EP2680843A2 - Dérivés de composés amino-hétéroaryles substitués par des pyrazoles - Google Patents

Dérivés de composés amino-hétéroaryles substitués par des pyrazoles

Info

Publication number
EP2680843A2
EP2680843A2 EP12751813.2A EP12751813A EP2680843A2 EP 2680843 A2 EP2680843 A2 EP 2680843A2 EP 12751813 A EP12751813 A EP 12751813A EP 2680843 A2 EP2680843 A2 EP 2680843A2
Authority
EP
European Patent Office
Prior art keywords
cancer
compound
deuterium
hydrogen
same
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.)
Withdrawn
Application number
EP12751813.2A
Other languages
German (de)
English (en)
Other versions
EP2680843A4 (fr
Inventor
Craig Masse
Bhaumik PANDYA
Ian Robert Silverman
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.)
Concert Pharmaceuticals Inc
Original Assignee
Concert Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Concert Pharmaceuticals Inc filed Critical Concert Pharmaceuticals Inc
Publication of EP2680843A2 publication Critical patent/EP2680843A2/fr
Publication of EP2680843A4 publication Critical patent/EP2680843A4/fr
Withdrawn legal-status Critical Current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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Definitions

  • ADME absorption, distribution, metabolism and/or excretion
  • ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites.
  • some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent.
  • modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
  • a metabolic inhibitor will be co-administered with a drug that is cleared too rapidly.
  • a drug that is cleared too rapidly.
  • the FDA recommends that these drugs be co-dosed with ritonavir, an inhibitor of cytochrome P450 enzyme 3A4 (CYP3A4), the enzyme typically responsible for their metabolism (see Kempf, D.J. et al., Antimicrobial agents and chemotherapy, 1997, 41(3): 654-60).
  • CYP3A4 cytochrome P450 enzyme 3A4
  • Ritonavir causes adverse effects and adds to the pill burden for HIV patients who must already take a combination of different drugs.
  • the CYP2D6 inhibitor quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism of dextromethorphan in a treatment of pseudobulbar affect.
  • Quinidine has unwanted side effects that greatly limit its use in potential combination therapy (see Wang, L et al., Clinical Pharmacology and Therapeutics, 1994, 56(6 Pt 1): 659-67; and FDA label for quinidine at www.accessdata.fda.gov).
  • a potentially attractive strategy for improving a drug's metabolic properties is deuterium modification.
  • Deuterium is a safe, stable, nonradioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability.
  • the size and shape of deuterium are essentially identical to those of hydrogen, replacement of hydrogen by deuterium would not be expected to affect the biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen.
  • Crizotinib also known as 3-[l(R)-(2,6-dichloro-3-fluorophenyl)ethoxy]-5-[l- (4-piperidinyl)-lH-pyrazol-4-yl]pyridin-2-amine is known to inhibit hepatocyte growth factor receptor (c-met/HGFR) kinase and also to block the tyrosine kinase of anaplastic lympohoma kinase (ALK).
  • a percentage of non- small cell lung cancer patients carry the echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase (EML4- ALK) fusion gene.
  • EML4- ALK echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase
  • EML4-ALK when inserted into a normal cell, causes the cell to become cancerous. Crizotinib blocks the tyrosine kinase of the ALK domain of this fusion gene. See Sasaki, t et al., The Biology and Treatment of EML4-ALK Non-Small Cell Lung Cancer, Eur. J. Cancer, 2010, July; 46(10): 1773-80.
  • Crizotinib currently is recommended for approval for non- small cell lung cancer (NSCLC) and is undergoing Phase I/II clinical trials for solid tumor cancer and for lymphoma.
  • This invention relates to novel pyrazole-substituted amino-heteroaryl compounds, and pharmaceutically acceptable salts thereof.
  • This invention also provides compositions comprising a compound of this invention and the use of such compositions in methods of treating diseases and conditions that are beneficially treated by administering an inhibitor of anaplastic lymphoma kinase (ALK) and hepatocyte growth factor receptor (c-met/HGFR) kinase.
  • ALK anaplastic lymphoma kinase
  • c-met/HGFR hepatocyte growth factor receptor
  • treat means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve or lessen the severity of one or more symptoms associated with the disease.
  • a disease e.g., a disease or disorder delineated herein
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen”
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • a position is designated specifically as “D” or “deuterium”
  • the position is understood to have deuterium at an abundance that is at least 3000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 45% incorporation of deuterium).
  • isotopic enrichment factor as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a compound of this invention 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), 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).
  • isotopologue refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
  • a compound represented by a particular chemical structure containing indicated deuterium atoms will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure.
  • the relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
  • the relative amount of such isotopologues in toto will be less than 49.9% of the compound. In other embodiments, the relative amount of such isotopologues in toto will be less than 47.5%, less than 40%, less than 32.5%, less than 25%, less than 17.5%, less than 10%, less than 5%, less than 3%, less than 1%, or less than 0.5% of the compound.
