WO2012151541A1 - Inhibiteurs de csf-1r pour le traitement de tumeurs cérébrales - Google Patents

Inhibiteurs de csf-1r pour le traitement de tumeurs cérébrales Download PDF

Info

Publication number
WO2012151541A1
WO2012151541A1 PCT/US2012/036630 US2012036630W WO2012151541A1 WO 2012151541 A1 WO2012151541 A1 WO 2012151541A1 US 2012036630 W US2012036630 W US 2012036630W WO 2012151541 A1 WO2012151541 A1 WO 2012151541A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
tumor
formula
csf
brain
Prior art date
Application number
PCT/US2012/036630
Other languages
English (en)
Inventor
Dylan DANIEL
Johanna JOYCE
James Sutton
Original Assignee
Novartis Ag
Sloan-Kettering Institute For Cancer Research
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 Novartis Ag, Sloan-Kettering Institute For Cancer Research filed Critical Novartis Ag
Publication of WO2012151541A1 publication Critical patent/WO2012151541A1/fr
Priority to PCT/US2013/036628 priority Critical patent/WO2013158559A1/fr
Priority to US14/394,765 priority patent/US20150119267A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • Cancers of the brain and nervous system are among the most difficult to treat. Prognosis for patients with these cancers depends on the type and location of the tumor as well as its stage of development. For many types of brain cancer, average life expectancy after symptom onset may be months or a year or two. Treatment consists primarily of surgical removal and radiation therapy; chemotherapy is also used, but the range of suitable chemotherapeutic agents is limited, perhaps because most therapeutic agents do not penetrate the blood-brain barrier adequately to treat brain tumors. Using known chemotherapeutics along with surgery and radiation rarely extends survival much beyond that produced by surgery and radiation alone. Thus improved therapeutic options are needed for brain tumors.
  • Gliomas are a common type of brain tumor. They arise from the supportive neuronal tissue comprised of glial cells (hence the name glioma), which maintain the position and function of neurons. Gliomas are classified according to the type of glial cells they resemble: astrocytomas (including glioblastomas) resemble star-shaped astrocyte glial cells, oligodendrogliomas resemble oligodendrocyte glial cells; and ependymomas resemble ependymal glial cells that form the lining of fluid cavities in the brain. In some cases, a tumor may contain a mixture of these cell types, and would be referred to as a mixed glioma.
  • astrocytomas including glioblastomas
  • oligodendrogliomas resemble oligodendrocyte glial cells
  • ependymomas resemble ependymal glial cells that form the lining of fluid cavities in the
  • the typical current treatment for brain cancers is surgical removal of the majority of the tumor tissue, which may be done by invasive surgery or using biopsy or extractive methods. Gliomas tend to disseminate irregularly, though, and are very difficult to remove completely. As a result, recurrence nearly always occurs soon after tumor removal. Radiation therapy and/or chemotherapy can be used in combination with surgical removal, but these generally provide only modest extension of survival time. For example, recent statistics showed that only about half of patients in the U.S. who are diagnosed with glioblastoma are alive one year after diagnosis, and only about 25% are still alive after two years, even when treated with the current standard of care combination treatments.
  • GBM Glioblastoma multiforme
  • TAMs Tumor-associated macrophages
  • TAMs are one of the prominent stromal cell types present, and often account for a substantial portion of the cells in the tumor tissues. Their origin is not certain: these TAMs may originate either from microglia, the resident macrophage population in the brain, or they may be recruited from the periphery.
  • TAMs can modulate tumor initiation and progression in a tissue-specific manner: they appear to suppress cancer development in some cases, but they enhance tumor progression in the majority of studies to date. Indeed, in approximately 80% of the cancers in which there is increased macrophage infiltration, the elevated TAM levels are associated with more aggressive disease and poor patient prognosis. Several studies have shown that human gliomas also exhibit a significant increase in TAM numbers, which correlates with advanced tumor grade, and TAMs are typically the predominant immune cell type in gliomas. However, the function of TAMs in gliomagenesis remains poorly understood, and it is not currently known whether targeting of these cells represents a viable therapeutic strategy.
  • TNF-a or integrin ⁇ 3 produced by TAMs have been implicated in the suppression of glioma growth, whereas in other reports CCL2 and MT1 -MMP have been proposed as enhancers of tumor development and invasion.
  • CSF-1 R signaling represents a novel, translationally relevant approach that has been used in several oncological contexts, including xenograft intratibial bone tumors. However, it has not yet been shown to be effective in brain tumors.
  • Some non-brain cancers have been targeted with compounds that affect a variety of cell types that are associated with, or support, tumor cells rather than directly targeting the tumor cells themselves.
  • PLX3397 is reported to co-inhibit three targets (FMS, Kit, and Flt3-ITD) and to down-modulate various cell types including macrophages, microglia, osteoclasts, and mast cells. PLX3397 has been tested for treating Hodgkin's lymphoma.
  • a CSF-1 R inhibitor had no direct effect on proliferation of glioblastoma cells in culture, though, and it did not reduce numbers of macrophage cells in tumors of treated animals. It is thus surprising that, as also demonstrated herein, a CSF-1 R inhibitor can effectively inhibit growth of brain tumors in vivo, cause reduction in tumor volume in advanced stage GBM, and even apparently eradicate some glioblastomas.
  • the present invention is based on demonstrations that brain tumors, particularly glioblastoma, can be treated with an inhibitor of CSF-1 R.
  • the effectiveness of the CSF- 1 R inhibitors described herein is believed to be due to their inhibition of certain activities of TAMs, even though it does not appear to significantly reduce the number of TAMs present, and is likely also a function of the demonstrated ability of these compounds to penetrate the blood-brain barrier effectively in subjects with a brain tumor.
  • Colony stimulating factor-1 also termed macrophage colony stimulating factor (M-CSF)
  • M-CSF macrophage colony stimulating factor
  • CSF-1 R also known as c-FMS
  • Small molecule inhibitors of CSF-1 R have been developed that block receptor phosphorylation by competing for ATP binding in the active site, as for other receptor tyrosine kinase inhibitors.
  • the present invention uses a potent, selective CSF-1 R inhibitor, which penetrates the blood-brain barrier (BBB), to block CSF-1 R signaling in glioma as illustrated in the RCAS-PDGF-B-HAINestin-Tv-a;lnk4a/ArT ⁇ mouse model of gliomagenesis.
  • BBB blood-brain barrier
  • This genetically engineered glioma model is ideal for preclinical testing as a model for human GBM, as it recapitulates all features of human GBM in an immunocompetent setting. Because it closely models human GBM, and proneural GBM in particular, efficacy in this model is expected to translate into clinical efficacy on human glioblastomas such as glioblastoma multiforme and mixed gliomas.
  • the invention can be practiced with any inhibitor of CSF-1 R capable of penetrating the brain.
  • Some such compounds are the 6-O-substituted benzoxazole and benzothiazole compounds disclosed in WO2007/121484, particularly the compounds of Formula lla and lib in that reference, and the compounds disclosed herein.
  • the invention provides a method to treat a brain tumor in a mammalian subject, comprising administering to the subject an effective amount of a compound of Formula (I):
  • R 1 is an alkyl pyrazole or an alkyl carboxamide
  • R 2 is a hydroxycycloalkyl
  • the method can be used to treat a patient, frequently a human subject, who has been diagnosed with a brain tumor. Further embodiments of the invention are described below.
