US20160184305A1 - 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions - Google Patents

1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions Download PDF

Info

Publication number
US20160184305A1
US20160184305A1 US14/909,052 US201414909052A US2016184305A1 US 20160184305 A1 US20160184305 A1 US 20160184305A1 US 201414909052 A US201414909052 A US 201414909052A US 2016184305 A1 US2016184305 A1 US 2016184305A1
Authority
US
United States
Prior art keywords
methyl
pyridazin
amino
tetramethylpiperidin
alkyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/909,052
Other languages
English (en)
Inventor
Atwood Kim Cheung
Natalie Dales
Timothy Brian Hurley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Novartis AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis AG filed Critical Novartis AG
Priority to US14/909,052 priority Critical patent/US20160184305A1/en
Publication of US20160184305A1 publication Critical patent/US20160184305A1/en
Assigned to NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH INC. reassignment NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEUNG, ATWOOD KIM, HURLEY, TIMOTHY BRIAN, DALES, NATALIE
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH INC.
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • 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
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • 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
    • C07D487/10Spiro-condensed systems

Definitions

  • Proximal spinal muscular atrophy is an inherited, clinically heterogeneous group of neuromuscular disorders characterized by degeneration of the anterior horn cells of the spinal cord. Patients suffer from symmetrical weakness of trunk and limb muscles, the legs being more affected than the arms and the proximal muscles weaker than the distal ones; diaphragm, facial and ocular muscles are spared.
  • SMA spinal muscular atrophy
  • Type I (Werdnig-Hoffmann disease) is the most acute and severe form, with onset before six months and death usually before two years; children are never able to sit without support. Symptoms of the disease can be present in utero, as reduction of fetal movements; at birth; or more often, within the first four months of life. Affected children are particularly floppy, experience feeding difficulties and diaphragmatic breathing, and are characterized by a general weakness in the intercostals and accessory respiratory muscles. Affected children never sit or stand and usually die before the age of 2; death is generally due to respiratory insufficiency.
  • Type II intermediate, chronic form
  • muscular fasciculations are common, and tendon reflexes progressively reduce.
  • Children are unable to stand or walk without aid.
  • Feeding and swallowing problems are not usually present in Type II SMA, although in some patients a feeding tube may become necessary.
  • Most patients generally develop a progressive muscular scoliosis which can require surgical correction.
  • clearing of tracheal secretions and coughing might become difficult because of poor bulbar function and weak intercostal muscles.
  • These patients have profound hypotonia, symmetrical flaccid paralysis, and no control of head movement.
  • Type III Kergelberg-Welander disease, or Juvenile Spinal Muscular Atrophy
  • motor milestones achievement is normal, and deambulation can be preserved until variable ages.
  • These patients often develop scoliosis, and symptoms of joint overuse, generally caused by weakness, are frequently seen. Life expectancy is almost normal but quality of life is markedly compromised.
  • Types I, II and III progress over time, accompanied by deterioration of the patient's condition.
  • Type IV SMA is characterized by weakness in the second or third decade of life, with mild motor impairment not accompanied by respiratory or nutritional problems.
  • Adult SMA is characterized by insidious onset and very slow progression. The bulbar muscles are rarely affected in Type IV. It is not clear that Type IV SMA is etiologically related to the Type I-III forms.
  • spinal muscular atrophy examples include X-linked disease, spinal muscular atrophy with respiratory distress (SMARD), spinal and bulbar muscular atrophy (Kennedy's disease, or Bulbo-Spinal Muscular Atrophy), and distal spinal muscular atrophy.
  • SMARD spinal muscular atrophy with respiratory distress
  • Kennedy's disease spinal and bulbar muscular atrophy
  • Bulbo-Spinal Muscular Atrophy spinal muscular atrophy
  • SMA is due to mutations in the Survival of Motor Neuron (SMN) gene, which exists in two forms in humans (SMN1 and SMN2). Loss of SMN is deleterious to motor neurons and results in neuromuscular insufficiency, a hallmark of the disease. From a genetic point of view, SMA is an autosomal recessive condition, caused by disruption of SMN1 gene, located in 5q13 (Lefebvre S., et al. (1995) Cell 80: 155-165). More than 98% of patients with spinal muscular atrophy have a homozygous disruption of SMN1 by deletion, rearrangement, or mutation. All these patients, however, retain at least one copy of SMN2.
  • the mRNA transcribed from the SMN1 gene is generally a full-length mRNA with only a small fraction of its transcripts spliced to remove exon 3, 5, or 7 (Gennarelli et al. (1995) Biochem. Biophys. Res. Commun. 213:342-348; Jong et al. (2000) J. Neurol. Sci. 173:147-153). All SMA subjects have at least one, and generally two to four copies of the SMN2 gene, which encodes the same protein as SMN1; however, the SMN2 gene produces only low levels of full-length SMN protein.
  • the SMN ⁇ 7 protein is non-functional and thought to be rapidly degraded. About 10% of SMN2 pre-mRNA is properly spliced and subsequently translated into full length SMN protein (FL-SMN), and the rest being the SMN ⁇ 7 copy.
  • FL-SMN full length SMN protein
  • the efficiency of SMN2 splicing might be dependent on severity of disease, and production of a full length transcript of SMN2 could range from 10% to 50%.
  • presence or absence of the SMN1 gene roughly 90% of which becomes the FL-SMN gene product and protein, influences the severity of SMA by whether or not it can compensate for the truncated SMN ⁇ 7 copies.
  • a low level of SMN protein allows embryonic development, but is not sufficient to sustain the survival of motor neurons of the spinal cord.
  • a subclass of neurogenic-type arthrogryposis multiplex congenita (congenital AMC) has separately been reported to involve SMN1 gene deletion, suggesting that some degree of pathology in those afflicted is likely due to low levels of motor neuron SMN.
  • Congenital AMC affects humans and animals, e.g., horses, cattle, sheep, goats, pigs, dogs, and cats.
  • ALS amyotrophic lateral sclerosis
  • the invention provides compounds, salts thereof, pharmaceutical formulations thereof and combinations thereof which compounds are Spinal Muscular Atrophy modulators.
  • the invention further provides methods of treating, preventing, or ameliorating Spinal Muscular Atrophy, comprising administering to a subject in need thereof an effective amount of an SMN modulator (e.g., a compound of the invention).
  • an SMN modulator e.g., a compound of the invention.
  • SMN modulators provided herein are compounds of Formula IA and salts thereof:
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound according to the definition of formula (I) or subformulae thereof and one or more pharmaceutically acceptable carriers.
  • the invention provides a combination, in particular a pharmaceutical combination, comprising a therapeutically effective amount of the compound according to the definition of formula (I) or subformulae thereof and one or more therapeutically active.
  • One embodiment of the invention is to provide a method for treating, preventing, or ameliorating an SMN-deficiency-related condition, comprising administering to a subject in need thereof an effective amount of an SMN modulator, or a pharmaceutical composition comprising the same.
  • Another embodiment of the invention is a method of modulating SMN protein through the administration of an SMN modulator.
  • said SMN modulator is capable of increasing one or more of FL-SMN or SMN ⁇ 7 levels.
  • said SMN modulator is capable of preventing exon 7 from being spliced from the SMN transcript.
  • the present invention is based on the discovery that the SMN modulators of the invention (e.g., compounds of formula (I) and/or compounds of formula (I-A) are capable of modulating SMN proteins, e.g., through SMN promoter activation, splicing modulation (e.g., preventing exon7 from being spliced out of the SMN gene), and/or SMN protein stability modulation.
  • the SMN modulators of the invention e.g., compounds of formula (I) and/or compounds of formula (I-A) are capable of modulating SMN proteins, e.g., through SMN promoter activation, splicing modulation (e.g., preventing exon7 from being spliced out of the SMN gene), and/or SMN protein stability modulation.
  • the present invention provides compounds that modulate SMN activity. Such compounds may be used in vitro or in vivo to modulate (preferably increase) SMN production and activity in a variety of contexts.
  • the invention provides compounds of Formula (IA) and pharmaceutically acceptable salts thereof, which modulate SMN activity.
  • Compounds of Formula I are represented by the structure (IA):
  • A is bicyclic heteroaryl or heterocyle having 9 or 10 ring atoms and 1 or 2 ring N atoms and or 1 O atoms, which bicyclic heteroaryl or heterocycle is substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from —C(O)NH 2 , —C(O)O—C 1 -C 4 alkyl, aryl, oxo, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 al
  • R, R 1 , R 2 , R 3 , and R 4 are independently selected from the group consisting of hydrogen, C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino;
  • R 5 and R 6 are independently selected from hydrogen and fluorine; or
  • R and R 3 taken in combination form a fused 5 or 6 member heterocyclic ring having 0 or 1 additional ring heteroatoms selected from N, O or S;
  • R, and R 3 taken in combination form a C 1 -C 3 alkylene group;
  • R 1 and R 5 taken in combination form a C 1 -C 3 alkylene group;
  • R 3 and R 4 taken in combination with the carbon atom to which they attach, form a spirocyclicC 3 -C 6 cycloalkyl;
  • X is CR A R B , O, NR 7 or
  • R 9 and R 13 are independently selected from hydrogen and C 1 -C 4 alkyl
  • R 10 and R 14 are independently selected from hydrogen, amino, mono- and di-C 1 -C 4 akylamino and C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino
  • R 11 is hydrogen, C 1 -C 4 alkyl, amino or mono- and di-C 1 -C 4 akylamino
  • R 12 is hydrogen or C 1 -C 4 alkyl
  • R 9 and R 11 taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a
  • the invention is a the compound, or salt thereof, according to the first embodiment wherein the compound is of Formula (I):
  • A is bicyclic heteroaryl having 10 ring atoms and 1 or 2 ring N atoms, which bicyclic heteroaryl is substituted with 0, 1, or 2 substituents independently selected from oxo, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di