  • the invention also provides salts of the compounds of the invention.
  • a salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • a salt of a provided compound is a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para- toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylprop
  • subject as used herein includes a human or a non-human animal, such as mouse, rat, guinea pig, dog, cat, horse, cow, pig, monkey (e.g., rhesus), chimpanzee, or baboon.
  • the subject is a non-human animal.
  • the subject is a human.
  • the compounds of the present invention may contain an asymmetric carbon atom, for example, as the result of deuterium substitution or otherwise.
  • compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers.
  • a compound of the present invention may exist as either a racemic mixture or a scalemic mixture, or as individual respective stereoisomers that are substantially free from another possible stereoisomer.
  • substantially free of other stereoisomers as used herein means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present.
  • stable compounds refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
  • Substituted with deuterium refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
  • variable may be referred to generally (e.g., "each R") or may be referred to specifically (e.g., R 1 , R 2 , R 3 , etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
  • the present invention rovides a compound of Formula I:
  • R 1 and R 2 are each independently selected from CI, CH 3 and CD 3 ;
  • R 3 is CH 3 or CD 3 ;
  • X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a , X 4b , and X 5 are each independently selected from hydrogen and deuterium;
  • Y 1 is hydrogen or deuterium
  • Y 2 is hydrogen or deuterium
  • R 3 is CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a , X 4b , and X 5 is hydrogen and each of Y 1 and Y 2 is hydrogen, then R 3 is CD 3 .
  • X la and X lb are the same, X 2a and X 2b are the same, X 3a and X 3b are the same, and X 4a and X 4b are the same.
  • R 1 and R 2 are independently selected from CI and CD 3 .
  • R 1 and R 2 are the same and are each CI.
  • R 1 and R 2 are the same and are each CD 3 .
  • X la , X lb , X 2a and X 2b are the same, X 3a , X 3b , X 4a and X 4b are the same and R 1 and R 2 are independently selected from CI and CD 3 .
  • each of X la , X lb , X 2a and X 2b is hydrogen; and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium; and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • R 1 and R 2 are the same and are each CI.
  • R 1 and R 2 are the same and are each CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CH 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CD 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CH 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium; R 1 and R 2 are the same and are selected from CI and CD 3 and R 3 is CD 3 . In one aspect, R 1 and R 2 are each CI. In one aspect, R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a and X 2b is deuterium
  • each of X 3a , X 3b , X 4a and X 4b is hydrogen
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CH 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a and X 2b is deuterium
  • each of X 3a , X 3b , X 4a and X 4b is hydrogen
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CD 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a and X 2b is hydrogen
  • each of X 3a , X 3b , X 4a and X 4b is deuterium
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CH 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • each of X la , X lb , X 2a and X 2b is hydrogen
  • each of X 3a , X 3b , X 4a and X 4b is deuterium
  • R 1 and R 2 are the same and are selected from CI
  • CD 3 and R 3 is CD 3 .
  • R 1 and R 2 are each CI.
  • R 1 and R 2 are each CD 3 .
  • X 5 is hydrogen
  • Y 1 is hydrogen
  • Y 2 is hydrogen
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is hydrogen
  • Y 1 is deuterium
  • Y 2 is hydrogen
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is hydrogen
  • Y 1 is hydrogen
  • Y 2 is deuterium
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is hydrogen
  • Y 1 is deuterium
  • Y 2 is deuterium
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is deuterium
  • Y 1 is hydrogen
  • Y 2 is hydrogen
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is deuterium
  • Y 1 is deuterium
  • Y 2 is hydrogen
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is deuterium
  • Y 1 is hydrogen
  • Y 2 is deuterium
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • X 5 is deuterium
  • Y 1 is deuterium
  • Y 2 is deuterium
  • R 1 and R 2 are the same and are selected from CI and CD 3 .
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is hydrogen.
  • each of X la , X lb , X 2a , X 2b , X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is hydrogen and each of X 3a , X 3b , X 4a and X 4b is deuterium.
  • each of X la , X lb , X 2a and X 2b is deuterium and each of X 3a , X 3b , X 4a and X 4b is hydrogen.
  • the compound is a compound of Formula I as defined above wherein any atom not designated as deuterium is present at its natural isotopic abundance.
  • the compound is selected from any one of the compounds (Cmpd) set forth in Table 1 (below):
  • Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure.
  • New chemical entities corresponding to Formula I can be accessed by multiple step organic synthesis routinely practiced by those skilled in the art and as depicted in Schemes la and lb, above.