  • Figure 1A is a graph showing the relative amounts of Live DAPI-positive cells in normal brain and glioblastoma tissue, as measured by the increased proportion of cells staining positive for CD45 (pan-leukocyte marker) and CD1 1 b (myeloid cell marker) in the tumor tissue.
  • the fluorescence activated cell sorting (FACS) data is shown, also.
  • Figure 1 B depicts CD68 stained brain cells from Normal Brain tissue and from a
  • GBM Grade IV glioblastoma
  • Figure 1C depicts the increased level of mRNAs for CD68, CSF-1 R and CSF-1 relative to the housekeeping gene Ubiquitin C (Ubc), for GBM tissue relative to normal brain tissue.
  • Ubc housekeeping gene Ubiquitin C
  • Figure 1 D shows the relative amounts of CD1 1 b, TVA, CSF-1 and CSF-1 R in TAMs relative to tumor cells.
  • Figure 2 depicts amounts of BLZ945 in Plasma, brain tissue from the left half of a brain containing GBM, and from the right half of the same brain with no visible GBM at several time points after treating cohorts of mice with BLZ945.
  • Figure 3A shows inhibition by BLZ945 of CSF-1 R phosphorylation, following CSF-1 stimulation, in bone-marrow derived macrophage cells (BMDM).
  • BMDM bone-marrow derived macrophage cells
  • Figure 3B shows the rate of population doubling of BMDM cells untreated, and demonstrates that treating the cells with 67 nM BLZ945 has the same effect on this rate as absence of CSF-1 stimulation.
  • Figures 3C-3E show rate of proliferation of BMDM cells from the Ink4a/Arf-/- mice, of CRL-2647 normal mouse brain cells, and for two mouse GBM cell cultures.
  • Figure 3F shows that the total number and size of neurospheres was unaffected by BLZ945 at 670 nM.
  • Figure 4A depicts symptom-free survival of
  • Figure 4B depicts tumor grade for treated and untreated mice at the 26 week study endpoint. All control mice had grade III or IV tumors.
  • Figure 5A shows tumor size data measured by MRI for treated and control animals during the first 6 days of treatment with BLZ945.
  • the BLZ945 treatment is the lower curve, showing no tumor growth over the course of 6 days.
  • Figure 5B shows tumor volume for individual control mice (upper graph) and treated mice (lower graph) during the first 6 days after dosing with BLZ945 started.
  • Figures 5C and 5D depict tumor volume measured by MRI in BLZ945-treated animals beginning with large tumors (>40 mm 3 volume), and Figure 5D shows that even with large tumors, tumor volume decreased in nearly all subjects over just 6 days.
  • Figure 5-2 shows data on tumor volume for individual animals in the control group for Example 5 (5-2A) and the treated group (5-2B), and Figure 5-2C shows the tumor size data for the large tumor subjects treated with BLZ945 in Example 5.
  • Figure 6 the first graph (6A) shows the percentage of Olig2+ cells in the brains of animals in the vehicle, treated, and 'Large tumor' groups in Example 5.
  • the second graph (6B) shows the fraction of tumor cells that were actively dividing, as measured by bromodeoxyuridine (BrdU) labeling.
  • the third graph (6C) shows the level of apoptosis in the tumor cells, as measured by cleaved caspase 3 (CC3) staining, and demonstrates that BLZ945 promotes apoptosis of tumor cells.
  • CC3 cleaved caspase 3
  • the invention provides compounds of Formula (I) for use to treat brain tumors, and methods of using compounds of Formula (I) for the treatment of brain tumors.
  • the compounds of Formula (I) have this formula:
  • R 1 is an alkyl pyrazole or an alkyl carboxamide
  • R 2 is a hydroxycycloalkyl
  • the treatment of a brain tumor can include inhibition of the rate of growth of a brain tumor (slowing tumor growth), or reversal of growth of a brain tumor (i.e., reduction in tumor volume), or substantial elimination of the tumor, which has been demonstrated by the treatment herein of mice having such tumors.
  • the treatment can slow progression or reverse progression of a glioblastoma. It may be used in conjunction with other treatments including removal of the bulk of a brain tumor, and may be used to slow or reverse regrowth or to reduce the volume or mass of residual tumorous tissue following brain tumor removal by surgical or biopsy methods.
  • the compounds may also be used in conjunction with other chemotherapeutics.
  • the compounds of formula (I) include compounds wherein R 1 is an alkyl- substituted pyrazole or carboxamide, e.g., a C1 -C4 alkyl pyrazole or a carboxamide of the formula -C(0)NHR, where R is a C1 -C4 alkyl group.
  • R 1 is an alkyl- substituted pyrazole or carboxamide, e.g., a C1 -C4 alkyl pyrazole or a carboxamide of the formula -C(0)NHR, where R is a C1 -C4 alkyl group.
  • the alkyl group is Me or Et.
  • R 1 is
  • R' is Me or Et.
  • R 2 can be a hydroxycyclohexyl group such as this:
  • Specifically preferred compounds include any of the following compounds, or a mixture of any two or more of these compounds, or a pharmaceutically acceptable salt of any one of these:
  • R' is Me, Et or Propyl, particularly methyl.
  • Specific embodiments of these compounds can be of (R,R) absolute stereochemistry or (S,S) absolute stereochemistry.
  • Compounds of Formula (I) are known in the art, and methods for making them are disclosed, for example, in WO2007/121484; their usefulness to treat glioma and their penetration of the blood-brain barrier were not previously known.
  • Compound (1 c) corresponds to BLZ945, which was utilized for in vitro and in vivo tests described herein.
  • R' is Me, Et or Propyl are another aspect of the present invention that provide a dual-inhibitor effect, which is expected to increase effectiveness in the treatment methods disclosed herein.
  • the compounds can be used alone or they can be formulated into a pharmaceutical composition that also contains at least one pharmaceutically acceptable excipient, and often contains at least two pharmaceutically acceptable excipients. It will be understood that pharmaceutically acceptable excipients are typically sterilized. Some suitable excipients are disclosed herein; in some embodiments, the compound is formulated as a composition comprising captisol, e.g, 20% captisol.
  • the brain tumor is selected from a brain metastasis, an astrocytoma (including glioblastoma), an oligodendroglioma, an ependymomas, and a mixed glioma.
  • the brain tumor is a glioma, particularly glioblastoma multiforme.
  • the brain tumor is a brain metastasis, i.e., a metastatic tumor arising from a cancer that originated elsewhere in the body.
  • the patient is one having glioblastoma or having a mixed glioma that comprises glioblastoma.
  • the subject is one diagnosed with proneural glioblastoma. See Verhaak, et al., Cancer Cell 17(1 ):98-1 10 (2010). This subtype of glioblastoma tends to occur in younger subjects and to involve mutations of TP53, IDH1 and PDGFRA. Verhaak, et al. reported that patients with proneural glioblastoma were less responsive than other subtypes (classical, neural, mesenchymal) to the aggressive chemotherapies in use in 2010, and even suggested that such treatment may be contraindicated for these patients.
  • the present methods are especially effective to treat proneural glioblastoma, as demonstrated by the proneural GBM animal model used herein.
  • the method is used to treat a subject before other treatment methods such as tumor removal.
  • the method is used to treat a subject in conjunction with other treatment methods such as tumor removal by either surgical or biopsy methods, or in conjunction with radiation therapy, or in conjunction with both tumor removal and radiation therapy.
  • other chemotherapeutic agents can be used along with the compounds and methods disclosed above. Suitable additional chemotherapeutic agents for use in these methods are those known in the art as conventional ones for use in treating glioblastoma.
  • chemotherapeutics include antiangiogenic agents, bevacizumab with or without irinotecan, nitrosoureas such as Carmustine (BCNU), platins such as c/ ' s -platinum (cisplatin), alkylating agents such as temozolomide, tyrosine kinase inhibitors (gefitinib or erlotinib), Ukrain, and cannabinoids.
  • additional therapeutic agents co-therapeutics
  • a preferred embodiment involves use of a compound selected from those of Formula I disclosed herein, (e.g., Formula la, lb, Ic, Id, le, If, Ig or Ih) in combination with temozolomide or a platin compound.