  • R, R 1 , R 2 , R 3 , and R 4 are independently selected from the group consisting of hydrogen, C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino;
  • R 5 and R 6 are independently selected from hydrogen and fluorine; or
  • R and R 3 taken in combination form a fused 5 or 6 member heterocyclic ring having 0 or 1 additional ring heteroatoms selected from N, O or S;
  • R, and R 3 taken in combination form a C 1 -C 3 alkylene group;
  • R 1 and R 5 taken in combination form a C 1 -C 3 alkylene group;
  • R 3 and R 4 taken in combination with the carbon atom to which they attach, form a spirocyclicC 3 -C 6 cycloalkyl;
  • X is CR A R B , O, NR 7 or
  • R 9 and R 13 are independently selected from hydrogen and C 1 -C 4 alkyl
  • R 10 and R 14 are independently selected from hydrogen, amino, mono- and di-C 1 -C 4 akylamino and C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino
  • R 11 is hydrogen, C 1 -C 4 alkyl, amino or mono- and di-C 1 -C 4 akylamino
  • R 12 is hydrogen or C 1 -C 4 alkyl
  • R 9 and R 11 taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a
  • the invention is the compound according to any one the first or second embodiments, or salt thereof, wherein A is selected from:
  • u and v are each, independently, 0, 1, 2 or 3; and each R a and R b are, independently, selected from, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, and C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound of any one of the first through third embodiments, or salt thereof, wherein A is selected from:
  • u and v are each, independently, 0, 1, 2 or 3; and each R a and R b are, independently, selected from, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, and C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound according to any one of the first through fourth embodiments, wherein A is substituted in the ortho position with a hydroxyl group.
  • the invention is the compound according to any one of the first through fourth embodiments, or salt thereof, wherein A is selected from:
  • the invention is the compound of any one of the first through fifth embodiments, or a salt thereof, wherein A has a single N atom
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula II:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, or salt thereof, according to any one of the first through sixth embodiments, wherein the compound is of formula III:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula IV:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula V:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula VI:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula VII:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound, according to any one of the first through sixth embodiments, or salt thereof, wherein the compound is of formula VIII:
  • R c and R d are each, independently, selected from hydrogen, cyano, halogen, hydroxy, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 3 -C 7 cycloalkyl, heterocyclyl, heteroaryl, heterocyclyl C 1 -C 4 alkyl, C 1 -C 4 alkyl aryl, C 1 -C 4 alkyl heterocyclyl, C 1 -C 4 alkyl heteroaryl, C 1 -C 4 alkoxy aryl, C 1 -C 4 alkoxy heterocyclyl, C 1 -C 4 alkoxy heteroaryl, C 1 -C 4 alkoxy substituted with hydroxy, C 1 -C 4 alkoxy, amino and mono- and di-C 1 -C 4 alkylamino.
  • the invention is the compound according to any one of the first through thirteenth embodiments, or a salt thereof, wherein B is a group of the formula:
  • R, R 1 , R 2 , R 3 , and R 4 are independently selected from the group consisting of hydrogen, C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino;
  • R 5 and R 6 are hydrogen; or
  • R and R 3 taken in combination form a fused 5 or 6 member heterocyclic ring having 0 or 1 additional ring heteroatoms selected from N, O or S;
  • R, and R 3 taken in combination form a C 1 -C 3 alkylene group;
  • R 1 and R 5 taken in combination form a C 1 -C 3 alkylene group;
  • R 3 and R 4 taken in combination with the carbon atom to which they attach, form a spirocyclicC 3 -C 6 cycloalkyl;
  • X is CR A R B , O, NR 7 or a bond;
  • R A and R 4 are independently selected from the group consisting of hydrogen,
  • the invention is the compound according to any one of the first through thirteenth embodiments, or a salt thereof, wherein B is a group of the formula:
  • R 9 and R 13 are independently selected from hydrogen and C 1 -C 4 alkyl
  • R 10 and R 14 are independently selected from hydrogen, amino, mono- and di-C 1 -C 4 akylamino and C 1 -C 4 alkyl, which alkyl is optionally substituted with hydroxy, amino or mono- and di-C 1 -C 4 akylamino
  • R 11 is hydrogen, C 1 -C 4 alkyl, amino or mono- and di-C 1 -C 4 akylamino
  • R 12 is hydrogen or C 1 -C 4 alkyl
  • R 9 and R 11 taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a saturated azacycle having 4 to 7 ring atoms which is optionally substituted with 1-3 C 1 -C 4 alkyl groups; or R 11 and R 12 , taken in combination form a
  • the invention is the compound of any one of the first through fifteenth embodiments, wherein B is selected from the group consisting of:
  • X is O or N(Me) or NH; and R 17 is hydrogen or methyl.
  • the invention is the compound, or salt thereof, according to any one of the first through sixteenth embodiments, wherein B is:
  • the invention is the compound, or salt thereof, according to any one of the first through seventeenth embodiments, wherein X is —O—.
  • the invention is the compound, or salt thereof, according to any one of the first through eighteenth embodiments, wherein X is N(Me).
  • the invention is a compound, or salt thereof, selected from the group consisting of:
  • the invention is a compound, or salt thereof, selected from the group consisting of:
  • the invention is a compound, or salt thereof, selected from the group consisting of:
  • the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of the first through twentythird embodiments, or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers.
  • the invention is a combination comprising a therapeutically effective amount of a compound according to any one of the first through twentysecond embodiments, or a pharmaceutically acceptable salt thereof and one or more therapeutically active co-agents.
  • the invention is a method to treat, prevent or ameliorate an SMN-deficiency-related condition, comprising administering to a subject in need thereof an effective amount of a compound or salt thereof of any one of the first through twentysecond embodiments.
  • the invention is the method of the twentyfifth embodiment, wherein said SMN-deficiency-related condition is Spinal Muscular Atrophy.
  • the invention is a compound according to any one of the first through twentysecond embodiments, or a pharmaceutically acceptable salt thereof, for use as a medicament.
  • the invention is a compound according to any one of the first through twentysecond embodiments, or a pharmaceutically acceptable salt thereof, for use in the treatment of an SMN-deficiency-related condition.
  • the invention is the compound according to the twentyeighth embodiment, or pharmaceutically acceptable salt thereof, for use in the treatment of spinal muscular atrophy.
  • the invention is the use of a compound according to any one of the first through twentysecond embodiments, or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of spinal muscular atrophy.
  • the term “SMN modulator” includes agents, such as the compounds of the invention, which possess the ability to modulate, e.g., increase, SMN protein levels by at least one of multiple possible mechanisms.
  • a non-limiting set of mechanisms includes SMN promoter activation, splicing modulation (e.g., preventing exon7 from being spliced out of the SMN gene), and SMN protein stability modulation.
  • SMN modulators can modulate, e.g., increase FL-SMN and/or SMN ⁇ 7 levels via any of said mechanisms, and/or can prevent SMN ⁇ 7 from being degraded.
  • SMA Spinal Muscular Atrophy
  • ALS amyotrophic lateral sclerosis
  • SMA Spinal Muscular Atrophy
  • Type I Wirednig-Hoffmann disease
  • Type II Intermediate, chronic form
  • Type III Kugelberg-Welander disease, or Juvenile Spinal Muscular Atrophy
  • SMA Spinal Muscular Atrophy
  • SMARD spinal muscular atrophy with respiratory distress
  • SMARD spinal and bulbar muscular atrophy
  • distal spinal muscular atrophy distal spinal muscular atrophy.
  • C 1-10 alkyl refers to a fully saturated branched or unbranched hydrocarbon moiety having 1 to 10 carbon atoms.
  • the terms “C 1-6 alkyl” and “C 1-4 alkyl” are to be construed accordingly.
  • C 1-10 alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl and n-decyl.
  • C 1-10 alkylene refers to divalent alkyl group as defined herein above having 1 to 10 carbon atoms.
  • the terms “C 1-6 alkylene” and “C 1-4 alkylene” are to be construed accordingly.
  • C 1-10 alkylene include, but are not limited to, methylene, ethylene, n-propylene, iso-propylene, n-butylene, sec-butylene, iso-butylene, tert-butylene, n-pentylene, isopentylene, neopentylene, n-hexylene, 3-methylhexylene, 2,2-dimethylpentylene, 2,3-dimethylpentylene, n-heptylene, n-octylene, n-nonylene and n-decylene.
  • haloC 1-10 alkyl refers to a C 1-10 alkyl group as defined herein, wherein at least one of the hydrogen atoms is replaced by a halo atom.
  • the haloC 1-10 alkyl group can be monohaloC 1-10 alkyl, dihaloC 1-10 alkyl or polyhaloC 1-10 alkyl including perhaloC 1-10 alkyl.
  • a monohaloC 1-10 alkyl can have one iodo, bromo, chloro or fluoro within the alkyl group.
  • DihaloC 1-10 alkyl and polyhaloC 1-10 alkyl groups can have two or more of the same halo atoms or a combination of different halo groups within the alkyl.
  • the polyhaloC 1-10 alkyl group contains up to 12, or 10, or 8, or 6, or 4, or 3, or 2 halo groups.
  • Non-limiting examples of haloC 1-10 alkyl include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • a perhaloC 1-10 alkyl group refers to an C 1-10 alkyl group having all hydrogen atoms replaced with halo atoms.
  • aryl refers to an aromatic hydrocarbon group having 6-20 carbon atoms in the ring portion. Typically, aryl is monocyclic, bicyclic or tricyclic aryl having 6-20 carbon atoms and includes one or more aromatic rings fused to one or more non-aromatic hydrocarbon rings. Non-limiting examples include phenyl, naphthyl or tetrahydronaphthyl.
  • C 1-10 alkoxy refers to C 1-10 alkyl-O—, wherein C 1-10 alkyl is defined herein above.
  • Representative examples of C 1-10 alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, hexyloxy, heptyloxy, octyloxy- and decyloxy-.
  • heterocyclyl or “heterocyclo” refers to a saturated or unsaturated non-aromatic ring or ring system, which is a 4-, 5-, 6-, or 7-membered monocyclic ring containing 1, or 3 heteroatoms selected from O, S and N, a 7-, 8-, 9-, 10-, 11-, or 12-membered bicyclic ring system containing 1, 2, 3, 4 or 5 heteroatoms selected from O, S and N, or a 10-, 11-, 12-, 13-, 14- or 15-membered tricyclic ring system and containing 1, 2, 3, 4, 5, 6 or 7 heteroatoms selected from O, S and N, where the N and S can also optionally be oxidized to various oxidation states.