  • ketones 10a or 10b Following carbonyl reduction of ketones 10a or 10b with borohydride or borodeuteride the resultant racemic benzylic alcohol is aceylated with acetic anhydride.
  • Enzymatic resolution of the mixture of enantiomers may be achieved with pig liver esterase (PLE) to provide chiral alcohols 11a or lib in greater than 97.5% enantiomeric excess (ee).
  • PLE pig liver esterase
  • boron-pinacolates, 15 may be united with aryl iodides 13a or 13b via palladium catalyzed cross coupling under alkaline biphasic conditions. Removal of the tert-butylcarbamate (BOC) protecting group with concentrated hydrochloric acid yields the desired active pharmaceutical ingredient as the free base. Preparation of appropriate pharmaceutical grade salt(s) will be necessary and may be accomplished using standard practices.
  • BOC tert-butylcarbamate
  • the invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier e.g., including any of the formulae herein
  • the carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • the present invention provides a pyrogen-free pharmaceutical composition
  • a pyrogen-free pharmaceutical composition comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutical salt of the compound or tautomer; and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene -block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as
  • the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques).
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit- dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol, and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
  • Application of the subject therapeutics may be local, so as to be administered at the site of interest.
  • Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • the compounds of this invention may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
  • the invention provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
  • the invention provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of this invention.
  • Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non-degradable, diffusible polymer capsules and biodegradable polymer wafers.
  • the invention provides an implantable medical device coated with a compound or a composition comprising a compound of this invention, such that said compound is therapeutically active.
  • the invention provides an implantable drug release device impregnated with or containing a compound or a composition comprising a compound of this invention, such that said compound is released from said device and is therapeutically active.
  • an organ or tissue is accessible because of removal from the subject, such organ or tissue may be bathed in a medium containing a composition of this invention, a composition of this invention may be painted onto the organ, or a composition of this invention may be applied in any other convenient way.
  • a composition of this invention further comprises a second therapeutic agent or a combination of second therapeutic agents.
  • the second therapeutic agent(s) may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound having the same mechanism of action as crizotinib.
  • Such agents include those indicated as being useful in combination with crizotinib, including but not limited to, those described in US 2011003805, and CN101836991.
  • the second therapeutic agent(s) is an agent useful in the treatment or prevention of a cancer, more specifically of prostate cancer, osteosarcomas, lung cancer, particularly non-small cell lung cancer, breast cancer, endometrial cancer, glioblastoma, colorectal cancer, ovarian cancer, pancreatic cancer, kidney cancer, small intestinal cancer, esophageal cancer or gastric cancer.
  • a cancer more specifically of prostate cancer, osteosarcomas, lung cancer, particularly non-small cell lung cancer, breast cancer, endometrial cancer, glioblastoma, colorectal cancer, ovarian cancer, pancreatic cancer, kidney cancer, small intestinal cancer, esophageal cancer or gastric cancer.
  • the second therapeutic agent is selected from kinase inhibitors.
  • the kinase inhibitor is selected from erlotinib, sorafenib, a deuterated form of erlotinib as disclosed in US Patent Application No. 11/957,442 and in US Patent Application No. 12/413,510, a deuterated form of sorafenib as disclosed in PCT Patent Application No.
  • erlotinib is compound A, another more specific embodiment, the deuterated form of erlotinib is compound
  • the deuterated form of sorafenib is compound C, (C).
  • the kinase inhibitor is erlotinib or sorafenib.
  • the kinase inhibitor is a deuterated form of erlotinib (as disclosed in the above-referenced patent applications) or a deuterated form of sorafenib (as disclosed in the above-referenced patent application).
  • a composition of this invention comprises a combination of the compound of Formula I with two second therapeutic agents selected from kinase inhibitors.
  • the combination is with erlotinib or a deuterated form of erlotinib as disclosed in US Patent Application No. 11/957,442 and in US Patent Application No. 12/413,510, and sorafenib or a deuterated form of sorafenib as disclosed in PCT Patent Application No. PCT/US2009/053595).
  • the combination is with erlotinib or compound A, and sorafenib or compound C.
  • the combination is with erlotinib or compound B, and sorafenib or compound C.
  • the combination is erlotinib and sorafenib.
  • the combination is a deuterated form of erlotinib and a deuterated form of sorafenib.
  • the combination is a deuterated form of erlotinib and sorafenib.
  • the combination is erlotinib and a deuterated form of sorafenib.
  • the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another.