  • macrophages have been implicated in reduced therapeutic responses in breast cancer and increased revascularization in glioblastoma xenografts following radiation therapy. Since these macrophage effects reduce the efficacy of other therapies, compounds of the invention, which inhibit macrophage activities in glioblastoma in vivo, may be expected to provide a synergistic effect when used in combination with other therapeutic agents or radiation therapy.
  • the methods described herein are practiced with a compound of Formula (Ic). In other embodiments, the methods may be practiced with a compound of Formula (I) that is not the compound of Formula (Ic), such as the other species disclosed herein.
  • the compound of Formula (I) also inhibits at least one other target to provide enhanced antitumor effects.
  • compounds of these formulas include:
  • Exemplary compounds of Formula Ig and Ih are included in the following table to illustrate the relative activities on CSF-1 R and PDGFR. Many such compounds are known in the art, see WO2007/066898, and methods to make these compounds are also well known.
  • the compounds of Formula I are quite active on CSF-1 R regardless of the stereochemistry at the cyclohexyl ring as shown in the table below.
  • the S,S isomers are also highly active on PDGFR- ⁇ as well as on CSF-1 R, and thus may act on GBM by two mechanisms to provide enhanced efficacy.
  • a method to treat a brain tumor in a mammalian subject comprising administering to the subject an effective amount of a compound of Formula (I):
  • R 1 is an alkyl pyrazole or an alkyl carboxamide
  • R 2 is a hydroxycycloalkyl
  • R' is Me or Et.
  • the brain tumor is a brain metastasis, astrocytoma (including glioblastoma), oligodendroglioma,
  • the method further comprises administering to the subject an effective amount of an additional cancer therapeutic an antiangiogenic agents, bevacizumab with or without irinotecan, nitrosoureas such as Carmustine (BCNU), platins such as c/ ' s -platinum (cisplatin), alkylating agents such as temozolomide, tyrosine kinase inhibitors (gefitinib or eriotinib), Ukrain, and cannabinoids.
  • an additional cancer therapeutic an antiangiogenic agents bevacizumab with or without irinotecan, nitrosoureas such as Carmustine (BCNU)
  • platins such as c/ ' s -platinum (cisplatin)
  • alkylating agents such as temozolomide, tyrosine kinase inhibitors (gefitinib or eriotinib), Ukrain, and cannabinoids.
  • the amount of the compound of Formula (I) administered to the subject is between about 50 mg/kg per day and about 500 mg/kg per day, or between 5-500 mg/kg, or between 100 and 300 mg/kg per day; suitably about 200 mg/kg per day.
  • R' is Methyl, Ethyl or Propyl; or a pharmaceutically acceptable salt thereof.
  • salt refers to an acid addition or base addition salt of a compound of the invention.
  • Salts include in particular “pharmaceutically acceptable salts”.
  • pharmaceutically acceptable salts refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present invention can be prepared by conventional chemical methods.
  • such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid.
  • a stoichiometric amount of the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • any formula given herein is intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 CI, 125 l respectively.
  • the compounds of the invention are unlabeled, i.e., they comprise approximately natural isotope abundances for all atoms.
  • the compounds of the invention are labeled by selective incorporation of an enriched non-natural isotope for one atom in the compound of Formula (I).
  • the invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H and 14 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • Such isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art in view of the description of synthesis of the compounds of Formula I in, for example, U.S. patent publication no. US2008/0045528 (WO2007/121484).
  • BLZ945 is described in that reference as well as several of its isomers.
  • Examples 173 and 174 in that reference describe synthesis of pyrazole compound (le) using 1 R,2R- aminocyclohexanol, and can be adapted for synthesis of other pyrazole compounds of Formula I, both labeled and unlabeled.
  • the same publication at page 163 describes synthesis of both 1 R,2R- and 1 S,2S-aminocyclohexanol, which can readily be substituted into the method of Example 173 to produce (If) and other compounds of Formula I having the (S,S) stereochemistry, both labeled and unlabeled.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • the term "pharmaceutically acceptable excipients” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289- 1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • a therapeutically effective amount of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease, etc.
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a subject, is effective to (1 ) at least partially alleviating, inhibiting, preventing and/or ameliorating a condition, or a disorder or a disease (i) mediated by CSF-1 R, or (ii) associated with CSF-1 R activity, or (iii) characterized by activity (normal or abnormal) of CSF-1 R; or (2) reducing or inhibiting the activity of CSF-1 R; or (3) reducing or inhibiting the expression of CSF-1 R.
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of CSF-1 R; or at least partially reducing or inhibiting the expression of CSF-1 R.
  • the meaning of the term "a therapeutically effective amount" as illustrated in the above embodiment for CSF-1 R also applies by the same means to any other relevant proteins/peptides/enzymes, such as PDGFR and the like.
  • the term "subject" refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In preferred embodiments, the subject is a human.
  • primates e.g., humans, male or female
  • the subject is a primate.
  • the subject is a human.
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • "treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • 'treating' typically includes either slowing rate of growth of a tumor or of regrowth of a tumor after the bulk of the tumor has been removed, or reducing the size of the tumor or of remnants of the tumor after the bulk of the tumor has been removed.
  • a subject is "in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • the subject has been diagnosed with a brain tumor, frequently a form of glioblastoma, and preferably with glioblastoma multiforme.
  • the term "a,” “an,” “the” and similar terms used in the context of the present invention are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context.
  • the present invention utilizes a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and rectal administration, etc.
  • the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions).
  • compositions can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifers and buffers, etc.
  • the pharmaceutical composition comprises at least one additional chemotherapeutic agent such as temozolomide, in an effective amount.
  • the pharmaceutical compositions are tablets or gelatin capsules comprising the active ingredient together with at least one excipient, such as captisol (used in the Examples herein) or one of the following:
  • diluents e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine;
  • lubricants e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also
  • lubricants e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol
  • binders e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired;
  • d) carriers such as an aqueous vehicle containing a co-solvating material such as captisol, PEG, glycerin, cyclodextrin, or the like;
  • disintegrants e.g., starches, agar, alginic acid or its sodium salt, or
  • Tablets may be either film coated or enteric coated according to methods known in the art.
  • the compound or composition is prepared for oral administration, as a tablet or capsule, for example, or as a solution or suspension of the compound of Formula (I), optionally packaged in a single-dose container such as a capsule.
  • compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin or olive oil.
  • the compound or composition is prepared to be administered by injection.
  • Certain injectable compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • Said compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 - 75%, or contain about 1-50%, of the active ingredient.