  • the heterocyclic group can be attached via a heteroatom or a carbon atom.
  • the heterocyclyl can include fused or bridged rings as well as spirocyclic rings.
  • heterocycles include tetrahydrofuran (THF), dihydrofuran, 1, 4-dioxane, morpholine, 1,4-dithiane, piperazine, piperidine, 1,3-dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane, dithiolane, 1,3-dioxane, 1,3-dithiane, oxathiane and thiomorpholine.
  • C 3-12 cycloalkyl refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms.
  • C 3-18 cycloalkyl refers to a fully saturated or unsaturated monocyclic hydrocarbon group of 3-8 carbon atoms.
  • Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl.
  • Exemplary bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 6,6-dimethylbicyclo[3.1.1]heptyl, 2,6,6-trimethylbicyclo[3.1.1]heptyl, bicyclo[2.2.2]octyl.
  • Exemplary tricyclic hydrocarbon groups include, for example, adamantyl.
  • C 3-12 cycloalklyoxy refers to C 3-12 cycloalkyl-O—, wherein C 3-12 cycloalkyl is defined herein above.
  • Representative examples of C 3-12 cycloalklyoxy include, but are not limited to monocyclic groups such as cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclopentenyloxy, cyclohexyloxy and cyclohexenyloxy and the like.
  • Exemplary bicyclic hydrocarbon groups include bornyloxy, indyloxy, hexahydroindyloxy, tetrahydronaphthyloxy, decahydronaphthyloxy, bicyclo[2.1.1]hexyloxy, bicyclo[2.2.1]heptyloxy, bicyclo[2.2.1]heptenyloxy, 6,6-dimethylbicyclo[3.1.1]heptyloxy, 2,6,6-trimethylbicyclo[3.1.1]heptyloxy, bicyclo[2.2.2]octyloxy and the like.
  • Exemplary tricyclic hydrocarbon groups include, for example, adamantyloxy.
  • aryloxy refers to both an —O-aryl and an —O-heteroaryl group, wherein aryl and heteroaryl are defined herein.
  • heteroaryl refers to a 5-, 6-, or 7-membered monocyclic aromatic ring containing 1, 2, 3 or 4 heteroatoms selected from O, S and N, an 8-, 9-, or 10-membered fused bicyclic ring system containing 1, 2, 3, 4 or 5 heteroatoms selected from O, S and N, or an 11-, 12-, 13-, or 14-membered fused tricyclic ring system containing 1, 2, 3, 4, 5 or 6 heteroatoms selected from O, S and N, wherein at least one of the rings of the bicyclic or tricyclic ring systems is fully aromatic.
  • Typical heteroaryl groups include 2- or 3-thienyl, 2- or 3-furyl, 2- or 3-pyrrolyl, 2-, 4-, or 5-imidazolyl, 3-, 4-, or 5-pyrazolyl, 2-, 4-, or 5-thiazolyl, 3-, 4-, or 5-isothiazolyl, 2-, 4-, or 5-oxazolyl, 3-, 4-, or 5-isoxazolyl, 3- or 5-1,2,4-triazolyl, 4- or 5-1,2,3-triazolyl, tetrazolyl, 2-, 3-, or 4-pyridyl, 3- or 4-pyridazinyl, 3-, 4-, or 5-pyrazinyl, 2-pyrazinyl, 2-, 4-, or 5-pyrimidinyl, 1-, 2-, 3-, 5-, 6-, 7-, or 8-indolizinyl, 1-, 3-, 4-, 5-, 6-, or 7-isoindolyl, 2-, 3-, 4-, 5-, 6-, or 7-indolyl, 2-
  • halogen refers to fluoro, chloro, bromo, and iodo.
  • the term “isomers” refers to different compounds that have the same molecular formula but differ in arrangement and configuration of the atoms.
  • an optical isomer or “a stereoisomer” refers to any of the various stereo isomeric configurations which may exist for a given compound of the present invention and includes geometric isomers. It is understood that a substituent may be attached at a chiral center of a carbon atom. Therefore, the invention includes enantiomers, diastereomers or racemates of the compound. “Enantiomers” are a pair of stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a “racemic” mixture.
  • Diastereoisomers are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other.
  • the absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system. When a compound is a pure enantiomer the stereochemistry at each chiral carbon may be specified by either R or S.
  • Resolved compounds whose absolute configuration is unknown can be designated (+) or ( ⁇ ) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line.
  • Certain of the compounds described herein contain one or more asymmetric centers or axes and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the present invention is meant to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures.
  • Optically active (R)- and (S)-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration. All tautomeric forms are also intended to be included.
  • salt refers to an acid addition or base addition salt of a compound of the invention.
  • Salts include in particular “pharmaceutical acceptable salts.”
  • pharmaceutically acceptable salts refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable.
  • the compounds of the present invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from a parent compound, a basic or acidic moiety, by conventional chemical methods.
  • such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid.
  • a stoichiometric amount of the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 Cl, 125 I respectively.
  • the invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 13 C, and 14 C, are present.
  • isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, d 6 -acetone, d 6 -DMSO.
  • Compounds of the invention i.e. compounds of formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers.
  • These co-crystals may be prepared from compounds of formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of formula (I) with the co-crystal former under crystallization conditions and isolating co-crystals thereby formed.
  • Suitable co-crystal formers include those described in WO 2004/078163.
  • the invention further provides co-crystals comprising a compound of formula (I).
  • a therapeutically effective amount of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease, etc.
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a subject, is effective to (1) at least partially alleviate, inhibit, prevent and/or ameliorate a condition, or a disorder or a disease (i) mediated by Survival of Motor Neuron (SMN) gene or gene product, or by SMN ⁇ 7 degradation, or by the relative levels of FL-SMN and SMN ⁇ 7 (ii) associated with SMN activity, or (iii) characterized by activity (normal or abnormal) of SMN; or (2) reducing or inhibiting the activity of SMN; or (3) reducing or inhibiting the expression of SMN1 or SMN2.
  • SSN Motor Neuron
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of SMN; or at least partially reducing or inhibiting the expression of SMN, in both cases by modulating the relative levels of FL-SMN and SMN ⁇ 7.
  • therapeutically effective amount and “effective amount” are used herein to mean an amount sufficient to reduce by at least about 15 percent, preferably by at least 50 percent, more preferably by at least 90 percent, and most preferably prevent, a clinically significant deficit in the activity, function and response of the host. Alternatively, a therapeutically effective amount is sufficient to cause an improvement in a clinically significant condition/symptom in the host.
  • the effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular compound of the invention. For example, the choice of the compound of the invention can affect what constitutes an “effective amount.”
  • One of ordinary skill in the art would be able to study the factors contained herein and make the determination regarding the effective amount of the compounds of the invention without undue experimentation.
  • the regimen of administration can affect what constitutes an effective amount.
  • the compound of the invention can be administered to the subject either prior to or after the onset of an SMN-deficiency-related condition. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the compound(s) of the invention can be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
  • the term “subject” refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
  • primates e.g., humans, male or female
  • the subject is a primate.
  • the subject is a human.
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat,” “treating,” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically (e.g., through stabilization of a discernible symptom), physiologically, (e.g., through stabilization of a physical parameter), or both.
  • “treat,” “treating,” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • any asymmetric atom (e.g., carbon or the like) of the compound(s) of the present invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)-configuration.
  • each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R)- or (S)-configuration.
  • Substituents at atoms with unsaturated bonds may, if possible, be present in cis- (Z)- or trans- (E)-form.
  • a compound of the present invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof.
  • Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
  • any resulting racemates of final products or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound.
  • a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-O,O′-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid.
  • Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high pressure liquid chromatography
  • the compounds of the present invention may also form internal salts, e.g., zwitterionic molecules.
  • the compounds of the present invention can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms.
  • solvate refers to a molecular complex of a compound of the present invention (including pharmaceutically acceptable salts thereof) with one or more solvent molecules.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • hydrate refers to the complex where the solvent molecule is water.
  • the compounds of the present invention including salts, hydrates and solvates thereof, may inherently or by design form polymorphs.
  • the invention further includes any variant of the present processes, in which an intermediate product obtainable at any stage thereof is used as starting material and the remaining steps are carried out, or in which the starting materials are formed in situ under the reaction conditions, or in which the reaction components are used in the form of their salts or optically pure material.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and rectal administration, etc.
  • the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions).
  • compositions can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers and buffers, etc.
  • the pharmaceutical compositions are tablets or gelatin capsules comprising the active ingredient together with
  • diluents e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine;
  • lubricants e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also
  • lubricants e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol
  • binders e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired
  • disintegrants e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or
  • Tablets may be either film coated or enteric coated according to methods known in the art.
  • compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin or olive oil.
  • compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • Said compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75%, or contain about 1-50%, of the active ingredient.
  • compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier.
  • Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • compositions for topical application include aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like.
  • topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like. They are thus particularly suited for use in topical, including cosmetic, formulations well-known in the art.
  • Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
  • a dry powder either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids
  • the present invention further provides anhydrous pharmaceutical compositions and dosage forms comprising the compounds of the present invention as active ingredients, since water may facilitate the degradation of certain compounds.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e. g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • the compounds of formula I in free form or in salt form exhibit valuable pharmacological properties, e.g. full length SMN protein production modulating properties, e.g. as indicated in in vitro and in vivo tests as provided in the next sections, and are therefore indicated for therapy or for use as research chemicals, e.g. as tool compounds.
  • valuable pharmacological properties e.g. full length SMN protein production modulating properties, e.g. as indicated in in vitro and in vivo tests as provided in the next sections, and are therefore indicated for therapy or for use as research chemicals, e.g. as tool compounds.
  • the present invention provides the use of a compound of formula (I) or a salt thereof in therapy.
  • the therapy is selected from a disease which may be treated by modulating full length SMN protein production.
  • the disease is selected from the afore-mentioned list, suitably spinal muscular atrophy.
  • the invention provides a method of treating a disease which is treated by modulating full length SMN protein production comprising administration of a therapeutically acceptable amount of a compound of formula (I) or salt thereof to a patient in need of such therapy.
  • the disease is selected from the afore-mentioned list, suitably spinal muscular atrophy.
  • the present invention provides the use of a compound of formula (I) or salt thereof for the manufacture of a medicament.
  • the medicament is for treatment of a disease which may be treated by modulation of SMN protein production.
  • the disease is selected from the afore-mentioned list, suitably spinal muscular atrophy.
  • the pharmaceutical composition or combination of the present invention can be in unit dosage of about 0.01-1000 mg of active ingredient(s) for a subject of about 0.05-70 kg or about 1-20 kg, or about 1-500 mg or about 1-250 mg or about 1-150 mg or about 0.5-100 mg, or about 0.01-1 mg or about 0.01-0.1 mg or about 1-50 mg of active ingredients.
  • the therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
  • the above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof.
  • the compounds of the present invention can be applied in vitro in the form of solutions, e.g., aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution.
  • the dosage in vitro may range between about 10 ⁇ 3 molar and 10 ⁇ 9 molar concentrations.
  • a therapeutically effective amount in vivo may range depending on the route of administration, between about 0.1-500 mg/kg, or between about 1-100 mg/kg.
  • the compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent.
  • the compound of the present invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents.
  • the invention provides a product comprising a compound of formula (I) and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy.
  • the therapy is the treatment of a spinal muscular atrophy.
  • Products provided as a combined preparation include a composition comprising the compound of formula (I) and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound of formula (I) and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) and another therapeutic agent(s).
  • the pharmaceutical composition may comprise a pharmaceutically acceptable carrier, as described above.
  • the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (I).
  • the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit of the invention typically comprises directions for administration.
  • the compound of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the compound of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the compound of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the compound of the invention and the other therapeutic agent.
  • Suitable protecting groups include hydroxy, phenol, amino and carboxylic acid.
  • Suitable protecting groups for hydroxy or phenol include trialkylsilyl or diarylalkylsilyl (e.g., t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, substituted benzyl, methyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
  • Protecting groups may be added or removed in accordance with standard techniques, which are well-known to those skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T. W. and P. G. M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley.
  • the protecting group may also be a polymer resin, such as a Wang resin or a 2-chlorotrityl-chloride resin.
  • reaction schemes illustrate methods to make compounds of this invention. It is understood that one skilled in the art would be able to make these compounds by similar methods or by methods known to one skilled in the art.
  • starting components and reagents may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, Strem, other commercial vendors, or synthesized according to sources known to those skilled in the art, or prepared as described in this invention.
  • A, B, X, R, R 1 , R 2 , R 3 , R 4 are defined as in the Specification unless specifically defined.
  • pyridazine compounds of Formula (I) of this invention can be synthesized following the general procedure described in Scheme 1.
  • Di-halopyridazine (1) reacts in a displacement reaction or a metal-mediated cross coupling reaction (Buchwald) with an alcohol or an amine (B) to provide pyridazine intermediate (2).
  • Transition metal-mediated cross coupling reaction such as a Suzuki reaction, between halide compound (2) and a substituted aryl or heteroaryl compound A, such as a boronate acid or boronate ester, provides compound (3) of Formula (I) of the invention.
  • Di-halopyridazine (1) reacts in a transition metal-mediated cross coupling reaction, such as a Suzuki reaction, with a substituted aryl or heteroaryl compound A, such as a boronate acid or ester, to provide pyridazine intermediate (4).
  • Pyridazine intermediate (4) reacts via a displacement reaction with an alcohol or an amine (B) to provide pyridazine (3) of Formula (I) of the invention.
  • Di-halopyridazine (1) reacts in a transition metal-mediated cross coupling reaction, such as a Suzuki reaction, with a substituted aryl or heteroaryl compound A, such as a boronate acid or ester, to provide pyridazine intermediate (4).
  • a transition metal-mediated cross coupling reaction such as a Suzuki reaction
  • a substituted aryl or heteroaryl compound A such as a boronate acid or ester
  • Pyridazine intermediate (4) reacts via second metal-mediated cross coupling, such as a Suzuki reaction, to provide pyridazine (3) of Formula (I) of the invention.
  • All starting materials, building blocks, reagents, acids, bases, dehydrating agents, solvents, catalysts and scavengers utilized to synthesize the compounds of the present invention are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art. Further, the compounds of the present invention can be produced by organic synthesis methods known to one of ordinary skill in the art as shown in the following examples.
  • Butyllithium (1.6 M in heptane, 4.90 mL, 7.90 mmol) was added dropwise to a solution of 6-bromo-7-methoxyquinoline (1.71 g, 7.18 mmol) cooled to ⁇ 78° C. The solution was stirred for 0.5 h after which time trimethyl borate (2.0 mL, 18 mmol) was added in a single portion. The reaction was allowed to warm to room temperature and stir overnight. The crude reaction was concentrated to dryness, then concentrated from heptane (2 ⁇ ).
  • Oxalyl chloride (4.18 ml, 47.7 mmol) was added dropwise to a suspension of 4-bromo-5-fluoro-2-nitrobenzoic acid (11.46 g, 43.4 mmol) in DCM (175 mL) and catalytic DMF (100 uL). After 3 h the volatiles were removed via rotary evaporation. The remaining residue was redissolved into DCM (25 mL) and added portion-wise to rapidly stirring methanol (100 mL). The solvent was again removed and the residue concentrated from toluene (2 ⁇ ) to afford methyl 4-bromo-5-fluoro-2-nitrobenzoate as a yellow liquid (12.07 g) which was taken on without purification.
  • Zinc powder (6.24 g, 95.4 mmol) was added portion-wise over 30 min (Caution! exotherm possible) to a solution of methyl 4-bromo-5-methoxy-2-nitrobenzoate (3.076 g, 10.60 mmol) in 3:1 DCM/AcOH (55 mL). The mixture was stirred at room temperature for three days. The mixture was filtered through Celite®, washing the filter pad with EtOAc, and concentrated. The crude product was redissolved into EtOAc, washed with saturated NaHCO 3 , brine, dried over MgSO 4 and concentrated to a yellow oil.
  • Step 5 (2-Amino-4-bromo-5-methoxyphenyl) methanol
  • Step 3 (7-Methoxy-2-methyl-4-oxo-1,4-dihydroquinolin-6-yl)boronic acid and 7-Methoxy-2-methyl-6-(4, 4, 5, 5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-4(1H)-one
  • Step 4 7-Methoxy-2-methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-4(1H)-one
  • Step 5 6-(4-Chloro-7-methoxy-2-methylquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 6 4-Chloro-2-methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol
  • 6-(4-Chloro-7-methoxy-2-methylquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (195 mg, 0.43 mmol) was subjected to methoxy deprotection conditions as described in GENERAL METHOD 2-3 using boron tribromide (1 M solution in DCM, 2.49 mL, 2.49 mmol).
  • Step 1 5-(((4-Bromo-3-methoxyphenyl)amino)methylene)-2,2-dimethyl-1,3-dioxane-4,6-dione
  • Step 3 7-Methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-4(1H)-one and, (7-methoxy-4-oxo-1,4-dihydroquinolin-6-yl)boronic acid
  • Step 4 7-Methoxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-4(1H)-one
  • the vessel was evacuated and back-filled with nitrogen (4 ⁇ ), and heated via microwave irradiation at 110° C. for 1.5 hour.
  • the mixture was cooled to room temperature, diluted with MeOH (3 mL), filtered through Celite®, and washed with 9:1 MeOH/DCM and then with 1:1 MeOH/DCM.
  • the resulting filtrate was concentrated and acidified to pH 3 with 1 M HCl in diethyl ether.
  • Step 5 6-(4-Chloro-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 6 4-chloro-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol
  • 6-(4-Chloro-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (62 mg, 0.14 mmol) was subjected to methoxy deprotection conditions as described in GENERAL METHOD 2-3 using boron tribromide (1 M solution in DCM, 0.70 mL, 0.70 mmol).
  • Step 1 5-(((3-Chloro-4-methoxyphenyl)amino)methylene)-2,2-dimethyl-1,3-dioxane-4,6-dione
  • Triethoxymethane (11 mL, 64 mmol) and 2,2-dimethyl-1,3-dioxane-4,6-dione (Meldrum's acid, 10.7 g, 74.2 mmol) were added to 3-chloro-4-methoxyaniline (10.0 g, 63.5 mmol) in ethanol (60 mL). The mixture was stirred at reflux overnight. An additional portion of triethoxymethane (2.1 mL, 13 mmol) and 2,2-dimethyl-1,3-dioxane-4,6-dione (2.74 g, 19.0 mmol) were added to the reaction and the mixture was stirred at reflux for 3 h.
  • Step 2 7-Chloro-6-methoxyquinolin-4(1H)-one and 5-chloro-6-methoxyquinolin-4(1H)-one
  • Step 3 6-Methoxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-4(1H)-one
  • Step 4 6-(4-Chloro-6-methoxyquinolin-7-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 5 4-Chloro-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-6-ol
  • a mixture of a chloropyridazine intermediate, such as Intermediate 1 (1.0 equivalents), potassium triphosphate (5.0 equivalents), and 2 nd generation XPhos precatalyst (0.07 equivalents), in DMF (0.3 M) is degassed via vacuum/N 2 purge (3 ⁇ ), and heated at 50° C.
  • a 50 or 100 mg/mL solution of a boronic acid, such as Intermediate 7 or Intermediate 8 in DMF is added in 1 mL aliquots every 2-3 h until Intermediate 1 is consumed.
  • the reaction is cooled to room temperature, filtered through Celite® washing with EtOAc, and partially concentrated.
  • the crude product is purified by SCX (GENERAL METHOD 3-1) and silica gel chromatography.
  • a mixture of a chloropyridazine intermediate, such as Intermediate 1 (1.0 equivalent), boronic acid reagent (1.5-2 equivalents), and Na 2 CO 3 (3 equivalents) in 3:1 DME/water (0.12 M) in a microwave vial is degassed with a stream of dry nitrogen.
  • Pd(PPh 3 ) 4 (0.1 equivalents) is added to the reaction mixture and the vial is sealed and heated in a Biotage® Initiator microwave reactor at 140° C. for 30 min.
  • the mixture is diluted with water and extracted with DCM (4 ⁇ ).
  • the organic extracts are dried over MgSO 4 , filtered, and concentrated to afford the crude product.
  • the crude product is purified via silica chromatography as described in GENERAL METHOD 4-1.
  • the methoxy substrate (1 equivalent) and pyridine hydrochloride (20-30 equivalents) are heated at 160-190° C. for 15-120 min in a Biotage® Initiator microwave reactor.
  • the reaction mixture is solubilized with MeOH/DMSO and purified via preparative reverse-phase HPLC (15 to 45% acetonitrile in water, 5 mM ammonium hydroxide modifier).
  • the methoxy substrate (1 equivalent) and pyridine hydrochloride (20-25 equivalents) are heated at 160-190° C. for 15-120 min in a Biotage® Initiator microwave reactor.
  • the reaction mixture is solubilized with MeOH and the solution loaded onto a mixture of solid sodium bicarbonate (30 equivalents) and silica gel ( ⁇ 6 g/mmol methoxy substrate), and concentrated to dryness.
  • Column chromatography as described in GENERAL METHOD 4-1 provided the product.
  • the methoxy substrate (1.0 equivalent) is dissolved in DCM (0.03 M) and the solution cooled to 0° C. Boron tribromide (1 M solution in DCM, 3-10 equivalents) is added dropwise.
  • the crude reaction mixture is stirred at room temperature overnight. If the reaction has not gone to completion, it can be heated at reflux for 4-24 h.
  • the reaction mixture is diluted with DCM and water.
  • the organic layer is diluted with EtOAc and washed with saturated NaHCO 3 (2 ⁇ ), water, brine and dried over Na 2 SO 4 or MgSO 4 .
  • the crude product is purified via preparative reverse-phase HPLC, silica gel chromatography (GENERAL METHODS 4-1), or by recrystallization.
  • the crude product is dissolved in MeOH and loaded onto a SiliaBond Propylsulfonic Acid® cartridge (Silicycle, Inc.) or a Bond Elut SCX cartridge (Agilent Technologies, Inc.) preconditioned with MeOH or acetonitrile.
  • the cartridge is flushed with MeOH or acetonitrile, and the product released by elution with 2-7 N ammonia in MeOH. Concentration of the eluent provides the purified product.
  • the crude product (either neat, dissolved into DCM, or loaded onto a solid support such as silica gel or Celite®) is subjected to normal phase flash column chromatography using RediSep® Rf pre-packed silica gel cartridges (Teledyne Isco, Inc.) with an elution gradient of 1-20% ammonia in MeOH (1.5 N, or 3.5 N concentration), in DCM.
  • Residual palladium, and certain other metal contaminants can be scavenged from reaction products by dissolution in an appropriate solvent (e.g., THF, DCM, or acetonitrile) and stirring with SiliaMetS® Dimercaptotriazine (DMT) resin (Silicycle, Inc.) for 4-24 h using 3-5 equivalents relative to the amount of the palladium catalyst (or other metal) used. Filtration, and washing of the resin, followed by concentration provides the purified product.
  • an appropriate solvent e.g., THF, DCM, or acetonitrile
  • SiliaMetS® Dimercaptotriazine (DMT) resin Silicycle, Inc.
  • the title compound (26 mg) was prepared from 6-(6-methoxyisoquinolin-7-yl)-N-methyl-N-(1,2,2,6,6-pentamethylpiperidin-4-yl)pyridazin-3-amine (63 mg, 0.150 mmol) following GENERAL METHOD 2-4 for demethylation using thiophenol.
  • Benzyl bromide (7 mL, 57.8 mmol) was added to a mixture of 3-hydroxy-4-iodobenzoic acid (5.13 g, 18.9 mmol) and K 2 CO 3 (8.0 g, 58 mmol) in acetone (200 mL). The mixture was then heated at reflux overnight. The mixture was cooled to room temperature then filtered through a small Celite® pad. The filtrate was concentrated in vacuo. MeOH (30 mL) was added to the residue followed by 1 M NaOH (94 mL, 94 mmol). The resulting mixture was heated in a 70° C. oil bath for 1 h.
  • p-Toluenesulfonic acid monohydrate (0.215 g, 1.129 mmol) was added to a solution of (3-(benzyloxy)-4-iodophenyl)methanol (1.92 g, 5.64 mmol) and 3,4-dihydro-2H-pyran (0.64 mL, 6.8 mmol) in DCM (50 mL) at room temperature. After 2 h, the solution was diluted with DCM (30 mL), then washed with saturated aqueous NaHCO 3 solution (2 ⁇ 20 mL). The organic layer was washed with brine, dried over MgSO 4 , filtered, and concentrated in vacuo. The resulting oil was stored under high vacuum overnight.
  • the reaction flask was equipped with a reflux condenser, and the flask evacuated and filled with N 2 (2 ⁇ ) then heated in a 100° C. oil bath overnight (18 h).
  • the mixture was cooled to room temperature then filtered through Celite® with a MeOH wash.
  • the filtrate was acidified to pH 2 using 1 M HCl then heated at 70° C. for 1 h.
  • the mixture was cooled to room temperature then filtered through Celite® washing with MeOH.
  • Step 4. 7-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)isoquinoline-1,6-diol
  • Step 3 6-(7-(Benzyloxy)quinolin-8-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 4 8-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol
  • Butyllithium (1.6 M in heptane, 1.90 mL, 3.04 mmol) was added drop-wise over the course of one hour to a solution of 7-bromo-6-methoxyquinoline (0.603 g, 2.53 mmol) and triisopropyl borate (0.823 ml, 3.55 mmol) in THF (12.6 mL) cooled to ⁇ 78° C. After the addition was complete, the cooling bath was removed and the slurry allowed to warm to room temperature. LC/MS analysis shows near quantitative conversion to the boronic acid.
  • Butyllithium (1.6 M in heptane, 0.41 mL, 0.65 mmol) was added dropwise over the course of 1 h to a solution of 7-bromo-6-methoxy-2-methylquinoline (150 mg, 0.60 mmol) and triisopropyl borate (0.180 mL, 0.77 mmol) cooled to ⁇ 78° C. After addition was complete, the reaction was stirred at ⁇ 78° C. for one hour and then allowed to warm to room temperature and stir overnight. LC/MS analysis shows a 9:1 ratio of the title boronic acid to the debrominated side product (6-methoxy-2-methylquinoline).
  • Step 3 6-(6-Methoxy-2-methylquinolin-7-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 1 6-(3-Chloro-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • 6-(7-Methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (Intermediate 9, 50 mg, 0.123 mmol), was borylated according to GENERAL METHOD 5-1, with the exception that the reaction was heated at 65 OC overnight.
  • To the crude boronic acid solution was added water (400 uL) and CuCl 2 (49.7 mg, 0.370 mmol). The mixture was heated at 65 OC for 1 h, then allowed to stir at room temperature overnight.
  • the mixture was partitioned between DCM/water, and was washed with DCM (6 ⁇ ).
  • the aqueous phase was acidified to pH 1 and concentrated to dryness.
  • Step 2 3-Chloro-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol
  • Step 1 6-(3-Bromo-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • 6-(7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (Intermediate 9, 100 mg, 0.247 mmol), was borylated according to GENERAL METHOD 5-1.
  • To the crude boronic acid solution was added water (800 uL) and CuBr 2 (165 mg, 0.740 mmol). The resulting mixture was stirred at room temperature overnight. The mixture was partitioned between DCM/water. The aqueous phase was extracted with EtOAc (5 ⁇ ), DCM (2 ⁇ ), and the combined organic extracts were concentrated.
  • Step 1 (7-Methoxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-3-yl)boronic acid
  • 6-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)-3-(1-methyl-1H-imidazol-4-yl)quinolin-7-ol was suspended in acetonitrile/H 2 O (3/1 mL). 1 M Aqueous HCl (3 equivalents) was then added and solvent was concentrated in vacuo to afford 6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)-3-(1-methyl-1H-imidazol-4-yl)quinolin-7-ol hydrochloride salt as a yellow solid (9.1 mg).
  • Benzophenone (0.765 g, 4.20 mmol) and potassium tert-butoxide (0.514 g, 4.58 mmol) were added and the mixture heated at 120° C. for 0.5 h.
  • the mixture was cooled to room temperature, diluted with EtOAc, washed with saturated aqueous sodium bicarbonate and brine.
  • the organic phase was extracted with 1 M HCl (3 ⁇ ).
  • the acidic extracts were basified to pH by addition of 2 M NaOH and extracted with EtOAc (3 ⁇ ) and diethyl ether (2 ⁇ ).
  • Step 3 6-(3-Ethyl-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • 6-(3-Ethyl-7-methoxyquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (100 mg, 0.231 mmol) was subjected to methoxy deprotection conditions as described in GENERAL METHOD 2-2.
  • the crude material was subjected to silica gel chromatography (GENERAL METHOD 4-1) to provide 3-ethyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol as a yellow solid (81 mg).
  • LC/MS Rt 0.44 min.
  • 6-bromo-7-methoxyquinoline PREPARATION 6 Step 1
  • DCM DCM
  • methyltrioxorhenium(VII) 0.209 g, 0.840 mmol
  • hydrogen peroxide 30 wt % in water, 1.03 mL, 10.1 mmol
  • the solution was allowed to stir overnight during which time a thick precipitate formed.
  • the mixture was diluted with heptane and the solids were isolated by filtration, washing with heptane. Drying of the solids in vacuo provided 6-bromo-7-methoxyquinoline 1-oxide (1.76 g).