  • association with one another means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • the compound of the present invention is present in an effective amount.
  • effective amount refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
  • an effective amount of a compound of this invention can range from 25 mg to 500 mg per treatment. Treatment is typically administered one to two times daily. In more specific embodiments the effective amount can be one of the following amounts or ranges:
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician. For example, guidance for selecting an effective dose can be determined by reference to the prescribing information for crizotinib.
  • an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
  • an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al, eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
  • the invention provides a method of modulating the activity of anaplastic lymphoma kinase (ALK) and hepatocyte growth factor receptor (c- met/HGFR) kinase in a cell, comprising contacting a cell with one or more compounds of Formula I herein, or a pharmaceutically acceptable salt thereof.
  • ALK anaplastic lymphoma kinase
  • c- met/HGFR hepatocyte growth factor receptor
  • the invention provides a method of treating a disease that is beneficially treated by inhibiting ALK and c-met/HGFR, e.g., crizotinib, in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound or a composition of this invention.
  • the subject is a patient in need of such treatment.
  • diseases are well known in the art and are disclosed in, but not limited to the following patents and published applications: WO 2006/021884.
  • Such diseases include, but are not limited to, cancer, in particular, lung cancer, non-small cell lung cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colorectal cancer, colon cancer, gastric cancer, breast cancer, endometrial cancer, carcinoma of the fallopian tubes, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, esophageal cancer, small intestinal cancer, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, cancer of the urethra, cancer of the penis, cancer of the prostate, chronic or acute leukemia, lymphoma, sarcoma of soft tissue, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system
  • the invention provides a method of treating abnormal cell growth in a mammal.
  • the method of this invention is used to treat a disease or condition selected from lymphoma, neuroblastoma, solid tumors and non-small cell lung cancer in a subject in need thereof.
  • Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
  • any of the above methods of treatment comprises the further step of co-administering to the subject in need thereof one or more second therapeutic agents.
  • the choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for co-administration with crizotinib.
  • the choice of second therapeutic agent is also dependent upon the particular disease or condition to be treated. Examples of second therapeutic agents that may be employed in the methods of this invention are those set forth above for use in combination compositions comprising a compound of this invention and a second therapeutic agent.
  • the combination therapies of this invention include coadministering a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a second therapeutic agent to a subject in need thereof for treatment of the following conditions (with the particular second therapeutic agent indicated in parentheses following the indication: non-small cell lung cancer (PF-00299804).
  • co-administered means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods.
  • composition of this invention comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • Effective amounts of these second therapeutic agents are well known to those skilled in the art and guidance for dosing may be found in patents and published patent applications referenced herein, as well as in Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), and other medical texts. However, it is well within the skilled artisan's purview to determine the second therapeutic agent's optimal effective-amount range.
  • the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • the invention provides the use of a compound of Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment or prevention in a subject of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention in a subject of a disease, disorder or symptom thereof delineated herein.
  • Microsomal Assay Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). ⁇ -nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCl 2 ), and dimethyl sulfoxide (DMSO) are purchased from Sigma-Aldrich.
  • 7.5 mM stock solutions of test compounds are prepared in DMSO.
  • the 7.5 mM stock solutions are diluted to 12.5-50 ⁇ in acetonitrile (ACN).
  • ACN acetonitrile
  • the 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl 2 .
  • the diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate.
  • a 10 aliquot of the 12.5-50 ⁇ test compound is added to the microsomes and the mixture is pre -warmed for 10 minutes. Reactions are initiated by addition of pre- warmed NADPH solution.
  • the final reaction volume is 0.5 mL and contains 0.5 mg/mL human liver microsomes, 0.25-1.0 ⁇ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl 2 .
  • the reaction mixtures are incubated at 37 °C, and 50 ⁇ , aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow- well 96-well plates which contain 50 ⁇ of ice-cold ACN with internal standard to stop the reactions.
  • the plates are stored at 4 °C for 20 minutes after which 100 ⁇ , of water is added to the wells of the plate before centrifugation to pellet precipitated proteins.

Landscapes

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

Abstract

La présente invention concerne de nouveaux composés amino-hétéroaryles substitués par des pyrazoles de formule (I) et leurs sels pharmaceutiquement acceptables. Cette invention concerne également des compositions comprenant un composé selon l'invention et l'utilisation de ces compositions dans des procédés pour traiter des maladies et des troubles qui sont avantageusement traités par l'administration d'un inhibiteur du récepteur anaplastic lymphoma kinase (ALK).
EP20120751813 2011-03-03 2012-03-01 Dérivés de composés amino-hétéroaryles substitués par des pyrazoles Withdrawn EP2680843A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161448887P 2011-03-03 2011-03-03
PCT/US2012/027341 WO2012119006A2 (fr) 2011-03-03 2012-03-01 Dérivés de composés amino-hétéroaryles substitués par des pyrazoles

Publications (2)

Publication Number Publication Date
EP2680843A2 true EP2680843A2 (fr) 2014-01-08
EP2680843A4 EP2680843A4 (fr) 2015-05-06

Family

ID=46758504

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20120751813 Withdrawn EP2680843A4 (fr) 2011-03-03 2012-03-01 Dérivés de composés amino-hétéroaryles substitués par des pyrazoles

Country Status (5)

Country Link
EP (1) EP2680843A4 (fr)
AU (1) AU2012223281A1 (fr)
CA (1) CA2829025A1 (fr)
MX (1) MX2013010067A (fr)
WO (1) WO2012119006A2 (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103304552B (zh) * 2012-03-09 2016-12-28 广东东阳光药业有限公司 取代的吡啶化合物及其使用方法和用途
EP2935251A1 (fr) 2012-12-20 2015-10-28 Concert Pharmaceuticals Inc. Inhibiteurs alk deutérés
CN104650049B (zh) * 2013-08-28 2018-06-08 广东东阳光药业有限公司 取代的吡啶化合物及其使用方法和用途
CA3063901A1 (fr) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Amides de pyridone deuteres et leurs promedicaments utilises en tant que modulateurs de canaux sodiques

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2341351T3 (es) * 2004-08-26 2010-06-18 Pfizer, Inc. Compuestos de aminoheteroarilo enantiomericamente puros como inhibidores de proteina cinasas.
SI1959955T1 (sl) * 2005-12-05 2011-02-28 Pfizer Prod Inc Postopek zdravljenja abnormalne celične rasti
MX2008012658A (es) * 2006-03-31 2008-12-16 Schering Corp Inhibidores de cinasa.
CN101967140A (zh) * 2010-09-14 2011-02-09 郑州泰基鸿诺药物科技有限公司 氘代克里唑蒂尼及其衍生物、制备方法和应用

Also Published As

Publication number Publication date
EP2680843A4 (fr) 2015-05-06
WO2012119006A3 (fr) 2014-04-17
AU2012223281A1 (en) 2013-09-19
MX2013010067A (es) 2014-01-31
CA2829025A1 (fr) 2012-09-07
WO2012119006A2 (fr) 2012-09-07

Similar Documents

Publication Publication Date Title
AU2014235462B2 (en) Deuterated palbociclib
US9776973B2 (en) Deuterated momelotinib
WO2012151361A1 (fr) Dérivés de carbamoylpyridone
US20150166601A1 (en) Deuterated carfilzomib
WO2018005328A1 (fr) Bictegravir deutérié
EP2680843A2 (fr) Dérivés de composés amino-hétéroaryles substitués par des pyrazoles
EP2542075A1 (fr) Dérivés de fluorouracile
JP2014005265A (ja) ピラゾール置換アミノ−ヘテロアリール化合物の誘導体
US8410082B2 (en) Fluorinated diaryl urea derivatives
US9676790B2 (en) Substituted thienotriazolodiazapines
US9707218B2 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
WO2012129381A1 (fr) Preladenant deutéré
WO2015009889A1 (fr) Dérivés deutériés d'intédanib et leur utilisation pour le traitement de troubles prolifératifs
EP2970213A1 (fr) Pacritinib deutérié
WO2013192512A1 (fr) Dérivés de composés amino-hétéroaryle substitués par un pyrazole
WO2014081816A1 (fr) Dérivés fluorés de composés amino-hétéroaryle substitués par pyrazole
WO2010068480A1 (fr) Dérivés deutérés de diméboline
WO2018013686A1 (fr) Idalopirdine deutérée

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20131002

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

R17D Deferred search report published (corrected)

Effective date: 20140417

RIC1 Information provided on ipc code assigned before grant

Ipc: A01N 43/56 20060101AFI20141211BHEP

Ipc: A61K 31/415 20060101ALI20141211BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20150407

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/00 20060101ALI20150330BHEP

Ipc: A61K 31/517 20060101ALI20150330BHEP

Ipc: A61K 31/4412 20060101ALI20150330BHEP

Ipc: C07D 401/14 20060101ALI20150330BHEP

Ipc: A01N 43/56 20060101AFI20150330BHEP

Ipc: A61K 31/415 20060101ALI20150330BHEP

Ipc: A61K 31/4545 20060101ALI20150330BHEP

Ipc: A61K 45/06 20060101ALI20150330BHEP

17Q First examination report despatched

Effective date: 20160418

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20160830