  • the compound or composition is prepared to be administered topically.
  • compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier.
  • Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • compositions for topical application include aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like.
  • topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like. They are thus particularly suited for use in topical, including cosmetic, formulations well-known in the art.
  • Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
  • a dry powder either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids
  • the effective amount of the compound of Formula (I) is between about 10 mg/kg per day, and about 500 mg/kg per day. In particular embodiments, the effective amount is between about 25 mg/kg per day and about 300 mg/kg per day, such as about 100 to about 250 mg/kg per day.
  • the dosage may be administered in 1 -4 doses per day, or it may be administered on alternating days. In a preferred embodiment, the dosage is about 200 mg/kg per day, and is administered in one or two oral doses per day.
  • the present invention further provides anhydrous pharmaceutical compositions and dosage forms comprising the compounds of the present invention as active ingredients, since water may facilitate the degradation of certain compounds.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e. g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • the compounds and methods described herein are useful to treat a variety of brain tumors, based on their demonstrated ability to penetrate the blood-brain barrier and to inhibit accumulation of TAMs in and/or around a tumor in the brain.
  • the brain tumor is a metastasis of a cancer that originated elsewhere in the body.
  • the brain tumor is a glioma such as glioblastoma multiforme.
  • the compounds of formula I in free form or in salt form exhibit valuable pharmacological properties, e.g. CSF-1 R and optionally PDGFR modulating properties, e.g. as indicated in in vitro and in vivo tests as provided in the next sections and are therefore indicated for therapy.
  • valuable pharmacological properties e.g. CSF-1 R and optionally PDGFR modulating properties, e.g. as indicated in in vitro and in vivo tests as provided in the next sections and are therefore indicated for therapy.
  • the present invention provides the use of a compound of formula (I) or in therapy.
  • the therapy is selected from a disease which may be treated by inhibition of CSF-1 R.
  • the disease is selected from the afore-mentioned list, suitably any brain tumor, more suitably a glioblastoma such as glioblastoma multiforme.
  • the invention provides a method of treating a disease which is treated by inhibition of CSF-1 R, comprising administration of a therapeutically acceptable amount of a compound of formula (I) or any of the embodiments of these compounds disclosed herein.
  • the disease is selected from the afore-mentioned list, suitably a brain tumor, such as one of the gliomas, specifically including glioblastoma multiforme.
  • the pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1-500 mg or about 1 -250 mg or about 1-150 mg or about 0.5-100 mg, or about 1-50 mg of active ingredients.
  • the therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to treat or inhibit the progress of the disorder or disease based on the present disclosure.
  • the above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof.
  • the compounds of the present invention can be applied in vitro in the form of solutions, e.g., aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution.
  • the dosage in vitro may range between about 10 "3 molar and 10 "9 molar concentrations.
  • a therapeutically effective amount in vivo may range depending on the route of administration, between about 0.1-500 mg/kg, typically 10-400 mg/kg, or between about 100-300 mg/kg, or between 1-100 mg/kg. In some embodiments, a dose of about 200 mg/kg is suitable for treatment of glioblastoma, and can be administered orally.
  • the activity of a compound according to the present invention can be assessed by the following in vitro & in vivo methods.
  • the compounds of the invention can be shown to inhibit CSF-1 R. As described herein these compounds readily traverse the blood-brain barrier, and also inhibit or reverse growth of a tumor in the brain.
  • the tumor is detectable by known methods, and progress of treatment can be monitored by known methods.
  • the progress of the treatment is monitored by using MRI (magnetic resonance imaging) to determine the size of the tumor and any metastases.
  • the compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agents such as the cotherapeutic agents described herein.
  • the compound of the present invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents.
  • the invention provides a product comprising a compound of formula (I) and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy.
  • the therapy is the treatment of a disease or condition mediated by inhibition of CSF-1 R.
  • Products provided as a combined preparation include a composition comprising the compound of formula (I) and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound of formula (I) and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
  • the invention provides a pharmaceutical composition comprising a compound of formula (I) and another therapeutic agent(s).
  • the pharmaceutical composition may comprise a pharmaceutically acceptable excipient, as described above, or more than one such cotherapeutic agent.
  • the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (I).
  • the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit of the invention typically comprises directions for administration.
  • the compound of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the compound of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the compound of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the compound of the invention and the other therapeutic agent.
  • the invention provides the use of a compound of formula (I) for treating a disease or condition mediated by CSF-1 R, wherein the medicament is prepared for administration with another therapeutic agent, including one of the additional chemotherapeutic agents disclosed herein as suitable for use in combination with compounds of Formula I .
  • the invention also provides the use of another therapeutic agent for treating a disease or condition mediated by CSF-1 R wherein the medicament is administered with a compound of formula (I).
  • the invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by CSF-1 R], wherein the compound of formula (I) is prepared for administration with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by CSF-1 R, wherein the other therapeutic agent is prepared for administration with a compound of formula (I).
  • the invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by CSF-1 R wherein the compound of formula (I) is administered with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by CSF-1 R, wherein the other therapeutic agent is administered with a compound of formula (I).
  • the invention also provides the use of a compound of formula (I) for treating a disease or condition mediated by CSF-1 R wherein the patient has previously (e.g. within 24 hours) been treated with another therapeutic agent.
  • the invention also provides the use of another therapeutic agent for treating a disease or condition mediated by CSF-1 R wherein the patient has previously (e.g. within 24 hours) been treated with a compound of formula (I).
  • the other therapeutic agent is selected from an antiangiogenic agents, bevacizumab with or without irinotecan, nitrosoureas such as Carmustine (BCN U), platins such as c/ ' s -platinum (cisplatin), alkylating agents such as temozolomide, tyrosine kinase inhibitors (gefitinib or erlotinib), Ukrain, and cannabinoids.
  • the other agent is a cotherapeutic agent selected from: an antiangiogenic compound, a cannabinoid, and temozolomide.
  • Specific individual combinations which may provide particular treatment benefits include compound la, lb, lc, Id, le, If, 1 g, or 1 h, in combination with temozolomide. This combination may be administered orally as described herein to treat various brain tumors, such as glioblastoma multiforme.
  • HPLC high performance liquid chromatography