  • Step 3 (7-Methoxy-2-oxo-1,2-dihydroquinolin-6-yl)boronic acid and 7-methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-2(1H)-one
  • Step 4 7-Methoxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-2(1H)-one
  • Step 5 7-Hydroxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-2(1H)-one
  • Methyltrioxorhenium (0.03 g, 0.12 mmol) was added to 6-chloro-7-methoxyquinoline (1.69 g, 8.73 mmol) in DCM (40 mL). A solution of hydrogen peroxide (50 wt % in water, 1.0 mL, 17 mmol) was then added at 5° C. and the mixture was stirred at room temperature overnight. An additional portion of methyltrioxorhenium (0.03 g, 0.12 mmol) was added followed by hydrogen peroxide (50 wt % in water, 1.0 mL, 17 mmol) and the reaction was stirred for 4 h.
  • Trifluoroacetic anhydride (6.0 mL, 42 mmol) was added dropwise to 6-chloro-7-methoxyquinoline 1-oxide (1.18 g, 5.63 mmol) in DMF (8.0 mL) at 5° C.
  • the reaction mixture was heated in a Biotage® Initiator microwave reactor at 100° C. for 30 min. Water (20 mL) was added to the reaction and the resulting solid was collected by filtration. The solid was triturated with hot acetonitrile and filtered to afford 6-chloro-7-methoxyquinolin-2(1H)-one (0.39 g).
  • MS (M+1) 210.1.
  • Potassium bis(trimethylsilyl)amide (0.5 M in toluene, 2.7 mL, 1.4 mmol) was added to 6-chloro-7-methoxyquinolin-2(1H)-one (0.20 g, 0.95 mmol) in toluene (5 mL) and the mixture was stirred for 10 min at room temperature.
  • Iodomethane (0.07 mL, 1 mmol) was added to the reaction and the mixture was stirred 2 h at 50° C.
  • An additional portion of potassium bis(trimethylsilyl)amide (0.5 M in toluene, 2.7 mL, 1.4 mmol) was added and the mixture was stirred for 10 min at room temperature.
  • the mixture was stirred at 80° C. for 2 h, then a 4% solution of Na 2 CO 3 was added to the reaction and the aqueous phase was extracted with DCM (3 ⁇ ).
  • the combined organic phases were extracted with 2 M HCl (3 ⁇ ).
  • the combined acidic aqueous phases were basified to pH 11 with 6 M NaOH then extracted with DCM (3 ⁇ ).
  • the combined organic phases were dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Example 10-1 or Example 10-2 The following example compounds were prepared from the Intermediate 11 and the appropriate sodium alkoxide or amine according to the preparations of Example 10-1 or Example 10-2.
  • Step 1 6-(7-Methoxy-2-methyl-4-(1-methyl-1H-pyrazol-4-yl)quinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 2 2-Methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)-4-(1-methyl-1H-pyrazol-4-yl)quinolin-7-ol
  • reaction mixture was diluted with DMSO (5 mL), acidified with formic acid (0.5 mL) and purified via preparative reverse-phase HPLC (5 to 20% acetonitrile in water, 7.5% formic acid as modifier) to afford 4-methoxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-6-ol formate salt (6 mg) as a yellow solid.
  • Step 3 6-(7-Ethoxyquinoxalin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Zn(OPiv) 2 .2 LiCl (1.64 g, 4.64 mmol) was placed in a Schlenk-flask equipped with a magnetic stir bar and a septum, and dried for 5 min under vaccuum with a heat gun.
  • the zinc salt was dissolved in dry THF (10 mL). 6-Bromo-7-ethoxyquinoxaline (0.94 g, 3.71 mmol) was added and the mixture was stirred for 5 min at room temperature.
  • Step 4 7-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoxalin-6-ol hydrochloride salt
  • Step 1 2-Hydroxy-4-methoxy-5-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)benzaldehyde
  • Step 2 2-Formyl-5-methoxy-4-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)phenyl trifluoromethanesulfonate
  • Step 1 2-Hydroxy-4-methoxy-5-(6-((2,2,6,6-tetramethylpiperidin-4-yl)oxy)pyridazin-3-yl)benzaldehyde
  • the trifluoroacetic acid salt of the product from preparatory reverse phase HPLC purification is dissolved in MeOH and loaded onto PL-HCO 3 MP® cartridge (Agilent Technologies) preconditionsed with MeOH. The cartridge is then flushed with excess MeOH to provided the product as a free base.
  • MP-carbonate a resin-bound base, is also available from Biotage (part number 800267 for 10 grams).
  • the chemical name for MP-carbonate is macroporous triethylammonium methyl polystyrene carbonate.
  • a mixture of a chloropyridazine intermediate, such as Intermediate 1 (1.0 equivalent), a boronic acid reagent (1.5-2 equivalents), SPhos (0.2 equivalents), Pd 2 (dba) 3 (0.05 equivalents), and K 3 PO 4 (3 equivalents) in 4:1 dioxane/water (0.2 M) was heated in an oil bath at 100° C. for 18 h.
  • the mixture was concentrated in vacuo and the crude residue was purified via silica gel chromatography (1-30% MeOH/DCM, column pretreated with TEA).
  • Step 1 6-methoxy-1-methyl-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-4(1H)-one
  • Step 2 6-Hydroxy-1-methyl-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-4(1H)-one
  • Step 3 6-(7-Ethoxy-2, 3-dimethylquinoxalin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 4 2, 3-Dimethyl-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoxalin-6-ol
  • Step 1 6-Bromo-7-ethoxy-2-methylquinoxaline and 7-bromo-6-ethoxy-2-methylquinoxaline
  • Step 2 6-Ethoxy-2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoxaline, (7-ethoxy-3-methylquinoxalin-6-yl)boronic acid, and 7-ethoxy-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoxaline and (7-ethoxy-2-methylquinoxalin-6-yl)boronic acid
  • Step 3 6-(7-Ethoxy-3-methylquinoxalin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine, and 6-(7-ethoxy-2-methylquinoxalin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 4 2-Methyl-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoxalin-6-ol, and 3-methyl-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoxalin-6-ol
  • the crude material was purified via preparative reverse-phase HPLC (15 to 40% acetonitrile in water, 5 mM ammonium hydroxide modifier) and the regioisomers were separated via preparative SFC (AS-H 21 ⁇ 250 mm column, 15% MeOH 10 mM NH 4 OH in CO 2 ). Residual palladium was scavenged from each using GENERAL METHOD 6-1. The two solids were separately suspended in 3:1 acetonitrile/water (8 mL). 1 M aqueous HCl (3 equivalents) was added and the solvent concentrated in vacuo to afford two yellow solids, the major regioisomer (56 mg) and the minor regioisomer (19 mg). NMR structure determination attempts were not able to conclusively distinguish the two regioisomers.
  • Step 1 6-(4-Cyclopropyl-7-methoxy-2-methylquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • a flask containing a mixture of 6-(4-chloro-7-methoxy-2-methylquinolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine PREPARATION 11 Step 5, 48 mg, 0.11 mmol), cyclopropyltrifluoroborate (24 mg, 0.16 mmol), n-butyl di-1-adamantylphosphine (5 mg, 0.013 mmol) and cesium carbonate (103 mg, 0.32 mmol) in toluene (1 mL) and water (0.1 mL) was evacuated, filled with N 2 (4 ⁇ ) and heated at 100° C. for 7 h.
  • Step 2 4-Cyclopropyl-2-methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinolin-7-ol
  • Example 20-4 A mixture of Example 20-4 (14 mg, 0.029 mmol), Pd-C (3 mg, 10 wt % on carbon, 3 ⁇ mol), Pd(OH) 2 (2.0 mg, 20 wt % on carbon, 3 ⁇ mol) and one drop of concentrated HCl aqueous solution in methanol (10 mL) was evacuated, filled with H 2 (4 ⁇ ) and shaken under H 2 (50 psi) on a Parr shaker hydrogenator at room temperature overnight. The reaction mixture was filtered through Celite®, washed with MeOH, concentrated, and subjected to SCX purification (GENERAL METHOD 3-1, 1 g SiliaBond Propylsulfonic Acid® cartridge).
  • Step 4 7-Methoxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinazolin-4(1H)-one
  • the vessel was evacuated and back-filled with nitrogen (4 ⁇ ), and heated via microwave irradiation at 110° C. for 1 h.
  • the mixture was cooled to room temperature and acidified to pH 2 with 4 M HCl in 1,4-dioxane.
  • SCX purification (GENERAL METHOD 3-1) followed by silica gel chromatography (0-20% MeOH in DCM) provided 7-methoxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinazolin-4(1H)-one (137 mg) as a light brown solid.
  • MS (M+1) 423.1.
  • Step 5 7-Hydroxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinazolin-4(1H)-one
  • Step 5 6-(7-Methoxyquinazolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 6 6-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinazolin-7-ol
  • 6-(7-Methoxyquinazolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine 28 mg, 0.069 mmol was subjected to methoxy deprotection conditions as described in GENERAL METHOD 2-3 using boron tribromide (1.0 M solution in DCM, 0.41 mL, 0.41 mmol).
  • Step 3 7-Methoxy-1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3, 4-dihydroquinolin-2(1H)-one
  • Step 4 7-Methoxy-1-methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)-3, 4-dihydroquinolin-2(1H)-one
  • Step 5 7-Hydroxy-1-methyl-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)-3, 4-dihydroquinolin-2(1H)-one
  • Step 4 6-(7-Methoxy-2-methylquinazolin-6-yl)-N-methyl-N-(2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine
  • Step 5 6-(6-(Methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinazolin-7-ol
  • 6-(7-Methoxy-2-methylquinazolin-6-yl)-N-methyl-N-(2,2,6,6-tetramethylpiperidin-4-yl)pyridazin-3-amine (92 mg, 0.21 mmol) was subjected to methoxy deprotection conditions as described in GENERAL METHOD 2-4 using thiophenol (0.028 mL, 0.27 mmol).
  • Step 2 7-Hydroxy-6-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)isoquinoline-1-carbonitrile
  • 6-Hydroxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoline-2-carbonitrile was prepared from 7-Bromo-6-methoxyquinoline according to the synthesis of Example 26-1.
  • LC/MS Rt 0.54.
  • Step 1 6-Methoxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)isoquinoline-1-carboxamide
  • 6-Hydroxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoline-2-carboxamide was isolated as a by-product from the methoxy deprotection of 6-methoxy-7-(6-(methyl(2,2,6,6-tetramethylpiperidin-4-yl)amino)pyridazin-3-yl)quinoline-2-carbonitrile (Example 26-3) using GENERAL METHOD 2-1.
  • LC/MS Rt 0.52.
US14/909,052 2013-07-31 2014-07-30 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions Abandoned US20160184305A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/909,052 US20160184305A1 (en) 2013-07-31 2014-07-30 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361860388P 2013-07-31 2013-07-31
US14/909,052 US20160184305A1 (en) 2013-07-31 2014-07-30 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions
PCT/US2014/048984 WO2015017589A1 (en) 2013-07-31 2014-07-30 1,4-disubstituted pyridazine derivatives and their use for treating smn-deficiency-related conditions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/048984 A-371-Of-International WO2015017589A1 (en) 2013-07-31 2014-07-30 1,4-disubstituted pyridazine derivatives and their use for treating smn-deficiency-related conditions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US201816030139A Continuation 2013-07-31 2018-07-09

