-C t-alkyl;
R3, R4 represent, independently from each other, hydrogen or methyl;
R7, R8 represent, independently from each other, hydrogen, Ci-C6-alkyl, C3-C6-cycloalkyl or phenyl, which phenyl is optionally substituted, one or more times, with halogen; or R7 and R8 together represent a 4-, 5-, 6- or 7-membered cyclic amine group, which is optionally substituted, one or more times, independently from each other, with a substituent selected from Ci-C6-alkyl, Ci-C6-haloalkyl, said 4-, 5-, 6- or 7-membered cyclic amine group optionally containing one further heteroatom selected from the group consisting of O, N and S;
R9 represents Ci-C t-alkyl or phenyl, wherein each Ci-C/t-alkyl or phenyl is optionally
substituted, one or more times, independently from each other, with R13;
R10 represents Ci-C/t-alkyl; or
R9 and R10 together, in case of -N=(SO)R9R10 group, represent a 5- to 8-membered
heterocycloalkyl group;
R11 represents hydrogen, Ci-C4-alkyl, -(CO)OR7, -(CO)NR7R8 or CN;
R12 represents hydrogen or Ci-C/t-alkyl;
R13 represents halogen, OH, -NR7R8, CN, N02, G-Ce-alkyl, G-Ce-haloalkyl, G-Ce-alkoxy,
Ci-Ce-haloalkoxy, C2-C6-alkenyl, C3-C6-cycloalkyl, -(CO)OR7 or -(CO)NR7R8;
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
In context with the present invention the term "VX-803" means 2-amino-6-fluoro-N-[5-fluoro-4- (4- { [4-(oxetan-3-yl)piperazin- l-yl]carbonyl Jpiperidin- l-yl)pyridin-3-yl]pyrazolo[ 1 ,5- a]pyrimidine-3-carboxamide. It has the following structure:
Figure imgf000019_0001
In context with the present invention the term "VX-970" means 3-(3 [(methylamino)methyl]phenyl } - 1 ,2-oxazol-5-yl)-5-[4-(propan-2-ylsulfonyl)phenyl]pyrazin-2 amine. It has the following structure:
Figure imgf000020_0001
In context with the present invention the term "AZD-6738" means 4-{4-[(3R)-3- methylmorpholin-4-yl] -6- [ 1 -(S-methylsulfonimidoyl)cyclopropyl]pyrimidin-2-yl } - 1 H- pyrrolo[2,3-b]pyridine. It has the following structure:
Figure imgf000020_0002
In another embodiment of the combination of the invention, said component A is selected from VX-803, VX-97 compound of general formula (lb)
Figure imgf000020_0003
(lb)
in which R1, R2, R4, R7, R8, R9, R10, R11, R12 and R13 are as defined for the compound of general formula (I) supra. In another embodiment of the invention, said component A is a compound of general formula (lb)
Figure imgf000021_0001
(lb) , in which
R
Figure imgf000021_0002
wherein * indicates the point of attachment of said group with the rest of the molecule; represents hydrogen, fluoro, chloro, CN, methyl, Ci-C/t-alkoxy, C2-C3-alkenyl, cyclopropyl, 3- to 6-membered heterocycloalkyl, 4- to 6-membered heterocycloalkenyl, phenyl, pyridinyl, thiazolyl, -(S02)R9, -SR9, -((SO)=NRu)R10, -N=(SO)R9R10, wherein each methyl, Ci-C t-alkoxy, C2-C3-alkenyl, cyclopropyl, 3- to 6-membered heterocycloalkyl, phenyl, pyridinyl or thiazolyl is optionally substituted, one or more times, independently from each other, with fluoro, chloro, OH, -NR7R8, methyl, 5-membered heterocycloalkyl, -NR8(CO)OR7,
-(S02)R9, -((SO)=NRU)R10,-(PO)(OR7)2, or with a group selected from:
Figure imgf000021_0003
wherein * indicates the point of attachment of said group with the rest of the molecule;
wherein each 4- to 6-membered heterocycloalkenyl is optionally substituted, one or more times, with methyl;
R4 represents hydrogen or methyl;
R7, R8 represent, independently from each other, hydrogen or Ci-C/t-alkyl;
R9 represents Ci-C/t-alkyl;
R10 represents Ci-C t-alkyl; or
R9 and R10 together, in case of -N=(SO)R9R10 group, represent a 6-membered heterocycloalkyl group;
R11 represents hydrogen, methyl, -(CO)OR7;
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
In another embodiment said component A is a compound selected from the group consisting of :
4- [(2-(morpholin-4-yl)- 8- [2H-pyrazol-3-yl] - [ 1 ,7]--naphthyridine-4-yl]phenyl-N- ethoxycarbonyl-S-methylsulphoximide
4- [(2-(morpholin-4-yl)- 8-(2H-pyrazol-3-yl)- [ 1 ,7]naphthyridine-4-yl]phenyl-S - methylsulphoximide
4-[6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3,6-dihydro-2H-pyran-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-[4-(N,S-dimethylsulfonimidoyl)phenyl]-2-[morpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[4-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(4-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l, 7] -naphthyridine
4-(2-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
dimethyl {4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl Jphosphonate
4-isopropenyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)- [1,7] naphthyridine
2- (morpholin-4-yl)-4-phenyl-8-(lH-pyrazol-3-yl)- [1,7] naphthyridine
4-[4-(S-ethylsulfonimidoyl)phenyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
3- [(2-(morpholin-4-yl)- 8- [2H-pyrazol-3-yl] - [ 1 ,7]naphthyridine-4-yl]phenyl-N- ethoxycarbonyl-S-methylsulphoximide
4- (l -methyl- l,2,3,6-tetrahydropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7- naphthyridine
4-(3-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-[5-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(l,2,3,6-tetrahydropyridin-4-yl)-l,7-naphthyridine 4-cyclopropyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
3- [(2-(morpholin-4-yl)- 8-(2H-pyrazol-3-yl)- [ 1 ,7]naphthyridine-4-yl]phenyl-S - methylsulphoximide
4- methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
4-[2-(methylsulfonyl)- 1 ,3-thiazol-4-yl] -2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]pyridin-2(lH)-one 5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]pyridin-2(lH)-one
4-[2-fluoro-4-(methylsulforiyl)pheriyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4- { 4-[S-(propan-2-yl)sulfonimidoyl]phenyl }-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine
4-(4-methanesulphonylphenyl)-2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)- [ 1 ,7] naphthyridine
2- ((R)-3-methylmorpholin-4-yl)-4-phenyl-8-(2H-pyrazol-3-yl)- [1,7] naphthyridine
4-(3-methanesulphonylphenyl)-2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)- [ 1 ,7] naphthyridine
4-cyclopropyl-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[ 1,7] -naphthyridine
4-[2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
3- [2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
4-methanesulphonyl-2-(morpholin-4-yl)-8-[2-(tetrahydropyran-2-yl)-2H-pyrazol-3-yl]- [ 1 ,7] naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(methylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine-4-carbonitrile
2-((R)-3-methylmorpholin-4-yl)-8-(-2H-pyrazol-3-yl]-[l,7]naphthyridine-4-carbonitrile 2-morpholin-4-yl-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine-4-carboxamide 4-methanesulphonylmethyl-2-morpholin-4-yl-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine
[2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]methanol
4-(l-methanesulphonylcyclopropyl)-2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)- [ 1 ,7] naphthyridine
4-isopropoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-(morpholin-4-yl)-4-(propan-2-yloxy)-8-(lH-pyrrol-2-yl)-l,7-naphthyridine
4-[3-(S-methylsulfonimidoyl)propoxy]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-ethoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-methoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-methyl-l-{ [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]oxy}propan-2-ol
2- (morpholin-4-yl)-8-(lH^yrazol-5-yl)-4-(tetrahydroi iran-2-ylmethoxy)-l,7-naphthyridine
3- { [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]oxy}dihydrofuran-2(3H)- one
4-[(3-methyl- 1 ,2-oxazol-5-yl)methoxy] -2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1 ,7- naphthyridine
4- [(5-methyl- 1 ,2-oxazol-3-yl)methoxy] -2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1 ,7- naphthyridine
4-benzyloxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l, 7] naphthyridine
4-isopropoxy-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine tert-butyl [4-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl }oxy)butyl] carbamate
4-methoxy-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine tert-butyl [3-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } oxy)propy 1] carbamate
2-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl }oxy)eth an amine
tert-butyl [2-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}oxy)ethyl]carbamate
4-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}oxy)butan- 1 - amine
2- [(3R,5S)-3,5-dimethylmorpholin-4-yl]-4-isopropoxy-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R,5R)-3,5-dimethylmorpholin-4-yl]-4-isopropoxy-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
4-chloro-2-morpholin-4-yl- 8-( 1 H-pyrazol-3-yl)- [ 1 ,7] naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(methylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N- { 2- [(3R)-3-methylmorpholin-4-yl] - 8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl } - 1 ,4□ 4- oxathian-4-imine 4-oxide
4-{ [dimethyl(oxido)- λ 6-sulfanylidene] amino }-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(piperazin-l-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-isopropoxy-2-((S)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-(morpholin-4-yl)-4-(propan-2-yloxy)-8-(lH-pyrrol-3-yl)-l,7-naphthyridine
4-(l-ethyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-methyl-lH-imidazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- { 2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl] aniline
4-(2,3-difluorophenyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
4-[2-fluoro-4-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1 ,7 -naphthyridine
4-fluoro-2-[2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl] aniline
4-(l-benzyl-lH-imidazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluorophenyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(2-methyl-l,3-thiazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4 4-methyl-6^methylsulfonyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
4-(l-cyclopropyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[2-fluoro-4-(piperazin-l-yl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[4-(methylsulfonyl)piperazin-l-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
N-(2,2-dimethylpropyl)-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
(l-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}piperidin-4- yl)methanol
N-cyclopropyl-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
4-(5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
N-(4-fluorophenyl)-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
2-[(3R)-3-methylmorpholin-4-yl]-4-(6-methylpyridin-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoro-4-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrrol-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoro-5-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-methoxypyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-methoxy-5-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(6-fluoro-2-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- [(3R)-3-methylmorpholin-4-yl] -4- [ 1 -methyl-3-(trifluoromethyl)- 1 H-pyrazol-5-yl] -8-( 1 H- pyrazol-5-yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methyl-2-thienyl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(5-methyl-2-thienyl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methyl-3-thienyl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-chloro-2-thienyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(2-methyl-3-thienyl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-l,7- naphthyridine
4-(3,5-dimethyl-l,2-oxazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-chloro-2-methoxypyridin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-yl)-l,7- naphthyridine 4-(3,6-dihydro-2H hiopyran -yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methylpiperidin-l-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-tert-butyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methyl-l,2-oxazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-ethyl-3-methyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(l,4-dimethyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-methyl-6-(methylsulfanyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
4-[2-methyl-6-(S-methylsulfonimidoyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8- (lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-propyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6,7-dihydro-5H-pyrrolo [ 1 ,2-a] imidazol-3-yl)-2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H- pyrazol-5-yl)- 1,7 -naphthyridine
4-[l-ethyl-3-(trifluoromethyl)-lH-pyrazol-5-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
methyl 5-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH- pyrrole-2-carboxylate
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2-thiazol-5-yl)-l,7-naphthyridine
N,N-dimethyl-2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl} aniline
4-(2,4-difluorophenyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine 4-(l -isopropyl- lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1,7- naphthyridine
ethyl methyl { 2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yljphosphinate
4- { [diethyl(oxido)- λ 6-sulfanylidene] amino } -2-[(3R)-3-methylmorpholin-4-yl]-8-( 1H- pyrazol-5-yl)- 1,7 -naphthyridine
isobutyl methyl} 2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yljphosphinate
2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}propan-2-ol 3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pentan-3-ol
4- (5-chloropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
5- fluoro-2- { 2-[(3R)-3-methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- l,7-naphthyridin-4- yl} aniline
4-[2-fluoro-3-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1 ,7 -naphthyridine
2- [(3R)-3-methylmorpholin-4-yl] -4- [ 1 -(oxetan-3-yl)- 1 H-pyrazol-5 -yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[2-fluoro-4-(pyrrolidin-l-yl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)-l,7-naphthyridine
4-[3-(methoxymethyl)-5-methyl-l,2-oxazol-4-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(5-methyl-l,3,4-oxadiazol-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N- { 2- [(3R)-3-methylmorpholin-4-yl] - 8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl } tetrahydro-
1 H- 1 4-thiophen- 1 -imine 1 -oxide
4- { [(4-fluorophenyl)(methyl)oxido- λ 6-sulf anylidene] amino } -2- [(3R)-3-methylmorpholin-4- yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine, mixture of 2 diastereoisomers
4- { [(2-fluorophenyl)(methyl)oxido- λ 6-sulf anylidene] amino } -2- [(3R)-3-methylmorpholin-4- yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine, mixture of 2 diastereoisomers
4-{ [(R)(2-fluorophenyl)(methyl)oxido- λ6-sulfanylidene]amino}-2-[(3R)-3- methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridine, diastereoisomer
4-{ [(S)(2-fluorophenyl)(methyl)oxido- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmorpholin- 4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine, diastereoisomer
4-(dimethylphosphoryl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(diethylphosphoryl)-2 (3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
ethyl isobutyl{2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-rlaphthyridirl-4- yljphosphinate
2-[(3R)-3-methylmo holirl-4-yl]-4-(morpholirl-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridine
4-(l-isobutyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4- [5-fluoro-6-(methylsulfonyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-
5- yl) - 1 ,7 -naphthyridine
4-[(3R)-3-methylmoφholin-4-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2- [(3R)-3-methylmoφholin-4-yl]-4-(4-methyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-fluoro-5-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1 ,7-naphthyridine
4-[4-(isopropylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoropyridin-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-ethyl-lH-imidazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
1 - { 2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl Jprolinamide
3- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pyridin-2- amine
2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[l-(2,2,2-trirluoroethyl)-lH-pyrazol- 5-yl]-l,7-naphthyridine
l-methyl-4-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}piperazin-2-one
4-[l-(2-fluoroethyl)-lH-pyrazol-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[l-(2-fluoroethyl)-lH-pyrazol-5-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1,7 -naphthyridine
2-(3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH- pyrazol- 1 -yl)ethanol
2-methyl-l-(3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } - 1 H-pyrazol- 1 -yl)propan-2-ol
4-[(2R)-2-methylmorpholin-4-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-fluoropyridin-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- [(3R)-3-methylmorpholin-4-yl]-4-(6-methylpyridin-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methylpyridin-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N-(2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl }phenyl)acetamide
3- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pyridin-2-ol 2-(3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl }phenyl)propan-2-ol
4- (5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[(2S)-2-methylmorpholin-4-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-[(trans)-2-methylcyclopropyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(difluoromethoxy)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]propan-2-ol
2-(morpholin-4-yl)-4-(3-oxa-8-azabicyclo[3.2.1]oct-8-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine 2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(pyrrolidin-l-yl)-l ,7-naphthyridine
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperazin-2-one
4-(dimethylphosphoryl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[(trans)-2,5-dimethylpiperazin-l-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[(cis)-3,5-dimethylpiperazin-l-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-3- (trifluoromethyl)azetidin-3-ol
methyl hydrogen {4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl Jphosphonate
4-(4-methylpiperazin- 1 -yl)-2-(morpholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[(3aR,6aS)-tetrahydro-lH-furo[3,4-c]pyrrol- 5(3H)-yl]- 1,7 -naphthyridine
4-(3-methoxy-3-methylazetidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4-[(lS,4S)-2-oxa-5-azabicyclo[2.2.1]hept-5-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[(methylsulfanyl)methyl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N,N-dimethyl-5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2- amine
4-(2-methylpyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
1 - { 2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yljcyclohexanol
2-fluoro-6- { 2-[(3R)-3-methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- l,7-naphthyridin-4- yl} aniline
(methyl} 4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}oxido- λ6- sulfanylidene)cyan amide
1 -ethyl-3-(methyl { 4- [2-(morpholin-4-yl)- 8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl]phenyl } oxido- λ 6-sulf anylidene)urea 3- ({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } oxy)propan- 1 -amine
4- (4-cyclopropyl-lH-l,2,3-triazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl) - 1 ,7 -naphthyridine
4-ethylsulfinyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-[propan-2-ylsulfinyl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[3-(methylsulfonyl)propoxy]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4-(phenylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(propan-2-ylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(ethylsulfonyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(phenylsulfinyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(methylsulfinyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmo holin-4-yl]-4-[l-oxidotetrahydro-2H-thiopyran-4-yl]-8-(lH-pyrazol-5- yl)- 1 ,7 -naphthyridine
4-(l,l-dioxidotetrahydro-2H-thiopyran-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4,8-di(lH-pyrazol-5-yl)-l,7-naphthyridine
N,N-dimethyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine
2-(morpholin-4-yl)-4-(phenylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-N-(propan-2-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine 4-(ethylsulfanyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(propan-2-ylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrol-2-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrol-3-yl)-l,7-naphthyridine
4-[(4-methoxyphenyl)sulfanyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(5-methyl-lH-pyrazol-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyrrolidin-2-one
4-(l , 1-dioxido- l,2-thiazolidin-2-yl)-2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-2-one
2- [(3R)-3-methylmorpholin-4-yl]-4-(2-methylpyridin-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[2-(propan-2-yloxy)pyridin-3-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(2-methoxypyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(pyridin-4-yl)-l,7-naphthyridine
4-[(4-methoxyphenyl)sulfanyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4- [3-fluoro-2-(morpholin-4-yl)pyridin-4-yl] -2- [(3R) -3-methylmorpholin-4-yl] - 8-( 1 H- pyrazol-5-yl)- 1,7 -naphthyridine
4-(6-fluoro-5-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 3-[2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl]- 1 ,3-oxazinan-2-one
3- [2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl]- 1 ,3-oxazolidin-2-one
4- (3-methoxypyridin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2,6-difluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5-chloro-2-fluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-fluoropyridin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-chloro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5,6-dimethylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5-fluoro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(5-methylthiophen-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-methoxythiophen-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-chlorothiophen-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(isoquinolin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-chlorothiophen-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methylthiophen-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4- (2,5-dimethylthiophen-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2 (3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-thiopyran-4-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l -methyl- l,2,5,6-tetrahydropyridin-3-yl)-8-(lH-pyrazol-
5- yl) - 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl] -4-(l -methyl- 1,2, 3,6-tetrahydropyridin-4-yl)-8-(lH-pyrazol- 5-yl) - 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-methylpiperidin-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2,3,6-tetrahydropyridin-4-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazol-3-yl]- 1,7 -naphthyridine
4-(4,6-difluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l,3-dimethyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l,5-dimethyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmo holin-4-yl]-4-(piperidin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[3-(trifluoromethyl)-lH-pyrazol-4- yl] - 1 ,7 -naphthyridine
4-(l-cyclobutyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-cyclopropyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-[l-(propan-2-yl)-lH-pyrazol-4-yl]-8-(lH-pyrazol-5-yl)- 1,7 -naphthyridine
4-[l-(difluoromethyl)-lH-pyrazol-4-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1 ,7 -naphthyridine
4-(l-tert-butyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,3,5-trimethyl-lH-pyrazol-4-yl)- 1 ,7 -naphthyridine
2- [(3R)-3-methylm(^holin-4-yl] -4- [ 1 -methyl-3-(trifluoromethyl)- 1 H-pyrazol-4-yl] -8-( 1 H- pyrazol-5-yl)- 1,7 -naphthyridine
2-(4-{2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH- pyrazol-l-yl)ethanol
4-(l-ethyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-(l-methyl-lH-pyrrol-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-[l-(propan-2-yl)-lH-pyrazol-3-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2,5-trimethyl-lH-pyrrol-3-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-(l-phenyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-(3-methyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine
2-[(3R)-3-methylmoφholin-4-yl]-4-[l-(2-methylpropyl)-lH-pyrazol-4-yl]-8-(lH-pyrazol-5- yl) - 1 ,7 -naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-(lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmoφholin-4-yl]-4-(l,3-oxazol-2-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(1 -dimethyl-lH-pyrazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l,5-dimethyl-lH-pyrazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morphoUn-4-yl)-8 lH-pyrazol-5-yl)-4-(l,3,5-trime l-lH-pyrazol-4-yl)-l,7- naphthyridine
4-{ [(2-methoxyethyl)(methyl)oxido- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmoφholin-4- yl] - 8 -( 1 H-pyrazol-5 -yl) - 1 ,7 -naphthyridine
4-{ [(4-bromophenyl)(oxido)propan-2-yl- λ 6-sulfanylidene] amino }-2-[(3R)-3- methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridine
2-(methyl-N-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } sulf onimidoyl)phenol
4-{ [(4-bromophenyl)(methyl)oxido- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmoφholin-4- yl] - 8 -( 1 H-pyrazol-5 -yl) - 1 ,7 -naphthyridine
4-{ [tert-butyl(methyl)oxido- λ 6-sulfanylidene]amino}-2-[(3R)-3-methylmoφholin-4-yl]-8- (lH-pyrazol-5-yl)-l,7-naphthyridine
formic acid - N-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-l,4 D4- oxathian-4-imine 4-oxide (1 : 1)
N-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]hexahydro-l D4-thiopyran-
1- imine 1 -oxide
3-methyl-2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}butan-2-ol
1 - { 2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl } - 1 - (tetrahydro-2H-pyran-4-yl)ethanol
3,3-dimethyl-2- { 2- [(3R)-3-methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl}butan-2-ol
2- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}hexan-2-ol 2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-3-yl)-l ,7-naphthyridine-4-carboxamide
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-(methylsulfonyl)cyclopropyl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-ylmethoxy)-l,7- naphthyridine
N,N-dimethyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide
{4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}(piperidin-l- yl)methanone
N,N-dimethyl-2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide N-cyclopropyl-4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide 4-(4-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(lH-indol-6-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(lH-indol-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
3- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benz amide
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide
N-methyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide
4- (3-fluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-chlorothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[2-(trifluoromethyl)phenyl]-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[4-(trifluoromethyl)phenyl]-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[3-(trifluoromethyl)phenyl]-l,7-naphthyridine 4-(3-chlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N- { 3- [2-(morpholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl]phenyl } acetamide 4-(3-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,5-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(3-methylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(furan-2-ylmethyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2,6-dimethyl-4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenol
4-(23-dimethylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
{3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}methanol
4-(4-fluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-chlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-fluoro-3-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-methylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,3-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N,N-dimethyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl] aniline
N,N-dimethyl-2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl] aniline
N- { 2- [2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl]phenyl } methanesulfonamide
N- { 4- [2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl]phenyl} methanesulfonamide
N,N-dimethyl-4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide
2-(morpholin-4-yl)-4- [( 1 E)-prop- 1 -en- 1 -yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridine
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenol
4-(2-fluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
{3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}(piperidin-l- yl)methanone
2- (morpholin-4-yl)-4-[4-(propan-2-yl)phenyl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N-cyclopropyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benz amide 4-(biphenyl-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,4-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2-chlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,5-dimethylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
3- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]aniline
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[3-(lH-pyrazol-l-yl)phenyl]-l,7-naphthyridine 3- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenol
4- (2-fluoro-5-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(5-fluoro-2-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2,4-difluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,3-difluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,6-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]aniline
4-(3,5-dichlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(biphenyl-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-chloropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(l-benzothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(l -methyl- lH-pyrazol-5-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(pyridin-3-yl)-l,7-naphthyridine
4-(2-methoxypyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
4-(5-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-3-yl)- 1,7-naphthyridine
2-(morpholin-4-yl)-4-[l-(phenylsulfonyl)-lH-indol-2-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
4-(6-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
{5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]thiophen-2-yl}methanol
4-(2-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine 4-(6-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
4-(2-chloro-6-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
4-(2-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine
4-(isoquinolin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- 1,7-naphthyridine 4-(3-chloropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-fluoropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,6-difluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l -methyl- lH-pyrazol-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine tert-butyl 5-methoxy-2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-lH- indole- 1 -carboxylate
2-(morpholin-4-yl)-4-[6-(morpholin-4-yl)pyridin-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(4-methylthiophen-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(thiophen-2-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(thiophen-3-yl)-l,7-naphthyridine
4-(3-methylthiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-chloro-5-methylpyridin-3-yl)-2-(morpholiri-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(5-chloro-2-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
tert-butyl 5-methyl-2- [2-(morpholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl] - 1 H- indole- 1 -carboxylate
4-(5-chloro-2-fluoropyridin-3-yl)-2-(morpholiri-4-yl)-8-(lH-pyrazol-5-yl)-l,7-riaphthyridirie 4-(3,5-dimethyl-l,2-oxazol-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-riaphthyridirie 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-8-yl)-l,7-naphthyridine
4-(5-methylthiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-riaphthyridirie 4-(6-ethoxypyridin-3-yl)-2-(morpholiri-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2-ethoxypyridin-3-yl)-2-(morpholiri-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-6-yl)-l,7-naphthyridine
4- (2-chlorothiophen-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
5- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]pyridin-2-amine
2-(morpholin-4-yl)-4-(lH-pyrazol-3-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(6-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-riaphthyridirie 4- (l -methyl- lH-pyrrol-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
5- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-ol
4-(5-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-chloro-2-methoxypyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-chlorothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[2-(methylsulfanyl)pyrimidin-5-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N-cyclopropyl-5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]pyrimidin-
2-amine
4-(isoquinolin-5-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-methyl-5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridine-2- carboxamide
N-tert-butyl-5-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridine-3- carboxamide
4-[5-(methylsulfanyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-l,7-naphthyridine
3- [2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-amine methyl 4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]thiophene-2- carboxylate
4- [2-methoxy-5-(trifluoromethyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4-[2-(propan-2-yloxy)pyridin-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5-chloro-6-ethoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l-tert-butyl-lH-pyrazol-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(piperidin-l-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-4-ol
N-methyl-2-(morpholin-4-yl)-N-phenyl-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine { l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]pyrrolidin-2-yl}methanol N-methyl-2-(morpholin-4-yl)-N^ropyl-8-(lH^yrazol-5-yl)-l,7-naphthyridin-4-amine 4-(azepan-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-methylpiperidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(4-methylpiperidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]piperidine-3-carboxamide
4-(2,5-dihydro-lH-pyrrol-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,4-dihydroquinolin-l(2H)-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,4-dihydroisoquinolin-2(lH)-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(13-dihydro-2H-isoindol-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[l,3,3-trimethyl-6-azabicyclo[3.2.1]oct-6-yl]-l,7- naphthyridine
tert-butyl l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-prolinate N-methyl-N-(2-methylpropyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)- l,7-naphthyridin-4- amine
N-(3-fluorophenyl)-N-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- amine
4-(l,l-dioxido-l-thia-6-azaspiro[3.3]hept-6-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-fluoropiperidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-(2-fluorophenyl)-N-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- amine
l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-prolinamide
{ l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-4-yl}methanol
4-(4-methoxypiperidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-(4-fluorophenyl)-N-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- amine
N-methyl- 1 - [2-(morpholin-4-yl)- 8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl] -prolinamide 4-[4-(ethylsulfonyl)piperazin-l-yl]-2-(morpholiri-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[4-(methylsulfonyl)piperazin- 1 -yl] -2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine
N-cyclopropyl-N-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-am
N-(2,2-dimethylpropyl)-N-methyl-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthy 4-amine
{ l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-3-yl}meth or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
The synthesis of the compounds of general formula (I) or (lb) of component A listed above is described in International Patent Publication WO2016020320 (Al). In a preferred embodiment, said component A is 2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl- lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine ("Compound A"), a tautomer, an N- oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, said component A is Compound A of structure
Figure imgf000044_0001
The synthesis of Compound A is described in Example 111 of WO2016020320 (Al).
The term "pharmaceutically acceptable salt" of component A, particularly of Compound A, refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," /. Pharm. Sci. 1911 , 66, 1-19. Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid. Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts. Those skilled in the art will further recognize that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods. Representative salts of a component A of this invention include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, sulfate, tartrate, thiocyanate, tosylate, and undecanoate.
Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, or butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl sulfate, or diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
Said component A may be in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially with component B and optionally component C as further described infra. The components A and B and optionally C may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route. The specific compounds of the lists as disclosed above are preferred as being component A of the combination, most preferred is "Compound A" used in the Experimental Section. It is to be understood that the present invention relates also to any combination of the embodiments of component A described above.
COMPONENT B OF THE COMBINATION
Component B is a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium- 223. A pharmaceutically acceptable salt of radium-223 can be, for example, an acid addition salt with an inorganic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, bisulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2 (4 hydroxybenzoyl) benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3 hydroxy 2 naphthoic, nicotinic, pamoic, pectinic, persulfuric, 3 phenylpropionic, picric, pivalic, 2 hydroxyethanesulfonate, itaconic, sulfamic, trifluoromethanesulfonic, dodecylsulfuric, ethansulfonic, benzenesulfonic, para toluenesulfonic, methansulfonic, 2 naphthalenesulfonic, naphthalinedisulfonic, camphorsulfonic acid, citric, tartaric, stearic, lactic, oxalic, malonic, succinic, malic, adipic, alginic, maleic, fumaric, D gluconic, mandelic, ascorbic, glucoheptanoic, glycerophosphoric, aspartic, sulfosalicylic, hemisulfuric, or thiocyanic acid, for example.
A preferred pharmaceutically acceptable salt of radium-223 is the dichloride (223RaCl2). Methods for preparation of a pharmaceutically acceptable solution comprising radium-223 are disclosed e.g. in WO 2000/40275(A2), WO 2011/134671(A1), and WO 2011/134672(A1).
Pharmaceutically acceptable solutions comprising radium-223 show a unique mechanism of action as a targeted radiopharmaceutical. They represent a new generation of alpha emitting therapeutic pharmaceuticals based on the natural bone-seeking nuclide radium-223. Preferably, an aqueous solution of radium-223 chloride (223R3Cl2) for intravenous injection, sterile and free from bacterial endotoxins is used.
Preferably, the solution is isotonic, containing a sodium citrate buffered saline to physiological pH. A preferred dosage regimen for radium-223 chloride injection is 50 kBq per kg body weight given at 4 week intervals, as a course consisting of 6 injections. Single radium-223 doses up to 250 kBq per kg body weight were evaluated in a phase I clinical trial. The observed adverse reactions at this dose were diarrhea and reversible myelosuppression (including one case (1/5) of grade 3 neutropenia).
As an example, the aqueous radium-223 dichloride solution may be supplied in a single-dose 10 ml vial which contains a fill volume of 6 ml. This product has a radioactivity concentration of radium-223 of 1,000 kBq/mL (0.03 mCi/mL), corresponding to 0.53 ng/mL of radium at reference date. The active moiety is the alpha particle emitting nuclide radium-223 (half-life is 11.4 days), present as a divalent cation (223Ra2+). The fraction of energy emitted from radium- 223 and its daughters as alpha-particles is 95.3%, the fraction emitted as beta-particles is 3.6%, and the fraction emitted as gamma-radiation is 1.1%. The combined energy from the emitted radiation from complete decay of radium-223 and its daughter nuclides is 28.2 MeV.
Radium-223 is to be administered intravenously by qualified personnel as a slow bolus injection. An intravenous access line should be used for administration of Radium-223. The line must be flushed with isotonic saline before and after injection of Radium-223. Radium-223 selectively targets areas of increased bone turnover, as in bone metastases, and concentrates by forming a complex with hydroxyapatite. Alpha emission contributes about 93% of the total radiation absorbed dose. The high linear energy alpha particle radiation induces double-strand DNA breaks, resulting in a potent and localized cytotoxic effect in the target areas containing metastatic cancer cells. The short path length (less than 100 micrometers) of the alpha particles minimizes the effect on adjacent healthy tissue such as the bone marrow.
COMBINATION In accordance with another aspect, the present invention provides combinations of at least two components, preferably two components, comprising component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium- 223. In accordance with another aspect, the present invention covers a combination of any component A mentioned herein with any component B mentioned herein, optionally with any component C mentioned herein. In accordance with a further aspect, the present invention concerns combinations of at least two components A and B, preferably of two components, comprising component A being an inhibitor of ATR kinase, particularly Compound A, and component B being pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223. In accordance with another aspect, the present invention provides combinations of at least two components A and B, preferably of two components, comprising component A being an inhibitor of ATR kinase, particularly Compound A, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable inorganic salt of the alkaline- earth radionuclide radium-223.
In accordance with a further aspect, the present invention provides combinations of at least two components, preferably of two components, comprising component A as described supra and component B as described supra, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
The combinations comprising at least two components A and B, preferably two components, as decribed and defined herein, are also referred to as "combinations of the present invention".
The synergistic behavior of a combination of the present invention is demonstrated herein with one of the ATR kinase inhibitors ("Compound A") specifically disclosed in the Examples section. In addition, a combination of the present invention comprising Compound A as mentioned above and a pharmaceutically acceptable salt of radium-223, particularly 223RaCh, is a preferred aspect of the invention.
In another aspect a combination of the present invention comprises Compound A or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, preferably the dichloride salt of radium-223. In a preferred embodiment the combination of the present invention comprises Compound A or a pharmaceutically acceptable salt thereof and the dichloride salt of radium-223.
Further, the present invention covers a kit comprising:
component A: one or more, preferably one, ATR kinase inhibitor(s) as described supra, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof;
component B: a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof as described supra.
In the kit optionally either or both of said components A and B in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially. The components A and B may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route. Preferably component A is administered by the oral route and component B is administered by the intravenous route.
Further, the present invention covers a kit comprising:
component A: one or more, preferably one, ATR kinase inhibitor(s) as described supra, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof;
component B: a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 or a solvate or a hydrate thereof as described supra; and, optionally, component C: one or more, preferably one, further pharmaceutical agent(s),
in which optionally either or all of said components A, B and C in any of the above-mentioned combinations are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially. The components A and B, optionally C, may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
The term "component C" being at least one pharmaceutical agent includes the effective compound itself as well as its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers as well as any pharmaceutical composition comprising such effective compound or its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers. A list of such pharmaceutical agents of component C is being provided further below. The combinations of component A and component B of this invention can be administered as the sole pharmaceutical agent or in combination with one or more further pharmaceutical agents C where the resulting combination of components A, B and C causes no unacceptable adverse effects. For example, the combinations of components A and B of this invention can be combined with component C, i.e. one or more further pharmaceutical agents, such as known anti-angiogenesis, anti-hyper-proliferative, antiinflammatory, analgesic, immunoregulatory, diuretic, antiarrhytmic, anti-hypercholsterolemia, anti-dyslipidemia, anti-diabetic or antiviral agents, and the like, as well as with admixtures and combinations thereof. Optional pharmaceutical agents which can be added as component C to the combination of components A and B can be one or more pharmaceutical agents such as 1311-chTNT, abarelix, abiraterone, aclarubicin, ado-trastuzumab emtansine, afatinib, aflibercept, aldesleukin, alectinib, alemtuzumab, alendronic acid, alitretinoin, altretamine, amifostine, aminoglutethimide, hexyl aminolevulinate, amrubicin, amsacrine, anastrozole, ancestim, anethole dithiolethione, anetumab ravtansine, angiotensin II, antithrombin III, aprepitant, arcitumomab, arglabin, arsenic trioxide, asparaginase, axitinib, azacitidine, basiliximab, belotecan, bendamustine, besilesomab, belinostat, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, blinatumomab, bortezomib, buserelin, bosutinib, brentuximab vedotin, busulfan, cabazitaxel, cabozantinib, calcitonine, calcium folinate, calcium levofolinate, capecitabine, capromab, carboplatin, carboquone, carfilzomib, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, ceritinib, cetuximab, chlorambucil, chlormadinone, chlormethine, cidofovir, cinacalcet, cisplatin, cladribine, clodronic acid, clofarabine, cobimetinib, copanlisib, crisantaspase, crizotinib, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daratumumab, darbepoetin alfa, dabrafenib, dasatinib, daunorubicin, decitabine, degarelix, denileukin diftitox, denosumab, depreotide, deslorelin, dianhydrogalactitol, dexrazoxane, dibrospidium chloride, dianhydrogalactitol, diclofenac, dinutuximab, docetaxel, dolasetron, doxifluridine, doxorubicin, doxorubicin + estrone, dronabinol, eculizumab, edrecolomab, elliptinium acetate, elotuzumab, eltrombopag, endostatin, enocitabine, enzalutamide, epirubicin, epitiostanol, epoetin alfa, epoetin beta, epoetin zeta, eptaplatin, eribulin, erlotinib, esomeprazole, estradiol, estramustine, ethinylestradiol, etoposide, everolimus, exemestane, fadrozole, fentanyl, filgrastim, fluoxymesterone, floxuridine, fludarabine, fluorouracil, flutamide, folinic acid, formestane, fosaprepitant, fotemustine, fulvestrant, gadobutrol, gadoteridol, gadoteric acid meglumine, gadoversetamide, gadoxetic acid, gallium nitrate, ganirelix, gefitinib, gemcitabine, gemtuzumab, Glucarpidase, glutoxim, GM-CSF, goserelin, granisetron, granulocyte colony stimulating factor, histamine dihydrochloride, histrelin, hydroxycarbamide, 1-125 seeds, lansoprazole, ibandronic acid, ibritumomab tiuxetan, ibrutinib, idarubicin, ifosfamide, imatinib, imiquimod, improsulfan, indisetron, incadronic acid, ingenol mebutate, interferon alfa, interferon beta, interferon gamma, iobitridol, iobenguane (1231), iomeprol, ipilimumab, irinotecan, Itraconazole, ixabepilone, ixazomib, lanreotide, lansoprazole, lapatinib, Iasocholine, lenalidomide, lenvatinib, lenograstim, lentinan, letrozole, leuprorelin, levamisole, levonorgestrel, levothyroxine sodium, lisuride, lobaplatin, lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melarsoprol, melphalan, mepitiostane, mercaptopurine, mesna, methadone, methotrexate, methoxsalen, methylaminolevulinate, methylprednisolone, methyltestosterone, metirosine, mifamurtide, miltefosine, miriplatin, mitobronitol, mitoguazone, mitolactol, mitomycin, mitotane, mitoxantrone, mogamulizumab, molgramostim, mopidamol, morphine hydrochloride, morphine sulfate, nabilone, nabiximols, nafarelin, naloxone + pentazocine, naltrexone, nartograstim, necitumumab, nedaplatin, nelarabine, neridronic acid, netupitant/palonosetron, nivolumabpentetreotide, nilotinib, nilutamide, nimorazole, nimotuzumab, nimustine, nintedanib, nitracrine, nivolumab, obinutuzumab, octreotide, ofatumumab, olaparib, omacetaxine mepesuccinate, omeprazole, ondansetron, oprelvekin, orgotein, orilotimod, osimertinib, oxaliplatin, oxycodone, oxymetholone, ozogamicine, p53 gene therapy, paclitaxel, palbociclib, palifermin, palladium- 103 seed, palonosetron, pamidronic acid, panitumumab, panobinostat, pantoprazole, pazopanib, pegaspargase, PEG-epoetin beta (methoxy PEG-epoetin beta), pembrolizumab, pegfilgrastim, peginterferon alfa-2b, pemetrexed, pentazocine, pentostatin, peplomycin, Perflubutane, perfosfamide, Pertuzumab, picibanil, pilocarpine, pirarubicin, pixantrone, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polyvinylpyrrolidone + sodium hyaluronate, polysaccharide-K, pomalidomide, ponatinib, porfimer sodium, pralatrexate, prednimustine, prednisone, procarbazine, procodazole, propranolol, quinagolide, rabeprazole, racotumomab, radotinib, raloxifene, raltitrexed, ramosetron, ramucirumab, ranimustine, rasburicase, razoxane, refametinib, regorafenib, risedronic acid, rhenium- 186 etidronate, rituximab, rolapitant, romidepsin, romiplostim, romurtide, roniciclib, samarium (153Sm) lexidronam, sargramostim, satumomab, secretin, siltuximab, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sonidegib, sorafenib, stanozolol, streptozocin, sunitinib, talaporfin, talimogene laherparepvec, tamibarotene, tamoxifen, tapentadol, tasonermin, teceleukin, technetium (99mTc) nofetumomab merpentan, 99mTc-HYNIC-[Tyr3] -octreotide, tegafur, tegafur + gimeracil + oteracil, temoporfin, temozolomide, temsirolimus, teniposide, testosterone, tetrofosmin, thalidomide, thiotepa, thymalfasin, thyrotropin alfa, tioguanine, tocilizumab, topotecan, toremifene, tositumomab, trabectedin, trametinib, tramadol, trastuzumab, trastuzumab emtansine, treosulfan, tretinoin, trifluridine + tipiracil, trilostane, triptorelin, trametinib, trofosfamide, thrombopoietin, tryptophan, ubenimex, valatinib, valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vismodegib, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimalamer, zoledronic acid, zorubicin or combinations thereof.
Preferred optional pharmaceutical agents which may be added as component C to the combination of components A and B is/ are one or more agents selected from enzalutamide, bicalutamide, flutamide, nilutamide, and/or abiraterone.
Generally, the use of cytotoxic and/or cytostatic agents as component C in combination with a combination of components A and B of the present invention may serve to:
(1) yield better efficacy in reducing the growth of a tumor and/or metastasis or even eliminate the tumor and/ or metastasis as compared to administration of either agent alone,
(2) provide for the administration of lesser amounts of the administered chemotherapeutic agents,
(3) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,
(4) provide for treating a broader spectrum of different cancer types in mammals, especially humans,
(5) provide for a higher response rate among treated patients,
(6) provide for a longer survival time among treated patients compared to standard chemotherapy treatments,
(7) provide a longer time for tumor progression, and/or
(8) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects.
Further, the present invention covers a pharmaceutical composition comprising a combination of the present invention as described supra together with one or more pharmaceutically acceptable excipients.
Further, the present invention covers a pharmaceutical composition comprising a combination of at least two components, component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 together with one or more pharmaceutically acceptable excipients.
Further, the present invention covers a pharmaceutical composition comprising a combination of at least two components, component A and component B, component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223, optionally with any component C mentioned herein, together with one or more pharmaceutically acceptable excipients.
In another embodiment the components A and B, and optionally component C, are present in separate formulations. In another embodiment the components A and B, and optionally component C, are present in a joint formulation.
Pharmaceutically acceptable excipients are non-toxic, preferably they are non-toxic and inert. Pharmaceutically acceptable excipients include, inter alia,
· fillers and excipients (for example cellulose, microcrystalline cellulose, such as, for example, Avicel®, lactose, mannitol, starch, calcium phosphate such as, for example, Di-Cafos®),
• ointment bases (for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols),
• bases for suppositories (for example polyethylene glycols, cacao butter, hard fat)
• solvents (for example water, ethanol, Isopropanol, glycerol, propylene glycol, medium chain-length triglycerides fatty oils, liquid polyethylene glycols, paraffins),
• surfactants, emulsifiers, dispersants or wetters (for example sodium dodecyle sulphate, lecithin, phospholipids, fatty alcohols such as, for example, Lanette®, sorbitan fatty acid esters such as, for example, Span®, polyoxyethylene sorbitan fatty acid esters such as, for example, Tween®, polyoxyethylene fatty acid glycerides such as, for example, Cremophor®, polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers such as, for example, Pluronic®),
• buffers and also acids and bases (for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine) isotonicity agents (for example glucose, sodium chloride),
adsorbents (for example highly-disperse silicas)
viscosity-increasing agents, gel formers, thickeners and/or binders (for example polyvinylpyrrolidon, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids such as, for example, Carbopol®, alginates, gelatine),
disintegrants (for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate such as, for example, Explotab®, cross- linked polyvinylpyrrolidon, croscarmellose-sodium such as, for example, AcDiSol®),
flow regulators, lubricants, glidant and mould release agents (for example magnesium stearate, stearic acid, talc, highly-disperse silicas such as, for example, Aerosil®), coating materials (for example sugar, shellac) and film formers for films or diffusion membranes which dissolve rapidly or in a modified manner (for example polyvinylpyrrolidones such as, for example, Kollidon®, polyvinyl alcohol, hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit®),
capsule materials (for example gelatine, hydroxypropylmethylcellulose),
synthetic polymers (for example polylactides, polyglycolides, polyacrylates, polymethacrylates such as, for example, Eudragit®, polyvinylpyrrolidones such as, for example, Kollidon®, polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers),
plasticizers (for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate),
penetration enhancers,
stabilisers (for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate), preservatives (for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate),
colourants (for example inorganic pigments such as, for example, iron oxides, titanium dioxide),
flavourings, sweeteners, flavour- and/or odour-masking agents.
Further excipients and procedures are described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al., "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311 ; Strickley, R.G "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1" PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324- 349 ; and Nema, S. et al., "Excipients and Their Use in Injectable Products" PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
The components A, B and C may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.
Component A is administered intravenously, intraperitoneally, preferably it is administered orally. Component B preferably is administered by the intravenous route. Component C is administered intravenously, intraperitoneally, preferably it is administered orally.
The pharmaceutical composition (formulation) varies by the route of administration. Components of this invention can be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both. Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present. Components of this invention can also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Oily suspensions can be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.
Syrups and elixirs can be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents. Components of this invention can also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in preferably a pharmaceutically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-l,l-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methycellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants.
Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta- aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
The pharmaceutical compositions of the present invention can be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate. The sterile injectable preparation can also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
Components of the invention can also be administered in the form of suppositories for rectal administration of the drug. These components can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.
Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.
It can be desirable or necessary to introduce a component of the present invention to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991.
In accordance with another aspect, the present invention concerns the use of the combination of the present invention as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone. In accordance with another aspect, the present invention concerns the kit as described herein for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone. In accordance with another aspect, the present invention concerns the pharmaceutical composition as described supra for the treatment or prophylaxis of a disease, preferably a hyper- proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
In accordance with another aspect, the present invention covers the use of such combinations as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
In accordance with another aspect, the present invention covers the use of such kit as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
In accordance with another aspect, the present invention covers the use of such pharmaceutical composition as described supra for the preparation of a medicament for the treatment or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone.
In accordance with another aspect, the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, using an effective amount of the combination as described supra.
In accordance with another aspect, the present invention concerns methods for the treatment and/or prophylaxis of a disease, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, using an effective amount of the kit or pharmaceutical composition as described supra.
In accordance with another aspect, the present invention concerns a method of treating a disease in a patient, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, comprising a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound
A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223,_wherein components A and B are administered simultaneously, concurrently, separately or sequentially.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223,_wherein components A and B are administered concurrently.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 2 hours to 96 hours prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 6 hours to 84 hours prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 12 hours to 72 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 24 hours to 48 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 18 hours to 30 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 20 hours to 28 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 22 hours to 26 hours prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 42 hours to 54 hours prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 44 hours to 52 hours prior to component A.
In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound
A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 46 hours to 50 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 24 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, wherein component B is administered 48 hours prior to component A. In accordance with another aspect, the present invention concerns a method of treating a disease in a patient, preferably a hyper-proliferative disease as described infra and/or metastases thereof, preferably metastases in bone, comprising
a) administering component A being an inhibitor of ATR kinase, particularly Compound A, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and
b) administering component B being a pharmaceutically acceptable salt of the alkaline- earth radionuclide radium-223, and optionally
c) administering component C being a pharmaceutical agent as described infra. The combinations, kits or pharmaceutical compositions of the present invention thus can be used for the treatment or prophylaxis of hyper-proliferative diseases, including diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, such as, for example, haematological tumors and/or metastases therof, solid tumors, and/or metastases thereof, e.g. leukemias, multiple myeloma thereof and myelodysplastic syndrome, malignant lymphomas, breast tumors including and bone metastases thereof, tumors of the thorax including non-small cell and small cell lung tumors and bone metastases thereof, gastrointestinal tumors, endocrine tumors, mammary and other gynaecological tumors and bone metastases thereof, urological tumors including renal, bladder and prostate tumors, skin tumors, and sarcomas, and/or metastases thereof.
The term "inappropriate" within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as preferably meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.
In particular, the present invention covers the treatment of lung carcinoma, in particular small- cell lung cancer, colorectal cancer, bladder cancer, lymphomas, in particular diffuse large B-cell lymphoma (DLBC) and mantle cell lymphoma (MCL), prostate cancer, in particular castration- resistant prostate cancer, gliomas, and ovarian cancer.
In one embodiment the invention covers combinations, kits or pharmaceutical compositions as described supra comprising component A or a pharmaceutically acceptable salt thereof and component B being a pharmaceutically acceptable salt of the alkaline earth radionuclide radium- 223 for use in the treatment of cancer indications particularly for such cancer type which form metastases in bone. Such cancer types are e.g. breast, prostate, lung, multiple myeloma, kidney or thyroid cancer. Another aspect of the invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of a hyper-proliferative disease and/or metastases thereof.
In accordance with another aspect, the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein components A and B are administered simultaneously, concurrently, separately or sequentially. In accordance with another aspect, the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein components A and B are administered concurrently.
In accordance with another aspect, the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein component B is administered prior to component A.
In accordance with another aspect, the present invention concerns the combination, the kit or the pharmaceutical composition according to the present invention for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, preferably metastases in bone, wherein component B is administered 2 hours to 96 hours, or 6 hours to 84 hours, or 12 hours to 72 hours, or 24 hours to 48 hours, or 18 hours to 30 hours, or 20 hours to 28 hours, or 22 hours to 26 hours, or 42 hours to 54 hours, or 44 hours to 52 hours, or 46 hours to 50 hours, or 24 hours, or 48 hours prior to component A. Another embodiment covers the use of a combination, kit or pharmaceutical composition according to the present invention for the preparation of a medicament for the treatment or prophylaxis of breast cancer, prostate cancer, multiple myeloma, non- small cell lung cancer and/or metastases thereof, especially wherein the metastases are bone metastases. In one embodiment the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer, in a subject, comprising administering to said subject a therapeutically effective amount of a combination according to the present invention. In another embodiment the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer, in a subject, comprising administering to said subject a therapeutically effective amount of a combination according to the present invention.
In another embodiment the invention covers a method of treatment or prophylaxis of a cancer, particularly breast cancer, prostate cancer, multiple myeloma, hepatocyte carcinoma, lung cancer, in particular non-small cell lung carcinoma, colorectal cancer, melanoma, or pancreatic cancer and/or metastases thereof in a subject, comprising administering to said subject a therapeutically effective amount of a combination accoring to the present invention.
Preferred uses of the combinations of the invention relate to the treatment of prostate cancer. The term "prostate cancer" as used herein means any histology type of prostate cancer including but not limited to acinar adenocarcinoma, ductal adenocarcinoma, transitional cell (or urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers, particularly acinar adenocarcinoma, stage MO castration-resistant prostate cancer (MO CRPC) or stage Ml castration-resistant prostate cancer (Ml CRPC), preferably Ml castration-resistant prostate cancer (Ml CRPC).
The terms "MO" and "Ml" (including Mia, Mlb, Mlc) are used in accordance with the "TNM staging system" for prostate cancer developed by the American Joint Committee on Cancer as further described in "TNM CLASSIFICATION OF MALIGNANT TUMORS", 7th edition Edited by James D. Brierley, Mary K. Gospodarowicz, Christian Wittekind, Published by UICC 2011. According to the TNM classification the term "MO" means that there are no distant metastases and that the cancer has not spread to other parts of the body. "Ml" means that there are distant metastases and that cancer has spread to distant parts of the body.
Preferred uses of the combinations of the invention are the treatment of castration-resistant prostate cancer (CRPC), particularly stage Ml CRPC, preferably stage Mlb castration-resistant prostate cancer (Mlb CRPC) or stage Mlc castration-resistant prostate cancer (Mlc CRPC).
The term "Mlb CRPC" as used herein means that the castration-resistant prostate cancer has spread to the bones. The term "Mlc CRPC" as used herein means that the castration-resistant prostate cancer has spread to other organs such as lungs, liver, or brain (with or without spread to the bones).
Preferred uses of the combinations of the invention are the treatment of prostate cancer, especially castration-resistant prostate cancer (CRPC), preferably of CRPC with symptomatic bone metastases and no known visceral metastatic disease.
One preferred embodiment is the use of the combinations, the kits or the pharmaceutical compositions of the invention for the treatment of prostate cancer, especially of castration- resistant prostate cancer (CRPC) with symptomatic bone metastases and no known visceral metastatic disease.
In another embodiment the use of the combinations, the kits or the pharmaceutical compositions of the invention concern the treatment of castration resistant prostate cancer (MO CRPC) or of Ml castration-resistant prostate cancer (Ml CRPC) in a subject, wherein the subject is chemotherapy-naive.
The term "chemotherapy-naive" as used herein means that the subject, prior to the treatment with the combinations, the kits or the pharmaceutical compositions of the present invention has not received a chemotherapy.
In another embodiment the use of the combinations, the kits or the pharmaceutical compositions of the present invention concern the treatment of castration resistant prostate cancer (MO CRPC) or of Ml castration-resistant prostate cancer (Ml CRPC) in a subject, wherein the subject has received a chemotherapy prior to the treatment with the combinations, the kits or the pharmaceutical compositions of the invention.
The term "chemotherapy" as used herein means a category of cancer treatment that uses one or more chemotherapeutic agents as part of a standardized chemotherapy regimen. Chemotherapeutic agents are rather non-specific agents such as for example alkylating agents, anthracyclines, taxanes, epothilones, histone deacetylase inhibitors, inhibitors of topoisomerase I, inhibitors of topoisomerase II, nucleotide analogues, platinum-based agents, vinca alkaloids, etc..
Another embodiment of the present invention concerns the use of the combinations of the invention for the treatment of breast cancer, in particular breast cancer with bone metastases (Amol Takalkar et al., Exp Hematol Oncol. 2014; 3: 23. "Radium-223 dichloride bone-targeted alpha particle therapy for hormone-refractory breast cancer metastatic to bone"; Suominen et al., J Natl Cancer Inst. 2013 Jun 19;105(12):908-16. "Survival benefit with radium-223 dichloride in a mouse model of breast cancer bone metastasis"). In another embodiment the present invention concerns the use of the combinations of the present invention for the treatment of multiple myeloma.
Combinations, kits or pharmaceutical compositions of the present invention might be utilized to inhibit, block, reduce, decrease, etc., cell proliferation and/or cell division, and/or produce apoptosis.
This invention includes a method comprising administering to a mammal in need thereof, including a human, an amount of a component A and an amount of component B of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof, which is effective to treat the disease. Hyper-proliferative diseases include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), as well as malignant neoplasia. Examples of malignant neoplasia treatable with the compounds according to the present invention include solid and hematological tumors. Solid tumors can be exemplified by tumors of the breast, bladder, bone, brain, central and peripheral nervous system, colon, anum, endocrine glands (e.g. thyroid and adrenal cortex), esophagus, endometrium, germ cells, head and neck, kidney, liver, lung, larynx and hypopharynx, mesothelioma, ovary, pancreas, prostate, rectum, renal, small intestine, soft tissue, testis, stomach, skin, ureter, vagina and vulva. Malignant neoplasias include inherited cancers exemplified by Retinoblastoma and Wilms tumor. In addition, malignant neoplasias include primary tumors in said organs and corresponding secondary tumors in distant organs ("tumor metastases"). Hematological tumors can be exemplified by aggressive and indolent forms of leukemia and lymphoma, namely non- Hodgkins disease, chronic and acute myeloid leukemia (CML / AML), acute lymphoblastic leukemia (ALL), Hodgkins disease, multiple myeloma and T-cell lymphoma. Also included are myelodysplastic syndrome, plasma cell neoplasia, paraneoplastic syndromes, and cancers of unknown primary site as well as AIDS related malignancies.
Examples of breast cancer include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ, particularly with bone metastases. Examples of cancers of the respiratory tract include, but are not limited to small-cell and non- small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
Examples of brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.
Examples of liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell. Lymphomas include, but are not limited to AIDS -related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin' s disease, and lymphoma of the central nervous system.
Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia. These diseases have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
Combinations of the present invention might also be used for treating diseases associated with excessive and/or abnormal angiogenesis.
Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity [Aiello et al. New Engl. J. Med. 1994, 331, 1480 ; Peer et al. Lab. Invest. 1995, 72, 638], age-related macular degeneration [AMD ; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855], neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc. In addition, the increased blood supply associated with cancerous and neoplastic tissue, encourages growth, leading to rapid tumor enlargement and metastases. Moreover, the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastases and the consequence spread of the cancer. Thus, combinations of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis diseases, e.g., by inhibiting and/or reducing blood vessel formation ; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.
DOSE AND ADMINISTRATION
Component A
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper-proliferative diseases and angiogenic diseases, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredients to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular component and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated. The total amount of the active ingredients to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 50 mg/kg body weight per day. Clinically useful dosing schedules of a compound will range from one to three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
Component B
A preferred dosage regimen for radium-223 injection is 50 kBq per kg body weight given at 4 week intervals, as a course consisting of 6 injections. Single radium-223 doses up to 250 kBq per kg body weight were evaluated in a phase I clinical trial. The observed adverse reactions at this dose were diarrhea and reversible myelosuppression (including one case (1/5) of grade 3 neutropenia).
As an example, the aqueous radium-223 dichloride solution may be supplied in a single-dose 10 ml vial which contains a fill volume of 6 ml. This product has a radioactivity concentration of radium-223 of 1,000 kBq/mL (0.03 mCi/mL), corresponding to 0.53 ng/mL of radium at reference date.
Radium-223 is to be administered intravenously by qualified personnel as a slow bolus injection. An intravenous access line should be used for administration of radium-223. The line must be flushed with isotonic saline before and after injection of radium-223.
Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compounds employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
EXPERIMENTAL SECTION
Example 1
Synergistic effect of the combination of Compound A and radium-223 dichloride Component A :
In this Experimental Section and in the Figures, the term "Compound A" is an example of component A. Compound A is described in Example 111 of International Patent Application WO2016020320 (Al). As shown herein Compound A is 2-[(3R)-3-methylmorpholin-4-yl]-4-(l- methyl-lH-pyrazol-5-yl -8-(lH-pyrazol-5-yl)-l,7-naphthyridine, of structure:
Figure imgf000073_0001
Compound A
Component B :
In this Experimental Section and in the Figures, component B is radium-223 dichloride, the synthesis of which is disclosed in WO2000/040275, which is incorporated herein in its entirety by reference.
Test system/ In vivo Model (applied in Experiments 1, 2 and 3):
The efficacy of Compound A in combination with radium-223 dichloride (Ra-223) was studied in the osteoblastic and mixed prostate cancer bone metastases model LNCaP-luc in male NOD/Scid mice (Minhong Zou et al., ONCOLOGY REPORTS 30: 615-622, 2013; Multiple metastases in a novel LNCaP model of human prostate cancer). LNCaP is an androgen-receptor (AR) positive CRPC cell line with several mutations/deletions in DNA damage or mismatch repair genes (APCR2714C, ARIDlAfs, ATG5fs, ATMA1119V/K1572N, ATRXEE2264- 2265E, BRCA2fs, CHEK2T430N, ERCC3A740T;R391W, ERCC5L1023I,
FANCAE369D,Q652*, HDAC2A62V, MLH3I541V, MSH3PPA66-68-;fs, POLBfs,
POLHD631G, PRKDCfs, PTENfs, RAD50fs, RAD54LL532M, RBlsplice_acceptor, SLX4S605N, TDP2T308S, TP53BP1R639Q;Q111*, TRRAPR2665W;P3554L, WDR48G107*, XRCC3P87L, XRCC4L70M) as well as in genes inducing replication stress or oncogenes (ATRK1379N, ERBB3K177N, MYCN45S,TOP2Afs, TC>P2BG323*/V889A) (fs: frame shift; del: deletion; *: stop codon; amp: gene amplification). LNCaP cells secrete prostate specific antigen (PSA) and are known to form osteoblastic and mixed lesions when inoculated to bone marrow cavity. Therefore this model can be effectively used for testing the effects of cancer drug candidates on the growth of prostate cancer cells in bone. In this experiment a luciferase transfected LNCaP cell line (LNCaP-luc) has been used to allow additional tumor growth monitoring by Bioluminescent Imaging (BLI) (Ruxana T. Sadikot and Timothy S. Blackwell; Proc Am Thorac Soc Vol 2. pp 537-540, 2005; Bioluminescence Imaging).
Mice were 5-8 weeks of age at the beginning of the study. On day 0 mice were given intratibial inoculation of LNCaP-luc human prostate cancer cells. The development of osteoblastic lesions were monitored by X-ray imaging before dosing was started and in 3 weeks intervals thereafter. The lesion area in hind limbs was determined from the images with MetaMorph image analysis software (Molecular Devices LLC, Sunnyvale, CA, USA) by drawing the outlines of radio- opaque and -lucent areas. Tumor burden was quantified by imaging the bioluminescence emitted by the LNCaP-luc cells. The obtained data were analyzed using Living Image® software (PerkinElmer, Waltham, MA, USA) version 4.2. Average radiance (p/s/cm2/sr) was determined from the inoculated tibia. Blood samples were taken every second week and the animals radiographed every three weeks, starting from week four. On week five, animals were stratified to treatment groups (n = 10-20) according to the BLI signal, PSA and radiography, and dosing was started. Body weights were determined twice a week. Changes in the body weight were a measure of treatment-related toxicity (> 10% = critical, stoppage in treatment until recovery, > 20% = toxic, termination). The animals were sacrificed six or seven weeks after dosing was started. PSA and N-terminal procollagen type I (PINP) were measured in plasma as markers of tumor growth and bone formation. PSA was measured using Human PSA ELISA assay (R&D Systems) (Hsing AW, Chokkalingam AP. Prostate cancer epidemiology. Front Biosci.
2006;11 : 1388-413; Schroder FH, Wildhagen MR Rotterdam Study Group of the 'European Randomised Study of Screening for Prostate Cancer' (ERSPC) Screening for prostate cancer: Evidence and perspectives. BJU Int. 2001;88:811-7), the levels of PINP were determined using VICTOR2 Multilabel Counter (PerkinElmer, Waltham, MA, USA) (N. Koopmans et al., THE JOURNAL OF UROLOGY, Vol. 178, 849-853, September 2007. Serum Bone Turnover Markers (PINP and ICTP) for the Early Detection of Bone Metastases in Patients With Prostate Cancer: A Longitudinal Approach; Windy Dean-Colomb et al.; Breast Cancer Res Treat. 2013 January ; 137(2). Elevated serum PINP predicts development of bone metastasis and survival in early-stage breast cancer). Statistical analysis was effected using single-factor ANOVA and comparison with the control group by means of pair-by-pair comparative analysis (Dunnett's test) or student's t-test.
Experiment 1:
The following experimental groups were assessed:
1) Vehicle for Ra-223: 28 mmol/L sodium citrate, 5 ml/kg, iv (=intraveneous) every 4 weeks (q4w)
2) Ra-223: 300 kBq/kg, 5 ml/kg, iv, q4w
3) Compound A dose 1 : 20 mg/kg 2 days on/ 5 days off po (= peroral) once per day (qd) + Vehicle for Ra-223,28 mmol/L sodium citrate, 5 ml/kg, iv, q4w
4) Compound A dose 2: 50 mg/kg 2 days on/ 5 days off po qd+ Vehicle for Ra-223, 28 mmol/L sodium citrate, 5 ml/kg, iv, q4w
5) Combo Ra-223 + Compound A dose 1: Ra-223 (300 kBq/kg, 5 ml/kg, iv, q4W, + Compound A (20 mg/kg 2 days on/ 5 days off po qd, started on week 4 after tumor cell inoculation) 6) Combo Ra-223 + Compound A dose 2: Ra-223 (300 kBq/kg, 5 ml/kg, iv, q4W, started on week 4 after tumor cell inoculation) + Compound A (50 mg/kg 2 days on/ 5 days off po qd, started on week 4 after tumor cell inoculation)
Results of Experiment 1:
Tumor growth in vehicle treated control group resulted in an increase of PSA level in plasma, increase of the BLI signal in bone and an enlarged bone lesion area. However, BLI analysis revealed high variation in the vehicle treated control group, whereby this parameter has been excluded in the final study analysis. Ra-223 single agent decreased PINP level in plasma compared to vehicle treated control group. Compound A treatment alone had no significant effect on tumor burden as analyzed by PSA or PINP. Combination therapies were significantly (P < 0.05) more effective in decreasing PSA (see Figure 1 A) and PINP (see Figure 1 B) level in plasma than vehicle, Ra-223 alone or the respective test dose of Compound A alone. The treatments were well tolerated, no critical body weight loss >10% was recorded (see Figure 2). In summary, our data indicate synergistic effects of the ATR inhibitor Compound A and radium- 223 in inhibiting tumor growth in the prostate cancer bone metastases model LNCaP-luc, indicating a strong potential as a future treatment of CRPC patients with bone metastases. To evaluate the cooperativity of the combination of Compound A with radium-223 dichloride expected additivity was calculated according to the Bliss model (C=A+B-A*B; wherein C is the expected T/C of the combination of drug A and drug B if they act additive, A is T/C of drug A, B is T/C of drug B). Excess >10% over the expected additive effect is assumed to indicate synergism of the two drugs, less than 10% of the expected additive effect is assumed to indicate antagonism (Bliss, C.I., The toxicity of poisons applied jointly. Ann. Appl. Biol. 26, 585-615, 1939) (see Table 1):
Table 1:
Anti-tumor activity of Compound A with Ra-223 in LNCaP-luc human prostate cancer bone metastases xenograft model in NOD/Scid mice
* P < 0.05 compared to vehicle control
* P < 0.05 compared to Ra-223
a) T/C = ratio of the PSA level of treatment versus control at the end of the study.
b) T/C = ratio of the PINP level of treatment versus control at the end of the study.
Experiment 2:
To evaluate different dosing schedules of Compound A in Ra-223 combination treatment the following experimental groups were assessed in Experiment 2:
1) Vehicle for Ra-223: 28 mmol/L sodium citrate, 5 ml/kg, iv (= intraveneous) every 4 weeks (q4w)
2) Ra-223: 150 kBq/kg, 5 ml/kg, iv, q4w
3) Compound A: 20 mg/kg 2 days on/ 5 days off po (= peroral) once per day (qd) 4) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (20 mg kg 2 days on/ 5 days off po, qd, start concurrently to Ra-223)
5) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (20 mg/kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223)
6) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (20 mg/kg 2 days on/ 5 days off po, qd, start 48 hours after Ra-223)
7) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (20 mg/kg 2 days on/ 26 days off po, qd, start 24 hours after Ra-223) Results of Experiment 2 :
Tumor growth in vehicle treated control group resulted in an increase of PSA level in plasma, increase of the BLI signal in bone and an enlarged bone lesion area. Ra-223 or Compound A single agent treatments had no significant effect on tumor burden as analysed by PSA level in plasma, BLI signal and total bone lesion area in comparison to vehicle treated control group. Combination therapies were significantly (P < 0.05) more effective in decreasing PSA level in plasma compared to vehicle control when Compound A was applied in weekly cycles for 2 days on and 5 days off starting concurrently with Ra-223 or 24h or 48h after Ra-223 (see Figure 3 A). Total bone lesion area was significantly reduced in combination therapy compared to vehicle control when Compound A was applied in weekly cycles for 2 days on and 5 days off starting concurrently with Ra-223 or 24h after Ra-223 (see Figure 3 B). Tumor burden determined by BLI signal in bone was significantly reduced by all tested combination treatment schedules in comparison to vehicle control (see Figure 3 C). Overall, treatments were well tolerated. No treatment related critical body weight loss (> 10%) or toxicities/death were recorded (data not shown).
In summary, our data indicate that, based on tumor burden (PSA, BLI) and bone lesions, Compound A treatment starting 24h after the first Ra-223 dose and applied in continued weekly cycles, achieves best anti-tumor activity in Ra-223 combination treatment in the prostate cancer bone metastases model LNCaP-luc. However, Compound A treatment starting concurrently or 48h after the first Ra-223 dose and applied in continued weekly cycles, as well as Compound A treatment starting 24h after each Ra-223 dose, applied in 4 weekly cycles, also show inhibitory effects in LNCaP-luc bone tumor growth in combination treatment with Ra-223. Table 2:
Anti-tumour activity of different dosing schedules of Compound A in combination treatment with Ra-223 in LNCaP-luc human prostate cancer bone metastases xenograft model in
NOD/Scid mice
Figure imgf000078_0001
* P < 0.05 compared to vehicle control
b) T/C = ratio of the PSA level of treatment versus control at the end of the study, b) T/C = ratio of the total bone lesion area of treatment versus control at the end of the study. c) T/C = ratio of the BLI signal of treatment versus control at the end of the study.
Experiment 3:
To evaluate different doses of Compound A in Ra-223 combination treatment the following experimental groups were assessed in Experiment 3:
1) Vehicle for Ra-223: 28 mmol/L sodium citrate, 5 ml/kg, iv (= intraveneous) every 4 weeks (q4w)
2) Ra-223: 150 kBq/kg, 5 ml/kg, iv, q4w
3) Compound A: 20 mg/kg 2 days on/ 5 days off po (= peroral) once per day (qd)
4) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (5 mg/kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223)
5) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (10 mg/kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223) 6) Combo Ra-223 (150 kBq kg, 5 ml/kg, iv, q4w) + Compound A (20 mg kg 2 days on/ 5 days off po, qd, start 24 hours after Ra-223)
7) Combo Ra-223 (150 kBq/kg, 5 ml/kg, iv, q4w) + Compound A (40 mg/kg 1 day on/ 6 days off po, qd, start 24 hours after Ra-223)
Results of Experiment 3:
Tumor growth in vehicle treated control group resulted in an increase of PSA level in plasma, increase of the BLI signal in bone and an enlarged bone lesion area. Ra-223 or Compound A single agent treatments had no significant effect on tumor burden as analysed by PSA level in plasma, BLI signal and total bone lesion area compared to vehicle treated control group, evaluation of bone growth marker PINP in plasma revealed reduction by Ra-223 alone compared to vehicle treated control. All tested Compound A doses evaluated in combination with Ra-223 achieved significant (P < 0.05) decrease in serum PSA or PINP level, BLI signal and bone lesion area compared to vehicle control group (see Table 3, Figure 4 A-D), except Ra-223 + Compound A 5 mg/kg achieving no significance with regards to PSA (see Figure 4 A). Overall, treatments were well tolerated. No treatment related critical body weight loss (> 10%) or toxicities/death were recorded (data not shown).
In summary, our data indicate that, based on tumor burden (PSA, BLI), bone growth (PINP) and bone lesions, Compound A doses higher than 5 mg/kg applied once daily for 2 days on and 5 days off starting 24h after the first Ra-223 dose and given in continued weekly cycles, demonstrate anti-tumor activity of the combination treatment in the prostate cancer bone metastases model LNCaP-luc and improve efficacy of respective monotherapies. Overall, best anti-tumor activity, at good tolerability, was achieved with Compound A doses of 20 mg/kg once daily for 2 days on and 5 days off or 40 mg/kg once daily for 1 day on and 6 days off starting 24h after the first Ra-223 dose and given in continued weekly cycles.
Table 3:
Anti-tumour activity of different doses of Compound A in combination treatment with Ra-223 in LNCaP-luc human prostate cancer bone metastases xenograft model in NOD/Scid mice
Figure imgf000080_0001
* P < 0.05 compared to vehicle control
* P < 0.05 compared to Ra-223
a) T/C = ratio of the PSA level of treatment versus control at the end of the study.
b) T/C = ratio of the PINP level of treatment versus control at the end of the study.
c) T/C = ratio of the total bone lesion area of treatment versus control at the end of the study, d) T/C = ratio of the BLI signal of treatment versus control at the end of the study.

Claims

1. A combination of at least two components, component A and component B, comprising a component A being an inhibitor of ATR kinase or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, and component B being a pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223.
2. The combination according to claim 1, in which said component A is selected from VX-803, VX-970, AZD-6738 and a compound of general formula I:
Figure imgf000081_0001
(') , in which:
represents a group selected from:
Figure imgf000081_0002
wherein * indicates the point of attachment of said group with the rest of the molecule; represents hydrogen, halogen, -NR7R8, CN, Ci-C6-alkyl, Ci-C6-alkoxy, 3- to 10- membered heterocycloalkoxy, C2-C6-alkenyl, C3-C6-cycloalkyl, 3- to 10-membered heterocycloalkyl, 4- to 10-membered heterocycloalkenyl, phenyl, heteroaryl, -(CO)OR7, -(CO)NR7R8, -(S02)R9, -(SO)R9, -SR9, -(S02)NR7R8, -NR7(S02)R9, -((SO)=NRu)R10, - N=(SO)R9R10, -SiR10RuR12, -(PO)(OR7)2, -(PO)(OR7)R10 or -(PO)(R10)2,
wherein each Ci-C6-alkyl, Ci-C6-alkoxy, 3- to 10-membered heterocycloalkoxy, d- e- alkenyl, C3-C6-cycloalkyl, 3- to 10-membered heterocycloalkyl, phenyl or heteroaryl is optionally substituted, one or more times, independently from each other, with halogen, OH, -NR7R8, Ci-C6-alkyl optionally substituted one or more times with hydroxyl or phenyl, Ci-C6-haloalkyl, Ci-C6-alkoxy, C3-C6-cycloalkyl, 3- to 6- membered heterocycloalkyl, phenyl, -(CO)OR7,
-(CO)NR7R8, -NR7(CO)R10, -NR8(CO)OR7, -NR8(CO) NR7R8, -(S02)R9, -(SO)R9, - SR9, -(S02)NR7R8, -NR7(S02)R9, -((SO)=NRu)R10, -N=(SO)R9R10, -(PO)(OR7)2, -(PO)(OR7)R10, -(PO)(R10)2 or with a heteroaryl group which is optionally substituted, one or more times, with G-C -alkyl;
wherein each 4- to 10-membered heterocycloalkenyl is optionally substituted, one or more times, independently from each other, with Ci-C -alkyl;
R3, R4 represent, independently from each other, hydrogen or methyl;
R7, R8 represent, independently from each other, hydrogen, Ci -alkyl, -cycloalkyl or phenyl, which phenyl is optionally substituted, one or more times, with halogen; or R7 and R8 together represent a 4-, 5-, 6- or 7-membered cyclic amine group, which is optionally substituted, one or more times, independently from each other, with a substituent selected from Ci -alkyl, Ci -haloalkyl, said 4-, 5-, 6- or 7-membered cyclic amine group optionally containing one further heteroatom selected from the group consisting of
O, N and S;
R9 represents Ci-C -alkyl or phenyl, wherein each Ci-C -alkyl or phenyl is optionally
substituted, one or more times, independently from each other, with R13;
R10 represents Ci-C -alkyl; or
R9 and R10 together, in case of -N=(SO)R9R10 group, represent a 5- to 8-membered
heterocycloalkyl group;
R11 represents hydrogen, Ci-C4-alkyl, -(CO)OR7, -(CO)NR7R8 or CN;
R12 represents hydrogen or Ci-C -alkyl;
R13 represents halogen, OH, -NR7R8, CN, N02, G-Ce-alkyl, G-Ce-haloalkyl, G-Ce-alkoxy, Ci-Ce-haloalkoxy, C2-C6-alkenyl, C3-C6-cycloalkyl, -(CO)OR7 or -(CO)NR7R8;
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, or a mixture of same.
3. The combination according to claim 1 or 2, in which said component A is selected from VX- 803, VX-970, AZD-6738 and a compound of formula (lb)
Figure imgf000082_0001
(lb) , in which
R1 represents:
Figure imgf000083_0001
, wherein * indicates the point of attachment of said group with the rest of the molecule;
represents hydrogen, fluoro, chloro, CN, methyl, Ci-C/t-alkoxy, C2-C3-alkenyl, cyclopropyl, 3- to 6-membered heterocycloalkyl, 4- to 6-membered heterocycloalkenyl, phenyl, pyridinyl, thiazolyl, -(S02)R9, -SR9, -((SO)=NRu)R10, -N=(SO)R9R10, wherein each methyl, Ci-C t-alkoxy, C2-C3-alkenyl, cyclopropyl, 3- to 6-membered heterocycloalkyl, phenyl, pyridinyl or thiazolyl is optionally substituted, one or more times, independently from each other, with fluoro, chloro, OH, -NR7R8, methyl, 5-membered heterocycloalkyl, -NR8(CO)OR7,
-(S02)R9, -((SO)=NRU)R10,-(PO)(OR7)2, or with a group selected from:
Figure imgf000083_0002
wherein * indicates the point of attachment of said group with the rest of the molecule;
wherein each 4- to 6-membered heterocycloalkenyl is optionally substituted, one or more times, with methyl;
R4 represents hydrogen or methyl;
R7, R8 represent, independently from each other, hydrogen or Ci-C/t-aikyl;
R9 represents Ci-C t-aikyl;
R10 represents Ci-C/t-aikyl; or
R9 and R10 together, in case of -N=(SO)R9R10 group, represent a 6-membered heterocycloalkyl group;
R11 represents hydrogen, methyl, -(CO)OR7;
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof, or a mixture of same.
4. The combination according to any one of claims 1 to 3, in which said component A is a compound selected from the group consisting of:
4-[(2-(morpholin-4-yl)-8-[2H-pyrazol-3-yl]-[l,7]-inaphthyridine-4-yl]phenyl-N-ethoxycarbonyl- S -methylsulphoximide
4-[(2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
4-[6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,6-dihydro-2H-pyran-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-[4-(N,S-dimethylsulfonimidoyl)phenyl]-2-[morpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[4-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(4-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[ 1,7] -naphthyridine
4-(2-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
dimethyl {4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl Jphosphonate
4-isopropenyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2- (morpholin-4-yl)-4-phenyl-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-[4-(S-ethylsulfonimidoyl)phenyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
3- [(2-(morpholin-4-yl)-8-[2H-pyrazol-3-yl]-[l,7]naphthyridine-4-yl]phenyl-N-ethoxycarbonyl- S -methylsulphoximide
4- (l-methyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-methanesulphonylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-[5-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(l,2,3,6-tetrahydropyridin-4-yl)-l,7-naphthyridine
4-cyclopropyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
3-[(2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
4-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
4-[2-(methylsulfonyl)-l,3-thiazol-4-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2(lH)-one 5 - [2- (morpholin-4-yl) - 8 -( 1 H-pyrazol-5 -yl) -1,7 -naphthyridin-4-yl] pyridin-2( 1 H) -one
4-[2-fluoro-4-(methylsulfonyl)phenyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4-{4-[S-(propan-2-yl)sulfonimidoyl]phenyl}-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(4-methanesulphonylphenyl)-2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)- [1,7] naphthyridine
2- ((R)-3-methylmorpholin-4-yl)-4-phenyl-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine
4-(3-methanesulphonylphenyl)-2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)- [1,7] naphthyridine
4-cyclopropyl-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]-naphthyridine
4-[2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
3- [2-((R)-3-methylmorpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]phenyl-S- methylsulphoximide
4- methanesulphonyl-2-(morpholin-4-yl)-8-[2-(tetrahydropyran-2-yl)-2H-pyrazol-3-yl]- [1,7] naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(methylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine-4-carbonitrile
2-((R)-3-methylmorpholin-4-yl)-8-(-2H-pyrazol-3-yl]-[l,7]naphthyridine-4-carbonitrile 2-morpholin-4-yl-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine-4-carboxamide
4-methanesulphonylmethyl-2-morpholin-4-yl-8-(2H-pyrazol-3-yl)-[l, 7] naphthyridine
[2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine-4-yl]methanol
4-(l-methanesulphonylcyclopropyl)-2-(morpholin-4-yl)-8-(2H-pyrazol-3-yl)-[l,7]naphthyridine 4-isopropoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-(morpholin-4-yl)-4-(propan-2-yloxy)-8-(lH-pyrrol-2-yl)-l,7-naphthyridine
4-[3-(S-methylsulfonimidoyl)propoxy]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-ethoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-methoxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine 2-methyl-l-{ [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl]oxy}propan-2-ol
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(tetrahydrofuran-2-ylmethoxy)-l,7-naphthyridine
3- { [2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]oxy}dihydrofuran-2(3H)-one
4- [(3-methyl-l,2-oxazol-5-yl)methoxy]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[(5-methyl-l,2-oxazol-3-yl)methoxy]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-benzyloxy-2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
4-isopropoxy-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine tert-butyl [4-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } oxy)butyl] carbamate
4-methoxy-2-((R)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine tert-butyl [3-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } oxy)propyl] carbamate
2-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } oxy)ethan amine
tert-butyl [2-({2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}oxy)ethyl]carbamate
4-( { 2-[(3R)-3-methylmorpholin-4-yl]-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl }oxy)butan- 1- amine
2-[(3R,5S)-3,5-dimethylmorpholin-4-yl]-4-isopropoxy-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R,5R)-3,5-dimethylmorpholin-4-yl]-4-isopropoxy-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine hydrochloride
4-chloro-2-morpholin-4-yl-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(methylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-l,4D4- oxathian-4-imine 4-oxide
4-{ [dimethyl(oxido)- λ 6-sulfanylidene] amino } -2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine 2-[(3R)-3-methylmorpholin-4-yl]-4-(piperazin-l-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-isopropoxy-2-((S)-3-methylmorpholin-4-yl)-8-(lH-pyrazol-3-yl)-[l,7]naphthyridine
2-(morpholin-4-yl)-4-(propan-2-yloxy)-8-(lH-pyrrol-3-yl)-l,7-naphthyridine
4-(l-ethyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-methyl-lH-imidazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl] aniline
4-(2 -difluorophenyl)-2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4 2-methyl-6^methylsulfonyl)pyridin-3-yl]-2 (3R)3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
4 2-fluoro-4-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-fluoro-2-[2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]aniline 4-(l-benzyl-lH-imidazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluorophenyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(2-methyl-l,3-thiazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[4-methyl-6-(methylsulfonyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
4-(l-cyclopropyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-iluoro-4-(piperazin- 1 -yl)phenyl] -2-[(3R)-3-methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[4-(methylsulfonyl)piperazin-l-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N-(2,2-dimethylpropyl)-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
(1 - { 2-[(3R)-3-methylmorpholin-4-yl]-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl }piperidin-4- yl)methanol N-cyclopropyl-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
4-(5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
N-(4-fluorophenyl)-N-methyl-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridin-4-amine
2-[(3R)-3-methylmorpholin-4-yl]-4-(6-methylpyridin-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoro-4-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrrol-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoro-5-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-fluoro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-methoxypyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-methoxy-5-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoro-2-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-methyl-3-(trifluoromethyl)-lH-pyrazol-5-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methyl-2-thienyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2 (3R)-3-methylmorpholin-4-yl]-4-(5-methyl-2 hienyl)-8-(m-pyrazol-5-yl)-l,7-naphthyridine 2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methyl-3-thienyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(3-chloro-2-thienyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l ,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(2-methyl-3-thienyl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-l,7- naphthyridine
4-(3,5-dimethyl-l,2-oxazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-chloro-2-methoxypyridin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-yl)-l,7- naphthyridine
4-(3,6-dihydro-2H-thiopyran-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methylpiperidin-l-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-tert-butyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methyl-l,2-oxazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-ethyl-3-methyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(l,4-dimethyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-methyl-6-(methylsulfanyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
4-[2-methyl-6-(S-methylsulfonimidoyl)pyridin-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-propyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6,7-dihydro-5H-pyrrolo[l,2-a]imidazol-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
4-[l-ethyl-3-(trifluoromethyl)-lH-pyrazol-5-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
methyl 5-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH- pyrrole-2-carboxylate
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2-thiazol-5-yl)-l,7-naphthyridine
N,N-dimethyl-2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl} aniline
4-(2,4-difluorophenyl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(l-isopropyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
ethyl methyl{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yljphosphinate
4- { [diethyl(oxido)- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)-l,7-naphthyridine
isobutyl methyl} 2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yljphosphinate
2- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}propan-2-ol
3- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pentan-3-ol 4-(5-chloropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
5- fluoro-2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}aniline
4- [2-fluoro-3-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-(oxetan-3-yl)-lH-pyrazol-5-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-fluoro-4-(pyrrolidin-l-yl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[3-(methoxymethyl)-5-methyl-l,2-oxazol-4-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(5-methyl-l,3,4-oxadiazol-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N-{2 (3R)-3-methylmoφholin -yl]-8-(lH yrazol-5-yl) -naphthyridin-4-yl}tetrahydro-lH- l 4-thiophen-l-imine 1 -oxide
4-{ [(4-fluorophenyl)(methyl)oxido- λ 6-sulfanylidene] amino }-2-[(3R)-3-methylmc^holin-4-yl]- 8-(lH-pyrazol-5-yl)-l,7-naphthyridine, mixture of 2 diastereoisomers
4-{ [(2-fluorophenyl)(methyl)oxido- λ6-sulfanylidene] amino }-2-[(3R)-3-methylmo holin-4-yl]- 8-(lH-pyrazol-5-yl)-l,7-naphthyridine, mixture of 2 diastereoisomers
4-{ [(R)(2-fluorophenyl)(methyl)oxido- λ6-sulfanylidene] amino }-2-[(3R)-3-methylmoφholin-4- yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine, diastereoisomer
4-{ [(S)(2-fluorophenyl)(methyl)oxido- λ6-sulfanylidene] amino } -2-[(3R)-3-methylmoφholin-4- yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine, diastereoisomer
4-(dimethylphosphoryl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(diethylphosphoryl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine ethyl isobutyl{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yljphosphinate
2-[(3R)-3-methylmorpholin-4-yl]-4-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l-isobutyl-lH-pyrazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[5-fluoro-6-(methylsulfonyl)pyridin-3-yl]-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
4-[(3R)-3-methylmorpholin-4-yl]-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methyl-lH-pyrazol-5-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[2-fluoro-5-(methylsulfonyl)phenyl]-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[4-(isopropylsulfonyl)phenyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(6-fluoropyridin-2-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-ethyl-lH-imidazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
1- {2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}prolinamide
3- {2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pyridin-2- amine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[l-(2,2,2-trifluoroethyl)-lH-pyrazol-5- yl] - 1 ,7 -naphthyridine
1 -methyl-4- { 2-[(3R)-3-methylmorpholin-4-yl] -8-(lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl }piperazin-2-one
4- [l-(2-fluoroethyl)-lH-pyrazol-3-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-[l-(2-fluoroethyl)-lH-pyrazol-5-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2- (3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH-pyrazol-
1- yl)ethanol
2-methyl- 1 -(3- { 2-[(3R)-3-methylmorpholin-4-yl] -8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl } - 1 H-pyrazol- 1 -yl)propan-2-ol
4-[(2R)-2-methylmorpholin-4-yl]-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-fluoropyridin-2-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(6-methylpyridin-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- [(3R)-3-methylmorpholin-4-yl]-4-(3-methylpyridin-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N-(2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}phenyl)acetamide
3- {2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}pyridin-2-ol
2-(3-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl }phenyl)propan-2-ol
4- (5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[(2S)-2-methylmoφholin-4-yl]-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-[(trans)-2-methylcyclopropyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(difluoromethoxy)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]propan-2-ol
2-(morpholin-4-yl)-4-(3-oxa-8-azabicyclo[3.2. l]oct-8-yl)-8-(lH-pyrazol-5-yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(pyrrolidin-l-yl)-l,7-naphthyridine
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperazin-2-one
4-(dimethylphosphoryl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[(trans)-2,5-dimethylpiperazin-l-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[(cis)-3,5-dimethylpiperazin-l-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-3-(trifluoromethyl)azetidin-3- ol
methyl hydrogen { 4- [2-(morpholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4- yl]phenyl Jphosphonate
4-(4-methylpiperazin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[(3aR,6aS)-tetrahydro-lH-furo[3,4-c]pyrrol-5(3H)- yl] - 1 ,7 -naphthyridine
4-(3-methoxy-3-methylazetidin-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(morpholin-4-yl)-4-[(lS,4S)-2-oxa-5-azabicyclo[2.2.1]hept-5-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[(methylsulfanyl)methyl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
N,N-dimethyl-5-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-amine 4-(2-methylpyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
1- {2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}cyclohexanol
2- fluoro-6-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}aniline
(methyl} 4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}oxido- λ6- sulfanylidene)cyanamide
l-ethyl-3-(methyl{4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl }oxido- λ 6-sulfanylidene)urea
3- ( { 2-[(3R)-3-methylmorpholin-4-yl]-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl }oxy)propan- 1- amine
4- (4-cyclopropyl-lH-l,2,3-triazol-5-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-ethylsulfinyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-[propan-2-ylsulfinyl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[3-(methylsulfonyl)propoxy]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-4-(phenylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(propan-2-ylsulfonyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(ethylsulfonyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(phenylsulfinyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(methylsulfinyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-oxidotetrahydro-2H-thiopyran-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(1 -dioxidotetrahydro-2H-thiopyran-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4,8-di(lH-pyrazol-5-yl)-l,7-naphthyridine
N,N-dimethyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine
2-(morpholin-4-yl)-4-(phenylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-N-(propan-2-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine
4-(ethylsulfanyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-(propan-2-ylsulfanyl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrol-2-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrol-3-yl)-l,7-naphthyridine
4-[(4-methoxyphenyl)sulfanyl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(5-methyl-lH-pyrazol-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine l-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyrrolidin-2-one 4-(l,l-dioxido-l,2-thiazolidin-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridine
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-2-one
2- [(3R)-3-methylmorpholin-4-yl]-4-(2-methylpyridin-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[2-(propan-2-yloxy)pyridin-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-methoxypyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2- (morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(pyridin-4-yl)-l,7-naphthyridine
4-[(4-methoxyphenyl)sulfanyl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[3-fluoro-2-(mo holin-4-yl)pyridin-4-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5- yl)- 1,7-naphthyridine
4-(6-fluoro-5-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-l,3-oxazinan-2-one
3- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-l,3-oxazolidin-2-one
4- (3-methoxypyridin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1,7- naphthyridine
4-(2,6-difluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5-chloro-2-fluoropyridin-3-yl)-2-[(3R)-3-methylmo holin-4-yl]-8-(lH-pyrazol-5-yl)- 1,7- naphthyridine
4-(3-rluoropyridin-4-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-chloro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5,6-dimethylpyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(5-rluoro-6-methylpyridin-3-yl)-2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(5-methylthiophen-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(3-methoxythiophen-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2-chlorothiophen-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(isoquinolin-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-chlorothiophen-2-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(4-methylthiophen-2-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(2,5-dimethylthiophen-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-thiopyran-4-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-l,2,5,6-tetrahydropyridin-3-yl)-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-l,2,3,6-tetrahydropyridin-4-yl)-8-(lH-pyrazol-5- yl)- 1,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-methylpiperidin-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2,3,6-tetrahydropyridin-4-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[l-(tetrahydro-2H-pyran-4-yl)-lH- pyrazol-3-yl] - 1 ,7-naphthyridine
4-(4,6-difluoropyridin-3-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l,3-dimethyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l,5-dimethyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(piperidin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-[3-(trifluoromethyl)-lH-pyrazol-4-yl]- 1 ,7 -naphthyridine
4-(l-cyclobutyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-(l-cyclopropyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-(propan-2-yl)-lH-pyrazol-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
4-[l-(difluoromethyl)-lH-pyrazol-4-yl]-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-(l-tert-butyl-lH-pyrazol-4-yl)-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,3,5-trimethyl-lH-pyrazol-4-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-methyl-3-(trifluoromethyl)-lH-pyrazol-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
2-(4-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-lH-pyrazol- l-yl)ethanol
4-( 1 -ethyl- lH-pyrazol-4-yl)-2- [(3R)-3-methylmorpholin-4-yl] -8-( 1 H-pyrazol-5-yl)- 1 ,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-methyl-lH-pyrrol-3-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-(propan-2-yl)-lH-pyrazol-3-yl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-4-(l,2,5-trimethyl-lH-pyrrol-3-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l-phenyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(3-methyl-lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-amine
2 (3R)-3-methylmorpholin -yl] 1-(2-methylpropyl) H-pyrazol-4-yl]-8-(lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(lH-pyrazol-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[(3R)-3-methylmorpholin-4-yl]-4-(l,3-oxazol-2-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(1 -dimethyl-lH-pyrazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l,5-dimethyl-lH yrazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(l,3, -trimethyl-lH-pyrazol-4-yl)-l,7-naphthyridine 4-{ [(2-methoxyethyl)(methyl)oxido- λ 6-sulfanylidene]amino}-2-[(3R)-3-methylmorpholin-4- yl]-8-( lH-pyrazol-5-yl)- 1 ,7 -naphthyridine
4-{ [(4-bromophenyl)(oxido)propan-2-yl- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmoφholin- 4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2- (methyl-N-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl } sulfonimidoyl)phenol
4- { [(4-bromophenyl)(methyl)oxido- λ 6-sulfanylidene] amino } -2- [(3R)-3-methylmorpholin-4-yl] - 8 -( 1 H-pyrazol-5 -yl) - 1 ,7-naphthyridine
4-{ [tert-butyl(methyl)oxido- λ6-sulfanylidene]amino}-2-[(3R)-3-methylmorpholin-4-yl]-8-(lH- pyrazol-5-yl)- 1 ,7-naphthyridine
formic acid - N-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-l,4□4-oxathian- 4-imine 4-oxide (1: 1)
N-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]hexahydro-l D4-thiopyran-l- imine 1 -oxide
3- methyl-2-{2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}butan-2-ol
l-{2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}-l-(tetrahydro- 2H-pyran-4-yl)ethanol
3,3-dimethyl-2-{2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl}butan-2-ol
2-{2-[(3R)-3-methylmoφholin-4-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl}hexan-2-ol 2-[(3R)-3-methylmorpholin-4-yl]-8-(lH-pyrazol-3-yl)-l ,7-naphthyridine-4-carboxamide
2-[(3R)-3-methylmorpholin-4-yl]-4-[l-(methylsulfonyl)cyclopropyl]-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(tetrahydro-2H-pyran-4-ylmethoxy)-l,7- naphthyridine
N,N-dimethyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benz amide
{4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}(piperidin-l- yl)methanone
N,N-dimethyl-2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benz amide N-cyclopropyl-4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide 4-(4-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(lH-indol-6-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(lH-indol-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
3 - [2- (morpholin-4-yl) - 8 -( 1 H-pyrazol-5 -yl) -1,7 -naphthyridin-4-yl] benzamide
4-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide
N-methyl-3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide 4-(3-fluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-chlorothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[2-(trifluoromethyl)phenyl]-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[4-(trifluoromethyl)phenyl]-l,7-naphthyridine 2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-[3-(trifluoromethyl)phenyl]-l,7-naphthyridine 4-(3-chlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-{3-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}acetamide 4-(3-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,5-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(3-methylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(furan-2-ylmethyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2,6-dimethyl-4-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenol
4-(2,3-dimethylphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
{3-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}methanol
4-(4-fluorophenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methylphenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-chlorophenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-fluoro-3-methoxyphenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-methylphenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,3-dimethoxyphenyl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N,N-dimethyl-3-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridin-4-yl] aniline
N,N-dimethyl-2-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]aniline
N-{2-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl Jmethanesulfonamide
N-{4-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- yl]phenyl}methanesulfonamide
N,N-dimethyl-4-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benz amide
2-(moφholin-4-yl)-4-[(lE)-prop-l-en-l-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenol
4-(2-fluorophenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
{3-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]phenyl}(piperidin-l- yl)methanone
2-(moφholin-4-yl)-4-[4-(propan-2-yl)phenyl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N-cyclopropyl-3-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]benzamide 4-(biphenyl-4-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,4-dimethoxyphenyl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(2-chlorophenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,5-dimethylphenyl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 3 - [2- (mo holin-4-yl) - 8 -( 1 H-pyrazol-5 -yl) -1,7 -naphthyridin-4-yl] aniline
2-(mc^holin-4-yl)-8-( 1 H-pyrazol-5-yl)-4- [3-( 1 H-pyrazol- 1 -yl)phenyl] - 1 ,7-naphthyridine 3 - [2- (morpholin-4-yl) - 8 -( 1 H-pyrazol-5 -yl) -1,7 -naphthyridin-4-yl] phenol
4-(2-fluoro-5-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-fluoro-2-methoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,4-difluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,3-difluorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,6-dimethoxyphenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl] aniline
4-(3,5-dichlorophenyl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(biphenyl-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-chloropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(l-benzothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-( 1-methyl- lH-pyrazol-5-yl)-2-(morpholin-4-yl)-8-( 1 H-pyrazol-5 -yl)- 1 ,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(pyridin-3-yl)-l,7-naphthyridine
4-(2-methoxypyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-3-yl)-l,7-naphthyridine
2-(morpholin-4-yl)-4-[ 1 -(phenylsulfonyl)- lH-indol-2-yl]-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridine
4-(2-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(6-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
{ 5- [2-(morpholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridin-4-yl] thiophen-2-yl } methanol
4-(2-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(6-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-chloro-6-methylpyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-methoxypyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(isoquinolin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-chloropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(3-fluoropyridin-4-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2,6-difluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-( 1-methyl- lH-pyrazol-4-yl)-2-(morpholin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7-naphthyridine tert-butyl 5-methoxy-2-[2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-lH- indole- 1-carboxylate
2-(mo holin-4-yl)-4-[6-(morpholin-4-yl)pyridin-3-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methylthiophen-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(thiophen-2-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(thiophen-3-yl)-l,7-naphthyridine
4-(3-methylthiophen-2-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-chloro-5-methylpyridin-3-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(4-methoxypyridin-3-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-chloro-2-methoxypyridin-3-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine tert-butyl 5-methyl-2-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-lH- indole- 1 -carboxylate
4-(5-chloro-2-fluoropyridin-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3,5-dimethyl-l,2-oxazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-8-yl)-l,7-naphthyridine
4-(5-methylthiophen-2-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(6-ethoxypyridin-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(2-ethoxypyridin-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(quinolin-6-yl)-l,7-naphthyridine
4- (2-chlorothiophen-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
5- [2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-amine
2-(moφholin-4-yl)-4-(lH-pyrazol-3-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(6-methylpyridin-3-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4- ( 1-methyl- 1 H-pyrrol-2-yl)-2-(moφholin-4-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridine
5- [2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-ol 4-(5-chloropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-chloro-2-methoxypyridin-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-chlorothiophen-2-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(5-fluoropyridin-3-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[2-(methylsulfanyl)pyrimidin-5-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l ,7-naphthyridine
N-cyclopropyl-5-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyrimidin-2- amine
4-(isoquinolin-5-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-methyl-5-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridine-2- carboxamide
N-tert-butyl-5-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridine-3- carboxamide
4-[5-(methylsulfanyl)pyridin-3-yl]-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-l,7-naphthyridine 3-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyridin-2-amine
methyl 4-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]thiophene-2- carboxylate
4-[2-methoxy-5-(trifluoromethyl)pyridin-3-yl]-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7- naphthyridine
2-(moφholin-4-yl)-4-[2-(propan-2-yloxy)pyridin-3-yl]-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(5-chloro-6-ethoxypyridin-3-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(l-tert-butyl-lH-pyrazol-4-yl)-2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 2-(moφholin-4-yl)-4-(piperidin-l-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
l-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-4-ol
N-methyl-2-(moφholin-4-yl)-N-phenyl-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine
{ l-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]pyrrolidin-2-yl}methanol N-methyl-2-(moφholin-4-yl)-N-propyl-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine 4-(azepan-l-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-(3-methylpiperidin-l-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(4-memylpiperidin- 1 -ν1)-2-^ο Μ
1- [2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridm^
4-(2,5-dihydro-lH^yrrol-l-yl)-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine 4-(3,4-dihydroquinolin-l(2H)-yl)-2-(moφholin-4-yl^^
4-(3,4-dihydroisoquinolin-2(lH)-yl)-2-(morpholin-4-y^
4-(l,3-dihydro-2H-isoindol-2-yl)-2-(morpholin-4-yl^
2- (moφholin -yl)-8 m-pyrazol-5-yl)-4-[l,3,3-trimethyl-6-azabicyclo[3.2.1]oct-6-yl]-l,7- naphthyridine
tert-butyl l-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]-prolinate
N-methyl-N-(2-methylpropyl)-2-(moφholirl-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridirl-4- amine
N-(3-fluorophenyl)-N-methyl-2-(moφholin-4-yl)-8-(lH yrazol-5-yl)-l,7-naphthyridin-4-amine
4-( 1, 1 -dioxido- 1 -thia-6-azaspiro[3.3]hept-6-yl)-2-(mo holin-4-yl)-8-( lH-pyrazol-5-yl)- 1 ,7- naphthyridine
4-(3-fluoropiperidin-l-yl)-2-(mo holirl-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-(2-fΊuorophenyl)-N-methyl-2-(moφholin-4-yl)-8-(lH yrazol-5-yl)-l,7-naphthyridin-4-amine
1 - [2- (morpholin-4-yl) - 8 -( 1 H-pyrazol-5 -yl) -1,7 -naphthyridin-4-yl] -prolinamide
{ l-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridirl-4-yl]piperidirl-4-yl}methanol
4-(4-methoxypiperidin-l-yl)-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine N-(4-fΊuorophenyl)-N-methyl-2-(moφholin-4-yl)-8-(lH yrazol-5-yl)-l,7-naphthyridin-4-amine
N-methyl-l-[2-(mo holin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl] -prolinamide
4-[4-(ethylsulfonyl)piperazin-l-yl]-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
4-[4-(methylsulfonyl)piperazin-l-yl]-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridine
N-cyclopropyl-N-methyl-2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-amine N-(2,2-dimethylpropyl)-N-methyl-2-(morpholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4- amine
{ l-[2-(moφholin-4-yl)-8-(lH-pyrazol-5-yl)-l,7-naphthyridin-4-yl]piperidin-3-yl}methanol or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
5. The combination according to any one of claims 1 to 4, in which said component A is 2-[(3R)- 3-methylmorpholin-4-yl] -4-( 1 -methyl- 1 H-pyrazol-5-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7- naphthyridineor a tautomer, an N-oxide, a hydrate, a solvate, or a pharmaceutically acceptable salt thereof.
6. The combination of any one of claims 1 to 5, in which said pharmaceutically acceptable salt of the alkaline-earth radionuclide radium-223 is radium- 223 dichloride.
7. The combination according to any one of claims 1 to 4, in which said component A is 2-[(3R)- 3-methylmorpholin-4-yl] -4-( 1 -methyl- 1 H-pyrazol-5-yl)-8-( 1 H-pyrazol-5-yl)- 1 ,7-naphthyridine and said component B is radium-223 dichloride.
8. A combination according to any one of claims 1 to 7 for use in the treatment or prophylaxis of a hyper-proliferative disease and/or metastases thereof.
9. Use of a combination according to any one of claims 1 to 7 for the preparation of a medicament for the treatment or prophylaxis of a hyper-proliferative disease and/or metastases thereof.
10. A combination for use according to claim 8 or a use according to claim 9, wherein the hyper- proliferative disease is selected from breast cancer or prostate cancer and/or metastases thereof.
11. Use according to claim 10, wherein the prostate cancer is castration resistant prostate cancer.
12. Use according to any one of claims 8 to 11, wherein the metastases are metastases in bone.
13. Use according to claim 11, wherein the castration resistant prostate cancer is castration resistant prostate cancer with symptomatic bone metastases and no known visceral metastatic disease.
14. The combination for use according to claim 8 for use in the treatment or prophylaxis of prostate cancer, particularly castration-resistant prostate cancer, and/or metastases thereof, wherein component B is administered prior to component A.
15. The combination for use according to claim 14, wherein component B is administered 2 hours to 96 hours, or 6 hours to 84 hours, or 12 hours to 72 hours, or 24 hours to 48 hours, or 18 hours to 30 hours, or 20 hours to 28 hours, or 22 hours to 26 hours, or 42 hours to 54 hours, or 44 hours to 52 hours, or 46 hours to 50 hours, or 24 hours, or 48 hours prior to component A.
16. A kit comprising
component A as defined in any one of claims 1 to 7;
component B as defined in claims 1 or 6; and, optionally,
component C being one or more further pharmaceutical agents.
17. A kit according to claim 16, in which optionally all, both or either of said components A and B and optionally C are in the form of a pharmaceutical composition which is ready for use to be administered simultaneously, concurrently, separately or sequentially.
18. A pharmaceutical composition comprising a combination as defined in any one of claims 1 to 7 together with one or more pharmaceutically acceptable excipients.
19. A pharmaceutical composition according to claim 18, in which the components A and B are present in a joint formulation.
20. A pharmaceutical composition according to claim 18, in which the components A and B are present in separate formulations.
PCT/EP2018/054362 2017-02-24 2018-02-22 Combination of atr kinase inhibitors with radium-223 salt WO2018153969A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17157764.6 2017-02-24
EP17157764 2017-02-24

Publications (1)

Publication Number Publication Date
WO2018153969A1 true WO2018153969A1 (en) 2018-08-30

Family

ID=58185318

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/054362 WO2018153969A1 (en) 2017-02-24 2018-02-22 Combination of atr kinase inhibitors with radium-223 salt

Country Status (4)

Country Link
AR (1) AR110995A1 (en)
TW (1) TW201840319A (en)
UY (1) UY37616A (en)
WO (1) WO2018153969A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10729680B2 (en) 2016-01-14 2020-08-04 Bayer Pharma Aktiengesellschaft 5-substituted 2-(morpholin-4-yl)-1,7-naphthyridines
WO2020165015A1 (en) * 2019-02-11 2020-08-20 Bayer Aktiengesellschaft The atr kinase inhibitor bay1895344 for use in the treatment of a hyper-proliferative disease
US10772893B2 (en) 2014-08-04 2020-09-15 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112773805B (en) * 2019-11-11 2024-05-10 中国科学院脑科学与智能技术卓越创新中心 Novel therapeutic agent for ischemic brain injury
CN114081848A (en) * 2021-11-19 2022-02-25 上海琪维生物科技有限公司 Composition for skin care and application thereof

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
WO2000040275A2 (en) 1999-01-04 2000-07-13 Anticancer Therapeutic Inventions As The preparation and use of radium-223 to target calcified tissues for pain palliation, bone cancer therapy, and bone surface conditioning
WO2010054398A1 (en) 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2010071837A1 (en) 2008-12-19 2010-06-24 Vertex Pharmaceuticals Incorporated Pyrazine derivatives useful as inhibitors of atr kinase
WO2010073034A1 (en) 2008-12-22 2010-07-01 Astrazeneca Ab Pyrimidine indole derivatives for treating cancer
WO2011134672A1 (en) 2010-04-30 2011-11-03 Algeta As Isotope production method
WO2011134671A1 (en) 2010-04-30 2011-11-03 Algeta As Isotope preparation method
WO2011143422A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2 -aminopyridine derivatives useful as inhibitors of atr kinase
WO2011143425A2 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143426A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
WO2011143399A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143423A2 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011154737A1 (en) 2010-06-11 2011-12-15 Astrazeneca Ab Morpholino pyrimidines and their use in therapy
WO2011163527A1 (en) 2010-06-23 2011-12-29 Vertex Pharmaceuticals Incorporated Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
WO2012138938A1 (en) 2011-04-05 2012-10-11 Vertex Pharmaceuticals Incorporated Aminopyrazine compounds useful as inhibitors of tra kinase
WO2012178124A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2012178125A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2012178123A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049720A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049719A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049722A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049859A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Treating pancreatic cancer and non-small cell lung cancer with atr inhibitors
WO2013071088A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071093A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
WO2013071090A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071094A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071085A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
WO2013152298A1 (en) 2012-04-05 2013-10-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase and combination therapies thereof
WO2014062604A1 (en) 2012-10-16 2014-04-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014089379A1 (en) 2012-12-07 2014-06-12 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014143242A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014143240A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Fused pyrazolopyrimidine derivatives useful as inhibitors of atr kinase
WO2014143241A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2015085132A1 (en) 2013-12-06 2015-06-11 Vertex Pharmaceuticals Incorporated 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
WO2015187451A1 (en) 2014-06-05 2015-12-10 Vertex Pharmacetucals Incorporated Radiolabelled derivatives of a 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]- pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, the preparation of said compound and different solid forms thereof
WO2016020320A1 (en) 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
WO2000040275A2 (en) 1999-01-04 2000-07-13 Anticancer Therapeutic Inventions As The preparation and use of radium-223 to target calcified tissues for pain palliation, bone cancer therapy, and bone surface conditioning
WO2010054398A1 (en) 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2010071837A1 (en) 2008-12-19 2010-06-24 Vertex Pharmaceuticals Incorporated Pyrazine derivatives useful as inhibitors of atr kinase
WO2010073034A1 (en) 2008-12-22 2010-07-01 Astrazeneca Ab Pyrimidine indole derivatives for treating cancer
WO2011134672A1 (en) 2010-04-30 2011-11-03 Algeta As Isotope production method
WO2011134671A1 (en) 2010-04-30 2011-11-03 Algeta As Isotope preparation method
WO2011143422A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2 -aminopyridine derivatives useful as inhibitors of atr kinase
WO2011143425A2 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143426A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
WO2011143399A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143423A2 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011154737A1 (en) 2010-06-11 2011-12-15 Astrazeneca Ab Morpholino pyrimidines and their use in therapy
WO2011163527A1 (en) 2010-06-23 2011-12-29 Vertex Pharmaceuticals Incorporated Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
WO2012138938A1 (en) 2011-04-05 2012-10-11 Vertex Pharmaceuticals Incorporated Aminopyrazine compounds useful as inhibitors of tra kinase
WO2012178125A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2012178124A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2012178123A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049720A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049719A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049722A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049859A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Treating pancreatic cancer and non-small cell lung cancer with atr inhibitors
WO2013071085A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
WO2013071093A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
WO2013071090A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071094A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071088A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013152298A1 (en) 2012-04-05 2013-10-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase and combination therapies thereof
WO2014062604A1 (en) 2012-10-16 2014-04-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014089379A1 (en) 2012-12-07 2014-06-12 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014143242A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014143240A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Fused pyrazolopyrimidine derivatives useful as inhibitors of atr kinase
WO2014143241A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2015085132A1 (en) 2013-12-06 2015-06-11 Vertex Pharmaceuticals Incorporated 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
WO2015187451A1 (en) 2014-06-05 2015-12-10 Vertex Pharmacetucals Incorporated Radiolabelled derivatives of a 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]- pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, the preparation of said compound and different solid forms thereof
WO2016020320A1 (en) 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines

Non-Patent Citations (49)

* Cited by examiner, † Cited by third party
Title
"TNM CLASSIFICATION OF MALIGNANT TUMORS", 2011, UICC
A. MARECHAL; L. ZOU: "DNA damage sensing by the ATM and ATR kinases", COLD SPRING HARB. PERSPECT. BIOL., vol. 5, 2013, pages a012716
ACS MED. CHEM. LETT., vol. 6, 2015, pages 37 - 41
ACS MED. CHEM. LETT., vol. 6, 2015, pages 42 - 46
AIELLO ET AL., NEW ENGL. J. MED., vol. 331, 1994, pages 1480
AMOL TAKALKAR ET AL.: "Radium-223 dichloride bone-targeted alpha particle therapy for hormone-refractory breast cancer metastatic to bone", EXP HEMATOL ONCOL., vol. 3, 2014, pages 23, XP021198273, DOI: doi:10.1186/2162-3619-3-23
AMOL TAKALKAR ET AL: "Radium-223 dichloride bone-targeted alpha particle therapy for hormone-refractory breast cancer metastatic to bone", EXPERIMENTAL HEMATOLOGY & ONCOLOGY, BIOMED CENTRAL LTD, LONDON, UK, vol. 3, no. 1, 8 September 2014 (2014-09-08), pages 23, XP021198273, ISSN: 2162-3619, DOI: 10.1186/2162-3619-3-23 *
BLISS, C.I.: "The toxicity of poisons applied jointly", ANN. APPL. BIOL., vol. 26, 1939, pages 585 - 615, XP055065740, DOI: doi:10.1111/j.1744-7348.1939.tb06990.x
BRULAND O. S. ET AL.: "High-linear energy transfer irradiation targeted to skeletal metastases by the alpha-emitter ''3Ra: adjuvant or alternative to conventional modalities?", CLIN. CANCER RES., vol. 12, 2006, pages 6250s - 7s
C. PARKER ET AL.: "Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 3, pages 213 - 23, XP055160177, DOI: doi:10.1056/NEJMoa1213755
C. PARKER ET AL: "Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer", NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 3, 18 July 2013 (2013-07-18), pages 213 - 223, XP055160177, ISSN: 0028-4793, DOI: 10.1056/NEJMoa1213755 *
D. WOODS; J.J. TUCHI: "Chemotherapy induced DNA damage response", CANCER BIOL. THERA., vol. 14, 2013, pages 379 - 389
E. FOKAS; R. PREVO ET AL.: "Targeting ATR in DNA damage response and cancer therapeutics", CANCER TREATMENT REV, vol. 40, 2014, pages 109 - 117, XP028765229, DOI: doi:10.1016/j.ctrv.2013.03.002
E.J. BROWN; D. BALTIMORE: "ATR disruption leads to chromosomal fragmentation and early embryonic lethality", GENES DEV, vol. 14, 2000, pages 397 - 402
EXP. REV. MOL. MED., vol. 16, 2014, pages e10
GARRET R., SEMIN. ONCOL., vol. 72, 1993, pages 3433 - 3435
HENRIKSEN G. ET AL.: "Significant antitumor effect from bone-seeking, alpha-particle-emitting (223)Ra demonstrated in an experimental skeletal metastases model", CANCER RES., vol. 62, 2002, pages 3120 - 3125
HENRIKSEN G. ET AL.: "Targeting of osseous sites with alpha-emitting 223Ra: comparison with the beta-emitter 89Sr in mice", J. NUCL. MED, vol. 44, 2003, pages 252 - 59
HSING AW; CHOKKALINGAM AP: "Prostate cancer epidemiology", FRONT BIOSCI., vol. 11, 2006, pages 1388 - 413
J. MED. CHEM., vol. 56, 2013, pages 2125 - 2138
J.M. WAGNER; S.H. KAUFMANN: "Prospects for the use of ATR inhibitors to treat cancer", PHARMACEUTICALS, vol. 3, 2010, pages 1311 - 1334
KANIS JA., BONE, vol. 17, 1995, pages 101s - 105s
KERR C.: "223)Ra targets skeletal metastases and spares normal tissue", LANCET ONCOL, vol. 3, 2002, pages 453, XP004813882, DOI: doi:10.1016/S1470-2045(02)00835-5
LEWINGTON V.J.: "Bone-seeking radionuclides for therapy", J. NUCL. MED, vol. 46, no. 1, 2005, pages 38S - 47S
LI Y.; RUSSELL P.J.; ALLEN B.J.: "Targeted alpha-therapy for control of micrometastatic prostate cancer", EXPERT REV. ANTICANCER THER., vol. 4, 2004, pages 459 - 68, XP008044031, DOI: doi:10.1586/14737140.4.3.459
LIEPE K.: "Alpharadin, a Ra-based alpha-particle-emitting pharmaceutical for the treatment of bone metastases in patients with cancer", CURR. OPIN. INVESTIG. DRUGS, vol. 10, 2009, pages 1346 - 58
LOPEZ ET AL., INVEST. OPTHTHALMOL. VIS. SCI., vol. 37, 1996, pages 855
M.K. ZEMAN; K.A. CIMPRICH: "Causes and consequences of replication stress", NAT. CELL BIOL., vol. 16, 2014, pages 2 - 9
MCDEVITT M.R. ET AL.: "Radioimmunotherapy with alpha-emitting nuclides", EUR. J. NUCL. MED., vol. 25, 1998, pages 1341 - 51, XP002901567, DOI: doi:10.1007/s002590050306
MINHONG ZOU ET AL., ONCOLOGY REPORTS, vol. 30, 2013, pages 615 - 622
N. KOOPMANS ET AL., THE JOURNAL OF UROLOGY, vol. 178, September 2007 (2007-09-01), pages 849 - 853
NEMA, S. ET AL.: "Excipients and Their Use in Injectable Products", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 51, no. 4, 1997, pages 166 - 171
NIELSEN, OS; MUNRO AJ; TANNOCK IF, J CLIN ONCOL, vol. 9, 1991, pages 509 - 5 24
NILSSON S. ET AL.: "A randomized, dose-response, multicenter phase II study of radium-223 chloride for the palliation of painful bone metastases in patients with castration-resistant prostate cancer", EUR. J. CANCER, vol. 48, 2012, pages 678 - 86
NILSSON S. ET AL.: "Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II study", LANCET ONCOL, vol. 8, 2007, pages 587 - 94, XP022137762, DOI: doi:10.1016/S1470-2045(07)70147-X
NILSSON S. ET AL.: "First clinical experience with alpha-emitting radium-223 in the treatment of skeletal metastases", CLIN. CANCER RES., vol. 11, 2005, pages 4451 - 59
O. GILAD; BY NABET ET AL.: "Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner", CANCER RES., vol. 70, 2010, pages 9693 - 9702
PARKER C. ET AL.: "A randomized, double-blind, dose-finding, multicenter, phase 2 study of radium chloride (Ra-223) in patients with bone metastases and castration-resistant prostate cancer", EUR UROL., vol. 63, no. 2, February 2013 (2013-02-01), pages 189 - 97
PEER ET AL., LAB. INVEST., vol. 72, 1995, pages 638
POWELL, M.F. ET AL.: "Compendium of Excipients for Parenteral Fonnulations", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 52, no. 5, 1998, pages 238 - 311, XP009119027
PURE APPL CHEM, vol. 45, 1976, pages 11 - 30
RUXANA T. SADIKOT; TIMOTHY S. BLACKWELL, PROC AM THORAC SOC, vol. 2, 2005, pages 537 - 540
S. LLONA-MINGUEZ; A. HOGLUND ET AL.: "Chemical strategies for development of ATR inhibitors", EXP. REV. MOL. MED., vol. 16, 2014, pages e10
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SCHRODER FH; WILDHAGEN MF: "Rotterdam Study Group of the 'European Randomised Study of Screening for Prostate Cancer' (ERSPC) Screening for prostate cancer: Evidence and perspectives", BJU INT., vol. 88, 2001, pages 811 - 7
STRICKLEY, R.G: "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 53, no. 6, 1999, pages 324 - 349
SUOMINEN ET AL., CLIN CANCER RES., vol. 23, no. 15, 1 August 2017 (2017-08-01), pages 4335 - 4346
SUOMINEN ET AL.: "Survival benefit with radium-223 dichloride in a mouse model of breast cancer bone metastasis", J NATL CANCER INST., vol. 105, no. 12, 19 June 2013 (2013-06-19), pages 908 - 16
WINDY DEAN-COLOMB ET AL., BREAST CANCER RES TREAT., vol. 137, no. 2, January 2013 (2013-01-01)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10772893B2 (en) 2014-08-04 2020-09-15 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
US11529356B2 (en) 2014-08-04 2022-12-20 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
US10729680B2 (en) 2016-01-14 2020-08-04 Bayer Pharma Aktiengesellschaft 5-substituted 2-(morpholin-4-yl)-1,7-naphthyridines
WO2020165015A1 (en) * 2019-02-11 2020-08-20 Bayer Aktiengesellschaft The atr kinase inhibitor bay1895344 for use in the treatment of a hyper-proliferative disease
CN113412114A (en) * 2019-02-11 2021-09-17 拜耳股份有限公司 ATR kinase inhibitor BAY1895344 for treating hyperproliferative diseases

Also Published As

Publication number Publication date
UY37616A (en) 2018-09-28
TW201840319A (en) 2018-11-16
AR110995A1 (en) 2019-05-22

Similar Documents

Publication Publication Date Title
US20230346927A1 (en) COMBINATION of ATR KINASE INHIBITORS and PD-1/PD-L1 INHIBITORS
EP3402795B1 (en) 5-substituted 2-(morpholin-4-yl)-1,7-naphthyridines
EP3585365B1 (en) Combination of atr kinase inhibitors with parp inhibitors
EP3268490B1 (en) Substituted 2,3-dihydroimidazo[1,2-c]quinazoline-containing combinations
WO2018153969A1 (en) Combination of atr kinase inhibitors with radium-223 salt
WO2018153971A1 (en) Combination of atr kinase inhibitors
CA2932221C (en) Combination of pi3k-inhibitors
WO2018153972A1 (en) Combination of atr kinase inhibitors and antiandrogens
WO2018206547A1 (en) Combination of bub1 and atr inhibitors
EP3077003A1 (en) Combination of pi3k-inhibitors
US20210369724A1 (en) Combination of atr kinase inhibitors with 2,3-dihydroimidazo[1,2-c]quinazoline compounds
WO2018054782A1 (en) Combination of pi3k-inhibitors
WO2018215282A1 (en) Combination of bub1 and pi3k inhibitors
US20220118123A1 (en) Combination of ar antagonists and targeted thorium conjugates
WO2017157418A1 (en) Combination of mknk1-inhibitors
US11185549B2 (en) Combination of a PI3K-inhibitor with an androgen receptor antagonist
TW201417816A (en) Combination of AKT-inhibitors
WO2020164997A1 (en) Combination of pi3k-inhibitors

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: 18707338

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: 18707338

Country of ref document: EP

Kind code of ref document: A1