  • HPLC high performance liquid chromatography
  • the analytical columns were reversed phase Phenomenex Luna C18 -5 , 4.6 x 50 mm, from Alltech (Deerfield, I L).
  • a gradient elution was used (flow 2.5 mL/min), typically starting with 5% acetonitrile/95% water and progressing to 100% acetonitrile over a period of 10 minutes.
  • All solvents contained 0.1 % trifluoroacetic acid (TFA).
  • UV ultraviolet light
  • HPLC solvents were from Burdick and Jackson (Muskegan, Ml), or Fisher Scientific (Pittsburgh, PA).
  • Mass spectrometric analysis was performed on one of two LCMS instruments: a Waters System (Alliance HT HPLC and a Micromass ZQ mass spectrometer; Column: Eclipse XDB-C18, 2.1 x 50 mm; gradient: 5-95% (or 35-95%, or 65-95% or 95-95%) acetonitrile in water with 0.05% TFA over a 4 min period ; flow rate 0.8 mL/min;
  • Example 1 Macrophage numbers are increased in a mouse model
  • TAMs gliomagenesis in the RCAS-PDGF-B-HNNestin-Tv-a;lnk4a/Arf / ⁇ mouse model.
  • GBM glioblastoma multiforme
  • CD1 1 b+ myeloid cells/ macrophages accounted for the overwhelming majority of leukocytes (89.9-98.5% of CD45+ cells), with a 3.8-fold increase in CD45+CD1 1 b+ cells in the tumors (12.7 ⁇ 2.0%) compared to normal brain (3.3 ⁇ 0.5%), and no differences in the populations of CD45+CD1 1 b- cells.
  • B Normal brain or GBM tissue sections from symptomatic PDG mice were immunofluorescently co-stained for CSF-1 R, CD68 (macrophages), and DAPI.
  • Assays were run in triplicate and expression normalized to ubiquitin C (Ubc) for each sample. Expression is depicted relative to normal brain.
  • D Normal brain or GBM tissue sections from symptomatic PDG mice were stained for CSF- 1 R in combination with the macrophage markers F4/80 and CD1 1 b as well as F4/80, CD1 1 b, and CD68 in combination with lba-1 (macrophages/ microglia). DAPI was used for the nuclear counterstain. Scale bar, 50 ⁇ . Data are presented as mean + SEM. P values were obtained using unpaired two-tailed Student's t-test; * P ⁇ 0.05; ** P ⁇ 0.01.
  • the different cell type-specific populations were also from GBMs to determine the source of CSF-1 and its receptor. The purity of the distinct populations was confirmed by expression of the TVA receptor only in the tumor cell fraction and CD1 1 b solely in the TAMs. While CSF-1 was expressed by both tumor cells and TAMs, CSF-1 R was only expressed by TAMs (Figure 1 D).
  • the first column in each group of three in Figure 1 D is Mixed cells, the second is FACS-purified tumor cells, and the third is FACS-purified TAMs; Mixed cells are set to 1 to normalize the data.
  • the graphs show no CD1 1 b expression in tumor cells and no CSF-1 R expression in tumor cells, while TVA stains tumor cells only, not TAMs, and CSF-1 is present in approximately equal amounts in both tumor and TAM cells.
  • Example 2 Analysis of the CSF-1 R inhibitor BLZ945: pharmacokinetics and cell-based assays.
  • BLZ945 (Compound lc) has been disclosed as a selective c-fms (CSF-1 R) kinase inhibitor for the suppression of tumor-induced osteolytic lesions in bone.
  • BLZ945 is an ATP competitive inhibitor that inhibits CSF-1 R in biochemical assays at 1 nM, and inhibits CSF-dependent cell proliferation at an IC-50 of about 67 nM.
  • the IC50 values for most of >200 miscellaneous kinases tested are >10 ⁇ (10,000 nM), and for cKIT and PDGFRp the IC-50's are 3.5 ⁇ (3500nM) and 5.9 ⁇ (5900 nM) respectively.
  • the compound When screened against several hundred kinases in the Ambit® kinase array, the compound showed activity lower than 50% of control only against CSF-1 R, PDGFRa and PDGFR3, and the activity on the two PDGFRs was far lower than its activity on CSF-1 R in direct inhibition assays.
  • compounds like BLZ945 but having the (S,S) stereochemistry exhibit activity against PDGFR3 at levels similar to their high level of activity on CSF-1 R.
  • mice having GBM detectable in only the right half of their brains were treated with BLZ945, and the concentration of compound in plasma, and in the right and left halves of the brain were then measured at various time points (15 mins, 2 hr, 8 hr, 24 hr).
  • the plasma concentration rises rapidly to a little over 100 uM and remains above 50 uM at 8 hr, then declines to a low level by 24 hr.
  • the concentration in brain tissue follows a similar pattern: it remains a little lower than the plasma level, but rises well above 50 uM at the 15 min and 2 hr time points.
  • BBB blood-brain barrier
  • Example 3 Inhibitory activity of BLZ945 against different cell types in vitro.
  • Bone marrow-derived macrophages were isolated and differentiated as previously described in the literature, and were then treated with 67 nM BLZ945.
  • BLZ945 caused a clear inhibition of CSF-1 R phosphorylation following CSF-1 stimulation (Figure 3A) at each time point (1 .5 min, 3 min, 5 min).
  • BMDMs from Ink4a/Arf null mice were also tested in the presence and absence of BLZ945.
  • Figure 3C shows that these BMDMs, like those from the wild-type mice, were substantially inhibited by concentrations of BLZ945 of 67 nM and above ( Figure 3D).
  • BLZ945 is an effective inhibitor of CSF-1 R signaling, which leads to a complete block in macrophage viability.
  • Figures 3C-3E demonstrate that proliferation of BMDM cells from the Ink4a/Arf-/- mice as strongly inhibited at concentrations of BLZ945 of 67 nM and above, as were CRL-2467 cells (normal mouse brain), while even at 6700 nM it has little or no effect on proliferation of four mouse and one human glioblastoma cell cultures
  • a human glioma cell line and a series of primary tumor cells and neurospheres were treated with BLZ945 at similar concentrations to those found effective against macrophage growth.
  • U87-MG cells derived from a human GBM, which have been shown to be dependent on PDGFR signaling in culture and in vivo, were not affected by BLZ945 treatment at the same doses as above ( Figure 3E).
  • the formation of secondary neurospheres from primary neurospheres (derived from mouse RCAS-PDGF-B-HA/Nestin-Tv-a;lnk4a/Ar ⁇ ⁇ GBMs) was not altered by BLZ945 treatment (Figure 3F).
  • Example 4 Treatment with the CSF-1R inhibitor BLZ945 blocks glioma progression.
  • mice were dosed via oral gavage daily with either 200 mg/kg BLZ945 in 20% captisol, or the vehicle (20% captisol) as a control. The mice were subsequently evaluated for symptom-free survival. The median survival in the vehicle treated cohort was 5.71 weeks (40 days), whereas 64.4% of the BLZ945 treated cohort were still alive at the trial endpoint of 26 weeks post-injection (31-32 weeks of age) (Figure 4A, P ⁇ 0.0001 ).
  • mice in the lnk4a/Ar ⁇ ⁇ background start developing spontaneous tumors, mostly lymphomas and sarcomas, around 30 weeks of age, which would complicate interpretation of the glioma phenotype in longer studies.
  • the data in Figure 4A shows that none of the control mice (vehicle only) were symptom free by 8 weeks after virus injection, while over half of the treated mice were symptom free at the endpoint of 26 weeks. Note: 4 treated mice were sacrificed at 12 weeks for histology studies. Of these, 3 were tumor free, and one had a grade II glioma.
  • Example 5 MRI Imaging to monitor effects of BLZ945 on tumor growth.
  • mice a;lnk4a/Ar ⁇ ⁇ mice was determined by MRI, and mice were added to the trial when this was at least 4.5 mm 3 or greater. Mice were treated with BLZ945 or the vehicle control for 7 days, as described above. MRI scans were performed on the day before treatment was initiated, at the mid-point of the treatment, and at the day before the end of the trial period. Vehicle-treated mice showed a progressive, dramatic increase in tumor volume over this short trial, as shown in Figure 5A, with the average tumor volume increasing about 5-fold. BLZ945 treatment blocked tumor progression as determined by MRI ( Figure 5A), with no increase in tumor size over the same short period.
  • FIG. 5B shows tumor volume for individual control mice (upper graph) and treated mice (lower graph) during the first 6 days after dosing with BLZ945 started. Nearly all of the BLZ945 treated animals show little or no increase in tumor size, while all of the control animals show large increases in tumor volume.