Publications (1)

Publication Number Publication Date
US20160184305A1 true US20160184305A1 (en) 2016-06-30

Family

ID=51352849

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/909,052 Abandoned US20160184305A1 (en) 2013-07-31 2014-07-30 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions
US16/259,441 Active 2035-04-22 US11672799B2 (en) 2013-07-31 2019-01-28 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating SMN-deficiency-related conditions
US18/322,521 Pending US20240009189A1 (en) 2013-07-31 2023-05-23 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/259,441 Active 2035-04-22 US11672799B2 (en) 2013-07-31 2019-01-28 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating SMN-deficiency-related conditions
US18/322,521 Pending US20240009189A1 (en) 2013-07-31 2023-05-23 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating smn-deficiency-related conditions

Country Status (14)

Country Link
US (3) US20160184305A1 (zh)
EP (2) EP3027600B1 (zh)
JP (3) JP6461150B2 (zh)
KR (2) KR102318727B1 (zh)
CN (1) CN105636953B (zh)
AU (1) AU2014296255B2 (zh)
BR (1) BR112016001899B1 (zh)
CA (1) CA2918805C (zh)
EA (1) EA030631B1 (zh)
ES (1) ES2917979T3 (zh)
MX (2) MX2016001400A (zh)
PL (1) PL3027600T3 (zh)
PT (1) PT3027600T (zh)
WO (1) WO2015017589A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10195196B2 (en) 2012-08-13 2019-02-05 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US11008572B2 (en) 2017-08-04 2021-05-18 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11845744B2 (en) 2019-02-05 2023-12-19 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11964971B2 (en) 2019-02-06 2024-04-23 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6461150B2 (ja) 2013-07-31 2019-01-30 ノバルティス アーゲー 1,4−二置換ピリダジン誘導体およびsmn欠損に関連する状態を処置するためのその使用
JP6884102B2 (ja) * 2015-02-09 2021-06-09 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト がんの治療のための化合物
EP3053577A1 (en) * 2015-02-09 2016-08-10 F. Hoffmann-La Roche AG Compounds for the treatment of cancer
WO2016184832A1 (en) * 2015-05-20 2016-11-24 F. Hoffmann-La Roche Ag Compounds for treating spinal muscular atrophy
CA3005434A1 (en) 2015-11-16 2017-05-26 Ohio State Innovation Foundation Methods and compositions for treating disorders and diseases using survival motor neuron (smn) protein
ES2879995T3 (es) * 2015-12-10 2021-11-23 Ptc Therapeutics Inc Métodos para el tratamiento de la enfermedad de Huntington
EA201992878A1 (ru) 2017-06-05 2020-05-08 ПиТиСи ТЕРАПЬЮТИКС, ИНК. Соединения для лечения болезни хантингтона
CN111372611A (zh) 2017-06-14 2020-07-03 Ptc医疗公司 修饰rna剪接的方法
WO2019000144A1 (zh) * 2017-06-26 2019-01-03 深圳市博奥康生物科技有限公司 一种表达aitr基因的cho细胞及其应用
WO2019000150A1 (zh) * 2017-06-26 2019-01-03 深圳市博奥康生物科技有限公司 一种tl6高表达的重组cho细胞的构建方法
MX2019015580A (es) 2017-06-28 2020-07-28 Ptc Therapeutics Inc Metodos para tratar la enfermedad de huntington.
BR112019027717A2 (pt) 2017-06-28 2020-07-28 Ptc Therapeutics, Inc. métodos para tratar a doença de huntington
CN112135815A (zh) 2018-03-27 2020-12-25 Ptc医疗公司 用于治疗亨廷顿氏病的化合物
AU2019294478B2 (en) 2018-06-27 2023-03-23 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating Huntington's disease
EP3814360A1 (en) 2018-06-27 2021-05-05 PTC Therapeutics, Inc. Heteroaryl compounds for treating huntington's disease
US11129829B2 (en) 2019-06-17 2021-09-28 Skyhawk Therapeutics, Inc. Methods for modulating splicing
CN110256342B (zh) * 2019-07-16 2022-06-07 河南省科学院化学研究所有限公司 一种2-氰基喹啉衍生物的合成方法
JP2022541070A (ja) 2019-07-25 2022-09-21 ノバルティス アーゲー 調節可能な発現系
AU2020377204A1 (en) 2019-11-01 2022-06-02 Novartis Ag The use of a splicing modulator for a treatment slowing progression of huntington's disease
TW202304446A (zh) 2021-03-29 2023-02-01 瑞士商諾華公司 剪接調節子用於減慢杭丁頓氏舞蹈症進展的治療之用途
EP4119151A4 (en) 2021-06-03 2023-10-25 MD Healthcare Inc. COMPOSITION FOR THE PREVENTION OR TREATMENT OF EYE DISEASES COMPRISING EXTRACELLULAR VESICLES FROM LACTOBACILLUS PARACASEI

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080247964A1 (en) * 2006-05-08 2008-10-09 Yuelian Xu Substituted azaspiro derivatives
WO2014028459A1 (en) * 2012-08-13 2014-02-20 Novartis Ag 1,4-disubstituted pyridazine analogs and methods for treating smn-deficiency-related conditions

