WO2015173181A1 - Compounds for treating spinal muscular atrophy - Google Patents

Compounds for treating spinal muscular atrophy Download PDF

Info

Publication number
WO2015173181A1
WO2015173181A1 PCT/EP2015/060343 EP2015060343W WO2015173181A1 WO 2015173181 A1 WO2015173181 A1 WO 2015173181A1 EP 2015060343 W EP2015060343 W EP 2015060343W WO 2015173181 A1 WO2015173181 A1 WO 2015173181A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrimidin
pyrido
pyridazin
hydrogen
methyl
Prior art date
Application number
PCT/EP2015/060343
Other languages
French (fr)
Inventor
Hasane Ratni
Luke Green
Nikolai A. Naryshkin
Marla L. Weetall
Original Assignee
F. Hoffmann-La Roche Ag
Hoffmann-La Roche Inc.
Ptc Therapeutics Inc.
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
Priority to CA2948561A priority Critical patent/CA2948561C/en
Priority to EA201692280A priority patent/EA035068B1/en
Priority to SG11201609497TA priority patent/SG11201609497TA/en
Priority to UAA201612716A priority patent/UA119670C2/en
Priority to EP23169721.0A priority patent/EP4241772A3/en
Priority to ES15721701T priority patent/ES2761423T3/en
Priority to MX2016014547A priority patent/MX371050B/en
Priority to AU2015261046A priority patent/AU2015261046C1/en
Application filed by F. Hoffmann-La Roche Ag, Hoffmann-La Roche Inc., Ptc Therapeutics Inc. filed Critical F. Hoffmann-La Roche Ag
Priority to MA39995A priority patent/MA39995B1/en
Priority to RS20191541A priority patent/RS59718B1/en
Priority to BR112016026205-0A priority patent/BR112016026205B1/en
Priority to DK15721701T priority patent/DK3143025T3/en
Priority to EP19200058.6A priority patent/EP3663296B1/en
Priority to KR1020217002966A priority patent/KR102256013B1/en
Priority to PL15721701T priority patent/PL3143025T3/en
Priority to CR20160518A priority patent/CR20160518A/en
Priority to SI201531021T priority patent/SI3143025T1/en
Priority to MYPI2016001903A priority patent/MY174284A/en
Priority to KR1020167034983A priority patent/KR102213740B1/en
Priority to CN201580027306.9A priority patent/CN106459092B/en
Priority to NZ72500815A priority patent/NZ725008A/en
Priority to JP2016567816A priority patent/JP6236173B2/en
Priority to EP15721701.9A priority patent/EP3143025B1/en
Publication of WO2015173181A1 publication Critical patent/WO2015173181A1/en
Priority to ZA2016/07026A priority patent/ZA201607026B/en
Priority to PH12016502081A priority patent/PH12016502081A1/en
Priority to IL24865316A priority patent/IL248653B/en
Priority to US15/351,267 priority patent/US9969754B2/en
Priority to US15/953,008 priority patent/US10882868B2/en
Priority to IL270027A priority patent/IL270027B/en
Priority to HRP20192159TT priority patent/HRP20192159T1/en
Priority to US17/099,578 priority patent/US11827646B2/en
Priority to FR21C1039C priority patent/FR21C1039I2/en
Priority to NO2021035C priority patent/NO2021035I1/en
Priority to LTPA2021010C priority patent/LTPA2021010I1/lt
Priority to NL301128C priority patent/NL301128I2/en
Priority to US18/496,034 priority patent/US20240076302A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides compounds which are SMN2 gene splicing modulators, their manufacture, pharmaceutical compositions comprising them and their use as medicaments for the treatment of spinal muscular atrophy (SMA).
  • SMA spinal muscular atrophy
  • R 1 , R2 and R 3 are as described herein, and pharmaceutically acceptable salts thereof.
  • SMA Spinal muscular atrophy
  • CNS central nervous system
  • Infantile SMA is the most severe form of this neurodegenerative disorder. Symptoms include muscle weakness, poor muscle tone, weak cry, limpness or a tendency to flop, difficulty sucking or swallowing, accumulation of secretions in the lungs or throat, feeding difficulties, and increased susceptibility to respiratory tract infections.
  • the legs tend to be weaker than the arms and developmental milestones, such as lifting the head or sitting up, cannot be reached. In general, the earlier the symptoms appear, the shorter the lifespan. As the motor neuron cells deteriorate, symptoms appear shortly afterward. The severe forms of the disease are fatal and all forms have no known cure.
  • the course of SMA is directly related to the rate of motor neuron cell deterioration and the resulting severity of weakness.
  • Type 0 SMA In Utero SMA is the most severe form of the disease and begins before birth. Usually, the first symptom of Type 0 SMA is reduced movement of the fetus that can first be observed between 30 and 36 weeks of pregnancy. After birth, these newborns have little movement and have difficulties with swallowing and breathing.
  • Type 1 SMA Infantile SMA or Werdnig-Hoffmann disease presents symptoms between 0 and 6 months, form of SMA is also very severe. Patients never achieve the ability to sit, and death usually occurs within the first 2 years without ventilatory support.
  • Type 2 SMA (Intermediate SMA) has an age of onset at 7-18 months. Patients achieve the ability to sit unsupported, but never stand or walk unaided. Prognosis in this group is largely dependent on the degree of respiratory involvement.
  • Type 3 SMA (Juvenile SMA or Kugelberg-Welander disease) is generally
  • Type 3 SMA individuals are able to walk independently at some point during their disease course but often become wheelchair-bound during youth or adulthood.
  • Type 4 SMA (Adult onset SMA). Weakness usually begins in late adolescence in the tongue, hands, or feet, then progresses to other areas of the body. The course of adult SMA is much slower and has little or no impact on life expectancy.
  • the SMN gene has been mapped by linkage analysis to a complex region in chromosome 5q. In humans, this region contains an approximately 500 thousand base pairs (kb) inverted duplication resulting in two nearly identical copies of the SMN gene. SMA is caused by an inactivating mutation or deletion of the telomeric copy of the gene (SMN1) in both
  • SMN RNA-protein complexes
  • SMN may have other functions in motor neurons, however its role in preventing the selective degeneration of motor neurons is not well established.
  • SMA is diagnosed based on clinical symptoms and by the presence of at least one copy of the SMN1 gene test. However, in approximately 5% of cases SMA is caused by mutation in genes other than the inactivation of SMN 1, some known and others not yet defined. In some cases, when the SMN 1 gene test is not feasible or does not show any abnormality, other tests such as an electromyography (EMG) or muscle biopsy may be indicated.
  • EMG electromyography
  • SMN delta exon 7 ( ⁇ 7 SMN) model (Le et al, Hum. Mol. Genet., 2005, 14:845) carries both the SMN2 gene and several copies of the ⁇ 7 SMN2 cDNA and recapitulates many of the phenotypic features of Type 1 SMA.
  • the ⁇ 7 SMN model can be used for both SMN2 expression studies as well as the evaluation of motor function and survival.
  • the C/C-allele mouse model ⁇ Jackson Laboratory strain #008714, The Jackson Laboratory, Bar Harbor, ME) provides a less severe SMA disease model, with mice having reduced levels of both SMN2 full length (FL SMN2) mRNA and SMN protein.
  • the C/C-allele mouse phenotype has the SMN2 gene and a hybrid mSMNl-SMN2 gene that undergoes alternative splicing, but does not have overt muscle weakness.
  • the C/C-allele mouse model is used for SMN2 expression studies.
  • HDAC histone deacetylase
  • mRNA stabilizers mRNA decapping inhibitor RG3039 from Repligen
  • neuroprotective agents such as Olesoxime have been chosen for investigation.
  • Such strategies are not aimed at SMN for the treatment of SMA, but instead are being explored to protect the SMN-deficient motor neurons from neurodegeneration.
  • WO2009/151546A A system designed for identifying compounds that cause ribosomal frameshifting to produce a stabilized SMN protein from ⁇ 7 SMN2 mRNA and certain
  • haloalkylheteroaryl refers to a Ci_7-alkyl which is substituted by amino
  • arylalkylheterocycloalkyl refers to a heterocycloalkyl-radical which is substituted by an alkyl which is substituted by an aryl.
  • moiety refers to an atom or group of chemically bonded atoms that is attached to another atom or molecule by one or more chemical bonds thereby forming part of a molecule.
  • variables A, R 1 , R2 and R 3 of formula (I) refer to moieties that are attached to the core structure of formula (I) by a covalent bond.
  • the term "one or more" refers to the range from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.
  • substituted denotes that a specified group bears one or more substituents. Where any group can carry multiple substituents and a variety of possible substituents is provided, the substituents are independently selected and need not to be the same.
  • unsubstituted means that the specified group bears no substituents.
  • optionally substituted means that the specified group is unsubstituted or substituted by one or more substituents, independently chosen from the group of possible substituents.
  • one or more means from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.
  • compound(s) of this invention and “compound(s) of the present invention” refer to compounds as disclosed herein and stereoisomers, tautomers, solvates, and salts (e.g., pharmaceutically acceptable salts) thereof.
  • pharmaceutically acceptable salts denotes salts which are not biologically or otherwise undesirable.
  • Pharmaceutically acceptable salts include both acid and base addition salts.
  • pharmaceutically acceptable acid addition salt denotes those pharmaceutically acceptable salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and organic acids selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene
  • pharmaceutically acceptable base addition salt denotes those pharmaceutically acceptable salts formed with an organic or inorganic base.
  • acceptable inorganic bases include sodium, potassium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts.
  • Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, and polyamine resins.
  • substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, trieth
  • chiral center denotes a carbon atom bonded to four nonidentical substituents.
  • chiral denotes the ability of non-superimposability with the mirror image, while the term “achiral” refers to embodiments which are superimposable with their mirror image.
  • Chiral molecules are optically active, i.e., they have the ability to rotate the plane of plane-polarized light.
  • Compounds of the present invention can have one or more chiral centers and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates.
  • optically pure enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates.
  • halo halogen
  • halogen halogen
  • halide halide
  • alkyl denotes a monovalent linear or branched saturated hydrocarbon group of 1 to 12 carbon atoms. In particular embodiments, alkyl has 1 to 7 carbon atoms, and in more particular embodiments 1 to 4 carbon atoms. Examples of alkyl include methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, or tert-butyl. Particular examples for alkyl are methyl and ethyl.
  • haloalkyl denotes an alkyl group wherein at least one of the hydrogen atoms of the alkyl group has been replaced by same or different halogen atoms, particularly fluoro atoms.
  • haloalkyl include monofluoro-, difluoro- or trifluoro-methyl, -ethyl or -propyl, for example 3,3,3-trifluoropropyl, 2-fluoroethyl, 2,2,2-trifluoroethyl, fluoromethyl, or
  • bicyclic ring system denotes two rings which are fused to each other via a common single or double bond (annelated bicyclic ring system), via a sequence of three or more common atoms (bridged bicyclic ring system) or via a common single atom (spiro bicyclic ring system).
  • Bicyclic ring systems can be saturated, partially unsaturated, unsaturated or aromatic.
  • Bicyclic ring systems can comprise heteroatoms selected from N, O and S.
  • cycloalkyl denotes a saturated monocyclic or bicyclic hydrocarbon group of 3 to 10 ring carbon atoms. In particular embodiments cycloalkyl denotes a monovalent saturated monocyclic hydrocarbon group of 3 to 8 ring carbon atoms. Bicyclic means consisting of two saturated carbocycles having one or more carbon atoms in common. Particular cycloalkyl groups are monocyclic. Examples for monocyclic cycloalkyl are cyclopropyl, cyclobutanyl, cyclopentyl, cyclohexyl or cycloheptyl. Examples for bicyclic cycloalkyl are bicyclo[2.2.1]heptanyl, or bicyclo[2.2.2]octanyl. One particular example of cycloalkyl is cyclopropyl.
  • heterocycloalkyl denotes a saturated or partly unsaturated mono-, bi- or tricyclic ring system of 3 to 9 ring atoms, comprising 1, 2, or 3 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • heterocycloalkyl is a monovalent saturated monocyclic ring system of 4 to 7 ring atoms, comprising 1, 2, or 3 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • Examples for monocyclic saturated heterocycloalkyl are aziridinyl, oxiranyl, azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydro-thienyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperazinyl, morpholinyl, thiomorpholinyl, l,l-dioxo-thiomorpholin-4-yl, azepanyl, diazepanyl, homopiperazinyl, or oxazepanyl.
  • bicyclic saturated heterocycloalkyl examples include 8-aza-bicyclo[3.2.1]octyl, quinuclidinyl, 8-oxa-3-aza-bicyclo[3.2.1]octyl, 9-aza-bicyclo[3.3.1]nonyl, 3-oxa-9-aza- bicyclo[3.3.1]nonyl, or 3-thia-9-aza-bicyclo[3.3.1]nonyl.
  • Examples of a partly unsaturated heterocycloalkyl are dihydrofuryl, imidazolinyl, dihydro-oxazolyl, tetrahydro-pyridinyl, or dihydropyranyl.
  • Particular examples of heterocycloalkyl are 1,4-diazepanyl,
  • heterocycloalkyl hexahydropyrrolo[l,2-a]pyrazinyl and piperazinyl.
  • N -heterocycloalkyl denotes a heterocycloalkyl radical containing at least one nitrogen ring atom and where the point of attachment of the heterocycloalkyl radical to the rest of the molecule is through a nitrogen ring atom.
  • Particular examples of N-heterocycloalkyl are 1,4-diazepanyl, hexahydropyrrolo[l,2-a]pyrazinyl, piperidinyl, piperazinyl and pyrrolidinyl. More particular examples of N-heterocycloalkyl are hexahydropyrrolo[l,2-a]pyrazinyl and piperazinyl.
  • an atom or functional group is denoted “basic” if it is suitable to accept a proton and if the calculated pKa of its conjugate acid is at least 7, more particularly if the calculated pKa of its conjugate acid is at least 7.8, most particularly if the calculated pKa of its conjugate acid is at least 8.
  • pKa values were calculated in-silico as described in F. Milletti et al., J. Chem. Inf. Model (2007) 47:2172-2181.
  • alkylene denotes a linear saturated divalent hydrocarbon group of 1 to 7 carbon atoms or a divalent branched saturated hydrocarbon group of 3 to 7 carbon atoms.
  • alkylene groups include methylene, ethylene, propylene, 2-methylpropylene, butylene, 2- ethylbutylene, pentylene, hexylene.
  • Particular examples for alkylene are ethylene, propylene, and butylene.
  • amino denotes a group of the formula -NR'R" wherein R' and R" are independently hydrogen, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl or as described herein. Alternatively, R' and R", together with the nitrogen to which they are attached, can form a heterocycloalkyl.
  • primary amino denotes a group wherein both R' and R" are hydrogen.
  • secondary amino denotes a group wherein R' is hydrogen and R" is a group other than hydrogen.
  • tertiary amino denotes a group wherein both R' and R" are other than hydrogen.
  • Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine.
  • active pharmaceutical ingredient (or “API") denotes the compound or molecule in a pharmaceutical composition that has a particular biological activity.
  • formulation are used interchangeably and denote a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with
  • pharmaceutically acceptable excipients to be administered to a mammal, e.g., a human in need thereof.
  • pharmaceutically acceptable denotes an attribute of a material which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and neither biologically nor otherwise undesirable and is acceptable for veterinary as well as human pharmaceutical use.
  • pharmaceutically acceptable ingredient in a pharmaceutical composition having no therapeutic activity and being non-toxic to the subject administered such as disintegrators, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, carriers, diluents or lubricants used in formulating pharmaceutical products.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • therapeutically effective amount denotes an amount of a compound or molecule of the present invention that, when administered to a subject, (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein.
  • the therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease treated, the age and relative health of the subject, the route and form of administration, the judgement of the attending medical or veterinary practitioner, and other factors.
  • treating or “treatment” of a disease state include inhibiting the disease state, i.e., arresting the development of the disease state or its clinical symptoms, or relieving the disease state, i.e., causing temporary or permanent regression of the disease state or its clinical symptoms.
  • SMA spinal muscular atrophy
  • Symptoms of SMA include muscle weakness, poor muscle tone, weak cry, weak cough, limpness or a tendency to flop, difficulty sucking or swallowing, difficulty breathing, accumulation of secretions in the lungs or throat, clenched fists with sweaty hand,
  • treating spinal muscular atrophy (SMA) or “treatment of spinal muscular atrophy (SMA)” includes one or more of the following effects: (i) reduction or amelioration of the severity of SMA; (ii) delay of the onset of SMA; (iii) inhibition of the progression of SMA; (iv) reduction of hospitalization of a subject; (v) reduction of hospitalization length for a subject;
  • amelioration of the severity of one or more symptoms associated with SMA comprises: (x) reduction of the duration of a symptom associated with SMA; (xi) prevention of the recurrence of a symptom associated with SMA; (xii) inhibition of the development or onset of a symptom of SMA; and/or (xiii) inhibition of the progression of a symptom associated with SMA.
  • treating SMA denotes one or more of the following beneficial effects: (i) a reduction in the loss of muscle strength; (ii) an increase in muscle strength; (iii) a reduction in muscle atrophy; (iv) a reduction in the loss of motor function; (v) an increase in motor neurons;
  • treating SMA refers to the functional ability or retention of the functional ability for a human infant or a human toddler to sit up unaided or for a human infant, a human toddler, a human child or a human adult to stand up unaided, to walk unaided, to run unaided, to breathe unaided, to turn during sleep unaided, or to swallow unaided.
  • ECi.5 X concentration for production of full length SMN2 minigene mRNA is defined as that concentration of test compound that is effective in increasing the amount of full length SMN2 minigene mRNA to a level 1.5-fold greater relative to that in vehicle-treated cells.
  • ECi.5 X concentration for SMN protein expression is defined as that concentration of test compound that is effective in producing 1.5 times the amount of SMN protein in an SMA patient fibroblast cell compared to the amount produced from the vehicle control.
  • the present invention relates to compounds of formula (I)
  • R 1 is hydrogen or Ci_7-alkyl
  • R is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C 3 _g-cycloalkyl;
  • R is hydrogen, Ci_7-alkyl, or C 3 _g-cycloalkyl
  • A is N-heterocycloalkyl or NR 12 R 13 , wherein N-heterocycloalkyl comprises 1 or 2 nitrogen ring atoms and is optionally substituted with 1, 2, 3 or 4 substituents selected from R 14 ;
  • R 12 is heterocycloalkyl comprising 1 nitrogen ring atom, wherein heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R 14 ;
  • R 13 is hydrogen, Ci_7-alkyl or C 3 _g-cycloalkyl
  • R 14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C 3 _ 8-cycloalkyl and heterocycloalkyl or two R 14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R 14 substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof.
  • Particular embodiments of the present invention are compounds of formula (I) and armaceutically acceptable salts thereof.
  • R 1 is hydrogen or Ci_7-alkyl
  • R is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C 3 _8-cycloalkyl;
  • R is hydrogen, Ci_7-alkyl, or C 3 _g-cycloalkyl
  • A is N-heterocycloalkyl comprising 1 or 2 nitrogen ring atoms, wherein N- heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R 14 ;
  • R 14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C 3 _ 8-cycloalkyl and heterocycloalkyl or two R 14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R 14 substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 1 is Ci_7-alkyl, particularly methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 12 is piperidinyl optionally substituted with 1, 2, 3 or 4 substituents selected from R 14 .
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 13 is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 14 is independently selected from Ci_7-alkyl and heterocycloalkyl or two R 14 together form Ci_7-alkylene.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 14 is independently selected from methyl, ethyl and pyrrolidinyl or two R 14 together form ethylene.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is a saturated mono- or bicyclic N-heterocycloalkyl comprising 1 or 2 nitrogen atoms and is optionally substituted with 1, 2, 3 or 4 substituents selected from R 14 .
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein the N-heterocycloalkyl in A or the heterocycloalkyl in R 12 as defined herein are substituted with 1 or 2 substituents selected from R 14 .
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein the N-heterocycloalkyl in A as defined herein is further characterized in that one ring nitrogen atoms is basic.
  • a particular embodiment of the present invention relates to compounds of formula (I),
  • X is N or CH
  • R 4 is hydrogen, Ci_ 7 -alkyl or -(CH 2 ) m -NR 9 R 10 ;
  • R 5 is hydrogen or Ci_ 7 -alkyl
  • R 6 is hydrogen or Ci_ 7 -alkyl
  • R 7 is hydrogen or Ci_ 7 -alkyl
  • R 8 is hydrogen or Ci_ 7 -alkyl
  • R 9 and R 10 are independently selected from hydrogen, Ci_ 7 -alkyl and C 3 _8-cycloalkyl;
  • R is hydrogen, Ci_ 7 -alkyl or C 3 _8-cycloalkyl; n is 0, 1 or 2; m is 0, 1, 2 or 3; or R 4 and R 5 together form a Ci_ 7 -alkylene; or R 4 and R 7 together form a Ci_ 7 -alkylene; or R 5 and R 6 together form a C 2 -7-alkylene;
  • R 7 and R 8 together form a C 2 _7-alkylene
  • R 9 and R 10 together form a C 2 _7-alkylene
  • a particular embodiment of the present invention relates to compounds of formula (I),
  • X is N or CH
  • R 4 is hydrogen, Ci_ 7 -alkyl or -(CH 2 ) m -NR 9 R 10 ;
  • R 5 is hydrogen or Ci-7-alkyl
  • R 6 is hydrogen or Ci-7-alkyl
  • R is hydrogen or Ci-7-alkyl
  • R is hydrogen or Ci-7-alkyl
  • R 9 and R 10 are independently selected from hydrogen, Ci-7-alkyl and C3_8-cycloalkyl; n is 0, 1 or 2;
  • n 0, 1, 2 or 3;
  • R 4 and R 7 together form Ci-7-alkylene; or R 5 and R 6 together form C 2 -7-alkylene; or R 5 and R 7 together form Ci_7-alkylene; or R 5 and R 9 together form Ci_7-alkylene; or R 7 and R 8 together form C 2 _7-alkylene; or R 7 and R 9 together form Ci_7-alkylene; or R 9 and R 10 together form C 2 _7-alkylene; with the proviso that if X is CH then R 4 is -(CH 2 ) m -NR 9 R 10 ; and with the proviso that if X is N and R 4 is -(CH 2 ) m -NR 9 R 10 then m is 2 or 3.
  • At least one of R 4 , R 5 , R 6 , R 7 and R 8 is other than hydrogen.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein X is N.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein n is i.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 4 is hydrogen, methyl or -(CH 2 ) m -NR 9 R 10 , more particularly hydrogen.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 5 is hydrogen, methyl or ethyl, more particularly methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 6 is hydrogen or methyl, more particularly hydrogen.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein m is 0.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 4 and R 5 together form propylene.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 5 and R 6 together form ethylene;
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein R 9 and R 10 together form butylene.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
  • R 4 , R 5 , R 6 , R 7 , R 8 and R 13 are as defined herein and wherein R 11 is hydrogen or C 1-7 - alkyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
  • R 4 , R 5 , R 6 , R 7 and R 8 are as defined herein and wherein R 11 is hydrogen or Ci-7-alkyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of piperazinyl, diazepanyl, pyrrolidinyl and
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of piperazin-l-yl, 1,4-diazepan-l-yl, pyrrolidin-l-yl and hexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl, each optionally substituted with 1 or 2 substituents selected from R 14 as defined herein.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is NR 12 R 13 , wherein R 12 and R 13 are as described herein.
  • a parti present invention relates to compounds of formula (I),
  • A is , wherein R 4 , R 5 , R 6 , R 7 , R 8 and R 13 are as described herein.
  • a particu e present invention relates to compounds of formula (I),
  • A is , wherein R 13 is hydrogen or methyl.
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
  • a particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
  • a particular embodiment of the present invention relates to a compound of formula (I), wherein R 1 is methyl, R2 is hydrogen or methyl, R 3 is hydrogen, and A is
  • Particular compounds of formula (I) of the present invention are those selected from the group consisting of:
  • Another embodiment of the invention relates to compounds of formula (VI)
  • R 1 , R2 and R 3 are as described herein;
  • Y is halogen or trifluoromethanesulfonate; and salts thereof.
  • a particular embodiment of the present invention relates to compounds of formula (VI), wherein Y is fluoro, chloro, bromo, iodo or trifluoromethanesulfonate, particularly fluoro.
  • Particular compounds of formula (VI) of the present invention are those selected from the group consisting of:
  • the commercially available amino-pyridine of formula (II) can be reacted with a malonic ester to afford the intermediate of formula (III), wherein Y and R are as described herein and R is Ci_ 2 -alkyl, particularly methyl.
  • the compound of formula (III) is then treated with a chlorinating reagent (such as POCl 3 and the like) to provide a compound of formula (IV).
  • the compound of formula (IV) is then reacted in a Suzuki cross-coupling reaction with a compound of formula (V), wherein R 1 and R 2 are as described herein and Z is B(OH) 2 or an Ci_7-alkyl boronic acid ester such as 4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl, in the presence of a catalyst (such as (l, r-bis(diphenylphosphino)ferrocene)palladium(II) dichloride (Pd(dppf)Cl 2 ) and the like) and a base (such as K 2 C0 3 and the like) in a suitable solvent (such as DMF and the like), to afford the compound of formula (VI).
  • a catalyst such as (l, r-bis(diphenylphosphino)ferrocene)palladium(II) dichloride (Pd(dppf)Cl 2 ) and the like
  • a base such as K 2
  • a Buchwald-Hartwig amination reaction in the presence of a palladium catalyst (e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ) or
  • a palladium catalyst e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ) or
  • DMF dimethylformamide
  • the invention relates to a process for the manufacture of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, comprising the reaction of a compound of formula (VI) with a compound M-A either in: a) an aromatic nucleophilic substitution reaction (particularly if Y is fluoro) by
  • a Buchwald-Hartwig amination reaction in the presence of a palladium catalyst (e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ) or
  • a palladium catalyst e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh 3 ) 4 ) or
  • DMF dimethylformamide
  • a particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, comprising an aromatic nucleophilic substitution reaction between a compound of formula (VI) as described above with a compound of formula M-A by heating in a solvent, wherein A, R 1 , R2 , R 3 and Y are as defined above, M is hydrogen, sodium or potassium, and wherein M is linked to A via a nitrogen atom of A.
  • a particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein the aromatic nucleophilic substitution reaction is performed at a temperature from 80°C to 200°C.
  • a particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein the solvent of the aromatic nucleophilic substitution reaction is selected from dimethyl sulfoxide (DMSO), N-methylpyrrolidone (NMP), and dimethylformamide (DMF).
  • DMSO dimethyl sulfoxide
  • NMP N-methylpyrrolidone
  • DMF dimethylformamide
  • a particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein M is hydrogen.
  • compositions or medicaments comprising the compounds of the invention and a therapeutically inert carrier, diluent or pharmaceutically acceptable excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments.
  • Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the compounds of the present invention may be administered in any convenient
  • compositions may comprise
  • components conventional in pharmaceutical preparations e.g., diluents, carriers, pH modifiers, preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents, antioxidants, and further active agents. They can also comprise still other therapeutically valuable substances.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel H.C. et ah, Ansel's Pharmaceutical Dosage Forms and
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • the dosage at which compounds of the invention can be administered can vary within wide limits and will, of course, be fitted to the individual requirements in each particular case.
  • a daily dosage of about 0.01 to 1000 mg per person of a compound of general formula (I) should be appropriate, although the above upper limit can also be exceeded when necessary.
  • An example of a suitable oral dosage form is a tablet comprising about 100 mg to 500 mg of the compound of the invention compounded with about 30 to 90 mg anhydrous lactose, about 5 to 40 mg sodium croscarmellose, about 5 to 30 mg polyvinylpyrrolidone (PVP) K30, and about 1 to 10 mg magnesium stearate.
  • the powdered ingredients are first mixed together and then mixed with a solution of the PVP.
  • the resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment.
  • An example of an aerosol formulation can be prepared by dissolving the compound, for example 10 to 100 mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such as sodium chloride, if desired.
  • a suitable buffer solution e.g. a phosphate buffer
  • a tonicifier e.g. a salt such as sodium chloride
  • the solution may be filtered, e.g., using a 0.2 ⁇ filter, to remove impurities and contaminants.
  • the compounds of formula (I) and their pharmaceutically acceptable salts possess valuable pharmacological properties and have been found to enhance inclusion of exon 7 of SMNl and/or SMN2 into mRNA transcribed from the SMNl and/or SMN2 gene, thereby increasing expression of SMN protein in a human subject in need thereof.
  • the compounds of the present invention can be used, either alone or in combination with other drugs, for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function.
  • diseases include, but are not limited to spinal muscular atrophy (SMA).
  • a particular embodiment of the present invention relates to pharmaceutical compositions comprising compounds of formula (I) as defined above or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients.
  • a particular embodiment of the present invention relates to pharmaceutical compositions comprising compounds of formula (I) or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of SMA.
  • a particular embodiment of the present invention relates to compounds of formula (I) or their pharmaceutically acceptable salts as defined above for use as therapeutically active substances, especially for use as therapeutically active substances for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of spinal muscular atrophy (SMA).
  • SMA spinal muscular atrophy
  • a particular embodiment of the present invention relates to compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the use in the treatment or
  • SMA spinal muscular atrophy
  • a particular embodiment of the present invention relates to a method for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or
  • SMA spinal muscular atrophy
  • a particular embodiment of the present invention relates to the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or
  • SMA spinal muscular atrophy
  • a particular embodiment of the present invention relates to the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the preparation of medicaments for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of spinal muscular atrophy (SMA).
  • SMA spinal muscular atrophy
  • medicaments comprise compounds of formula (I) or their pharmaceutically acceptable salts as defined above.
  • ACN Acetonitrile
  • CH 2 C1 2 dichloromethane (DCM)
  • DIPEA diisopropyl ethylamine
  • DMA dimethyl acetamide
  • TEA triethylamine
  • RT room temperature
  • B 2 (pin) 2
  • Pd(dppf)Cl 2 (l,l'-Bis(diphenylphosphino)ferrocene)palladium(II) dichloride
  • PPTS Pyridinium p-toluenesulfonate.
  • Compounds of formula (I) enhance inclusion of exon 7 of SMN2 into mRNA transcribed from the SMN2 gene and increase levels of SMN protein produced from the SMN2 gene, and thus can be used to treat SMA in a human subject in need thereof.
  • These examples further illustrate the testing of certain compounds described herein in vitro and/or in vivo and demonstrate the usefulness of the compounds for enhancing the inclusion of exon 7 of SMNI into mRNA transcribed from the SMNl gene. Accordingly, compounds of formula (I) also enhance the inclusion of exon 7 of SMNl into mRNA transcribed from the SMNl gene and increase levels of SMN protein produced from the SMNl gene.
  • RT-qPCR reverse transcription-quantitative PCR-based assay
  • FL SMN2mini full length SMN2 minigene
  • FL SMN2mini full length SMN2 minigene
  • Materials used and respective sources are listed below in Table 1.
  • the SMN2-A minigene construct was prepared as described in International Patent
  • HEK293H cells stably transfected with the SMN2-A minigene construct (10,000 cells/well) are seeded in 200 of cell culture medium (DMEM plus 10% FBS, with 200 ⁇ g/mL
  • test compounds are serially diluted 3.16-fold in 100% DMSO to generate a 7-point concentration curve.
  • a solution of test compound (1 ⁇ , 200x in DMSO) is added to each cell- containing well and the plate is incubated for 24 hours in a cell culture incubator (37 °C, 5% C0 2 , 100% relative humidity). 2 replicates are prepared for each test compound concentration.
  • the cells are then lysed in the Cells-To-Ct lysis buffer and the lysate is stored at -80°C.
  • SMN2-A minigene and GAPDH mRNA are quantified using the primers and probes referenced in Table 2.
  • Primer SMN Forward A (SEQ ID NO. l) hybridizes to a nucleotide sequence in exon 7 (nucleotide 22 to nucleotide 40)
  • primer SMN Reverse A (SEQ ID NO.2) hybridizes to a nucleotide sequence in the coding sequence of Firefly luciferase
  • SMN Probe A (SEQ ID NO.3) hybridizes to a nucleotide sequence in exon 7 (nucleotide 50 to nucleotide 54) and exon 8 (nucleotide 1 to nucleotide 21).
  • the combination of these three oligonucleotides detects only SMN1 or SMN2 minigenes (RT-qPCR) and will not detect endogenous SMN1 or SMN2 genes.
  • SEQ ID NO. 2 TCTTTATGTTTTTGGCGTCTTC PTC 1
  • SEQ ID NO. 3 6FAM- AAGGAGAAATGCTGGCAT PTC 1
  • TAMRA hGAPDH Forward Primer SEQ ID NO. 5 CAACGGATTTGGTCGTATTGG LTI 2
  • hGAPDH Reverse Primer SEQ ID NO. 6 TGATGGCAACAATATCCACTTTACC LTI 2
  • the SMN forward and reverse primers are used at final concentrations of 0.4 ⁇ .
  • the SMN probe is used at a final concentration of 0.15 ⁇ .
  • the GAPDH primers are used at final concentrations of 0.2 ⁇ and the probe at 0.15 ⁇ .
  • the SMN2-minigene GAPDH mix (15 ⁇ ⁇ total volume) is prepared by combining 7.5 ⁇ ⁇ of 2x RT-PCR buffer, 0.4 ⁇ , of 25x RT-PCR enzyme mix, 0.75 ⁇ , of 20x GAPDH primer- probe mix, 4.0075 ⁇ , of water, 2 ⁇ , of 10-fold diluted cell lysate, 0.06 ⁇ , of 100 ⁇ SMN forward primer, 0.06 ⁇ , of 100 ⁇ SMN reverse primer, and 0.225 ⁇ , of 100 ⁇ SMN probe.
  • PCR is carried out at the following temperatures for the indicated time: Step 1: 48°C (15 min); Step 2: 95°C (10 min); Step 3: 95°C (15 sec); Step 4: 60°C (1 min); then repeat Steps 3 and 4 for a total of 40 cycles.
  • Each reaction mixture contains both SMN2-A minigene and GAPDH primers/probe sets (multiplex design), allowing simultaneous measurement of the levels of two transcripts.
  • the increase in the abundance of the FL SMN2mini mRNA relative to that in cells treated with vehicle control is determined from real-time PCR data using a modified AACt method (as described in Livak and Schmittgen, Methods, 2001, 25:402-8).
  • the amplification efficiency E is calculated from the slope of the amplification curve for FL SMN2mini and GAPDH individually.
  • the abundance of FL SMN2mini and GAPDH mRNA are then calculated as (1 + E) "Ct , where Ct is the threshold value for each amplicon.
  • the abundance of FL SMN2mini mRNA is normalized to GAPDH mRNA abundance.
  • the normalized FL SMN2mini mRNA abundance from test compound-treated samples is then divided by normalized FL SMN2mini mRNA abundance from vehicle-treated cells to determine the level of FL SMN2mini mRNA relative to vehicle control.
  • Table 3 provides ECi.5 X concentrations for production of full length SMN2 minigene mRNA that was obtained from the 7-point concentration data generated according to the above procedure for particular compounds of the present invention.
  • Particular compounds of the present invention exhibit an ECi.5 X concentration for production of full length SMN2 minigene mRNA ⁇ 1 ⁇ .
  • More particular compounds of the present invention exhibit an ECi.5 X concentration for production of full length SMN2 minigene mRNA ⁇ 0.1 ⁇ .
  • Most particular compounds of the present invention exhibit an EC1.5x concentration for production of full length SMN2 minigene mRNA ⁇ 0.02 ⁇ .
  • the SMN HTRF (homogeneous time resolved fluorescence) assay is used to quantify the level of SMN protein in SMA patient fibroblast cells treated with test compounds. Materials used and respective sources are listed below in Table 4.
  • Cells are thawed and cultured in DMEM- 10% FBS for 72 hours. Cells are trypsinized, counted and re-suspended to a concentration of 25,000 cells/mL in DMEM-10% FBS. The cell suspensions are plated at 5,000 cells per well in a 96 well microtiter plate and incubated for 3 to 5 hours. Test compounds are serially diluted 3.16-fold in 100% DMSO to generate a 7-point concentration curve. 1 ⁇ L ⁇ of test compound solution is transferred to cell-containing wells and cells are incubated for 48 hours in a cell culture incubator (37°C, 5% C0 2 , 100% relative humidity). Triplicate samples are set up for each test compound concentration.
  • the supernatant is removed from the wells and 25 ⁇ ⁇ of the RIPA lysis buffer, containing protease inhibitors, is added to the wells and incubated with shaking at room temperature for 1 hour. 25 ⁇ ⁇ of the diluent is added and then 35 ⁇ ⁇ of the resulting lysate is transferred to a 384- well plate, where each well contains 5 ⁇ ⁇ of the antibody solution (1 : 100 dilution of anti-SMN d2 and anti-SMN kryptate in SMN reconstitution buffer). The plate is centrifuged for 1 minute to bring the solution to the bottom of the wells, then incubated overnight at room temperature. Fluorescence for each well of the plate at 665 nm and 620 nm is measured on an En Vision multilabel plate reader (Perkin-Elmer).
  • the normalized fluorescence signal is calculated for each sample, Blank and vehicle control well by dividing the signal at 665 nm by the signal at 620 nm. Normalizing the signal accounts for possible fluorescence quenching due to the matrix effect of the lysate.
  • the AF value (a measurement of SMN protein abundance as a percent value) for each sample well is calculated by subtracting the normalized average fluorescence for the Blank control wells from the normalized fluorescence for each sample well, then dividing this difference by the
  • the AF value for each sample well represents the SMN protein abundance from test compound-treated samples.
  • the AF value for each sample well is divided by the AF value for the vehicle control wells to calculate the fold increase in SMN protein abundance relative to the vehicle control.
  • Table 5 provides ECi.5 X concentrations for SMN protein expression that was obtained from the 7-point concentration data generated according to the above procedure for particular compounds of the present invention.
  • Particular compounds of the present invention exhibit an ECi.5 X concentration for SMN protein expression ⁇ 1 ⁇ .
  • More particular compounds of the present invention exhibit an ECi.5 X concentration for SMN protein expression ⁇ 100 nM.
  • Particular compounds of the present invention exhibit a maximum fold increase > 1.5. More particular compounds of the present invention exhibit a maximum fold increase > 1.7. Most particular compounds of the present invention exhibit a maximum fold increase > 1.8.

Abstract

The present invention provides compounds of formula (I) wherein A, R1, R2 and R3 are as described herein, as well as pharmaceutically acceptable salts thereof. Further the present invention is concerned with the manufacture of the compounds of formula (I), pharmaceutical compositions comprising them and their use as medicaments.

Description

Compounds for treating spinal muscular atrophy
Introduction
The present invention provides compounds which are SMN2 gene splicing modulators, their manufacture, pharmaceutical compositions comprising them and their use as medicaments for the treatment of spinal muscular atrophy (SMA).
In particular, the present invention relates to compounds of formula (I)
Figure imgf000002_0001
wherein A, R 1 , R2 and R 3 are as described herein, and pharmaceutically acceptable salts thereof.
Background Spinal muscular atrophy (SMA), in its broadest sense, describes a collection of inherited and acquired central nervous system (CNS) diseases characterized by progressive motor neuron loss in the spinal cord and brainstem causing muscle weakness and muscle atrophy. The most common form of SMA is caused by mutations in the Survival Motor Neuron (SMN) gene and manifests over a wide range of severity affecting infants through adults {Crawford and Pardo, Neurobiol. Dis., 1996, 3:97).
Infantile SMA is the most severe form of this neurodegenerative disorder. Symptoms include muscle weakness, poor muscle tone, weak cry, limpness or a tendency to flop, difficulty sucking or swallowing, accumulation of secretions in the lungs or throat, feeding difficulties, and increased susceptibility to respiratory tract infections. The legs tend to be weaker than the arms and developmental milestones, such as lifting the head or sitting up, cannot be reached. In general, the earlier the symptoms appear, the shorter the lifespan. As the motor neuron cells deteriorate, symptoms appear shortly afterward. The severe forms of the disease are fatal and all forms have no known cure. The course of SMA is directly related to the rate of motor neuron cell deterioration and the resulting severity of weakness. Infants with a severe form of SMA frequently succumb to respiratory disease due to weakness in the muscles that support breathing. Children with milder forms of SMA live much longer, although they may need extensive medical support, especially those at the more severe end of the spectrum. The clinical spectrum of SMA disorders has been divided into the following five groups.
(a) Type 0 SMA (In Utero SMA) is the most severe form of the disease and begins before birth. Usually, the first symptom of Type 0 SMA is reduced movement of the fetus that can first be observed between 30 and 36 weeks of pregnancy. After birth, these newborns have little movement and have difficulties with swallowing and breathing.
(b) Type 1 SMA (Infantile SMA or Werdnig-Hoffmann disease) presents symptoms between 0 and 6 months, form of SMA is also very severe. Patients never achieve the ability to sit, and death usually occurs within the first 2 years without ventilatory support.
(c) Type 2 SMA (Intermediate SMA) has an age of onset at 7-18 months. Patients achieve the ability to sit unsupported, but never stand or walk unaided. Prognosis in this group is largely dependent on the degree of respiratory involvement.
(d) Type 3 SMA (Juvenile SMA or Kugelberg-Welander disease) is generally
diagnosed after 18 months. Type 3 SMA individuals are able to walk independently at some point during their disease course but often become wheelchair-bound during youth or adulthood.
(e) Type 4 SMA (Adult onset SMA). Weakness usually begins in late adolescence in the tongue, hands, or feet, then progresses to other areas of the body. The course of adult SMA is much slower and has little or no impact on life expectancy.
The SMN gene has been mapped by linkage analysis to a complex region in chromosome 5q. In humans, this region contains an approximately 500 thousand base pairs (kb) inverted duplication resulting in two nearly identical copies of the SMN gene. SMA is caused by an inactivating mutation or deletion of the telomeric copy of the gene (SMN1) in both
chromosomes, resulting in the loss of SMN1 gene function. However, all patients retain the centromeric copy of the gene (SMN2), and the copy number of the SMN2 gene in SMA patients generally correlates inversely with the disease severity; i.e., patients with less severe SMA have more copies of SMN2. Nevertheless, SMN2 is unable to compensate completely for the loss of SMN1 function due to alternative splicing of exon 7 caused by a translationally silent C to T mutation in exon 7. As a result, the majority of transcripts produced from SMN2 lack exon 7 (Δ7 SMN2), and encode a truncated SMN protein that has an impaired function and is rapidly degraded. The SMN protein is thought to play a role in RNA processing and metabolism, having a well characterized function of mediating the assembly of a specific class of RNA-protein complexes termed snRNPs. SMN may have other functions in motor neurons, however its role in preventing the selective degeneration of motor neurons is not well established. In most cases, SMA is diagnosed based on clinical symptoms and by the presence of at least one copy of the SMN1 gene test. However, in approximately 5% of cases SMA is caused by mutation in genes other than the inactivation of SMN 1, some known and others not yet defined. In some cases, when the SMN 1 gene test is not feasible or does not show any abnormality, other tests such as an electromyography (EMG) or muscle biopsy may be indicated. Medical care for SMA patients at present is limited to supportive therapy including respiratory, nutritional and rehabilitation care; there is no drug known to address the underlying cause of the disease. Current treatment for SMA consists of prevention and management of the secondary effects of chronic motor unit loss. The major management issue in Type 1 SMA is the prevention and early treatment of pulmonary problems, which are the cause of death in the majority of the cases. While some infants afflicted with SMA grow to be adults, those with Type 1 SMA have a life expectancy of less than two years.
Several mouse models of SMA have been developed. In particular, the SMN delta exon 7 (Δ7 SMN) model (Le et al, Hum. Mol. Genet., 2005, 14:845) carries both the SMN2 gene and several copies of the Δ7 SMN2 cDNA and recapitulates many of the phenotypic features of Type 1 SMA. The Δ7 SMN model can be used for both SMN2 expression studies as well as the evaluation of motor function and survival. The C/C-allele mouse model {Jackson Laboratory strain #008714, The Jackson Laboratory, Bar Harbor, ME) provides a less severe SMA disease model, with mice having reduced levels of both SMN2 full length (FL SMN2) mRNA and SMN protein. The C/C-allele mouse phenotype has the SMN2 gene and a hybrid mSMNl-SMN2 gene that undergoes alternative splicing, but does not have overt muscle weakness. The C/C-allele mouse model is used for SMN2 expression studies.
As a result of improved understanding of the genetic basis and pathophysiology of SMA, several strategies for treatment have been explored, but none have yet demonstrated success in the clinic. Gene replacement of SMN1, using viral delivery vectors, and cell replacement, using differentiated SMN1+/+ stem cells, have demonstrated efficacy in animal models of SMA. More research is needed to determine the safety and immune response and to address the requirement for the initiation of treatment at the neonatal stage before these approaches can be applied to humans. Correction of alternative splicing of SMN2 in cultured cells has also been achieved using synthetic nucleic acids as therapeutic agents: (i) antisense oligonucleotides that target sequence elements in SMN2 pre-mRNA and shift the outcome of the splicing reaction toward the generation of full length SMN2 mRNA (Passini et ah, Sci. Transl. Med., 2011, 3:72ral8; and, Hua et ah, Nature, 2011, 478:123) and (ii) trans-splicing RNA molecules that provide a fully functional RNA sequence that replace the mutant fragment during splicing and generate a full length SMN1 mRNA (Coady and Lorson, J NeuroscL, 2010, 30:126).
Other approaches under exploration include searching for drugs that increase SMN levels, enhance residual SMN function, or compensate for its loss. Aminoglycosides have been shown to enhance expression of a stabilized SMN protein produced from Δ7 SMN2 mRNA by promoting the translational read-through of the aberrant stop codon, but have poor central nervous system penetration and are toxic after repeat dosing. Chemotherapeutic agents, such as aclarubicin, have been shown to increase SMN protein in cell culture; however, the toxicity profile of these drugs prohibits long-term use in SMA patients. Some drugs under clinical investigation for the treatment of SMA include transcription activators such as histone
deacetylase ("HDAC") inhibitors (e.g., butyrates, valproic acid, and hydroxyurea), and mRNA stabilizers (mRNA decapping inhibitor RG3039 from Repligen), the goal being to increase the amount of total RNA transcribed from the SMN2 gene. However, the use of the HDAC inhibitors or mRNA stabilizers does not address the underlying cause of SMA and may result in a global increase in transcription and gene expression with potential safety problems in humans.
In an alternative approach, neuroprotective agents such as Olesoxime have been chosen for investigation. Such strategies are not aimed at SMN for the treatment of SMA, but instead are being explored to protect the SMN-deficient motor neurons from neurodegeneration.
A system designed for identifying compounds that increase the inclusion of exon 7 of SMN into RNA transcribed from the SMN2 gene and certain benzooxazole and benzoisoxazole compounds identified thereby have been described in International Patent Application
WO2009/151546A1. A system designed for identifying compounds that cause ribosomal frameshifting to produce a stabilized SMN protein from Δ7 SMN2 mRNA and certain
isoindolinone compounds identified thereby have been described in International Patent
Applications 02010/019236 Al and WO2013/119916A2.
Despite the progress made in understanding the genetic basis and pathophysiology of SMA, there remains a need to identify compounds that alter the course of spinal muscular atrophy, one of the most devastating childhood neurological diseases. Detailed description of the invention
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, suitable methods and materials are described below.
All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
The nomenclature used in this Application is based on IUPAC systematic nomenclature, unless indicated otherwise.
Any open valency appearing on a carbon, oxygen, sulfur or nitrogen atom in the structures herein indicates the presence of a hydrogen, unless indicated otherwise.
The definitions described herein apply irrespective of whether the terms in question appear alone or in combination. It is contemplated that the definitions described herein can be appended to form chemically-relevant combinations, such as e.g. "heterocycloalkylaryl",
"haloalkylheteroaryl", "arylalkylheterocycloalkyl", or "alkoxyalkyl". The last member of the combination is the radical which is binding to the rest of the molecule. The other members of the combination are attached to the binding radical in reversed order in respect of the literal sequence, e.g. the combination amino-Ci_7-alkyl refers to a Ci_7-alkyl which is substituted by amino, or e.g. the combination arylalkylheterocycloalkyl refers to a heterocycloalkyl-radical which is substituted by an alkyl which is substituted by an aryl.
The term "moiety" refers to an atom or group of chemically bonded atoms that is attached to another atom or molecule by one or more chemical bonds thereby forming part of a molecule.
For example, the variables A, R 1 , R2 and R 3 of formula (I) refer to moieties that are attached to the core structure of formula (I) by a covalent bond.
When indicating the number of substituents, the term "one or more" refers to the range from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.
The term "optional" or "optionally" denotes that a subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. The term "substituent" denotes an atom or a group of atoms replacing a hydrogen atom on the parent molecule.
The term "substituted" denotes that a specified group bears one or more substituents. Where any group can carry multiple substituents and a variety of possible substituents is provided, the substituents are independently selected and need not to be the same. The term "unsubstituted" means that the specified group bears no substituents. The term "optionally substituted" means that the specified group is unsubstituted or substituted by one or more substituents, independently chosen from the group of possible substituents. When indicating the number of substituents, the term "one or more" means from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.
The terms "compound(s) of this invention" and "compound(s) of the present invention" refer to compounds as disclosed herein and stereoisomers, tautomers, solvates, and salts (e.g., pharmaceutically acceptable salts) thereof.
When the compounds of the invention are solids, it is understood by those skilled in the art that these compounds, and their solvates and salts, may exist in different solid forms, particularly different crystal forms, all of which are intended to be within the scope of the present invention and specified formulae.
The term "pharmaceutically acceptable salts" denotes salts which are not biologically or otherwise undesirable. Pharmaceutically acceptable salts include both acid and base addition salts.
The term "pharmaceutically acceptable acid addition salt" denotes those pharmaceutically acceptable salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and organic acids selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicyclic acid.
The term "pharmaceutically acceptable base addition salt" denotes those pharmaceutically acceptable salts formed with an organic or inorganic base. Examples of acceptable inorganic bases include sodium, potassium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts. Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, and polyamine resins.
Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al. Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511). The prefixes D and L or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or L designating that the compound is levorotatory. A compound prefixed with (+) or D is dextrorotatory.
The term "chiral center" denotes a carbon atom bonded to four nonidentical substituents. The term "chiral" denotes the ability of non-superimposability with the mirror image, while the term "achiral" refers to embodiments which are superimposable with their mirror image. Chiral molecules are optically active, i.e., they have the ability to rotate the plane of plane-polarized light.
Compounds of the present invention can have one or more chiral centers and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates. Whenever a chiral center is present in a chemical structure, it is intended that all stereoisomers associated with that chiral center are encompassed by the present invention.
The terms "halo", "halogen", and "halide" are used interchangeably herein and denote fluoro, chloro, bromo, or iodo. One particular example of halogen is fluoro.
The term "alkyl" denotes a monovalent linear or branched saturated hydrocarbon group of 1 to 12 carbon atoms. In particular embodiments, alkyl has 1 to 7 carbon atoms, and in more particular embodiments 1 to 4 carbon atoms. Examples of alkyl include methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, or tert-butyl. Particular examples for alkyl are methyl and ethyl.
The term "haloalkyl" denotes an alkyl group wherein at least one of the hydrogen atoms of the alkyl group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of haloalkyl include monofluoro-, difluoro- or trifluoro-methyl, -ethyl or -propyl, for example 3,3,3-trifluoropropyl, 2-fluoroethyl, 2,2,2-trifluoroethyl, fluoromethyl, or
trifluoromethyl and the like. The term "perhaloalkyl" denotes an alkyl group where all hydrogen atoms of the alkyl group have been replaced by the same or different halogen atoms. The term "bicyclic ring system" denotes two rings which are fused to each other via a common single or double bond (annelated bicyclic ring system), via a sequence of three or more common atoms (bridged bicyclic ring system) or via a common single atom (spiro bicyclic ring system). Bicyclic ring systems can be saturated, partially unsaturated, unsaturated or aromatic. Bicyclic ring systems can comprise heteroatoms selected from N, O and S. The term "cycloalkyl" denotes a saturated monocyclic or bicyclic hydrocarbon group of 3 to 10 ring carbon atoms. In particular embodiments cycloalkyl denotes a monovalent saturated monocyclic hydrocarbon group of 3 to 8 ring carbon atoms. Bicyclic means consisting of two saturated carbocycles having one or more carbon atoms in common. Particular cycloalkyl groups are monocyclic. Examples for monocyclic cycloalkyl are cyclopropyl, cyclobutanyl, cyclopentyl, cyclohexyl or cycloheptyl. Examples for bicyclic cycloalkyl are bicyclo[2.2.1]heptanyl, or bicyclo[2.2.2]octanyl. One particular example of cycloalkyl is cyclopropyl.
The term "heterocycloalkyl" denotes a saturated or partly unsaturated mono-, bi- or tricyclic ring system of 3 to 9 ring atoms, comprising 1, 2, or 3 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon. In particular embodiments, heterocycloalkyl is a monovalent saturated monocyclic ring system of 4 to 7 ring atoms, comprising 1, 2, or 3 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon. Examples for monocyclic saturated heterocycloalkyl are aziridinyl, oxiranyl, azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydro-thienyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperazinyl, morpholinyl, thiomorpholinyl, l,l-dioxo-thiomorpholin-4-yl, azepanyl, diazepanyl, homopiperazinyl, or oxazepanyl. Examples for bicyclic saturated heterocycloalkyl are 8-aza-bicyclo[3.2.1]octyl, quinuclidinyl, 8-oxa-3-aza-bicyclo[3.2.1]octyl, 9-aza-bicyclo[3.3.1]nonyl, 3-oxa-9-aza- bicyclo[3.3.1]nonyl, or 3-thia-9-aza-bicyclo[3.3.1]nonyl. Examples of a partly unsaturated heterocycloalkyl are dihydrofuryl, imidazolinyl, dihydro-oxazolyl, tetrahydro-pyridinyl, or dihydropyranyl. Particular examples of heterocycloalkyl are 1,4-diazepanyl,
hexahydropyrrolo[l,2-a]pyrazinyl, piperidinyl, piperazinyl and pyrrolidinyl. More particular examples of heterocycloalkyl are hexahydropyrrolo[l,2-a]pyrazinyl and piperazinyl.
The term "N -heterocycloalkyl" denotes a heterocycloalkyl radical containing at least one nitrogen ring atom and where the point of attachment of the heterocycloalkyl radical to the rest of the molecule is through a nitrogen ring atom. Particular examples of N-heterocycloalkyl are 1,4-diazepanyl, hexahydropyrrolo[l,2-a]pyrazinyl, piperidinyl, piperazinyl and pyrrolidinyl. More particular examples of N-heterocycloalkyl are hexahydropyrrolo[l,2-a]pyrazinyl and piperazinyl.
The term "basicity" in reference to a compound is expressed herein by the negative decadic logarithm of the acidity constant of the conjugate acid (pKa = -log Ka). The larger the pKa of the conjugate acid, the stronger the base (pKa + pKb = 14). In this application, an atom or functional group is denoted "basic" if it is suitable to accept a proton and if the calculated pKa of its conjugate acid is at least 7, more particularly if the calculated pKa of its conjugate acid is at least 7.8, most particularly if the calculated pKa of its conjugate acid is at least 8. pKa values were calculated in-silico as described in F. Milletti et al., J. Chem. Inf. Model (2007) 47:2172-2181.
The term "alkylene" denotes a linear saturated divalent hydrocarbon group of 1 to 7 carbon atoms or a divalent branched saturated hydrocarbon group of 3 to 7 carbon atoms. Examples of alkylene groups include methylene, ethylene, propylene, 2-methylpropylene, butylene, 2- ethylbutylene, pentylene, hexylene. Particular examples for alkylene are ethylene, propylene, and butylene.
The term "amino" denotes a group of the formula -NR'R" wherein R' and R" are independently hydrogen, alkyl, alkoxy, cycloalkyl, heterocycloalkyl, aryl, heteroaryl or as described herein. Alternatively, R' and R", together with the nitrogen to which they are attached, can form a heterocycloalkyl. The term "primary amino" denotes a group wherein both R' and R" are hydrogen. The term "secondary amino" denotes a group wherein R' is hydrogen and R" is a group other than hydrogen. The term "tertiary amino" denotes a group wherein both R' and R" are other than hydrogen. Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine. The term "active pharmaceutical ingredient" (or "API") denotes the compound or molecule in a pharmaceutical composition that has a particular biological activity.
The terms "pharmaceutical composition" and "pharmaceutical formulation" (or
"formulation") are used interchangeably and denote a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with
pharmaceutically acceptable excipients to be administered to a mammal, e.g., a human in need thereof.
The term "pharmaceutically acceptable" denotes an attribute of a material which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and neither biologically nor otherwise undesirable and is acceptable for veterinary as well as human pharmaceutical use.
The terms "pharmaceutically acceptable excipient", "pharmaceutically acceptable carrier" and "therapeutically inert excipient" can be used interchangeably and denote any
pharmaceutically acceptable ingredient in a pharmaceutical composition having no therapeutic activity and being non-toxic to the subject administered, such as disintegrators, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, carriers, diluents or lubricants used in formulating pharmaceutical products.
The terms "individual" or "subject" refer to a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human.
The term "therapeutically effective amount" denotes an amount of a compound or molecule of the present invention that, when administered to a subject, (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein. The therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease treated, the age and relative health of the subject, the route and form of administration, the judgement of the attending medical or veterinary practitioner, and other factors.
The terms "treating" or "treatment" of a disease state include inhibiting the disease state, i.e., arresting the development of the disease state or its clinical symptoms, or relieving the disease state, i.e., causing temporary or permanent regression of the disease state or its clinical symptoms.
The term "spinal muscular atrophy" (or SMA) relates to a disease caused by an
inactivating mutation or deletion in the SMN1 gene on both chromosomes, resulting in a loss of SMN1 gene function.
Symptoms of SMA include muscle weakness, poor muscle tone, weak cry, weak cough, limpness or a tendency to flop, difficulty sucking or swallowing, difficulty breathing, accumulation of secretions in the lungs or throat, clenched fists with sweaty hand,
flickering/vibrating of the tongue, head often tilted to one side, even when lying down, legs that tend to be weaker than the arms, legs frequently assuming a "frog legs" position, feeding difficulties, increased susceptibility to respiratory tract infections, bowel/bladder weakness, lower-than-normal weight, inability to sit without support, failure to walk, failure to crawl, and hypotonia, areflexia, and multiple congenital contractures (arthrogryposis) associated with loss of anterior horn cells.
The term "treating spinal muscular atrophy (SMA)" or "treatment of spinal muscular atrophy (SMA)" includes one or more of the following effects: (i) reduction or amelioration of the severity of SMA; (ii) delay of the onset of SMA; (iii) inhibition of the progression of SMA; (iv) reduction of hospitalization of a subject; (v) reduction of hospitalization length for a subject;
(vi) increase of the survival of a subject; (vii) improvement of the quality of life of a subject; (viii) reduction of the number of symptoms associated with SMA; (ix) reduction of or
amelioration of the severity of one or more symptoms associated with SMA; (x) reduction of the duration of a symptom associated with SMA; (xi) prevention of the recurrence of a symptom associated with SMA; (xii) inhibition of the development or onset of a symptom of SMA; and/or (xiii) inhibition of the progression of a symptom associated with SMA.
More particular, the term "treating SMA" denotes one or more of the following beneficial effects: (i) a reduction in the loss of muscle strength; (ii) an increase in muscle strength; (iii) a reduction in muscle atrophy; (iv) a reduction in the loss of motor function; (v) an increase in motor neurons;
(vii) a reduction in the loss of motor neurons; (viii) protection of SMN deficient motor neurons from degeneration; (ix) an increase in motor function; (x) an increase in pulmonary function; and/or (xi) a reduction in the loss of pulmonary function.
In further detail, the term "treating SMA" refers to the functional ability or retention of the functional ability for a human infant or a human toddler to sit up unaided or for a human infant, a human toddler, a human child or a human adult to stand up unaided, to walk unaided, to run unaided, to breathe unaided, to turn during sleep unaided, or to swallow unaided.
The term "ECi.5X concentration for production of full length SMN2 minigene mRNA" (or "ECi.5x minigene") is defined as that concentration of test compound that is effective in increasing the amount of full length SMN2 minigene mRNA to a level 1.5-fold greater relative to that in vehicle-treated cells.
The term "ECi.5X concentration for SMN protein expression" (or "ECi.5X SMN protein") is defined as that concentration of test compound that is effective in producing 1.5 times the amount of SMN protein in an SMA patient fibroblast cell compared to the amount produced from the vehicle control. In detail, the present invention relates to compounds of formula (I)
Figure imgf000013_0001
wherein
R1 is hydrogen or Ci_7-alkyl;
R is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C3_g-cycloalkyl;
R is hydrogen, Ci_7-alkyl, or C3_g-cycloalkyl;
A is N-heterocycloalkyl or NR 12 R 13 , wherein N-heterocycloalkyl comprises 1 or 2 nitrogen ring atoms and is optionally substituted with 1, 2, 3 or 4 substituents selected from R14;
R 12 is heterocycloalkyl comprising 1 nitrogen ring atom, wherein heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R14;
R 13 is hydrogen, Ci_7-alkyl or C3_g-cycloalkyl;
R14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C3_ 8-cycloalkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R14 substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof.
Particular embodiments of the present invention are compounds of formula (I) and armaceutically acceptable salts thereof.
Further, it is to be understood that every embodiment relating to a specific A, R 1 , R2 or R 3 disclosed herein may be combined with any other embodiment relating to another A, R 1 , R 2 or as disclosed herein. A particular embodiment of the present invention relates to compounds of formula (I) wherein
R1 is hydrogen or Ci_7-alkyl;
R is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C3_8-cycloalkyl;
R is hydrogen, Ci_7-alkyl, or C3_g-cycloalkyl;
A is N-heterocycloalkyl comprising 1 or 2 nitrogen ring atoms, wherein N- heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R14;
R14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C3_ 8-cycloalkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R14 substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R1 is Ci_7-alkyl, particularly methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R12 is piperidinyl optionally substituted with 1, 2, 3 or 4 substituents selected from R14.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R 13 is hydrogen or Ci_7-alkyl, particularly hydrogen or methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R14 is independently selected from Ci_7-alkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R14 is independently selected from methyl, ethyl and pyrrolidinyl or two R14 together form ethylene. A particular embodiment of the present invention relates to compounds of formula (I), wherein A is a saturated mono- or bicyclic N-heterocycloalkyl comprising 1 or 2 nitrogen atoms and is optionally substituted with 1, 2, 3 or 4 substituents selected from R14.
A particular embodiment of the present invention relates to compounds of formula (I), wherein the N-heterocycloalkyl in A or the heterocycloalkyl in R 12 as defined herein are substituted with 1 or 2 substituents selected from R14.
A particular embodiment of the present invention relates to compounds of formula (I), wherein the N-heterocycloalkyl in A as defined herein is further characterized in that one ring nitrogen atoms is basic.
A particular embodiment of the present invention relates to compounds of formula (I),
wherein A is
Figure imgf000015_0001
wherein
X is N or CH;
R4 is hydrogen, Ci_7-alkyl or -(CH2)m-NR9R10;
R5 is hydrogen or Ci_7-alkyl;
R6 is hydrogen or Ci_7-alkyl;
R7 is hydrogen or Ci_7-alkyl;
R8 is hydrogen or Ci_7-alkyl;
R9 and R10 are independently selected from hydrogen, Ci_7-alkyl and C3_8-cycloalkyl;
R is hydrogen, Ci_7-alkyl or C3_8-cycloalkyl; n is 0, 1 or 2; m is 0, 1, 2 or 3; or R4 and R5 together form a Ci_7-alkylene; or R4 and R7 together form a Ci_7-alkylene; or R5 and R6 together form a C2-7-alkylene;
or R 5 and R 7 together form a Ci_7-alkylene;
or R5 and R9 together form a Ci_7-alkylene;
or R 7 and R 8 together form a C2_7-alkylene;
or R 7 and R 9 together form a Ci_7-alkylene;
or R9 and R10 together form a C2_7-alkylene;
with the proviso that if X is CH then R4 is -(CH2)m-NR9R10; and
with the proviso that if X is N and R4 is -(CH2)m-NR9R10 then m is 2 or 3.
A particular embodiment of the present invention relates to compounds of formula (I),
wherein A is
Figure imgf000016_0001
; wherein
X is N or CH;
R4 is hydrogen, Ci_7-alkyl or -(CH2)m-NR9R10;
R5 is hydrogen or Ci-7-alkyl;
R6 is hydrogen or Ci-7-alkyl;
R is hydrogen or Ci-7-alkyl;
R is hydrogen or Ci-7-alkyl;
R9 and R10 are independently selected from hydrogen, Ci-7-alkyl and C3_8-cycloalkyl; n is 0, 1 or 2;
m is 0, 1, 2 or 3;
or R4 and R5 together form Ci-7-alkylene;
or R4 and R7 together form Ci-7-alkylene; or R5 and R6 together form C2-7-alkylene; or R 5 and R 7 together form Ci_7-alkylene; or R5 and R9 together form Ci_7-alkylene; or R 7 and R 8 together form C2_7-alkylene; or R 7 and R 9 together form Ci_7-alkylene; or R9 and R10 together form C2_7-alkylene; with the proviso that if X is CH then R4 is -(CH2)m-NR9R10; and with the proviso that if X is N and R4 is -(CH2)m-NR9R10 then m is 2 or 3.
It has been found that brain penetration is improved when at least one of R4, R5, R6, R7 and R is not hydrogen.
In a particular embodiment of the invention at least one of R4, R5, R6, R7 and R8 is other than hydrogen.
A particular embodiment of the present invention relates to compounds of formula (I), wherein X is N. A particular embodiment of the present invention relates to compounds of formula (I), wherein n is i.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R4 is hydrogen, methyl or -(CH2)m-NR9R10, more particularly hydrogen.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R5 is hydrogen, methyl or ethyl, more particularly methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R6 is hydrogen or methyl, more particularly hydrogen.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen or methyl. A particular embodiment of the present invention relates to compounds of formula (I), wherein R is hydrogen. A particular embodiment of the present invention relates to compounds of formula (I), wherein m is 0.
A particular embodiment of the present invention relates to compounds of formula (I), wherein R4 and R5 together form propylene. A particular embodiment of the present invention relates to compounds of formula (I), wherein R5 and R6 together form ethylene;
A particular embodiment of the present invention relates to compounds of formula (I), wherein R9 and R10 together form butylene.
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
Figure imgf000018_0001
wherein R4, R5, R6, R7, R8 and R13 are as defined herein and wherein R11 is hydrogen or C1-7- alkyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
Figure imgf000019_0001
wherein R4, R5, R6, R7 and R8 are as defined herein and wherein R11 is hydrogen or Ci-7-alkyl. A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of piperazinyl, diazepanyl, pyrrolidinyl and
hexahydropyrrolo[l,2-a]pyrazinyl, each optionally substituted with 1, 2, 3 or 4 substituents selected from R14 as defined herein.
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of piperazin-l-yl, 1,4-diazepan-l-yl, pyrrolidin-l-yl and hexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl, each optionally substituted with 1 or 2 substituents selected from R14 as defined herein.
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is NR 12 R 13 , wherein R 12 and R 13 are as described herein. A parti present invention relates to compounds of formula (I),
wherein A is
Figure imgf000020_0001
, wherein R4, R5, R6, R7, R8 and R13 are as described herein.
A particu e present invention relates to compounds of formula (I),
wherein A is
Figure imgf000020_0002
, wherein R13 is hydrogen or methyl.
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
Figure imgf000020_0003
A particular embodiment of the present invention relates to compounds of formula (I), wherein A is selected from the group of:
Figure imgf000021_0001
A particular embodiment of the present invention relates to a compound of formula (I), wherein R 1 is methyl, R2 is hydrogen or methyl, R 3 is hydrogen, and A is
Figure imgf000021_0002
or Particular compounds of formula (I) of the present invention are those selected from the group consisting of:
2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-(4-methylpiperazin- l-yl)pyrido[ 1 ,2-a]pyrimidin-4- one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8-dimethylimidazo[l,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8-dimethylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one;
7-[(8aS)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5R)-3,5-dimethylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3S)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3R)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
7-( 1 ,4-diazepan- 1 -yl)-2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4- one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3R)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
7-( 1 ,4-diazepan- 1 -yl)-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one; 7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a] pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b] pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[ 1,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[ 1,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-pyrrolidin-l-ylpyrrolidin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin- l-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
7-(3,3-dimethylpiperazin- l-yl)-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3S)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3R)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-meth pyrido[l,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin-l-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]pyrido[l^ a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-pyrrolidin-l-ylpyrrolidin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2 a]pyrimidin-4-one;
9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l-yl]pyrid a]pyrimidin-4-one;
9-methyl-2-(2-methylimidazo[l,2-b]pyri^
a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(3,3-dimethylpiperazin- l-yl)-9-methyl-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R)-3-ethylpiperazin-l-yl]-2-(2-methylim
4-one;
and pharmaceutically acceptable salts thereof. Particular compounds of formula (I) of the present invention are those selected from the group consisting of:
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8-dimethylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8-dimethylimidazo[l,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5R)-3,5-dimethylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a] pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b] pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3S)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
and pharmaceutically acceptable salts thereof. Compounds of formula (VI) are suitable as intermediates in the manufacture of compounds of formula (I).
Another embodiment of the invention relates to compounds of formula (VI)
Figure imgf000025_0001
wherein R 1 , R2 and R 3 are as described herein;
Y is halogen or trifluoromethanesulfonate; and salts thereof.
A particular embodiment of the present invention relates to compounds of formula (VI), wherein Y is fluoro, chloro, bromo, iodo or trifluoromethanesulfonate, particularly fluoro. Particular compounds of formula (VI) of the present invention are those selected from the group consisting of:
7-fluoro-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-fluoro-pyrido[ 1 ,2-a]pyrimidin-4-one;
7-fluoro-9-methyl-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one; 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl-pyrido[l,2-a]pyrimidin-4-one; and salts thereof.
Manufacturing Processes
Compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above can be prepared following standard methods known in the art.
As illustrated in Scheme 1, the commercially available amino-pyridine of formula (II) can be reacted with a malonic ester to afford the intermediate of formula (III), wherein Y and R are as described herein and R is Ci_2-alkyl, particularly methyl. The compound of formula (III) is then treated with a chlorinating reagent (such as POCl3 and the like) to provide a compound of formula (IV). The compound of formula (IV) is then reacted in a Suzuki cross-coupling reaction with a compound of formula (V), wherein R 1 and R 2 are as described herein and Z is B(OH)2 or an Ci_7-alkyl boronic acid ester such as 4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl, in the presence of a catalyst (such as (l, r-bis(diphenylphosphino)ferrocene)palladium(II) dichloride (Pd(dppf)Cl2) and the like) and a base (such as K2C03 and the like) in a suitable solvent (such as DMF and the like), to afford the compound of formula (VI). Finally, the compound of formula (VI) is reacted with a compound M-A either in: a) an aromatic nucleophilic substitution reaction (particularly if Y is fluoro) by
heating at a temperature from 80°C to 200°C; or
b) a Buchwald-Hartwig amination reaction in the presence of a palladium catalyst (e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh3)4) or
bis(dibenzylideneacetone)palladium (Pd(dba)2) by heating at a temperature from 20°C to 100°C; in a solvent (e.g. dimethyl sulfoxide (DMSO), N-methylpyrrolidone (NMP), or
dimethylformamide (DMF)) to give a compound of formula (I), wherein A is as defined herein, M is hydrogen, sodium or potassium, particularly hydrogen, and wherein M is linked to A via a nitrogen atom of A.
Figure imgf000027_0001
Scheme 1.
In one embodiment, the invention relates to a process for the manufacture of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, comprising the reaction of a compound of formula (VI) with a compound M-A either in: a) an aromatic nucleophilic substitution reaction (particularly if Y is fluoro) by
heating at a temperature from 80°C to 200°C; or
b) a Buchwald-Hartwig amination reaction in the presence of a palladium catalyst (e.g. tetrakis(triphenylphosphine)palladium (Pd(PPh3)4) or
bis(dibenzylideneacetone)palladium Pd(dba)2) by heating at a temperature from 20°C to 100°C; in a solvent (e.g. dimethyl sulfoxide (DMSO), N-methylpyrrolidone (NMP), or
dimethylformamide (DMF)), wherein A, Y, R 1 , R2 and R 3 are as defined herein, M is hydrogen, sodium or potassium, particularly hydrogen, and wherein M is linked to A via a nitrogen atom of A.
Figure imgf000028_0001
A particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, comprising an aromatic nucleophilic substitution reaction between a compound of formula (VI) as described above with a compound of formula M-A by heating in a solvent, wherein A, R 1 , R2 , R 3 and Y are as defined above, M is hydrogen, sodium or potassium, and wherein M is linked to A via a nitrogen atom of A.
A particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein the aromatic nucleophilic substitution reaction is performed at a temperature from 80°C to 200°C.
A particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein the solvent of the aromatic nucleophilic substitution reaction is selected from dimethyl sulfoxide (DMSO), N-methylpyrrolidone (NMP), and dimethylformamide (DMF).
A particular embodiment of the invention relates to a process for the preparation of compounds of formula (I) and pharmaceutically acceptable salts thereof as defined above, wherein M is hydrogen.
Particularly, compounds of formula (I) and pharmaceutically acceptable salts thereof can be prepared in accordance to the methods described in the examples herein.
Pharmaceutical Compositions
Another embodiment provides pharmaceutical compositions or medicaments comprising the compounds of the invention and a therapeutically inert carrier, diluent or pharmaceutically acceptable excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments. Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
The compounds of the present invention may be administered in any convenient
administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions may comprise
components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents, antioxidants, and further active agents. They can also comprise still other therapeutically valuable substances.
A typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel H.C. et ah, Ansel's Pharmaceutical Dosage Forms and
Drug Delivery Systems (2004) Lippincott, Williams & Wilkins, Philadelphia; Gennaro A.R. et ah, Remington: The Science and Practice of Pharmacy (2000) Lippincott, Williams & Wilkins, Philadelphia; and Rowe R.C, Handbook of Pharmaceutical Excipients (2005) Pharmaceutical Press, Chicago. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). The dosage at which compounds of the invention can be administered can vary within wide limits and will, of course, be fitted to the individual requirements in each particular case. In general, in the case of oral administration a daily dosage of about 0.01 to 1000 mg per person of a compound of general formula (I) should be appropriate, although the above upper limit can also be exceeded when necessary. An example of a suitable oral dosage form is a tablet comprising about 100 mg to 500 mg of the compound of the invention compounded with about 30 to 90 mg anhydrous lactose, about 5 to 40 mg sodium croscarmellose, about 5 to 30 mg polyvinylpyrrolidone (PVP) K30, and about 1 to 10 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment.
An example of an aerosol formulation can be prepared by dissolving the compound, for example 10 to 100 mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such as sodium chloride, if desired. The solution may be filtered, e.g., using a 0.2 μιη filter, to remove impurities and contaminants.
Uses
As described above, the compounds of formula (I) and their pharmaceutically acceptable salts possess valuable pharmacological properties and have been found to enhance inclusion of exon 7 of SMNl and/or SMN2 into mRNA transcribed from the SMNl and/or SMN2 gene, thereby increasing expression of SMN protein in a human subject in need thereof.
The compounds of the present invention can be used, either alone or in combination with other drugs, for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function. These diseases include, but are not limited to spinal muscular atrophy (SMA).
A particular embodiment of the present invention relates to pharmaceutical compositions comprising compounds of formula (I) as defined above or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients. A particular embodiment of the present invention relates to pharmaceutical compositions comprising compounds of formula (I) or their pharmaceutically acceptable salts as defined above and one or more pharmaceutically acceptable excipients for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of SMA. A particular embodiment of the present invention relates to compounds of formula (I) or their pharmaceutically acceptable salts as defined above for use as therapeutically active substances, especially for use as therapeutically active substances for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of spinal muscular atrophy (SMA).
A particular embodiment of the present invention relates to compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the use in the treatment or
prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for use in the treatment or prevention of spinal muscular atrophy (SMA).
A particular embodiment of the present invention relates to a method for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or
prevention of spinal muscular atrophy (SMA), which method comprises administering
compounds of formula (I) or their pharmaceutically acceptable salts as defined above to a subject.
A particular embodiment of the present invention relates to the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or
prevention of spinal muscular atrophy (SMA).
A particular embodiment of the present invention relates to the use of compounds of formula (I) or their pharmaceutically acceptable salts as defined above for the preparation of medicaments for the treatment or prevention of diseases caused by an inactivating mutation or deletion in the SMNl gene and/or associated with loss or defect of SMNl gene function, particularly for the treatment or prevention of spinal muscular atrophy (SMA). Such
medicaments comprise compounds of formula (I) or their pharmaceutically acceptable salts as defined above.
Examples
The invention will be more fully understood by reference to the following examples. They should however not be construed as limiting the scope of the invention.
Abbreviations used
ACN: Acetonitrile; CH2C12: dichloromethane (DCM); DIPEA: diisopropyl ethylamine; DMA: dimethyl acetamide; TEA: triethylamine; RT: room temperature; B2(pin)2:
bis(pinacolato)diboron; Pd(dppf)Cl2: (l,l'-Bis(diphenylphosphino)ferrocene)palladium(II) dichloride; PPTS: Pyridinium p-toluenesulfonate.
Intermediate 1
7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one a) 2-chloro-7-fluoro-pyridor 1 ,2-a1pyrimidin-4-one
Figure imgf000032_0001
A mixture of 2-amino-5-fluoropyridine (11.20 g, 0.10 mol) and dimethyl malonate (57.0 mL, 0.50 mol) was heated at 230 °C for 1.5 h. After cooling to room temperature, the precipitate was filtered and washed with ACN (3x) to give 7-fluoro-2-hydroxy-4H-pyrido[l,2-a]pyrimidin- 4-one as a dark solid (14 g), which was used directly in the next step. MS m/z 181.3 [M+H]+.
A dark mixture of crude 7-fluoro-2-hydroxy-4H-pyrido[l,2-a]pyrimidin-4-one (14g, -77 mmol) in POCl3 (50 mL) and DIPEA (13.3 mL, 77 mmol) was heated at 110 °C for 15 hours. The solvent was removed and the dark residue was treated with ice-water, washed with water (3x) and dried to give a brown solid. The crude brown solid was chromatographed (5% MeOH in CH2C12) to give 2-chloro-7-fluoro-4H-pyrido[l,2-a]pyrimidin-4-one as a yellow solid (9.84 g, 50%, 2 steps), MS m/z 199.2 [M+H]+. b) 2-methyl-6-(4^,5,5-tetramethyl-1 ,2-dioxaborolan-2-yl)imidazori,2-blpyridazine
Figure imgf000033_0001
A mixture of 6-chloro-2-methylimidazo[l,2-b]pyridazine (900 mg, 5.37 mmol), 4,4,4\4 5,5,5\5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (1.36 g, 5.37 mmol, 1.0 eq.), KOAc (1.05 g, 10.7 mmol) and Pd(dppf)Cl2«CH2Cl2 (393 mg, 0.54 mmol) in dioxane ( 50 mL) was degassed and heated under N2 at 95 °C. After 15 hours, the mixture was diluted with EtOAc, filtered through celite and concentrated under vacuum to give 2-methyl-6-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine which was used directly in the next step. c) 7-fluoro-2-(2-methylimidazor 1 ,2-b1pyridazin-6-yl)pyridor 1 ,2-alpyrimidin-4-one
Figure imgf000033_0002
To a solution of 2-chloro-7-fluoro-4H-pyrido[l,2-a]pyrimidin-4-one (750 mg, 3.78 mmol) in ACN (36 mL) was added 2-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)imidazo[l,2-b]pyridazine (1.17 g, 4.53 mmol, Eq: 1.2), Pd(Ph3P)4 (218 mg, 0.189 mmol, 0.05 eq.) and an aqueous solution of K2C03 (3.78 mL, 7.55 mmol, 2.0 eq.). The mixture was degassed and heated under argon at 105 °C overnight. The reaction was cooled to RT, and filtered. The precipitate was washed with Et20 and then water, dried in vacuo to give 250 mg (22%) of 7- fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one as a light brown solid. MS m/z 296.1 [M+H]+.
Intermediate 2
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fl^ a) 2,8-dimethyl-6-(4 A5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)imidazor 1 ,2-blpyridazine
Figure imgf000034_0001
In a sealed flask, 3,6-dichloro-4-methylpyridazine (27 g, 161 mmol) was suspended in aqueous ammonia (25%, 300 mL). The reaction mixture was heated at 110 °C for 48 hours (turned into solution after 1 hour). After cooling to room temperature, the reaction was poured into CH2CI2, and the organic phase was separated, dried over Na2S04, and concentrated under vacuum, to give 22.4 g of 6-chloro-4-methyl-pyridazin-3-amine and 6-chloro-5-methyl- pyridazin-3-amine as a mixture of regioisomers which were used directly in the next step.
The mixture of regioisomers 6-chloro-4-methyl-pyridazin-3-amine and 6-chloro-5-methyl- pyridazin-3-amine (22.4 g) was suspended in 2-propanol (300 mL). l-bromo-2,2- dimethoxypropane (36.0 g, 26.6 mL, 193 mmol, 1.2 eq.) and PPTS (2.96 g, 11.6 mmol, 0.0725 eq.) were added, and the resulting solution was heated at 105°C overnight. The solvent was removed in vacuo and the residue was taken up in CH2C12 and washed with NaHC03. The organic phases were dried over Na2S04, concentrated in vacuo and the crude light brown solid was chromatographed (EtOAc / Heptane 1/2 -1/1) to give separately 6.1 g of 6-chloro-2,8- dimethyl-imidazo[l,2-b]pyridazine MS m/z 182.1 [M+H]+ (21%) as a white solid and 5.9 g of 6- chloro-2,7-dimethyl-imidazo[l,2-b]pyridazine MS m/z 182.1 [M+H]+ (20%) as a white solid. A mixture of 6-chloro-2,8-dimethylimidazo[l,2-b]pyridazine (0.9 g, 4.96 mmol),
4,4,4,,4,,5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (1.26 g, 4.96 mmol, 1.0 eq.), KOAc (0.97 g, 9.91 mmol) and Pd(dppf)Cl2«CH2Cl2 (363 mg, 0.49 mmol) in dioxane ( 50 mL) was degassed and heated under N2 at 110 °C. After 15 hours, the mixture was diluted with EtOAc, filtered through celite and concentrated under vacuum to give 2,8-dimethyl-6-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine which was used directly in the next step. b) 2-(2,8-dimethylimidazor 1 ,2-blpyridazin-6-yl)-7-fruoro-pyridor 1 ,2-alpyrimidin-4-one
Figure imgf000035_0001
To a solution of 2-chloro-7-fluoro-4H-pyrido[l,2-a]pyrimidin-4-one (750 mg, 3.78 mmol, described herein above) in ACN (36 mL) was added 2,8-dimethyl-6-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)imidazo[l,2-b]pyridazine (1.24 g, 4.53 mmol, 1.2 eq.), Pd(Ph3P)4 (218 mg, 0.189 mmol, 0.05 eq.) and an aqueous solution of K2C03 (3.78 mL, 7.55 mmol, 2.0 eq.). The mixture was degassed and heated under argon at 100 °C for 6 hours. The reaction was cooled to RT, and filtered. The precipitate was washed with Et20 and then water, dried in vacuo to give 700 mg (60%) of 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2-a]pyrimidin- 4-one as a light brown solid. MS m/z 310.1 [M+H]+.
Intermediate 3
7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyri^ a) 2-chloro-7-fluoro-9-methyl-pyridor 1 ,2-alpyrimidin-4-one
Figure imgf000035_0002
A mixture of 5-fluoro-3-methylpyridin-2-amine (3.3 g, 26.2 mmol) and dimethyl malonate (15.0 mL, 0.13 mol, 5.0 eq.) was heated at 210 °C for 1.5 hours. After cooling to room
temperature, the precipitate was filtered and washed with ACN (3x) to give 7-fluoro-2-hydroxy- 9-methyl-pyrido[l,2-a]pyrimidin-4-one as a dark solid (2.3 g), which was used directly in the next step. MS m/z 195.1 [M+H]+.
A mixture of crude 7-fluoro-2-hydroxy-9-methyl-pyrido[l,2-a]pyrimidin-4-one (2.3 g, 11.8 mmol) in POCl3 (7.7 mL, 82.9 mmol) and DIEA (2.07 mL, 11.8 mmol) was heated at 110 °C for 15 hours. The solvent was removed and the residue was treated with ice-water, washed with water (3x) and dried to give a brown solid. The crude brown solid was chromatographed (5% MeOH in CH2C12) to give 2-chloro-7-fluoro-9-methyl-pyrido[l,2- a]pyrimidin-4-one as a yellow solid (1.77 g, 70% over 2 steps), MS m/z 213.1 [M+H]+. b) 7-fluoro-9-methyl-2-(2-methylimidazor 1 ,2-b1pyridazin-6-yl)pyridor 1 ,2-alpyrimidin-4- one
Figure imgf000036_0001
To a solution of 2-chloro-7-fluoro-9-methyl-4H-pyrido[l,2-a]pyrimidin-4-one (2.2 g, 10.3 mmol) in ACN (80 mL) was added 2-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)imidazo[l,2-b]pyridazine (3.22 g, 12.4 mmol, 1.2 eq., described herein above), Pd(Ph3P)4 (1.20 g, 1.03 mmol, 0.1 eq.) and an aqueous solution of K2C03 (10.3 mL, 20.7 mmol, 2.0 eq.). The mixture was degassed and heated under argon at 100 °C for 6 hours. The reaction was cooled to RT, and filtered. The precipitate was washed with Et20 and then water, dried in vacuo to give 1.80 g (56%) of 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one as a light brown solid. MS m/z 310.1 [M+H]+.
Intermediate 4
2-(2,8-dimethylimidazo[l,2-b]pyridazm^
one
Figure imgf000036_0002
To a solution of 2-chloro-7-fluoro-9-methyl-4H-pyrido[l,2-a]pyrimidin-4-one (0.98 g, 4.61 mmol, described herein above) in ACN (50 mL) was added 2,8-dimethyl-6-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine (1.51 g, 5.53 mmol, 1.2 eq., described herein above), Pd(Ph3P)4 (0.32 g, 0.277 mmol, 0.06 eq.) and an aqueous solution of K2C03 (4.61 mL, 9.22 mmol, 2.0 eq.). The mixture was degassed and heated under argon at 100 °C for 6 hours. The reaction was cooled to RT, and filtered. The precipitate was washed with Et20 and water, then dried in vacuo to give 0.89 g (60%) of 2-(2,8-dimethylimidazo[l,2- b]pyridazin-6-yl)-7-fluoro-9-methyl-pyrido[l,2-a]pyrimidin-4-one as a light brown solid. MS m/z 324.4 [M+H]+.
Example 1
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-(4-methylpiperazin-l-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000037_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 35 mg, 0.119 mmol) and 1-methylpiperazine (47.5 mg, 0.474 mmol, 4 eq.) were stirred in DMSO (1 mL) at 120°C overnight. LC-MS showed total convertion. The solvent was removed under high vacuum. The crude product was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 9/1) to afford the title product (25 mg, 56%) as a light yellow solid. MS m/z 376.3 [M+H+].
Example 2
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000037_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 125 mg, 0.426 mmol) and (R)-octahydropyrrolo-[l,2- a]pyrazine (160 mg, 1.27 mmol, 3 eq.) were stirred in DMSO (5 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=98/2 to 95/5) to afford the title product (65 mg, 38%) as a light yellow solid. MS m/z 402.5 [M+H+]. Example 3
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000038_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 200 mg, 0.647 mmol) and (S)-octahydropyrrolo-[l,2- a]pyrazine (286 mg, 2.26 mmol, 3.5 eq.) were stirred in DMSO (5 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=98/2 to 95/5) to afford the title product (115 mg, 43%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 4
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000038_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 200 mg, 0.647 mmol), DIPEA (0.113 mL, 0.67 mmol, leq.) and (R)-octahydropyrrolo-[l,2-a]pyrazine (245 mg, 1.95 mmol, 3.0 eq.) were stirred in DMSO (2.5 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=98/2 to 95/5) to afford the title product (132 mg, 49%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 5
7-[(8aS)-8a-methyl-l,3,4,6,7,8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000039_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 90 mg, 0.291 mmol), DIPEA (0.05 mL, 0.29 mmol, leq.) and (S)-8a-methyloctahydropyrrolo[l,2-a]pyrazine (81 mg, 0.58 mmol, 2.0 eq.) were stirred in DMSO (2.5 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (55 mg, 44%) as a light yellow solid. MS m/z 430.3 [M+H+].
Example 6
7-[(8aR)-8a-methyl-l,3,4,6,7,8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000039_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 90 mg, 0.291 mmol), DIPEA (0.05 mL, 0.29 mmol, leq.) and (R)-8a-methyloctahydropyrrolo[l,2-a]pyrazine (81 mg, 0.58 mmol, 2.0 eq.) were stirred in DMSO (2.5 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (50 mg, 40%) as a light yellow solid. MS m/z 430.4 [M+H+].
Example 7
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5R)-3,5-dimethylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000040_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 50 mg, 0.162 mmol), and cis-2,6-dimethylpiperazine (74 mg, 0.647 mmol, 4.0 eq.) were stirred in DMSO (1.5 mL) at 110°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (32 mg, 49%) as a light yellow solid. MS m/z 404.4 [M+H+].
Example 8
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l-yl]pyrido[l,2 a]pyrimidin-4-one
Figure imgf000041_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 33 mg, 0.107 mmol), and (S)-2-methylpiperazine (43 mg, 0.427 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 120°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (18 mg, 43%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 9
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-methylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one
Figure imgf000041_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 85 mg, 0.275 mmol), and (R)-2-methylpiperazine (110 mg, 1.10 mmol, 4.0 eq.) were stirred in DMSO (5 mL) at 120°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (35 mg, 33%) as a light yellow solid. MS m/z 390.3 [M+H+]. Example 10
7-(l,4-diazepan-l-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrim
4-one
Figure imgf000042_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 33 mg, 0.107 mmol), and 1,4-diazepane (32 mg, 0.320 mmol, 3.0 eq.) were stirred in DMSO (2 mL) at 120°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (20 mg, 48%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 11
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one
Figure imgf000042_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), and (S)-2-methylpiperazine (68 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 110°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (40 mg, 63%) as a light yellow solid. MS m/z 376.2 [M+H+].
Example 12
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-methylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one
Figure imgf000043_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), and (R)-2-methylpiperazine (68 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 110°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (48 mg, 75%) as a light yellow solid. MS m/z 376.3 [M+H+].
Example 13
7-(l,4-diazepan-l-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4- one
Figure imgf000043_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), and 1,4-diazepane (68 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 110°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (41 mg, 65%) as a light yellow solid. MS mJz 376.2 [M+H+].
Example 14
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000044_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), and cis-2,6-dimethylpiperazine (77 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 110°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (41 mg, 62%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 15
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000044_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), and (S)-octahydropyrrolo[l,2- a]pyrazine (85 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (36 mg, 53%) as a light yellow solid. MS m/z 402.3 [M+H+].
Example 16
7-[(8aS)-8a-methyl-l,3,4,6,7,8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2- methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000045_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol) and (S)-8a- methyloctahydropyrrolo[l,2-a]pyrazine (95 mg, 0.677 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (45 mg, 64%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 17
7-[(8aR)-8a-methyl-l,3,4,6,7,8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2- methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000045_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 100 mg, 0.339 mmol) and (R)-8a- methyloctahydropyrrolo[l,2-a]pyrazine (190 mg, 1.35 mmol, 4.0 eq.) were stirred in DMSO (4 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (45 mg, 64%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 18
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3^yrrolidin-l-ylpyrrolidin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000046_0001
In a microwave reactor, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 45 mg, 0.145 mmol), (R)-l,3'-bipyrrolidine dihydrochloride (62 mg, 0.291 mmol, 2.0 eq.) and DIPEA (0.20 mL, 1.16 mmol, 8 eq.) were stirred in NMP (3 mL) at 220°C for 1 hour. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=98/2 to 90/10) to afford the title product (25 mg, 40%) as a light yellow solid. MS m/z 430.3 [M+H+]. Example 19
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000047_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 50 mg, 0.169 mmol), DIPEA (0.24 mL, 1.35 mmol, 8 eq.) and 4,7-diazaspiro[2.5]octane dihydrochloride (62.7 mg, 0.339 mmol, 2.0 eq.) were stirred in DMSO (2 mL) at 125°C for 2 days. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (22 mg, 33%) as a light yellow solid. MS m/z 388.3 [M+H+].
Example 20 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6- yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000047_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 50 mg, 0.162 mmol), DIPEA (0.22 mL, 1.29 mmol, 4 eq.) and 4,7-diazaspiro[2.5]octane dihydrochloride (32 mg, 0.320 mmol, 3.0 eq.) were stirred in
DMSO (2 mL) at 130°C for 48 hours. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=98/2 to 95/5) to afford the title product (12 mg, 18%) as a light yellow solid. MS m/z 402.3 [M+H+].
Example 21
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3^yrrolidin-l-ylpyrrolidin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000048_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 40 mg, 0.135 mmol), DIPEA (0.19 mL, 1.08 mmol, 8 eq.) and (R)- 1,3 '-bipyrrolidine dihydrochloride (58 mg, 0.271 mmol, 2.0 eq.) were stirred in DMSO (4 mL) and heated at 220°C for 40 minutes in a microwave. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=98/2 to 90/10) to afford the title product (30 mg, 53%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 22
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin-l-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000048_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 40 mg, 0.129 mmol) and 2,2-dimethylpiperazine (59 mg, 0.517 mmol, 4.0 eq.) were stirred in DMSO (1.6 mL) at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 9/1) to afford the title product (29 mg, 55%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 23
7-(3,3-dimethylpiperazin-l-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000049_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 40 mg, 0.135 mmol) and 2,2-dimethylpiperazine (62 mg, 0.542 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (26 mg, 49%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 24
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3S)-3-methylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000049_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl- pyrido[l,2-a]pyrimidin-4-one (Intermediate 4; 50 mg, 0.155 mmol) and (S)-2-methylpiperazine (62 mg, 0.619 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (45 mg, 72%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 25
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3R)-3-methylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000050_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl- pyrido[l,2-a]pyrimidin-4-one (Intermediate 4; 50 mg, 0.155 mmol) and (R)-2-methylpiperazine (62 mg, 0.619 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (40 mg, 70%) as a light yellow solid. MS m/z 404.3 [M+H+]. Example 26
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-y ^
methyl-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000051_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl- pyrido[l,2-a]pyrimidin-4-one (Intermediate 4; 50 mg, 0.155 mmol) and cis-2,6- dimethylpiperazine (70 mg, 0.619 mmol, 4.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column
chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (26 mg, 40%) as a light yellow solid. MS m/z 418.3 [M+H+].
Example 27
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin-l-yl)-9-methyl- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000051_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl- pyrido[l,2-a]pyrimidin-4-one (Intermediate 4; 50 mg, 0.155 mmol) and 2,2-dimethylpiperazine (35 mg, 0.309 mmol, 2.0 eq.) were stirred in DMSO (2 mL) at 125°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (36 mg, 56%) as a light yellow solid. MS m/z 418.3 [M+H+].
Example 28
7 4,7-diazaspiro[2.5]octan-7-yl)-2^2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000052_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-9-methyl- pyrido[l,2-a]pyrimidin-4-one (Intermediate 4; 50 mg, 0.155 mmol), DIPEA (0.21 mL, 1.24 mmol, 8 eq.) and 4,7-diazaspiro[2.5]octane dihydrochloride (57 mg, 0.309 mmol, 2.0 eq.) were stirred in DMSO (2 mL) at 125°C for 2 days. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (17 mg, 26%) as a light yellow solid. MS m/z 416.3 [M+H+].
Example 29
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5S)-3,5-dimethylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000052_0002
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 50 mg, 0.162 mmol), TEA (0.18 mL, 1.29 mmol, 8 eq.) and (2S,6S)-2,6-dimethylpiperazine dihydrochloride (90 mg, 0.485 mmol, 3.0 eq.) were stirred in DMSO (2 mL) at 140°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 9/1) to afford the title product (20 mg, 30%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 30
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3^yrrolidin-l-ylpyrrolidin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000053_0001
In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2- a]pyrimidin-4-one (Intermediate 2; 50 mg, 0.162 mmol), DIPEA (0.22 mL, 1.29 mmol, 8 eq.) and (S)-l,3'-bipyrrolidine dihydrochloride (103 mg, 0.485 mmol, 3.0 eq.) were stirred in NMP (2 mL) at 140°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 9/1) to afford the title product (22 mg, 32%) as a light yellow solid. MS m/z 430.3 [M+H+]. Example 31
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3^yrrolidin-l-ylpyrrolidin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000054_0001
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 75 mg, 0.254 mmol), TEA (0.28 mL, 2.03 mmol, 8 eq.) and (S)-l,3'-bipyrrolidine dihydrochloride (162 mg, 0.762 mmol, 3.0 eq.) were stirred in NMP (4 mL) and heated at 220°C for 1 hour in a microwave. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (12 mg, 11%) as a light yellow solid. MS m/z 416.2 [M+H+].
Example 32
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000054_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 75 mg, 0.254 mmol), TEA (0.28 mL, 2.03 mmol, 8 eq.) and (2S,6S)-2,6-dimethylpiperazine dihydrochloride (143 mg, 0.762 mmol, 3.0 eq.) were stirred in DMSO (3 mL) and heated at 140°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (10 mg, 10%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 33
9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000055_0001
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 250 mg, 0.808 mmol), and (S)-2-methylpiperazine (405 mg, 4.04 mmol, 5.0 eq.) were stirred in DMSO (6 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02,
CH2Cl2/MeOH=95/5 to 85/15) to afford the title product (135 mg, 43%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 34
9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-methylpiperazin-l- yl]pyrido[l,2-a]pyrimidin-4-one
Figure imgf000055_0002
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 250 mg, 0.808 mmol), and (R)-2-methylpiperazine (405 mg, 4.04 mmol, 5.0 eq.) were stirred in DMSO (6 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02,
CH2Cl2/MeOH=95/5 to 85/15) to afford the title product (100 mg, 32%) as a light yellow solid. MS m/z 390.3 [M+H+].
Example 35
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-^ yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000056_0001
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 250 mg, 0.808 mmol), and (2S,6R)-2,6-dimethylpiperazine (461 mg, 4.04 mmol, 5.0 eq.) were stirred in DMSO (6 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 85/15) to afford the title product (101 mg, 31%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 36
7-(3,3-dimethylpiperazin-l-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000056_0002
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 250 mg, 0.808 mmol), and 2,2-dimethylpiperazine (461 mg, 4.04 mmol, 5.0 eq.) were stirred in DMSO (6 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (S1O2,
CH2Cl2/MeOH=95/5 to 85/15) to afford the title product (120 mg, 36%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 37
7-(4,7-diazaspiro[2.5]octan-7-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000057_0001
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 125 mg, 0.404 mmol), K2C03 (223 mg, 1.62 mmol, 4 eq.) and 4,7-diazaspiro[2.5]octane dihydrochloride (112 mg, 0.606 mmol, 1.5 eq.) were stirred in DMA (2 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (75 mg, 46%) as a light yellow solid. MS m/z 402.2 [M+H+].
Example 38
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6 yl)pyrido[l,2-a]pyrimidin-4-one
Figure imgf000058_0001
In a sealed tube, 7-fluoro-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one (Intermediate 3; 125 mg, 0.404 mmol), K2C03 (223 mg, 1.62 mmol, 4 eq.) and (2S,6S)-2,6-dimethylpiperazine dihydrochloride (113 mg, 0.606 mmol, 1.5 eq.) were stirred in DMA (2 mL) and heated at 130°C overnight. The solvent was removed under high vacuum. The residue was taken up in CH2C12 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 90/10) to afford the title product (50 mg, 31%) as a light yellow solid. MS m/z 404.3 [M+H+].
Example 39
7-[(3R)-3-ethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one
Figure imgf000058_0002
In a sealed tube, 7-fluoro-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-4H-pyrido[l,2- a]pyrimidin-4-one (Intermediate 1; 200 mg, 0.677 mmol), K2C03 (374 mg, 2.71 mmol, 4 eq.) and (R)-2-ethylpiperazine dihydrochloride (238 mg, 0.606 mmol, 1.5 eq.) were stirred in DMA (3 mL) at 100°C for 4 days. The solvent was removed under high vacuum. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=95/5 to 8/2) to afford the title product (168 mg, 64%) as a light yellow solid. MS m/z 390.2 [M+H+]. Biological Assays
To describe in more detail and assist in understanding the present description, the following non-limiting biological examples are offered to more fully illustrate the scope of the description and are not to be construed as specifically limiting the scope thereof. Such variations of the present description that may be now known or later developed, which would be within the purview of one skilled in the art to ascertain, are considered to fall within the scope of the present description and as hereinafter claimed. These examples illustrate the testing of certain compounds described herein in vitro and/or in vivo and demonstrate the usefulness of the compounds for treating of SMA by enhancing the inclusion of exon 7 of SMN2 into mRNA transcribed from the SMN2 gene. Compounds of formula (I) enhance inclusion of exon 7 of SMN2 into mRNA transcribed from the SMN2 gene and increase levels of SMN protein produced from the SMN2 gene, and thus can be used to treat SMA in a human subject in need thereof. These examples further illustrate the testing of certain compounds described herein in vitro and/or in vivo and demonstrate the usefulness of the compounds for enhancing the inclusion of exon 7 of SMNI into mRNA transcribed from the SMNl gene. Accordingly, compounds of formula (I) also enhance the inclusion of exon 7 of SMNl into mRNA transcribed from the SMNl gene and increase levels of SMN protein produced from the SMNl gene.
Assay 1
SMN2 minigene mRNA splicing RT-qPCR assay in cultured cells
The reverse transcription-quantitative PCR-based (RT-qPCR) assay is used to quantify the level of the full length SMN2 minigene (referred to herein by the term "FL SMN2mini") mRNA containing SMN2 exon 7 in a HEK293H cell line stably transfected with said minigene and treated with a test compound. Materials used and respective sources are listed below in Table 1.
Material Source
HEK293H cells Life Technologies, Inc. (formerly Invitrogen) Catalog No. 11631-017
Cells-To-Ct lysis Life Technologies, Inc. (formerly Applied Biosystems) part No. 4399002 buffer
DMEM Life Technologies, Inc. (formerly Invitrogen) Catalog No. 11960-044
96-well flat-bottom Becton Dickinson Catalog No. 353072
plates RT-PCR Enzyme Life Technologies, Inc. (formerly Applied Biosystems) part No. 4388520 Mix
RT-PCR buffer Life Technologies, Inc. (formerly Applied Biosystems) part No. 4388519
AgPath-ID One- Life Technologies, Inc. (formerly Applied Biosystems) part No. 4387391 Step RT-PCR kit
Thermocycler Life Technologies, Inc. (formerly Applied Biosystems) 7900HT
Table 1. Materials and their respective sources used in the SMN2 minigene mRNA splicing RT-qPCR assay in cultured cells.
The SMN2-A minigene construct was prepared as described in International Patent
Application WO2009/151546A1 page 145 paragraph [00400] to page 147 paragraph [00412] (incl. Figure 1 and Figure 3 therein).
HEK293H cells stably transfected with the SMN2-A minigene construct (10,000 cells/well) are seeded in 200 of cell culture medium (DMEM plus 10% FBS, with 200 μg/mL
hygromycin) in 96-well flat-bottom plates and the plate is immediately swirled to ensure proper dispersal of cells and the formation of an even monolayer of cells. Cells are allowed to attach for 6 hours. Test compounds are serially diluted 3.16-fold in 100% DMSO to generate a 7-point concentration curve. A solution of test compound (1 μί, 200x in DMSO) is added to each cell- containing well and the plate is incubated for 24 hours in a cell culture incubator (37 °C, 5% C02, 100% relative humidity). 2 replicates are prepared for each test compound concentration. The cells are then lysed in the Cells-To-Ct lysis buffer and the lysate is stored at -80°C. Full length SMN2-A minigene and GAPDH mRNA are quantified using the primers and probes referenced in Table 2. Primer SMN Forward A (SEQ ID NO. l) hybridizes to a nucleotide sequence in exon 7 (nucleotide 22 to nucleotide 40), primer SMN Reverse A (SEQ ID NO.2) hybridizes to a nucleotide sequence in the coding sequence of Firefly luciferase, SMN Probe A (SEQ ID NO.3) hybridizes to a nucleotide sequence in exon 7 (nucleotide 50 to nucleotide 54) and exon 8 (nucleotide 1 to nucleotide 21). The combination of these three oligonucleotides detects only SMN1 or SMN2 minigenes (RT-qPCR) and will not detect endogenous SMN1 or SMN2 genes.
Primers/Probes Sequences Source
SMN Forward Primer A SEQ ID NO. 1: GAAGGAAGGTGCTCACATT PTC1
SMN Reverse Primer A SEQ ID NO. 2: TCTTTATGTTTTTGGCGTCTTC PTC1 SMN Forward Probe A SEQ ID NO. 3: 6FAM- AAGGAGAAATGCTGGCAT PTC1
AGAGCAGC-TAMRA hGAPDH Forward Probe SEQ ID NO. 4: VIC-CGCCTGGTCACCAGGGCTGCT- LTI2
TAMRA hGAPDH Forward Primer SEQ ID NO. 5: CAACGGATTTGGTCGTATTGG LTI2 hGAPDH Reverse Primer SEQ ID NO. 6: TGATGGCAACAATATCCACTTTACC LTI2
Table 2. 1 Primers and probes designed by PTC Therapeutics, Inc.; 2 Commercially available from Life Technologies, Inc. (formerly Invitrogen).
The SMN forward and reverse primers are used at final concentrations of 0.4 μΜ. The SMN probe is used at a final concentration of 0.15 μΜ. The GAPDH primers are used at final concentrations of 0.2 μΜ and the probe at 0.15 μΜ.
The SMN2-minigene GAPDH mix (15 μΐ^ total volume) is prepared by combining 7.5 μΐ^ of 2x RT-PCR buffer, 0.4 μΐ, of 25x RT-PCR enzyme mix, 0.75 μΐ, of 20x GAPDH primer- probe mix, 4.0075 μΐ, of water, 2 μΐ, of 10-fold diluted cell lysate, 0.06 μΐ, of 100 μΜ SMN forward primer, 0.06 μΐ, of 100 μΜ SMN reverse primer, and 0.225 μΐ, of 100 μΜ SMN probe. PCR is carried out at the following temperatures for the indicated time: Step 1: 48°C (15 min); Step 2: 95°C (10 min); Step 3: 95°C (15 sec); Step 4: 60°C (1 min); then repeat Steps 3 and 4 for a total of 40 cycles.
Each reaction mixture contains both SMN2-A minigene and GAPDH primers/probe sets (multiplex design), allowing simultaneous measurement of the levels of two transcripts. The increase in the abundance of the FL SMN2mini mRNA relative to that in cells treated with vehicle control is determined from real-time PCR data using a modified AACt method (as described in Livak and Schmittgen, Methods, 2001, 25:402-8). The amplification efficiency E is calculated from the slope of the amplification curve for FL SMN2mini and GAPDH individually. The abundance of FL SMN2mini and GAPDH mRNA are then calculated as (1 + E)"Ct, where Ct is the threshold value for each amplicon. The abundance of FL SMN2mini mRNA is normalized to GAPDH mRNA abundance. The normalized FL SMN2mini mRNA abundance from test compound-treated samples is then divided by normalized FL SMN2mini mRNA abundance from vehicle-treated cells to determine the level of FL SMN2mini mRNA relative to vehicle control. Table 3 provides ECi.5X concentrations for production of full length SMN2 minigene mRNA that was obtained from the 7-point concentration data generated according to the above procedure for particular compounds of the present invention.
Particular compounds of the present invention exhibit an ECi.5X concentration for production of full length SMN2 minigene mRNA < 1 μΜ.
More particular compounds of the present invention exhibit an ECi.5X concentration for production of full length SMN2 minigene mRNA < 0.1 μΜ.
Most particular compounds of the present invention exhibit an EC1.5x concentration for production of full length SMN2 minigene mRNA < 0.02 μΜ.
Figure imgf000062_0001
Table 3. ECi.5X concentrations for production of full length SMN2 minigene mRNA.
Assay 2
SMN protein assay in cultured cells
The SMN HTRF (homogeneous time resolved fluorescence) assay is used to quantify the level of SMN protein in SMA patient fibroblast cells treated with test compounds. Materials used and respective sources are listed below in Table 4.
Figure imgf000063_0001
Table 4. Materials and their respective sources used in the SMN protein assay in cultured cells.
Cells are thawed and cultured in DMEM- 10% FBS for 72 hours. Cells are trypsinized, counted and re-suspended to a concentration of 25,000 cells/mL in DMEM-10% FBS. The cell suspensions are plated at 5,000 cells per well in a 96 well microtiter plate and incubated for 3 to 5 hours. Test compounds are serially diluted 3.16-fold in 100% DMSO to generate a 7-point concentration curve. 1 μL· of test compound solution is transferred to cell-containing wells and cells are incubated for 48 hours in a cell culture incubator (37°C, 5% C02, 100% relative humidity). Triplicate samples are set up for each test compound concentration. After 48 hours, the supernatant is removed from the wells and 25 μΐ^ of the RIPA lysis buffer, containing protease inhibitors, is added to the wells and incubated with shaking at room temperature for 1 hour. 25 μΐ^ of the diluent is added and then 35 μΐ^ of the resulting lysate is transferred to a 384- well plate, where each well contains 5 μΐ^ of the antibody solution (1 : 100 dilution of anti-SMN d2 and anti-SMN kryptate in SMN reconstitution buffer). The plate is centrifuged for 1 minute to bring the solution to the bottom of the wells, then incubated overnight at room temperature. Fluorescence for each well of the plate at 665 nm and 620 nm is measured on an En Vision multilabel plate reader (Perkin-Elmer).
The normalized fluorescence signal is calculated for each sample, Blank and vehicle control well by dividing the signal at 665 nm by the signal at 620 nm. Normalizing the signal accounts for possible fluorescence quenching due to the matrix effect of the lysate. The AF value (a measurement of SMN protein abundance as a percent value) for each sample well is calculated by subtracting the normalized average fluorescence for the Blank control wells from the normalized fluorescence for each sample well, then dividing this difference by the
normalized average fluorescence for the Blank control wells and multiplying the resulting value by 100. The AF value for each sample well represents the SMN protein abundance from test compound-treated samples. The AF value for each sample well is divided by the AF value for the vehicle control wells to calculate the fold increase in SMN protein abundance relative to the vehicle control. Table 5 provides ECi.5X concentrations for SMN protein expression that was obtained from the 7-point concentration data generated according to the above procedure for particular compounds of the present invention.
Particular compounds of the present invention exhibit an ECi.5X concentration for SMN protein expression < 1 μΜ.
More particular compounds of the present invention exhibit an ECi.5X concentration for SMN protein expression < 100 nM.
Most particular compounds of the present invention exhibit an ECi.5X concentration for SMN protein expression < 30 nM. Table 6 provides the maximum fold increase of SMN protein that was obtained from the 7- point concentration data generated according to the above procedure for particular compounds of the present invention
Particular compounds of the present invention exhibit a maximum fold increase > 1.5. More particular compounds of the present invention exhibit a maximum fold increase > 1.7. Most particular compounds of the present invention exhibit a maximum fold increase > 1.8. EC1.5x EC1.5x EC1.5x SMN SMN SMN
Example Example Example
protein protein protein (nM) (nM) (nM)
1 10.8 14 17.6 27 126.5
2 19.8 15 21.2 28 49.7
3 25.6 16 3 29 2.1
4 15.7 17 20.2 30 13.6
5 4.1 18 25 31 27.7
6 11 19 29.8 32 4
7 15.5 20 37 33 4
8 5.9 21 68.7 34 4.4
9 2.5 22 13.8 35 19.5
10 22.8 23 23.9 36 34.4
11 7 24 4.7 37 45
12 7.5 25 11.9 38 3.1
13 3 26 1230 39 15.8
Table 5. ECi.5X concentrations for SMN protein expression.
Figure imgf000065_0001
Table 6. Maximum fold increase of SMN protein.

Claims

Claims
The compound of formula (I)
Figure imgf000066_0001
wherein R1 is hydrogen or Ci_7-alkyl; R2 is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C3_g-cycloalkyl; R3 is hydrogen, Ci_7-alkyl, or C3_g-cycloalkyl; A is N-heterocycloalkyl or NR 12 R 13 , wherein N-heterocycloalkyl comprises 1 or 2 nitrogen ring atoms and is optionally substituted with 1, 2, 3 or 4 substituents selected from R14;
R 12 is heterocycloalkyl comprising 1 nitrogen ring atom, wherein heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R14;
R 13 is hydrogen, Ci_7-alkyl or C3_g-cycloalkyl;
R 14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C3_ 8-cycloalkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof. A compound according to claim 1, wherein R1 is hydrogen or Ci_7-alkyl;
R is hydrogen, cyano, Ci_7-alkyl, Ci_7-haloalkyl or C3_g-cycloalkyl; R is hydrogen, Ci_7-alkyl, or C3_8-cycloalkyl;
A is N-heterocycloalkyl comprising 1 or 2 nitrogen ring atoms, wherein N- heterocycloalkyl is optionally substituted with 1, 2, 3 or 4 substituents selected from R14; R14 is independently selected from hydrogen, Ci_7-alkyl, amino, amino-Ci_7-alkyl, C3_
8-cycloalkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene; with the proviso that if A is N-heterocycloalkyl comprising only 1 nitrogen ring atom, then at least one R14 substituent is amino or amino-Ci_7-alkyl; and pharmaceutically acceptable salts thereof. 3. A compound according to any of claims 1 or 2, wherein R1 is Ci_7-alkyl.
4. A compound according to any of claims 1 to 3, wherein R1 is methyl.
5. A compound according to any of claims 1 to 4, wherein R is hydrogen or Ci_7-alkyl.
6. A compound according to any of claims 1 to 5, wherein R is hydrogen or methyl.
7. A compound according to any of claims 1 to 6, wherein R is hydrogen or Ci_7-alkyl. 8. A compound according to any of claims 1 to 7, wherein R is hydrogen or methyl.
9. A compound according to any of claims 1 to 8, wherein R 12 is piperidinyl optionally
substituted with 1, 2, 3 or 4 substituents selected from R14.
10. A compound according to any of claims 1 to 9, wherein R 13 is hydrogen or Ci_7-alkyl.
11. A compound according to any of claims 1 to 10, wherein R 13 is hydrogen or methyl. 12. A compound according to any of claims 1 to 11, wherein R14 is independently selected from Ci_7-alkyl and heterocycloalkyl or two R14 together form Ci_7-alkylene.
13. A compound according to any of claims 1 to 12, wherein R14 is independently selected from methyl, ethyl and pyrrolidinyl or two R14 together form ethylene
14. A compound according to any of claims 1 to 13, wherein the N-heterocycloalkyl in A or tthhee hheetteerrooccyyccllooaallkkyyll iinn R as defined in Claim 1 are further characterized in that one ring nitrogen atom is basic.
15. A compound according to any of claims 1 to 14, wherein
Figure imgf000068_0001
, wherein
X is N or CH;
R4 is hydrogen, Ci_7-alkyl or -(CH2)m-NR9R10;
R5 is hydrogen or Ci_7-alkyl;
R6 is hydrogen or Ci_7-alkyl;
R is hydrogen or Ci_7-alkyl;
R is hydrogen or Ci_7-alkyl;
R9 and R10 are independently selected from hydrogen, Ci_7-alkyl and C3_8-cycloalkyl;
R 13 is hydrogen, Ci_7-alkyl or C3_8-cycloalkyl; n is 0, 1 or 2; m is 0, 1, 2 or 3; or R4 and R5 together form Ci_7-alkylene; or R4 and R7 together form Ci_7-alkylene; or R5 and R6 together form C2_7-alkylene; or R 5 and R 7 together form Ci_7-alkylene; or R5 and R9 together form Ci_7-alkylene;
7 Q
or R and R together form C2_7-alkylene;
7 Q
or R and R together form Ci_7-alkylene; or R9 and R10 together form C2_7-alkylene; with the proviso that if X is CH then R4 is -(CH2)m-NR9R10; and with the proviso that if X is N and R4 is -(CH2)m-NR9R10 then m is 2 or 3.
A compound according to any of claims 1 to 15, wherein A
Figure imgf000069_0001
wherein
X is N or CH;
R4 is hydrogen, Ci_7-alkyl or -(CH2)m-NRyRlu;
R5 is hydrogen or Ci_7-alkyl;
R6 is hydrogen or Ci_7-alkyl;
R7 is hydrogen or Ci_7-alkyl;
R is hydrogen or Ci_7-alkyl;
Ry and R are independently selected from hydrogen, Ci_7-alkyl and C3_8-cycloalkyl; is 0, 1 or 2; m is 0, 1, 2 or 3; or R4 and R5 together form Ci_7-alkylene; or R4 and R7 together form Ci_7-alkylene; or R5 and R6 together form C2_7-alkylene; or R 5 and R 7 together form Ci_7-alkylene; or R5 and R9 together form Ci_7-alkylene; or R 7 and R 8 together form C2_7-alkylene; or R 7 and R 9 together form Ci_7-alkylene; or R and R together form C2_7-alkylene; with the proviso that if X is CH then R4 is -(CH2)m-NR9R10; and with the proviso that if X is N and R4 is - (CH2)m -NR9R10 then m is 2 or 3.
17. A compound according to any of claims 1 to 16, wherein X is N.
18. A compound according to any of claims 1 to 17, wherein n is 1.
19. A compound according to any of claims 1 to 18, wherein R4 is hydrogen, methyl or
-(CH2)m-NR9R10.
20. A compound according to any of claims 1 to 19, wherein R4 is hydrogen.
21. A compound according to any of claims 1 to 20, wherein R5 is hydrogen, methyl or ethyl.
22. A compound according to any of claims 1 to 21, wherein R5 is methyl.
23. A compound according to any of claims 1 to 22, wherein R6 is hydrogen or methyl.
24. A compound according to any of claims 1 to 23, wherein R6 is hydrogen.
25. A compound according to any of claims 1 to 24, wherein R is hydrogen or methyl.
26. A compound according to any of claims 1 to 25, wherein R is hydrogen.
27. A compound according to any of claims 1 to 19, wherein m is 0.
28. A compound according to any of claims 1 to 18, wherein R4 and R5 together form
propylene.
29. A compound according to any of claims 1 to 18, wherein R5 and R6 together form ethylene
30. A compound according to any of claims 1 to 18, wherein R9 and R10 together form
butylene.
31. A compound according to any of claims 1 to 30, wherein A is selected from the group of:
Figure imgf000070_0001
Figure imgf000071_0001
wherein R4, R5, R6, R7, R8 and R13 are as defined in any of claims 1 to 30 and wherein R is hydrogen or Ci_7-alkyl.
A compound according to any of claims 1 to 31, wherein A is selected from the group of:
Figure imgf000071_0002
wherein R4, R5, R6, R7 and R8 are as defined in any of claims 1 to 31 and wherein R hydrogen or Ci_7-alkyl.
33. A compound according to any of claims 1 to 32, wherein A is selected from the group of piperazinyl, diazepanyl, pyrrolidinyl and hexahydropyrrolo[l,2-a]pyrazinyl, each optionally substituted with 1, 2, 3 or 4 substituents selected from R14 as defined in any of claims 1 to 32. 34. A compound according to any of claims 1 to 33, wherein A is selected from the group of piperazin-l-yl, 1,4-diazepan-l-yl, pyrrolidin-l-yl and hexahydropyrrolo[l,2-a]pyrazin- 2(lH)-yl, each optionally substituted with 1 or 2 substituents selected from R14 as defined in any of claims 1 to 33. 35. A compound according to any of claims 1 to 31, wherein A is NR 12 R 13 , wherein R 12 and
R 13 are as described in any of claims 1 to 31.
36. A compound according to any of claims 1 to 31, wherein A is
Figure imgf000072_0001
, wherein R4, R5, R6, R7, R8 and R13 are as defined in any of claims 1 to 31. 37. A compound according to any of claims 1 to 34, wherein A is selected from the group of:
Figure imgf000072_0002
A compound according to any of claims 1 to 34, wherein A is selected from the group of
Figure imgf000073_0001
39. A compound according to any one of claims 1 to 38, selected from the group consisting of:
2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-(4-methylpiperazin- l-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one; 7-[(8aR)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5R)-3,5-dimethylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3S)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3R)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
7-( 1 ,4-diazepan- 1 -yl)-2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l-yl]pyrido[l,2- a]pyrimidin-4-one;
2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)-7- [(3R)-3-methylpiperazin- 1 -yl]pyrido[ 1 ,2- a]pyrimidin-4-one;
7-( 1 ,4-diazepan- 1 -yl)-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4- one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one;
7-[(8aS)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2- methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-8a-methyl- 1,3,4,6,7, 8-hexahydropyrrolo[l,2-a]pyrazin-2-yl]-2-(2- methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin- l-yl)pyrido[ 1 ,2- a]pyrimidin-4-one;
7-(3,3-dimethylpiperazin-l-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3S)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one; 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3R)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9- methyl-pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-(3,3-dimethylpiperazin-l-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5S)-3,5-dimethylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-pyrrolidin-l-ylpyrrolidin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)-7-[(3R)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(3,3-dimethylpiperazin-l-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3S,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R)-3-ethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a] pyrimidin-4-one;
and pharmaceutically acceptable salts thereof.
A compound according to any one of claims 1 to 39, selected from the group consisting of: 7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b] pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aR)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2,8- dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-[(3S,5R)-3,5-dimethylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(8aS)-3,4,6,7,8,8a-hexahydro-lH-pyrrolo[l,2-a]pyrazin-2-yl]-2-(2-methylimidazo[l,2- b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2-methylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a] pyrimidin-4- one ;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2- a] pyrimidin-4- one ;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl-7-[(3S)-3-methylpiperazin-l- yl]pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-9-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one;
7-[(3R,5S)-3,5-dimethylpiperazin-l-yl]-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
7-(4,7-diazaspiro[2.5]octan-7-yl)-9-methyl-2-(2-methylimidazo[l,2-b]pyridazin-6- yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
and pharmaceutically acceptable salts thereof.
The compound of formula (VI)
Figure imgf000076_0001
wherein R 1 , R2 and R 3 are as described in any of claims 1 to 7; Y is halogen or trifluoromethanesulfonate; and salts thereof.
A compound of formula (VI) according to claim 41, wherein Y is fluoro, chloro, bromo, iodo or trifluoromethanesulfonate.
43. A compound of formula (VI) according to claim 41, wherein Y is fluoro.
44. A compound of formula (VI) according to any one of claims 41 to 43, selected from the group consisting of:
7-fluoro-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one;
2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2-a]pyrimidin-4-one;
7-fluoro-9-methyl-2-(2-methylimidazo[ 1 ,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one; 2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-7-fluoro-9-methyl-pyrido[ 1 ,2-a]pyrimidin-4- one;
and salts thereof. 45. A process for the preparation of compounds of formula (I) according to any of claims 1 to 40, comprising an aromatic nucleophilic substitution reaction between a compound of formula (VI) as described in any of claims 41 to 44 with a compound of formula M-A by heating in a solvent, wherein A, R 1 , R2 and R 3 are as defined in any of claims 1 to 40, Y is as defined in any of claims 41 to 44, M is hydrogen, sodium or potassium, and wherein M is linked to A via a nitrogen atom of A.
46. The process of claim 45, wherein the aromatic nucleophilic substitution reaction is
performed at a temperature from 80°C to 200°C.
47. The process according to any of claims 45 or 46, wherein the solvent of the aromatic
nucleophilic substitution reaction is selected from dimethyl sulfoxide, N-methylpyrrolidone, and dimethylformamide.
48. The process according to any of claims 45 to 47, wherein M is hydrogen.
49. Compounds of formula (I) according to any of claims 1 - 40, obtainable by a process
according to any of claims 45 to 48.
50. Pharmaceutical compositions comprising compounds of formula (I) according to any of claims 1 - 40 or their pharmaceutically acceptable salts and one or more pharmaceutically acceptable excipients.
51. Compounds of formula (I) according to any of claims 1 - 40 or their pharmaceutically
acceptable salts above for use as therapeutically active substances.
52. Compounds of formula (I) according to any of claims 1 - 40 or their pharmaceutically
acceptable salts for the use in the treatment or prevention of spinal muscular atrophy (SMA).
53. A method for the treatment or prevention of spinal muscular atrophy (SMA), which method comprises administering compounds of formula (I) according to any of claims 1 - 40 or their pharmaceutically acceptable salts as defined above to a subject.
54. The use of compounds of formula (I) according to any of claims 1 - 40 or their
pharmaceutically acceptable salts for the treatment or prevention of spinal muscular atrophy (SMA).
55. The use of compounds of formula (I) according to any of claims 1 - 40 or their
pharmaceutically acceptable salts for the preparation of medicaments for the treatment or prevention of spinal muscular atrophy (SMA). 56. The invention as described hereinbefore.
PCT/EP2015/060343 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy WO2015173181A1 (en)

Priority Applications (36)

Application Number Priority Date Filing Date Title
KR1020167034983A KR102213740B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
SG11201609497TA SG11201609497TA (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
UAA201612716A UA119670C2 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
EP23169721.0A EP4241772A3 (en) 2014-05-15 2015-05-11 Process for the preparation of compounds useful for treating spinal muscular atrophy
ES15721701T ES2761423T3 (en) 2014-05-15 2015-05-11 Compounds to treat spinal muscular atrophy
MX2016014547A MX371050B (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy.
AU2015261046A AU2015261046C1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
MYPI2016001903A MY174284A (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
MA39995A MA39995B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
RS20191541A RS59718B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
BR112016026205-0A BR112016026205B1 (en) 2014-05-15 2015-05-11 SMN2 GENE TWISTING MODULATORS, THEIR USE AND THEIR PREPARATION PROCESS, AND PHARMACEUTICAL COMPOSITIONS
DK15721701T DK3143025T3 (en) 2014-05-15 2015-05-11 Compounds for the treatment of spinal muscle atrophy
EP19200058.6A EP3663296B1 (en) 2014-05-15 2015-05-11 Process for the preparation of compounds useful for treating spinal muscular atrophy
KR1020217002966A KR102256013B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
EA201692280A EA035068B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
CR20160518A CR20160518A (en) 2014-05-15 2015-05-11 COMPOUNDS TO TREAT SPINAL MUSCLE ATROPHY
SI201531021T SI3143025T1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
CA2948561A CA2948561C (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
PL15721701T PL3143025T3 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
CN201580027306.9A CN106459092B (en) 2014-05-15 2015-05-11 For treating the compound of Duchenne-Arandisease
NZ72500815A NZ725008A (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
JP2016567816A JP6236173B2 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
EP15721701.9A EP3143025B1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy
ZA2016/07026A ZA201607026B (en) 2014-05-15 2016-10-12 Compounds for treating spinal muscular atrophy
PH12016502081A PH12016502081A1 (en) 2014-05-15 2016-10-19 Compounds for treating spinal muscular atrophy
IL24865316A IL248653B (en) 2014-05-15 2016-10-31 Compounds for treating spinal muscular atrophy
US15/351,267 US9969754B2 (en) 2014-05-15 2016-11-14 Compounds for treating spinal muscular atrophy
US15/953,008 US10882868B2 (en) 2014-05-15 2018-04-13 Compounds for treating spinal muscular atrophy
IL270027A IL270027B (en) 2014-05-15 2019-10-17 Compounds for treating spinal muscular atrophy
HRP20192159TT HRP20192159T1 (en) 2014-05-15 2019-11-29 Compounds for treating spinal muscular atrophy
US17/099,578 US11827646B2 (en) 2014-05-15 2020-11-16 Compounds for treating spinal muscular atrophy
FR21C1039C FR21C1039I2 (en) 2014-05-15 2021-09-09 COMPOUNDS FOR THE TREATMENT OF SPINAL TUSKIVE ATROPHY
NO2021035C NO2021035I1 (en) 2014-05-15 2021-09-10 Risdiplam or a pharmaceutically acceptable salt thereof
LTPA2021010C LTPA2021010I1 (en) 2014-05-15 2021-09-14
NL301128C NL301128I2 (en) 2014-05-15 2021-09-15 Risdiplam or a pharmaceutically acceptable salt thereof
US18/496,034 US20240076302A1 (en) 2014-05-15 2023-10-27 Compounds for treating spinal muscular atrophy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461993839P 2014-05-15 2014-05-15
US61/993,839 2014-05-15

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/351,267 Continuation US9969754B2 (en) 2014-05-15 2016-11-14 Compounds for treating spinal muscular atrophy

Publications (1)

Publication Number Publication Date
WO2015173181A1 true WO2015173181A1 (en) 2015-11-19

Family

ID=53175048

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/060343 WO2015173181A1 (en) 2014-05-15 2015-05-11 Compounds for treating spinal muscular atrophy

Country Status (36)

Country Link
US (1) US9969754B2 (en)
EP (3) EP3143025B1 (en)
JP (1) JP6236173B2 (en)
KR (2) KR102213740B1 (en)
CN (1) CN106459092B (en)
AR (1) AR100442A1 (en)
AU (1) AU2015261046C1 (en)
BR (1) BR112016026205B1 (en)
CA (1) CA2948561C (en)
CL (1) CL2016002836A1 (en)
CR (1) CR20160518A (en)
DK (1) DK3143025T3 (en)
EA (2) EA035068B1 (en)
ES (2) ES2761423T3 (en)
FR (1) FR21C1039I2 (en)
HR (2) HRP20230637T1 (en)
HU (2) HUE046491T2 (en)
IL (2) IL248653B (en)
LT (1) LTPA2021010I1 (en)
MA (2) MA39995B1 (en)
MX (1) MX371050B (en)
MY (1) MY174284A (en)
NL (1) NL301128I2 (en)
NO (1) NO2021035I1 (en)
NZ (1) NZ725008A (en)
PE (1) PE20170128A1 (en)
PH (1) PH12016502081A1 (en)
PL (2) PL3663296T3 (en)
PT (1) PT3143025T (en)
RS (1) RS59718B1 (en)
SG (1) SG11201609497TA (en)
SI (2) SI3143025T1 (en)
TW (1) TWI667239B (en)
UA (1) UA119670C2 (en)
WO (1) WO2015173181A1 (en)
ZA (1) ZA201607026B (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017080967A1 (en) * 2015-11-12 2017-05-18 F. Hoffmann-La Roche Ag Compositions for treating spinal muscular atrophy
US9956223B2 (en) 2013-08-19 2018-05-01 Hoffmann-La Roche Inc. Screening method
WO2018098446A1 (en) * 2016-11-28 2018-05-31 Ptc Therapeutics, Inc Methods for modulating rna splicing
CN108137601A (en) * 2015-11-12 2018-06-08 豪夫迈·罗氏有限公司 For treating the compound of amyotrophic lateral sclerosis
US10208067B2 (en) 2015-05-20 2019-02-19 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
WO2019057740A1 (en) 2017-09-22 2019-03-28 F. Hoffmann-La Roche Ag Process for the prepration of 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one derivatives
WO2019068604A1 (en) * 2017-10-03 2019-04-11 F. Hoffmann-La Roche Ag New treatment of sma
US10562903B2 (en) 2015-12-10 2020-02-18 Hoffmann-La Roche Inc. Bridged piperidine derivatives
WO2020079203A1 (en) 2018-10-19 2020-04-23 F. Hoffmann-La Roche Ag New forms of pyrido[1,2-a]pyrimidin-4-one derivatives, its formulation and its process of making
US10668171B2 (en) 2015-05-30 2020-06-02 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US10874672B2 (en) 2015-12-10 2020-12-29 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US10882868B2 (en) * 2014-05-15 2021-01-05 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
WO2021021775A1 (en) 2019-07-31 2021-02-04 Teva Pharmaceuticals International Gmbh Solid state forms of risdiplam and process for preparation thereof
US11382918B2 (en) 2017-06-28 2022-07-12 Ptc Therapeutics, Inc. Methods for treating Huntington's Disease
US11395822B2 (en) 2017-06-28 2022-07-26 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
WO2022162107A1 (en) 2021-02-01 2022-08-04 Sandoz Ag Crystalline form of risdiplam
US11407753B2 (en) 2017-06-05 2022-08-09 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
WO2022194801A1 (en) 2021-03-17 2022-09-22 F. Hoffmann-La Roche Ag New thiazolopyrimidinone derivatives
WO2022194802A1 (en) * 2021-03-17 2022-09-22 F. Hoffmann-La Roche Ag New thiadiazolopyrimidone derivatives
WO2022194909A2 (en) 2021-03-18 2022-09-22 F. Hoffmann-La Roche Ag Novel process
US11608501B2 (en) 2017-06-14 2023-03-21 Ptc Therapeutics, Inc. Methods for modifying RNA splicing
US11685746B2 (en) 2018-06-27 2023-06-27 Ptc Therapeutics, Inc. Heteroaryl compounds for treating Huntington's disease
US11780839B2 (en) 2018-03-27 2023-10-10 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11858941B2 (en) 2018-06-27 2024-01-02 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating Huntington's disease
WO2024069646A1 (en) * 2022-09-26 2024-04-04 Natco Pharma Limited Improved process for the preparation of risdiplam and its intermediates
WO2024081932A1 (en) 2022-10-14 2024-04-18 Genentech, Inc. Methods for treating spinal muscular atrophy

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX354074B (en) 2012-02-10 2018-02-12 Ptc Therapeutics Inc Compounds for treating spinal muscular atrophy.
CN113966219A (en) 2019-06-12 2022-01-21 豪夫迈·罗氏有限公司 Novel treatment of SMA
WO2022048675A1 (en) * 2020-09-07 2022-03-10 苏州科睿思制药有限公司 Crystal form of risdiplam, preparation method therefor, and use thereof
WO2023057404A1 (en) 2021-10-06 2023-04-13 F. Hoffmann-La Roche Ag Novel combined administration
WO2023202501A1 (en) * 2022-04-18 2023-10-26 深圳市塔吉瑞生物医药有限公司 Substituted pyridopyrimidinone compound, composition comprising same, and use thereof
WO2023240236A1 (en) 2022-06-10 2023-12-14 Voyager Therapeutics, Inc. Compositions and methods for the treatment of spinal muscular atrophy related disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009151546A2 (en) * 2008-05-27 2009-12-17 Ptc Therapeutics, Inc. Methods for treating spinal muscular atrophy
WO2010019326A1 (en) * 2008-08-14 2010-02-18 Cardiac Pacemakers, Inc. Performance assessment and adaptation of an acoustic communication link
WO2013119916A2 (en) * 2012-02-10 2013-08-15 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20000029843A (en) * 1996-08-06 2000-05-25 디. 제이. 우드;스피겔 알렌 제이 Substituted pyrido- or pyrimido-containing 6,6- or 6,7-bicyclic derivatives
CA2700841A1 (en) 2007-09-27 2009-04-02 The United States Of America, As Represented By The Secretary, Departmen T Of Health And Human Services Isoindoline compounds for the treatment of spinal muscular atrophy and other uses
KR20100012134A (en) * 2008-07-28 2010-02-08 신호열 A mouse for notebook-pc
US8986935B2 (en) 2008-08-13 2015-03-24 Ptc Therapeutics, Inc. Methods for treating spinal muscular atrophy
MX352861B (en) * 2011-12-30 2017-12-13 Ptc Therapeutics Inc Compounds for treating spinal muscular atrophy.
AU2013289938A1 (en) 2012-07-13 2015-01-29 Indiana University Research & Technology Corporation Compounds for treatment of spinal muscular atrophy
MX2018005890A (en) 2015-11-12 2018-08-15 Hoffmann La Roche Compositions for treating spinal muscular atrophy.

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009151546A2 (en) * 2008-05-27 2009-12-17 Ptc Therapeutics, Inc. Methods for treating spinal muscular atrophy
WO2010019326A1 (en) * 2008-08-14 2010-02-18 Cardiac Pacemakers, Inc. Performance assessment and adaptation of an acoustic communication link
WO2013119916A2 (en) * 2012-02-10 2013-08-15 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9956223B2 (en) 2013-08-19 2018-05-01 Hoffmann-La Roche Inc. Screening method
US10702530B2 (en) 2013-08-19 2020-07-07 Hoffmann-La Roche Inc. Screening method
US10882868B2 (en) * 2014-05-15 2021-01-05 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
US20210147447A1 (en) * 2014-05-15 2021-05-20 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
US11827646B2 (en) 2014-05-15 2023-11-28 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
US10208067B2 (en) 2015-05-20 2019-02-19 Hoffmann-La Roche Inc. Compounds for treating spinal muscular atrophy
US11602567B2 (en) 2015-05-30 2023-03-14 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
US10668171B2 (en) 2015-05-30 2020-06-02 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
KR20180080317A (en) * 2015-11-12 2018-07-11 에프. 호프만-라 로슈 아게 Composition for treating spinal muscular atrophy
AU2016353961B2 (en) * 2015-11-12 2019-08-29 F. Hoffmann-La Roche Ag Compositions for treating spinal muscular atrophy
WO2017080967A1 (en) * 2015-11-12 2017-05-18 F. Hoffmann-La Roche Ag Compositions for treating spinal muscular atrophy
US20180289713A1 (en) * 2015-11-12 2018-10-11 Hoffmann-La Roche Inc. Compounds for treating amyotrophic lateral sclerosis
CN108137601A (en) * 2015-11-12 2018-06-08 豪夫迈·罗氏有限公司 For treating the compound of amyotrophic lateral sclerosis
US11938136B2 (en) 2015-11-12 2024-03-26 Hoffmann-La Roche Inc. Compositions for treating spinal muscular atrophy
KR102162062B1 (en) 2015-11-12 2020-10-07 에프. 호프만-라 로슈 아게 Composition for treatment of spinal muscular dystrophy
EA036399B1 (en) * 2015-11-12 2020-11-06 Ф. Хоффманн-Ля Рош Аг Compositions for treating spinal muscular atrophy
US10874672B2 (en) 2015-12-10 2020-12-29 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US10562903B2 (en) 2015-12-10 2020-02-18 Hoffmann-La Roche Inc. Bridged piperidine derivatives
US10881658B2 (en) 2015-12-10 2021-01-05 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11638706B2 (en) 2015-12-10 2023-05-02 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
JP2019535789A (en) * 2016-11-28 2019-12-12 ピーティーシー セラピューティクス,インコーポレーテッド Methods for modulating RNA splicing
WO2018098446A1 (en) * 2016-11-28 2018-05-31 Ptc Therapeutics, Inc Methods for modulating rna splicing
US11702646B2 (en) 2016-11-28 2023-07-18 Ptc Therapeutics, Inc. Methods for modulating RNA splicing
CN110352007A (en) * 2016-11-28 2019-10-18 Ptc医疗公司 Method for adjusting RNA montage
US11407753B2 (en) 2017-06-05 2022-08-09 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11608501B2 (en) 2017-06-14 2023-03-21 Ptc Therapeutics, Inc. Methods for modifying RNA splicing
US11395822B2 (en) 2017-06-28 2022-07-26 Ptc Therapeutics, Inc. Methods for treating Huntington's disease
US11382918B2 (en) 2017-06-28 2022-07-12 Ptc Therapeutics, Inc. Methods for treating Huntington's Disease
IL273268B2 (en) * 2017-09-22 2023-05-01 Hoffmann La Roche Process for the prepration of 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one derivatives
CN111132981A (en) * 2017-09-22 2020-05-08 豪夫迈·罗氏有限公司 Process for the preparation of 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (2, 8-dimethylimidazo [1,2-B ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one derivatives
WO2019057740A1 (en) 2017-09-22 2019-03-28 F. Hoffmann-La Roche Ag Process for the prepration of 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one derivatives
CN111132981B (en) * 2017-09-22 2023-10-31 豪夫迈·罗氏有限公司 Process for preparing pyrido [1,2-A ] pyrimidin-4-one derivatives
TWI812646B (en) * 2017-09-22 2023-08-21 瑞士商赫孚孟拉羅股份公司 Novel process
IL273268A (en) * 2017-09-22 2020-04-30 Hoffmann La Roche Process for the prepration of 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one derivatives
US11390632B2 (en) 2017-09-22 2022-07-19 Hoffmann-La Roche Inc. Process for the preparation of 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo [1,2-B]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one derivatives
WO2019068604A1 (en) * 2017-10-03 2019-04-11 F. Hoffmann-La Roche Ag New treatment of sma
JP2020536090A (en) * 2017-10-03 2020-12-10 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft New treatment for SMA
US11534444B2 (en) 2017-10-03 2022-12-27 Hoffmann-La Roche Inc. Treatment of SMA
JP7423515B2 (en) 2017-10-03 2024-01-29 エフ. ホフマン-ラ ロシュ アーゲー New treatment for SMA
US11780839B2 (en) 2018-03-27 2023-10-10 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
US11685746B2 (en) 2018-06-27 2023-06-27 Ptc Therapeutics, Inc. Heteroaryl compounds for treating Huntington's disease
US11858941B2 (en) 2018-06-27 2024-01-02 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating Huntington's disease
WO2020079203A1 (en) 2018-10-19 2020-04-23 F. Hoffmann-La Roche Ag New forms of pyrido[1,2-a]pyrimidin-4-one derivatives, its formulation and its process of making
WO2021021775A1 (en) 2019-07-31 2021-02-04 Teva Pharmaceuticals International Gmbh Solid state forms of risdiplam and process for preparation thereof
WO2022162107A1 (en) 2021-02-01 2022-08-04 Sandoz Ag Crystalline form of risdiplam
WO2022194801A1 (en) 2021-03-17 2022-09-22 F. Hoffmann-La Roche Ag New thiazolopyrimidinone derivatives
WO2022194802A1 (en) * 2021-03-17 2022-09-22 F. Hoffmann-La Roche Ag New thiadiazolopyrimidone derivatives
WO2022194909A2 (en) 2021-03-18 2022-09-22 F. Hoffmann-La Roche Ag Novel process
WO2022194909A3 (en) * 2021-03-18 2023-04-06 F. Hoffmann-La Roche Ag Process for preparing risdiplam
WO2024069646A1 (en) * 2022-09-26 2024-04-04 Natco Pharma Limited Improved process for the preparation of risdiplam and its intermediates
WO2024081932A1 (en) 2022-10-14 2024-04-18 Genentech, Inc. Methods for treating spinal muscular atrophy

Also Published As

Publication number Publication date
CN106459092B (en) 2019-10-15
MA39995B1 (en) 2019-11-29
KR20170003687A (en) 2017-01-09
NL301128I2 (en) 2021-10-25
ES2949660T3 (en) 2023-10-02
MX2016014547A (en) 2017-02-23
US20170197990A1 (en) 2017-07-13
CR20160518A (en) 2017-02-21
US9969754B2 (en) 2018-05-15
LTPA2021010I1 (en) 2021-10-11
FR21C1039I2 (en) 2022-09-02
EP4241772A3 (en) 2023-11-15
BR112016026205B1 (en) 2021-12-07
ES2761423T3 (en) 2020-05-19
IL248653B (en) 2019-11-28
NZ725008A (en) 2019-11-29
IL270027B (en) 2020-08-31
KR102256013B1 (en) 2021-05-26
CA2948561C (en) 2019-10-22
EP4241772A2 (en) 2023-09-13
AU2015261046B2 (en) 2019-04-18
HRP20230637T1 (en) 2023-09-29
CA2948561A1 (en) 2015-11-19
MX371050B (en) 2020-01-14
ZA201607026B (en) 2022-05-25
KR20210014219A (en) 2021-02-08
EA201692280A1 (en) 2018-07-31
EA035068B1 (en) 2020-04-23
TWI667239B (en) 2019-08-01
CN106459092A (en) 2017-02-22
UA119670C2 (en) 2019-07-25
EA202090486A2 (en) 2020-06-30
HRP20192159T1 (en) 2020-02-21
NO2021035I1 (en) 2021-09-10
JP6236173B2 (en) 2017-11-22
PL3663296T3 (en) 2023-08-07
HUS2100037I1 (en) 2021-10-28
PT3143025T (en) 2019-12-03
AU2015261046C1 (en) 2019-07-25
MA51988A (en) 2021-01-20
EP3663296B1 (en) 2023-05-17
PL3143025T3 (en) 2020-03-31
PE20170128A1 (en) 2017-03-16
TW201609738A (en) 2016-03-16
BR112016026205A8 (en) 2021-07-20
RS59718B1 (en) 2020-01-31
AR100442A1 (en) 2016-10-05
DK3143025T3 (en) 2019-12-09
BR112016026205A2 (en) 2017-08-15
EP3663296A1 (en) 2020-06-10
MY174284A (en) 2020-04-01
FR21C1039I1 (en) 2021-10-15
CL2016002836A1 (en) 2017-04-21
AU2015261046A1 (en) 2016-10-27
IL248653A0 (en) 2017-01-31
JP2017515863A (en) 2017-06-15
EP3143025A1 (en) 2017-03-22
PH12016502081A1 (en) 2017-01-09
SG11201609497TA (en) 2016-12-29
EP3143025B1 (en) 2019-10-09
SI3663296T1 (en) 2023-08-31
SI3143025T1 (en) 2020-01-31
EA202090486A3 (en) 2020-08-31
KR102213740B1 (en) 2021-02-09
HUE046491T2 (en) 2020-03-30
NL301128I1 (en) 2021-09-22

Similar Documents

Publication Publication Date Title
AU2015261046B2 (en) Compounds for treating spinal muscular atrophy
US10208067B2 (en) Compounds for treating spinal muscular atrophy
CA2952895C (en) Imidazo[1,2-a]pyrazin-1yl-benzamide compounds for treating spinal muscular atrophy
WO2017081111A1 (en) Compounds for treating amyotrophic lateral sclerosis
US20240076302A1 (en) Compounds for treating spinal muscular atrophy
EA040467B1 (en) METHOD FOR PRODUCING COMPOUNDS FOR THE TREATMENT OF SPINAL MUSCLE ATROPHY

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: 15721701

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015721701

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015721701

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12016502081

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: NC2016/0003234

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 002128-2016

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2015261046

Country of ref document: AU

Date of ref document: 20150511

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 248653

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/014547

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: CR2016-000518

Country of ref document: CR

ENP Entry into the national phase

Ref document number: 2948561

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016567816

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016026205

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 201692280

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20167034983

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016026205

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20161109