  • mice with tumor volumes of 48.7 to 132 mm 3 were treated with BLZ945, and changes in tumor volume were monitored by MRI over a span of 6 days. Tumor volume actually decreased in nearly all test animals, and 6 of 18 treated mice had a reduction of at least 30% in tumor size ( Figures 5D and 5-2C). Control animals were not included in this test, because they would not have been expected to survive to the endpoint.
  • Example 6 Analysis of hallmark capabilities of cancer in BLZ945 treated tumors.
  • apoptosis in these cells was assessed, also. Apoptotic cells were counted as those that had cytoplasmic cleaved caspase-3 (CC3)+ staining and condensed nuclei. As shown in Figure 6C, the CSF-1 R inhibitor treatment caused an increase in apoptosis at the earlier time point in particular, although little staining was observed in the Day 7 large tumor cohort.
  • Tumor volume change is volume at endpoint (day 6) relative to day one, and the reported changes are relative to the control (vehicle) group.
  • BLZ945 is a potent new therapy that blocks tumor progression in a very aggressive glioma model in mice.
  • the compound dramatically enhanced survival in a preclinical mouse model of gliomagenesis, and sharply reduced tumor growth rates and also reduced tumor size over a short and longer test period.
  • BLZ945 appears to eliminate visible tumors in significant numbers of mice, and sharply reduces the tumor grade in most of the treated mice.
  • compound (Id) inhibits PDGFR with an IC50 only about 4-fold higher than its IC50 for CSF-1 R.
  • a therapeutically effective concentration of either of these compounds is expected to affect both target sites, and to exhibit synergistic activity on gliomas.
  • mice were fully anesthetized with 10 mg/ml ketamine/1 mg/ml xylazine and were subcutaneously injected with 50 ⁇ of the local anesthetic 0.25% bupivacaine at the surgical site.
  • Mice were intracranially injected with 1 ⁇ containing 2 x 105 DF-1 :RCAS-PDGF-B-HA cells between 5-6 weeks of age using a fixed stereotactic apparatus (Stoelting). Injections were made to the right frontal cortex, approximately 1.5 mm lateral and 1 mm caudal from bregma, and at a depth of 2 mm.
  • CSF-1 and CSF-1 R were initiated in mice with RCAS-PDGF-B-HA- SV40- eGFP (RCAS-PDGF-GFP) as previously described (E. I. Fomchenko et al., PloS ONE 6, e20605 (201 1 ).).
  • Nestin-Tv-a;lnk4a/Arf-/- pups were injected with 1 ⁇ of DF- 1 :RCAS-PDGF-B-GFP cells on post-natal day 2 into the left cortex between the eye and ear.
  • the CSF-1 R inhibitor BLZ945 was formulated in 20% captisol at a concentration of 12.5 mg/ml.
  • the vehicle control, 20% captisol was processed in the same manner.
  • mice were dosed with 200 mg/kg BLZ945 or vehicle (20% captisol) by oral gavage once per day.
  • tumor-bearing mice were treated with a single dose of BLZ945 and sacrificed at different time points post treatment. Plasma, and the left (contralateral) and right (tumor-bearing) hemispheres of the brain were snap frozen in liquid nitrogen for subsequent analysis of BLZ945 concentrations in the tissue. For long-term survival studies, dosing was begun at 17 days/ 2.5 weeks post-injection of RCAS-PDGF-B-HA. For the fixed time-point studies, mice underwent MRI scans at 4-5 weeks post-injection of RCAS-PDGF-B-HA, as previously described (Transl Oncol 2, 89 (2009)).
  • regions of interest were circumscribed on T2 weighted images and their corresponding area in mm 2 was multiplied by the slice height of 0.7 mm.
  • the total tumor volume is the sum of the ROI volume in each slice, and the volume for the first and last slice in which the tumor appear is halved to approximate the volume of a trapezoid.
  • animals were randomly assigned to treatment groups.
  • a third cohort of mice with tumors larger than 40 mm 3 was also treated with BLZ945 (denoted as BLZ945 Large).
  • a size-matched vehicle treated cohort was not included for this cohort having the larger starting tumor burden because these mice would not have been able to survive to the trial endpoint.
  • mice were euthanized at defined time points as described in the figure legends or when they became symptomatic from their tumors, which included signs of poor grooming, lethargy, weight loss, hunching, macrocephaly, or seizures.
  • mice were euthanized by carbon dioxide asphyxiation or fully anesthetized with avertin (2,2,2-tribromoethanol, Sigma) and cervically dislocated prior to tissue harvest.
  • avertin 2,2,2-tribromoethanol, Sigma
  • mice were fully anesthetized with avertin and transcardially perfused with 20 ml of PBS.
  • the brain was then isolated and the tumor macrodissected from the surrounding normal tissue.
  • mice were injected intraperitoneally with 100 mg/g of bromodeoxyuridine (BrdU; Sigma) 2 hours prior to sacrifice.
  • mice were fully anesthetized with avertin, transcardially perfused with 10 ml of PBS, followed by 10 ml of 4% paraformaldehyde in PBS (PFA).
  • PFA paraformaldehyde
  • the brain was postfixed in PFA overnight at 4°C while other tissues were cryopreserved in 30% sucrose at 4°C. After post-fixation, the brain was then transferred to 30% sucrose and incubated at 4°C until the brain was fully equilibrated and sank to the bottom of the tube (typically 2 to 3 days). All tissues were then embedded in OCT (Tissue-Tek) and 10 ⁇ cryostat tissue sections were used for all subsequent analysis.
  • H&E hematoxylin and eosin
  • tissue sections were counterstained with DAPI (5 mg/ml stock diluted 1 :5000 in PBS) for 5 minutes prior to mounting with ProLong Gold Antifade mounting media (Invitrogen).
  • DAPI ProLong Gold Antifade mounting media
  • tissue sections were first subjected to citrate buffer based antigen retrieval by submerging in antigen unmasking solution (0.94% v/v in distilled water; Vector Laboratories) and microwaving for 10 minutes on half power, followed by cooling to room temperature for at least 30 minutes.
  • tissue were then washed in PBS and blocked with mouse Ig blocking reagent (Vector Laboratories) according to the manufacturer's instructions for 1 hour at room temperature.
  • tissue sections were incubated with 2M HCI for 15 minutes at room temperature to denature DNA and then in neutralizing 0.1 M sodium borate buffer (pH 8.5) for 5 minutes. After PBS washes, the rest of the staining was performed according to the standard protocol.
  • Tissue sections were then washed 6 times for 5 minutes in PBS and blocked overnight at 4°C in a new buffer of 5% donkey serum, 3% bovine serum albumin, and 0.5% PNB in PBS. The following day, slides were incubated for 2 hours at room temperature with the next set of primary antibodies: rabbit anti-Olig2 (1 :200) and rat anti-CD1 1 b (1 :200) diluted in 5% donkey serum, 3% bovine serum albumin, and 0.5% PNB in PBS.
  • Tissue sections were visualized under a Carl Zeiss Axioimager Z1 microscope equipped with an Apotome.
  • the analysis of immunofluorescence staining, cell number, proliferation, apoptosis, and colocalization studies were performed using TissueQuest analysis software (TissueGnostics) as previously described (Journal Immunol Methods 237, 39 (2000)).
  • mice analyzed For analysis of phagocytosis, 15 randomly selected fields of view from within the tumor were acquired using the 63x oil immersion objective (total magnification 630x) and the Apotome to ensure cells were in the same optical section. Positive cells were counted manually using Volocity (PerkinElmer) and were discriminated by the presence of a DAPI+ nucleus. Apoptotic cells were counted as those that had cytoplasmic cleaved caspase-3 (CC3)+ staining and condensed nuclei. A cell was considered to have been engulfed by a macrophage when it was surrounded by a contiguous CD1 1 b+ ring that encircled at least two-thirds of the cell border. The numbers of mice analyzed are specified in the figure legends.
  • mice were treated with BLZ945 or vehicle and sacrificed 1 hour following the final dose and tumors were harvested.
  • Samples were biochemically fractionated as described previously. Synaptosomal membrane fractions were lysed in NP-40 lysis buffer (0.5% NP-40, 50 mM Tris-HCI [pH 7.5], 50 mM NaCI, 1 x complete Mini protease inhibitor cocktail (Roche), 1 x PhosSTOP phosphatase inhibitor cocktail (Roche)) and protein quantified using the BCA assay (Pierce). Protein lysates were loaded (90 ⁇ g/lane) onto SDS-PAGE gels and transferred to PVDF membranes for immunoblotting.
  • Membranes were probed with antibodies against phospho-CSF-1 R Y721 (1 :1000;
  • BMDMs Primary bone marrow derived macrophages
  • the tumor was digested to a single cell suspension by incubation with 5 ml of papain digestion solution (0.94 mg/ml papain [Worthington], 0.48 mM EDTA, 0.18 mg/ml NAcety-L-cysteine [Sigma], 0.06 mg/ml DNase I [Sigma], diluted in Earl's Balanced Salt Solution and allowed to activate at room temperature for at least 30 minutes).
  • papain digestion solution 0.94 mg/ml papain [Worthington], 0.48 mM EDTA, 0.18 mg/ml NAcety-L-cysteine [Sigma], 0.06 mg/ml DNase I [Sigma]
  • the enzyme was inactivated by the addition of 2 ml of 0.71 mg/ml ovomucoid (Worthington).
  • the cell suspension was then passed through a 40 ⁇ mesh to remove undigested tissue, washed with FACS buffer (1 % IgG Free BSA in PBS [Jackson Immunoresearch]), and centrifuged at a low speed of 750 rpm (Sorvall Legend RT), to remove debris and obtain the cell pellet.
  • FACS buffer 1 % IgG Free BSA in PBS [Jackson Immunoresearch]
  • centrifuged at a low speed of 750 rpm Sorvall Legend RT
  • tumors were digested to a single cell suspension by incubation for 10 minutes at 37°C with 5 mL of 1 .5 mg/ml collagenase III (Worthington) and 0.06 mg/mL DNase I in 1 x Hanks Balanced Salt Solution (HBSS) with calcium and magnesium.
  • HBSS Hanks Balanced Salt Solution
  • the cell suspension was then washed with PBS and passed through a 40 ⁇ mesh to remove undigested tissue.
  • the cell pellet was resuspended in 15 ml of room temperature 25% Percoll prepared from stock isotonic Percoll (90% Percoll [Sigma], 10% 10x HBSS), and then spun for 15 minutes at 1500 rpm (Sorvall Legend RT) with accelerator and brake set to 1 .
  • the cell pellet was then washed with 1 x HBSS prior to being resuspended in FACS buffer. After counting, cells were incubated with 1 ⁇ of Fc Block for every million cells for at least 15 minutes at 4°C.
  • samples were run on a BD LSR II (Becton Dickstein), and all subsequent compensation and gating performed with FlowJo analysis software (TreeStar).
  • samples were run on a BD FACSAria (Becton Dickstein) cell sorter and cells were collected into FACS buffer. Cells were then centrifuged and resuspended in 500 ⁇ Trizol (Invitrogen) before snap freezing in liquid nitrogen and storage at -80°C.
  • Macrodissected tumors were digested to a single cell suspension by incubation for 8-12 minutes at 37°C as described above.
  • the cell suspension was washed with Neural Stem Cell (NSC) Basal Media (Stem Cell Technologies), and centrifuged at low speed (750 rpm Sorvall Legend RT), to remove debris.
  • NSC Neural Stem Cell
  • DMEM fetal bovine serum
  • FBS FBS
  • glioma cultures were grown for 24 hours on poly-L-lysine coated coverslips (BD Biocoat). Cells were then fixed with 4% PFA in 0.1 M phosphate buffer overnight at 4°C, permeabilized with 0.1 % Triton-X for 5 minutes and blocked with 0.5% PNB for at least one hour. The presence of macrophages, tumor cells and astrocytes were examined by immunofluorescent staining of CD1 1 b (1 :200), Nestin (1 :500) and GFAP (1 :1000), respectively (Table 7).
  • the cell pellet was resuspended in neurosphere media consisting of mouse NSC Basal Media, NSC proliferation supplements, 10 ng/ml EGF, 20 ng/ml basic-FGF and 1 mg/ml Heparin (Stem Cell Technologies). Fresh media was added every 72 hours for 2 weeks. Primary neurospheres were collected, mechanically disaggregated to a single cell suspension and propagated by serial passaging. To generate glioma cell lines, secondary neurospheres were dissociated to single cell suspensions and cultivated in DMEM+10% FBS as a monolayer. Multiple glioma cell lines were derived from independent mice, denoted GBM1-4 herein. Glioma cells were infected with a pBabe-H2B-mCherry construct as described previously (O. Florey, et al., Nat Cell Biol 13. 1335 (201 11).
  • BMDMs bone marrow-derived macrophages
  • C57BL/6 WT C57BL/6 ⁇ -actin-GFP or Nestin-Tv-a
  • Ink4a/Arf-/- mice were anesthetized with Avertin (Sigma) and then sacrificed via cervical dislocation.
  • Femurs and tibiae were harvested under sterile conditions from both legs and flushed.
  • the marrow was passed through a 40 ⁇ strainer and cultured in 30 ml Teflon® bags (PermaLife PL-30) with 10 ng/ml recombinant mouse CSF-1 (R&D Systems).
  • Bone marrow cells were cultured in Teflon® bags for 7 days, with fresh CSF-1 -containing media replacing old media every other day to induce macrophage differentiation.
  • U-87 MG HTB-14 glioma and CRL-2467 microglia cell lines were purchased from the ATCC.
  • the U-87 MG cell line was cultured in DMEM+10% FBS.
  • the CRL-2467 cell line was cultured in DMEM+10% FBS with 30 ng/ml recombinant mouse CSF-1.
  • GCM Glioma cell-conditioned media experiments
  • Media that had been conditioned by glioma tumor cell lines grown in serum free media for 24 hours was passed through 0.22 ⁇ filters to remove cellular debris, and is referred to herein as glioma cell-conditioned media (GCM).
  • GCM was used to stimulate differentiated C57BL/6 WT or ⁇ -3 ⁇ - ⁇ + BMDMs.
  • Control macrophages received fresh media containing 10% FBS and 10 ng/ml recombinant mouse CSF-1.
  • differentiated BMDMs were cultivated in GCM containing either DMSO as vehicle, or 67nM BLZ945, 670nM BLZ945, or in regular media containing 10 ng/ml mouse recombinant CSF-1 and 10 ng/ml IL-4 (R&D Systems) for 24 hours or 48 hours prior to experimental analysis. Analysis of Mrc1/ CD206 expression by flow cytometry
  • mice primary glioma cultures (containing a mixed population of tumor cells, TAMs, astrocytes etc.), 1 x 106 cells were cultivated in DMEM+10% FBS in the presence of BLZ945 or DMSO as vehicle.
  • BMDMs 1 x 10 6 cells were cultivated in DMEM supplemented with recombinant mouse CSF-1 or GCM in the presence of
  • BLZ945 or DMSO as vehicle. After 48 hours, cells were scraped and washed with FACS buffer. Cells were counted and incubated with 1 ⁇ of Fc Block (BD Pharmingen) per 106 cells for at least 15 minutes at 4°C. Cells were then stained with CD45 and CD1 1 b antibodies for 10 minutes at 4°C and washed with FACS buffer. Cells were fixed and permeabilized using the BD Cytofix/CytopermTM kit (BD Biosciences) according to the manufacturer's instructions. Subsequently cells were stained with anti- CD206 antibody. For analysis, samples were run on a BD LSR II (Becton Dickstein), and all subsequent compensation and gating performed with FlowJo analysis software (TreeStar).
  • Fc Block Fc Block
  • Control or GCM pre-stimulated macrophages derived from ⁇ -8 ⁇ . ⁇ - ⁇ + mice were cocultured in a 1 :1 ratio with 1 x 105 serum starved mCherry-positive glioma cells (from the cell lines derived above) for 48 hours in the presence of 670nM BLZ945 or DMSO as vehicle. Following collection of trypsinized co-cultured cells, wells were rinsed in additional media and this volume was collected to ensure harvesting of all macrophages, which adhered tightly to cell culture dishes.
  • Cell growth rate was determined using the MTT cell proliferation kit (Roche). Briefly, cells were plated in triplicate in 96-well plates (1 x 10 3 cells/well for glioma cell lines and 5 x 10 3 cells/well for BMDM and CRL-2467 cells) in the presence or absence of 6.7- 6700 nM of BLZ945. Media was changed every 48 hours. BMDM and CRL-2467 cells were supplemented with 10 ng/ml and 30 ng/ml recombinant mouse CSF-1 respectively unless otherwise indicated. 10 ⁇ of MTT labeling reagent was added to each well and then incubated for 4 hours at 37°C, followed by the addition of 100 ⁇ MTT solubilization reagent overnight. The mixture was gently resuspended and absorbance was measured at 595 nm and 750 nm on a spectraMax 340pc plate reader (Molecular Devices).
  • Primary neurospheres were disaggregated to a single cell suspension and 5 x 10 3 cells were plated in a 6 well plate in neurosphere media in the presence of BLZ945 or DMSO as vehicle. Media was changed every 48 hours. Secondary neurosphere formation was assayed by counting the number of neurospheres obtained after 2 weeks.
  • Taqman probes (Applied Biosystems) for Cd1 1 b (Mm00434455_m1 ), Cd68 (Mm03047343_m1 ), Csf-1 (Mm00432688_m1 ), Csf-1 r (Mm00432689_m1 ), II34 (Mm00712774_m1 ), Mrc1 (Mm00485148_m1 ), and Tv-a (custom), were used for qPCR. Assays were run in triplicate and expression was normalized to ubiquitin C (Mm01201237_m1 ) for each sample.

Abstract

La présente invention concerne un composé de la formule (I) dans laquelle R1 est un alkyle pyrazole ou un alkyle carboxamide et R2 est un hydroxycycloalkyle, ou un sel de qualité pharmaceutique de celui-ci, et des compositions contenant ces composés, afin de les utiliser pour traiter une tumeur cérébrale, en particulier un glioblastome. L'invention concerne un traitement efficace d'une tumeur cérébrale et peut être utilisée par une administration par voie orale d'un composé de la formule (I), tel que décrit par les présentes.
PCT/US2012/036630 2011-05-05 2012-05-04 Inhibiteurs de csf-1r pour le traitement de tumeurs cérébrales WO2012151541A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/US2013/036628 WO2013158559A1 (fr) 2012-04-16 2013-04-15 Inhibition de la signalisation du récepteur du facteur 1 de stimulation des colonies pour le traitement du cancer du cerveau
US14/394,765 US20150119267A1 (en) 2012-04-16 2013-04-15 Inhibition of colony stimulating factor-1 receptor signaling for the treatment of brain cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161482723P 2011-05-05 2011-05-05
US61/482,723 2011-05-05

Publications (1)

Publication Number Publication Date
WO2012151541A1 true WO2012151541A1 (fr) 2012-11-08

Family

ID=46298660

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/036630 WO2012151541A1 (fr) 2011-05-05 2012-05-04 Inhibiteurs de csf-1r pour le traitement de tumeurs cérébrales

Country Status (1)

Country Link
WO (1) WO2012151541A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014147631A1 (fr) * 2013-03-22 2014-09-25 Natco Pharma Limited Formulation comprenant du géfitinib comme suspension orale
WO2016182988A1 (fr) 2015-05-08 2016-11-17 Memorial Sloan Kettering Cancer Center Compositions et méthodes pour le traitement des gliomes
KR20170022699A (ko) * 2015-08-21 2017-03-02 한국과학기술연구원 우레이도벤조티아졸 유도체 및 이를 포함하는 암의 예방 또는 치료용 약학 조성물
RU2765820C2 (ru) * 2017-03-28 2022-02-03 Новартис Аг Новые способы лечения рассеянного склероза
WO2022057895A1 (fr) * 2020-09-21 2022-03-24 Hutchison Medipharma Limited Composés hétéroaromatiques et leurs utilisations

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5601674A (en) 1989-04-14 1997-02-11 General Electric Company Fiber reinforced ceramic matrix composite member and method for making
WO2007119046A1 (fr) * 2006-04-14 2007-10-25 Astrazeneca Ab 4-anilinoquinoleine-3-carboxamides en tant qu'inhibiteurs de la csf-ir kinase
WO2007121484A2 (fr) * 2006-04-19 2007-10-25 Novartis Ag Composés à base de benzoxazole et de benzothiazole 6-0 substitués et procédés d'inhibition de signalisation csf-1r
WO2008144062A1 (fr) * 2007-05-21 2008-11-27 Novartis Ag Inhibiteurs du csf-1r, compositions et procédés d'utilisation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5601674A (en) 1989-04-14 1997-02-11 General Electric Company Fiber reinforced ceramic matrix composite member and method for making
WO2007119046A1 (fr) * 2006-04-14 2007-10-25 Astrazeneca Ab 4-anilinoquinoleine-3-carboxamides en tant qu'inhibiteurs de la csf-ir kinase
WO2007121484A2 (fr) * 2006-04-19 2007-10-25 Novartis Ag Composés à base de benzoxazole et de benzothiazole 6-0 substitués et procédés d'inhibition de signalisation csf-1r
WO2008144062A1 (fr) * 2007-05-21 2008-11-27 Novartis Ag Inhibiteurs du csf-1r, compositions et procédés d'utilisation

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CONIGLIO SALVATORE J ET AL: "Microglial Stimulation of Glioblastoma Invasion Involves Epidermal Growth Factor Receptor (EGFR) and Colony Stimulating Factor 1 Receptor (CSF-1R) Signaling", MOLECULAR MEDICINE, vol. 18, no. 3, 27 January 2012 (2012-01-27), (BALTIMORE), pages 519 - 527, XP002679581, ISSN: 1076-1551 *
KOMOHARA Y ET AL: "Possible involvement of the M2 anti-inflammatory macrophage phenotype in growth of human gliomas", JOURNAL OF PATHOLOGY, vol. 216, no. 1, September 2008 (2008-09-01), pages 15 - 24, XP002679579, ISSN: 0022-3417 *
W. R. TRACEY, A.B. JEFFERSON: "NIBR Postdoctoral Program", 21 September 2010 (2010-09-21), pages 1 - 2,40, XP002679580, Retrieved from the Internet <URL:http://med.stanford.edu/careercenter/highlights/files/Novartis%20PowerPoint.pdf> [retrieved on 20120709] *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014147631A1 (fr) * 2013-03-22 2014-09-25 Natco Pharma Limited Formulation comprenant du géfitinib comme suspension orale
WO2016182988A1 (fr) 2015-05-08 2016-11-17 Memorial Sloan Kettering Cancer Center Compositions et méthodes pour le traitement des gliomes
EP3294417A4 (fr) * 2015-05-08 2019-02-06 Memorial Sloan Kettering Cancer Center Compositions et méthodes pour le traitement des gliomes
US10722517B2 (en) 2015-05-08 2020-07-28 Memorial Sloan Kettering Cancer Center Compositions and methods for treatment of glioma
KR20170022699A (ko) * 2015-08-21 2017-03-02 한국과학기술연구원 우레이도벤조티아졸 유도체 및 이를 포함하는 암의 예방 또는 치료용 약학 조성물
KR101721128B1 (ko) 2015-08-21 2017-03-29 한국과학기술연구원 우레이도벤조티아졸 유도체 및 이를 포함하는 암의 예방 또는 치료용 약학 조성물
RU2765820C2 (ru) * 2017-03-28 2022-02-03 Новартис Аг Новые способы лечения рассеянного склероза
WO2022057895A1 (fr) * 2020-09-21 2022-03-24 Hutchison Medipharma Limited Composés hétéroaromatiques et leurs utilisations

Similar Documents

Publication Publication Date Title
EP2704713B1 (fr) Inhibiteurs de csf-1r pour traitement de tumeurs cérébrales
US20170259081A1 (en) Methods for regulating cell mitosis by inhibiting serine/threonine phosphatase
US20100029683A1 (en) Methods for regulating cell mitosis by inhibiting serine/threonine phosphateses
WO2012151541A1 (fr) Inhibiteurs de csf-1r pour le traitement de tumeurs cérébrales
JP2024012493A (ja) 消化管間質腫瘍の治療のための併用療法
EA032345B1 (ru) Способ лечения рака с использованием кофермента q10
JP7025094B2 (ja) ヘテロシクリデンアセトアミド誘導体含有医薬
WO2019109074A1 (fr) Thérapies anticancéreuses à base de mébendazole et méthodes d&#39;utilisation
EP3880207B1 (fr) Combinaison d&#39;un inhibiteur de mcl-1 et de midostaurine, utilisations et compositions pharmaceutiques associées
EP1711230B1 (fr) Utilisation d&#39;epothilone pour le traitement de defauts de connectivite neuronale comme par exemple la schizophrenie et l&#39;autisme
US11357766B2 (en) Compositions and methods for treating cancer
JP2022529949A (ja) ミトコンドリア標的化及び抗癌治療のためのアルキルtpp化合物
US20130345231A1 (en) Anticancer therapeutic agents
KR20140145604A (ko) 암 치료용 키나제 저해제
AU2012250574B8 (en) CSF-1R inhibitors for treatment of brain tumors
JP6630285B2 (ja) Erg発癌遺伝子陽性癌のための新規阻害剤
US20220339145A1 (en) Compositions and Methods for Treating Cancer
EP4175639B1 (fr) Masitinib pour le traitement du cancer de la prostate résistant à la castration

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: 12727698

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: 12727698

Country of ref document: EP

Kind code of ref document: A1