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2427943C2 (de) * 1974-06-10 1984-08-02 Dr. Karl Thomae Gmbh, 7950 Biberach Benzimidazole, Verfahren zu deren Herstellung und diese enthaltende Arzneimittel
DE2516040C2 (de) * 1974-06-10 1984-12-20 Dr. Karl Thomae Gmbh, 7950 Biberach Benzimidazole, Verfahren zu deren Herstellung und diese enthaltende Arzneimittel
HUT74690A (en) 1993-11-24 1997-01-28 Du Pont Merck Pharma Novel isoxazoline and isoxazole fibrinogen receptor antagonists process for producing them and pharmaceutical compositions containing them
KR20020073586A (ko) * 2000-02-11 2002-09-27 버텍스 파마슈티칼스 인코포레이티드 피페라진 및 피페리딘 유도체
EP1133993A1 (en) * 2000-03-10 2001-09-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Substances for the treatment of spinal muscular atrophy
IL152845A0 (en) * 2000-06-07 2003-06-24 Vertex Pharma Fused heterocyclic compounds and pharmaceutical compositions containing the same
US6376508B1 (en) * 2000-12-13 2002-04-23 Academia Sinica Treatments for spinal muscular atrophy
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US6982259B2 (en) 2002-04-30 2006-01-03 Schering Aktiengesellschaft N-heterocyclic derivatives as NOS inhibitors
WO2004014881A2 (en) 2002-08-09 2004-02-19 Astra Zeneca Ab '1,2,4'oxadiazoles as modulators of metabotropic glutamate receptor-5
EP1536790A2 (en) 2002-08-09 2005-06-08 AstraZeneca AB Oxadiazoles as modulators of metabotropic glutamate receptor-5
EP1581525A2 (en) 2002-08-09 2005-10-05 AstraZeneca AB Compounds having an activity at metabotropic glutamate receptors
DE60309740T2 (de) 2002-11-11 2007-03-29 Neurosearch A/S 1,4-dizabicyclo(3,2,2)nonane derivative, verfahren zu ihrer herstellung und therapeutical verwendung
WO2004110351A2 (en) 2003-05-14 2004-12-23 Anadys Pharmaceuticals, Inc. Heterocyclic compounds for treating hepatitis c virus
JP4735261B2 (ja) 2003-06-27 2011-07-27 Msd株式会社 ヘテロアリールオキシ含窒素飽和ヘテロ環誘導体
CA2533889A1 (en) 2003-08-15 2005-03-03 Merck & Co., Inc. Mitotic kinesin inhibitors
US7399765B2 (en) * 2003-09-19 2008-07-15 Abbott Laboratories Substituted diazabicycloalkane derivatives
US7655657B2 (en) * 2003-12-22 2010-02-02 Abbott Laboratories Fused bicycloheterocycle substituted quinuclidine derivatives
WO2005077368A2 (en) 2004-02-03 2005-08-25 Astrazeneca Ab Treatment of gastro-esophageal reflux disease (gerd)
WO2005077373A2 (en) 2004-02-03 2005-08-25 Astrazeneca Ab Treatment of gastro-esophageal reflux disease (gerd)
FR2868780B1 (fr) 2004-04-13 2008-10-17 Sanofi Synthelabo Derives de la 1-amino-phthalazine, leur preparation et leur application en therapeutique
EP1753740A2 (en) 2004-05-21 2007-02-21 Merck & Co., Inc. Amino cyclopentyl heterocyclic and carbocyclic modulators of chemokine receptor activity
AU2005283085B2 (en) 2004-06-18 2012-06-21 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
EP2316458A1 (en) 2004-09-20 2011-05-04 Xenon Pharmaceuticals Inc. Pyridazine derivatives for inhibiting human stearoyl-coa-desaturase
PL1637141T3 (pl) 2004-09-21 2012-04-30 Trobio Ab Stabilizowana kompozycja proteazy zawierająca proteazę serynową, pochodne morfoliny i odwracalne inhibitory tej proteazy serynowej
US7354944B2 (en) 2004-10-18 2008-04-08 Amgen Inc. Thiadiazole compounds and methods of use
MX2008000017A (es) 2005-06-28 2008-03-26 Sanofi Aventis Derivados de isoquinolina como inhibidores de rho-cinasa.
AU2006264966B2 (en) 2005-07-04 2013-02-21 High Point Pharmaceuticals, Llc Histamine H3 receptor antagonists
GB0517184D0 (en) * 2005-08-22 2005-09-28 Glaxo Group Ltd Compounds
TW200813051A (en) * 2006-05-08 2008-03-16 Neurogen Corp Substituted azaspiro derivatives
ES2525076T3 (es) * 2006-05-19 2014-12-17 Abbvie Bahamas Ltd. Derivados de alcanos azabicíclicos sustituidos con bicicloheterociclos condensados activos en el SNC
US20080076924A1 (en) * 2006-06-30 2008-03-27 Patrick Betschmann Piperazines as P2X7 antagonists
US20080139598A1 (en) 2006-11-07 2008-06-12 Joseph Barbosa Amine-linked Multicyclic Compounds and Methods of Their Use
KR20090092287A (ko) 2006-12-22 2009-08-31 노파르티스 아게 Pdk1 억제를 위한 퀴나졸린
GB0704394D0 (en) 2007-03-07 2007-04-11 Senexis Ltd Compounds
GB0705803D0 (en) * 2007-03-28 2007-05-02 Glaxo Group Ltd Novel compounds
EP2014656A3 (en) 2007-06-11 2011-08-24 High Point Pharmaceuticals, LLC New heteocyclic h3 antagonists
CN101910131B (zh) 2007-11-16 2014-04-23 里格尔药品股份有限公司 甲酰胺、磺酰胺和胺化合物及其使用方法
NZ602030A (en) * 2008-02-28 2014-02-28 Vertex Pharma Heteroaryl derivatives as cftr modulators
WO2009137503A1 (en) * 2008-05-05 2009-11-12 Envivo Pharmaceuticals, Inc. Hdac inhibitors and uses thereof
US20110142758A1 (en) * 2008-06-10 2011-06-16 Neurosearch A/S Indolyl-pyridazinyl-diazabicyclononane derivatives in labelled and unlabelled form and their use in diagnostic methods
GB0810902D0 (en) 2008-06-13 2008-07-23 Astex Therapeutics Ltd New compounds
JP2011524917A (ja) 2008-06-20 2011-09-08 メタボレックス, インコーポレイテッド アリールgpr119作動薬およびその使用
EP2330894B8 (en) * 2008-09-03 2017-04-19 BioMarin Pharmaceutical Inc. Compositions including 6-aminohexanoic acid derivatives as hdac inhibitors
WO2010037210A1 (en) 2008-10-03 2010-04-08 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
JP2012505899A (ja) 2008-10-16 2012-03-08 シェーリング コーポレイション アジン誘導体およびそれの使用方法
BRPI0921496A2 (pt) 2008-11-04 2016-01-19 Chemocentryx Inc composto, composição farmacêutica, e, métodos para tratar doença ou distúrbio, para inibir ligação de quimiocinas a um receptor, para formar imagem de um dito tumor, orgão ou tecido, e para detectar níveis elevados de cxcr7 em uma amostra
WO2010129053A2 (en) 2009-05-05 2010-11-11 Dana Farber Cancer Institute Egfr inhibitors and methods of treating disorders
CN102573842A (zh) * 2009-07-23 2012-07-11 诺瓦提斯公司 氮杂双环烷基衍生物或吡咯烷-2-酮衍生物的用途
KR101481872B1 (ko) 2009-09-10 2015-01-12 에프. 호프만-라 로슈 아게 Jak의 억제제
JO3250B1 (ar) * 2009-09-22 2018-09-16 Novartis Ag إستعمال منشطات مستقبل نيكوتينيك أسيتيل كولين ألفا 7
ES2627911T3 (es) * 2009-11-18 2017-08-01 Plexxikon, Inc. Derivados de N-[2-fluoro-3-(4-amino-7H-pirrolo[2,3-d]pirimidin-5-carbonil)-fenil]-4-bencenosulfonamida como moduladores de la proteína quinasa Raf para el tratamiento del cáncer
WO2011078143A1 (ja) 2009-12-22 2011-06-30 塩野義製薬株式会社 ピリミジン誘導体およびそれらを含有する医薬組成物
JP6026284B2 (ja) 2010-03-03 2016-11-16 プロビオドルグ エージー グルタミニルシクラーゼの阻害剤
US20130096160A1 (en) 2010-04-14 2013-04-18 Secretary, Department Of Health And Human Services Arylthiazolyl piperidines and related compounds as modulators of survival motor neuron (smn) protein production
AR081626A1 (es) 2010-04-23 2012-10-10 Cytokinetics Inc Compuestos amino-piridazinicos, composiciones farmaceuticas que los contienen y uso de los mismos para tratar trastornos musculares cardiacos y esqueleticos
WO2012022467A2 (en) * 2010-08-16 2012-02-23 Santhera Pharmaceuticals (Schweiz) Ag Novel benzoquinone derivatives and use thereof as modulators of mitochondrial function
KR20140018286A (ko) * 2011-03-18 2014-02-12 노파르티스 아게 파킨슨병에서의 도파민 유발 이상운동증에서 사용하기 위한 알파 7 니코틴성 아세틸콜린 수용체 활성화제 및 mGluR5 길항제의 조합물
JP6461150B2 (ja) * 2013-07-31 2019-01-30 ノバルティス アーゲー 1,4−二置換ピリダジン誘導体およびsmn欠損に関連する状態を処置するためのその使用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080247964A1 (en) * 2006-05-08 2008-10-09 Yuelian Xu Substituted azaspiro derivatives
WO2014028459A1 (en) * 2012-08-13 2014-02-20 Novartis Ag 1,4-disubstituted pyridazine analogs and methods for treating smn-deficiency-related conditions

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Bunnelle et al.; Octahydropyrrolo[3,4-c]pyrrole: A Diamine Scaffold for Construction of Either α4β2 or α7-Selective Nictotinic Acetylcholine Receptor (nAChR) Ligands. Substitutions that Switch Subtype Selectivity"; 2009; J. Med. Chem.; 52: 4126-4141 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10195196B2 (en) 2012-08-13 2019-02-05 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US10758533B2 (en) 2012-08-13 2020-09-01 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US11229648B2 (en) 2012-08-13 2022-01-25 Novartis Ag 1,4-disubstituted pyridazine analogs thereof and methods for treating SMN-deficiency-related conditions
US11008572B2 (en) 2017-08-04 2021-05-18 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11021708B2 (en) 2017-08-04 2021-06-01 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11091475B2 (en) 2017-08-04 2021-08-17 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11162101B2 (en) 2017-08-04 2021-11-02 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11326165B1 (en) 2017-08-04 2022-05-10 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11434489B1 (en) 2017-08-04 2022-09-06 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11603531B1 (en) 2017-08-04 2023-03-14 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11845744B2 (en) 2019-02-05 2023-12-19 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11964971B2 (en) 2019-02-06 2024-04-23 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing

Also Published As

Publication number Publication date
ES2917979T3 (es) 2022-07-12
EP3027600B1 (en) 2022-04-06
US11672799B2 (en) 2023-06-13
JP2021098753A (ja) 2021-07-01
MX2016001400A (es) 2016-05-05
KR102318727B1 (ko) 2021-10-28
KR20160037199A (ko) 2016-04-05
KR20210130843A (ko) 2021-11-01
EA030631B1 (ru) 2018-09-28
PT3027600T (pt) 2022-06-23
CA2918805C (en) 2023-02-28
JP2016527272A (ja) 2016-09-08
JP6856614B2 (ja) 2021-04-07
MX2020011652A (es) 2022-09-27
AU2014296255B2 (en) 2017-08-03
CN105636953A (zh) 2016-06-01
CN105636953B (zh) 2018-01-02
US20190160062A1 (en) 2019-05-30
CA2918805A1 (en) 2015-02-05
JP6461150B2 (ja) 2019-01-30
JP7208285B2 (ja) 2023-01-18
EP4105208A1 (en) 2022-12-21
PL3027600T3 (pl) 2022-08-08
WO2015017589A1 (en) 2015-02-05
BR112016001899B1 (pt) 2022-03-15
AU2014296255A1 (en) 2016-02-18
EA201690287A1 (ru) 2016-07-29
US20240009189A1 (en) 2024-01-11
JP2019059783A (ja) 2019-04-18
BR112016001899A2 (zh) 2017-09-05
EP3027600A1 (en) 2016-06-08

Similar Documents

Publication Publication Date Title
US11672799B2 (en) 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating SMN-deficiency-related conditions
US10758533B2 (en) 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US9040712B2 (en) Thiadiazole analogs thereof and methods for treating SMN-deficiency-related-conditions

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH INC.,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEUNG, ATWOOD KIM;DALES, NATALIE;HURLEY, TIMOTHY BRIAN;SIGNING DATES FROM 20131106 TO 20131118;REEL/FRAME:039106/0814

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH INC.;REEL/FRAME:039106/0860

Effective date: 20140124

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION