AU2012223281A1 - Derivatives of pyrazole-substituted amino-heteroaryl compounds - Google Patents

Derivatives of pyrazole-substituted amino-heteroaryl compounds Download PDF

Info

Publication number
AU2012223281A1
AU2012223281A1 AU2012223281A AU2012223281A AU2012223281A1 AU 2012223281 A1 AU2012223281 A1 AU 2012223281A1 AU 2012223281 A AU2012223281 A AU 2012223281A AU 2012223281 A AU2012223281 A AU 2012223281A AU 2012223281 A1 AU2012223281 A1 AU 2012223281A1
Authority
AU
Australia
Prior art keywords
cancer
compound
deuterium
hydrogen
pct
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
AU2012223281A
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 AU2012223281A1 publication Critical patent/AU2012223281A1/en
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
    • A61K31/435Heterocyclic 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

Landscapes

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

Abstract

This invention relates to novel pyrazole-substituted amino-heteroaryl compounds of Fomula (I) 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).

Description

WO 2012/119006 PCT/US2012/027341 1 DERIVATIVES OF PYRAZOLE-SUBSTITUTED AMINO-HE TEROARYL COMPOUNDS Cross Reference to Related Applications [0001] The present application claims priority to United States Provisional Application Number 61/448,887, filed March 3, 2011, which is hereby incorporated by reference in its entirety. Background of the Invention [0002] Many current medicines suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties that prevent their wider use or limit their use in certain indications. Poor ADME properties are also a major reason for the failure of drug candidates in clinical trials. While formulation technologies and prodrug strategies can be employed in some cases to improve certain ADME properties, these approaches often fail to address the underlying ADME problems that exist for many drugs and drug candidates. One such problem is rapid metabolism that causes a number of drugs, which otherwise would be highly effective in treating a disease, to be cleared too rapidly from the body. A possible solution to rapid drug clearance is frequent or high dosing to attain a sufficiently high plasma level of drug. This, however, introduces a number of potential treatment problems such as poor patient compliance with the dosing regimen, side effects that become more acute with higher doses, and increased cost of treatment. A rapidly metabolized drug may also expose patients to undesirable toxic or reactive metabolites. [0003] Another ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites. As a result, 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. In certain cases, 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. [0004] In some select cases, a metabolic inhibitor will be co-administered with a drug that is cleared too rapidly. Such is the case with the protease inhibitor class of drugs that are used to treat HIV infection. The FDA recommends that these drugs be co-dosed with WO 2012/119006 PCT/US2012/027341 2 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). Ritonavir, however, causes adverse effects and adds to the pill burden for HIV patients who must already take a combination of different drugs. Similarly, 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, however, 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). [0005] In general, combining drugs with cytochrome P450 inhibitors is not a satisfactory strategy for decreasing drug clearance. The inhibition of a CYP enzyme's activity can affect the metabolism and clearance of other drugs metabolized by that same enzyme. CYP inhibition can cause other drugs to accumulate in the body to toxic levels. [0006] A potentially attractive strategy for improving a drug's metabolic properties is deuterium modification. In this approach, one attempts to slow the CYP-mediated metabolism of a drug or to reduce the formation of undesirable metabolites by replacing one or more hydrogen atoms with deuterium atoms. Deuterium is a safe, stable, non radioactive 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. At the same time, because 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. [0007] Over the past 35 years, the effects of deuterium substitution on the rate of metabolism have been reported for a very small percentage of approved drugs (see, e.g., Blake, MI et al, J Pharm Sci, 1975, 64:367-91; Foster, AB, Adv Drug Res 1985, 14:1-40 ("Foster"); Kushner, DJ et al, Can J Physiol Pharmacol 1999, 79-88; Fisher, MB et al, Curr Opin Drug Discov Devel, 2006, 9:101-09 ("Fisher")). The results have been variable and unpredictable. For some compounds deuteration caused decreased WO 2012/119006 PCT/US2012/027341 3 metabolic clearance in vivo. For others, there was no change in metabolism. Still others demonstrated increased metabolic clearance. The variability in deuterium effects has also led experts to question or dismiss deuterium modification as a viable drug design strategy for inhibiting adverse metabolism (see Foster at p. 35 and Fisher at p. 101). [0008] The effects of deuterium modification on a drug's metabolic properties are not predictable even when deuterium atoms are incorporated at known sites of metabolism. Only by actually preparing and testing a deuterated drug can one determine if and how the rate of metabolism will differ from that of its non-deuterated counterpart. See, for example, Fukuto et al. (J. Med. Chem. 1991, 34, 2871-76). Many drugs have multiple sites where metabolism is possible. The site(s) where deuterium substitution is required and the extent of deuteration necessary to see an effect on metabolism, if any, will be different for each drug. [0009] Crizotinib also known as 3-[1(R)-(2,6-dichloro-3-fluorophenyl)ethoxy]-5-[1 (4-piperidinyl)-1H-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, 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. [0010] 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. [0011] Treatment with crizotinib has been associated with mild to moderate gastrointestinal-related events and fatigue. [0012] Despite the beneficial activities of crizotinib, there is a continuing need for new compounds to treat the aforementioned diseases and conditions.
WO 2012/119006 PCT/US2012/027341 4 Summary of the Invention [0013] 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. Definitions [0014] The term "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. [00151 "Disease" means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. [0016] It will be recognized that some variation of natural isotopic abundance occurs in a synthesized compound depending upon the origin of chemical materials used in the synthesis. Thus, a preparation of crizotinib will inherently contain small amounts of deuterated isotopologues. The concentration of naturally abundant stable hydrogen and carbon isotopes, notwithstanding this variation, is small and immaterial as compared to the degree of stable isotopic substitution of compounds of this invention. See, for instance, Wada, E et al., Seikagaku, 1994, 66:15; Gannes, LZ et al., Comp Biochem Physiol Mol Integr Physiol, 1998, 119:725. [0017] In the compounds of this invention any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Also unless otherwise stated, when 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).
WO 2012/119006 PCT/US2012/027341 5 [0018] The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. [0019] In other embodiments, 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). [0020] The term "isotopologue" refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof. [0021] The term "compound," when referring to a compound of this invention, refers to a collection of molecules having an identical chemical structure, except that there may be isotopic variation among the constituent atoms of the molecules. Thus, it will be clear to those of skill in the art that 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. However, as set forth above 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 1 7 .5%, less than 10%, less than 5%, less than 3%, less than 1%, or less than 0.5% of the compound. [0022] The invention also provides salts of the compounds of the invention. [0023] 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. According to another WO 2012/119006 PCT/US2012/027341 6 embodiment, a salt of a provided compound is a pharmaceutically acceptable acid addition salt. [0024] The term "pharmaceutically acceptable," as used herein, 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. A "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. A "pharmaceutically acceptable counterion" is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient. [0025] 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. 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-1,4-dioate, hexyne-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, 0-hydroxybutyrate, glycolate, maleate, tartrate, methanesulfonate, propanesulfonate, naphthalene- 1 -sulfonate, naphthalene-2 sulfonate, mandelate and other salts. In one embodiment, pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid. [0026] Unless otherwise indicated, when a disclosed compound is named or depicted WO 2012/119006 PCT/US2012/027341 7 by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound. [0027] The term "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. In one embodiment, the subject is a non-human animal. In another embodiment, the subject is a human. [0028] The compounds of the present invention (e.g., compounds of Formula I), may contain an asymmetric carbon atom, for example, as the result of deuterium substitution or otherwise. As such, compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers. Accordingly, 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. The term "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. Methods of obtaining or synthesizing an individual stereoisomer for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates. [0029] Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound. [0030] The term "stable compounds," as used herein, 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). [0031] "D" and "d" both refer to deuterium. "Stereoisomer" refers to both enantiomers and diastereomers. "Tert" and "t-" each refer to tertiary. "US" refers to the United States of America. [0032] "Substituted with deuterium" refers to the replacement of one or more WO 2012/119006 PCT/US2012/027341 8 hydrogen atoms with a corresponding number of deuterium atoms. [0033] Throughout this specification, a variable may be referred to generally (e.g.,"each R") or may be referred to specifically (e.g., R', R2, R', etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable. Therapeutic Compounds [0034] The present invention provides a compound of Formula I: X2b2 x 3 a NHX1a
X
3 b X5 Xlb N-N X 4 b X 4 a
R
1
R
3 A y2O NN 4 R2
NH
2 F (I), or a pharmaceutically acceptable salt thereof, wherein: RI and R 2 are each independently selected from Cl, CH 3 and CD 3 ; R3 is CH 3 or CD 3 ; la lb 2a 2b 3a 3b 4a 4b X , X , Xa, X2, Xa, X , Xa, X , and X 5 are each independently selected from hydrogen and deuterium;
Y
1 is hydrogen or deuterium; and
Y
2 is hydrogen or deuterium; provided that when each of R 1 and R2 are Cl, each of Xia, Xb, X2a, X 2, X 3a, X 3, X4a X 4, and X 5 is hydrogen and each of Y' and Y 2 is hydrogen, then R3 is CD 3 . [0035] In one embodiment of the compound of Formula I, when R 1 and R2 are each independently selected from Cl and CH 3 , each of Xia, X b, x 2 a, x2b, x 3 a, x3b, x 4 a, x4b, and X 5 is hydrogen and each of Y' and Y 2 is hydrogen, then R 3 is CD 3 . [0036] In one embodiment of a compound of Formula I, Xia and Xlb are the same, x 2 a and X2b are the same, X 3 a and X 3 b are the same, and X 4 a and X 4 b are the same. In one WO 2012/119006 PCT/US2012/027341 9 aspect of this embodiment R 1 and R2 are independently selected from Cl and CD 3 . In a further aspect of this embodiment, R 1 and R2 are the same and are each Cl. In another further aspect of this embodiment, R 1 and R2 are the same and are each CD 3 . [0037] In one embodiment of a compound of Formula I, Xi, X b, X2a and X 2 b are the same, X3a, X3 , X 4 a and X 4 b are the same and R 1 and R2 are independently selected from Cl and CD 3 . In one aspect, each of Xa, X b, X 2 a and X 2 b is hydrogen; and each of X 3 a, X 3, X4a and X 4 b is deuterium. In one aspect, each of X 1 a, X b, X2a and X 2 b is deuterium; and each of X 3 a, X3 , X4a and X 4 b is hydrogen. In one aspect, each of Xa, x 1, x 2a, x2b x 3a, x 3b, x 4 a and X 4 b is deuterium. In one aspect, each of Xa, x 1, x 2a, x 2, x 3a, x3, x 4 a and X 4 b is hydrogen. In one aspect of this embodiment, R 1 and R2 are the same and are each Cl. In one aspect of this embodiment, R 1 and R2 are the same and are each CD 3 . [0038] In one embodiment of a compound of Formula I, each of Xia, Xb, X 2a, X2b x 3a, x 3b, x 4 a and X4b is hydrogen, R1 and R2 are the same and are selected from Cl and
CD
3 and R 3 is CH 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, RI and R2 are each CD 3 . [0039] In one embodiment of a compound of Formula I, each of Xia, Xb, X 2a, X2b x 3a, x 3b, x 4 a and X4b is hydrogen, R1 and R2 are the same and are selected from Cl and
CD
3 and R 3 is CD 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, RI and R2 are each CD 3 . [0040] In one embodiment of a compound of Formula I, each of Xia, Xb, X 2a, X2b x 3a, x 3b, x 4 a and X4b is deuterium, R and R2 are the same and are selected from Cl and
CD
3 and R 3 is CH 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, RI and R2 are each CD 3 . [0041] In one embodiment of a compound of Formula I, each of Xia, Xb, X 2a, X2b x 3a, x 3b, x 4 a and X4b is deuterium; R and R2 are the same and are selected from Cl and
CD
3 and R 3 is CD 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, RI and R2 are each CD 3 . [0042] In one embodiment of a compound of Formula I, each of Xia, Xb, x 2 a and X 2 b is deuterium, each of X 3 a, X3b, x 4 a and X 4 b is hydrogen, R 1 and R2 are the same and are selected from Cl and CD 3 and R 3 is CH 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, R 1 and R2 are each CD 3
.
WO 2012/119006 PCT/US2012/027341 10 [0043] In one embodiment of a compound of Formula I, each of Xia, Xlb, x 2 a and X 2 b is deuterium, each of X 3 a, X3b, x 4 a and X 4 b is hydrogen, R 1 and R2 are the same and are selected from Cl and CD 3 and R 3 is CD 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, R 1 and R2 are each CD 3 . [0044] In one embodiment of a compound of Formula I, each of Xia, Xlb, x 2 a and X 2 b is hydrogen, each of X 3 a, x3b, x 4 a and X 4 b is deuterium, R 1 and R 2 are the same and are selected from Cl and CD 3 and R 3 is CH 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, R 1 and R2 are each CD 3 . [0045] In one embodiment of a compound of Formula I, each of Xia, Xb, x 2 a and X 2 b is hydrogen, each of X 3 a, X3b, x 4 a and X 4 b is deuterium, R 1 and R2 are the same and are selected from Cl and CD 3 and R 3 is CD 3 . In one aspect, R 1 and R2 are each Cl. In one aspect, R 1 and R2 are each CD 3 . [0046] In one embodiment of a compound of Formula I, X 5 is hydrogen, Y is hydrogen, Y 2 is hydrogen and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0047] In one embodiment of a compound of Formula I, X 5 is hydrogen, Y' is deuterium, Y 2 is hydrogen and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0048] In one embodiment of a compound of Formula I, X 5 is hydrogen, Y' is hydrogen, Y 2 is deuterium and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one WO 2012/119006 PCT/US2012/027341 11 aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0049] In one embodiment of a compound of Formula I, X 5 is hydrogen, Y is deuterium, Y 2 is deuterium and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, Xi, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X , Xa, X2 , X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0050] In one embodiment of a compound of Formula I, X 5 is deuterium, Y' is hydrogen, Y 2 is hydrogen and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0051] In one embodiment of a compound of Formula I, X 5 is deuterium, Y' is deuterium, Y 2 is hydrogen and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen. [0052] In one embodiment of a compound of Formula I, X 5 is deuterium, Y' is hydrogen, Y 2 is deuterium and R 1 and R2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xia, X, X 2a, X 2, X 3a, X 3, X4a and X 4 b is hydrogen. In one aspect each of , X, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is deuterium. In one aspect each of Xia X 1, X2a and X 2 b is hydrogen and each of X 3 a, X3b, X4a and X 4 b is deuterium. In one aspect each of Xia, X b, x 2 a and X 2 b is deuterium and each of X 3 a, X3b, X 4 a and X 4 b is hydrogen.
WO 2012/119006 PCT/US2012/027341 12 In one embodiment of a compound of Formula I, X 5 is deuterium, Y is deuterium, Y 2 is deuterium and R 1 and R 2 are the same and are selected from Cl and CD 3 . In one aspect, each of Xi, X b, X 2a, X 2, X 3a, X 3, X 4 a and X 4 b is hydrogen. In one aspect each of Xia, X 1, X 2a, X 2, X 3a, X 3, X4a and X 4 b is deuterium. In one aspect each of Xia, X 1, X2a and X2b is hydrogen and each of X 3 a, X 3 b, X 4 a and X 4 b is deuterium. In one aspect each of X a, X b, X 2 a and X 2 b is deuterium and each of X3a, X3b, X 4 a and X 4 b is hydrogen. [0053] In an example of any of the foregoing embodiments, 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. [0054] In one embodiment, the compound is selected from any one of the compounds (Cmpd) set forth in Table 1 (below): Table 1. Compounds of Formula I Cmpd Xia/ X 2 a/ X3a/ X4a/ X5 Y Y 2
R
1
R
2
R
3 X l
X
2 b X 3 b
X
4 b 100 D D D D D D D Cl Cl CD 3 101 D D D D D H D Cl Cl CD 3 102 D D D D H D H Cl Cl CH 3 103 D D H H D H D Cl Cl CD 3 104 D D H H D H H Cl Cl CH 3 105 D D H H D D H Cl Cl CH 3 106 D D H H H D H Cl Cl CH 3 107 D D H H H D D Cl Cl CD 3 108 H H D D D D H Cl Cl CD 3 109 H H D D D H H Cl Cl CH 3 110 H H D D D H D Cl Cl CD 3 111 H H D D D D H Cl Cl CH 3 112 H H H H H H H Cl Cl CD 3 113 H H H H H H H Cl Cl CD 3 114 D D D D D D D CD 3
CD
3
CD
3 115 D D D D D H D CD 3
CD
3
CD
3 116 D D D D H D H CD 3
CD
3
CH
3 117 D D H H D H D CD 3
CD
3
CD
3 118 D D H H D H H CD 3
CD
3
CH
3 119 D D H H D D H CD 3
CD
3
CH
3 120 D D H H H D H CD 3
CD
3
CH
3 121 D D H H H D D CD 3
CD
3
CD
3 122 H H D D D D H CD 3
CD
3
CD
3 123 H H D D D H H CD 3
CD
3
CH
3 WO 2012/119006 PCT/US2012/027341 13 Cmpd Xia/ I X 2 a/ I X 3 a/ X 4 a/ X Y Y 2 R R2 R 3 XlIb
X
2 b
X
3 b
X
4 b 124 H H D D D H D CD 3
CD
3
CD
3 125 H H D D D D H CD 3
CD
3
CH
3 126 H H H H H H H CD3 CD3 CD3 127 H H H H H H H CD 3
CD
3
CD
3 wherein any atom not designated as deuterium is present at its natural isotopic abundance, or a pharmaceutically acceptable salt thereof. [0055] The synthesis of compounds of Formula I may be readily achieved by synthetic chemists of ordinary skill by reference to the Exemplary Synthesis and Examples disclosed herein. Relevant procedures analogous to those of use for the preparation of compounds of Formula I and intermediates thereof are disclosed, for instance in Cui, J., WO 2006/021881, Cui, J. WO 2006/021884, Lui, J. WO 2010/108103, O'Donnell, C. J.; J. Med. Chem. 2010, 53, 1222-1237, and Shimizu, H. Tetrahedron Lett. 2006, 47, 5 927-5 93 1. [0056] 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. Exemplary Synthesis [0057] A convenient method for synthesizing compounds of Formula I is depicted in Scheme 1. [0058] Scheme la: Synthetic Route to Compounds of Formula I (R 1 and R 2 = Cl) Y1 CI O CI 1R N 1) DCI, D 2 0 (for R 3
CD
3 ) HO (12)
CH
3 (skip step for R 3 = CH 3 ) 2 0H NO 2 2) NaBD 4 or NaBH 4 CDIAD, PPh 3 F 3) Ac2O F 10a 11a WO 2012/119006 PCT/US2012/027341 14 CI R 3 I R31 CI R 3 O N 1) Fe, EtOH, HCI 0 N I NH 2 2) H10 4 , H 2
SO
4 Y2 NH 2 F AcOH, H 2 0 CI 13a F 14a v2a X2bX" N X~bx~a 3 a- NHxl x4O x l X x 5 X1a 1) pxla N-N X 4 b 1x 3 b x-Ba 7- ~ (15) xa X 2 b
Y
1 - CI R 3 Pd(ddpf)C1 2 , N KOAc/DMSO y2 0 2) 4M HCI/dioxane CI NH 2 CI F Formula I (R 1
=R
2 =CI) [0059] Scheme 1b: Synthetic Route to Compounds of Formula I (R1 and R 2 = CD3) CI O B-CD 3
CD
3 0 1) HCI, H 2 0 or CD 3 R
OH
3 HO CH 3 DCI, D 2 0 2 OH CI Pd(ddpf)C1 2 , CD3 NaBD 4 or NaBH 4 CD3
CD
3 3) AC 2 0OC F KOAc/DMSO F 4) PLE F 10a 10b 11b
I"Y
1 11 1 HO _1 (12) CD 3 R3 CD3 R3
NO
2 y2 0 N 1) Fe, EtOH, HCI 0 2 o N y2~ y2 N DIAD, PPh 3 CD3 NH 2 2) H10 4 , H 2
SO
4 CD3 NH 2 F AcOH, H 2 0 F 13b 14b WO 2012/119006 PCT/US2012/027341 15 v2a x 2 b /xN (15) X X 2 b 3 CD 3
NR
3 Pd(ddpf)Cl 2 ,N KOAc/DMSO O2 . 2) 4M HX/dioxane / NH 2
C
3 F Formula I (R 1
=R
2
=CD
3 ) [00601 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 Ta and Ib, above. Commercially available 2',6'-dichloro-3' fluoroacetophenone 10a may first be subjected to hydrogen-deuterium exchange in the presence of deuterium chloride in deuterium oxide to provide entities where Ri = CD 3 . [00611 Alternatively 10a may be subjected to Suzuki-Miyaura cross coupling with trideuteromethylboronic acid (commercially available) to provide ketones 10b where R 1 = R 2 = CD 3 . 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 11b in greater than 97.5% enantiomeric excess (ee). [00621 Mitsunobu inversion of the secondary alcohol with 2-nitropyridin-3-ols (12 where Y 1 = H or D) may be achieved with diisopropyl azidodicarboxylate (DIAD) and triphenylphosphine to provide biarylethers 13a or 13b. Following two functional group inter-conversions involving reduction of the nitro group and introduction of iodine at the five position on the pyridine, the scaffold is ready for union with heterocycle 15. [00631 Appropriately deuterated 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 WO 2012/119006 PCT/US2012/027341 16 free base. Preparation of appropriate pharmaceutical grade salt(s) will be necessary and may be accomplished using standard practices. [0064] Scheme 2: Synthetic Route to Intermediate 15. X2bxa Boc X X2a Boc NH x 3 a N x1a x 3 a N x1a
X
3 b X5 X1b MsCI X 3 b X5 X1 _ HO X 4a NEt 3 MsO NaH 16 17 X2bX2 ,Boc X 2 b x 2 a ,Boc xa N 1la o, o ~ 3 a N xla x 3 b X b B-B 3 b xlb
X
4 a 0 0X 4 a N-N X 4 b Pd(dppf) 2 Cl 2 N-N X 4 b 1/ KOAc / 18 OBO 15 WO 2012/119006 PCT/US2012/027341 17 [0065] The preparation of functionalized piperidines such as 16a, 16b, and the precursor to 16c and 16d, containing high levels of isotope abundance, has been previously disclosed (Lui, J. WO 2010/108103). Intermediates 16c and 16d may be prepared from the ketone precursor through reduction with NaBD 4 or NaBH 4 respectively. D HBoc Boc D D Boc D ND D N HH N D DDN D H H D D D H HO DD HO DD 0 HH 16a 16b Precursorto6cand6d D D Boc DD Boc H N D H N D D D H D H H HO HH HO HH 16c 16d Conversion of the secondary alcohol to the corresponding mesylate allows for installation of the 3-iodo-1-H-pyrazole moiety by direct displacement under anionic conditions. Elaboration of the iodo moiety of 18 to the boron pinacolate, to give 15, is achieved by reaction of dioxoboralane under palladium catalysis. [0066] Scheme 3a: Synthetic Route to Intermediate 12b. H i) NaOMe Br n-BuLi D N MeH'q HO MeOH HO MeOD HO
NO
2 ii) Br 2
NO
2
NO
2 12a 20 12b [0067] Scheme 3b: Alternate Synthetic Route to Intermediate 12b. HD O N N a O M e , M e O D D H N HO N HO- Z
NO
2
NO
2 12a 12b WO 2012/119006 PCT/US2012/027341 18 [0068] Isotopologues of 2-nitropyridin-3-ol such as 12b where Y 1 = D may be prepared by regioselective bromination on 2-nitropyridin-3-ol 12a as reported by O'Donnell ( J. Med. Chem. 2010, 53, 1222-1237). Halogen metal exchange followed by low temperature quench with appropriate isotope containing electrophile then inserts the desired isotope in correct position. Alternatively, direct hydrogen to deuterium exchange under alkaline conditions and deuterium oxide would yield 12b directly from 12a. [0069] The specific approaches and compounds shown above are not intended to be limiting. The chemical structures in the schemes herein depict variables that are hereby defined commensurately with chemical group definitions (moieties, atoms, etc.) of the corresponding position in the compound formulae herein, whether identified by the same variable name (i.e., R1, R2, R3, etc.) or not. The suitability of a chemical group in a compound structure for use in the synthesis of another compound is within the knowledge of one of ordinary skill in the art. [0070] Additional methods of synthesizing compounds of Formula I and their synthetic precursors, including those within routes not explicitly shown in schemes herein, are within the means of chemists of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the applicable compounds are known in the art and include, for example, those described in Larock R, Comprehensive Organic Transformations, VCH Publishers (1989); Greene, TW et al., Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley and Sons (1999); Fieser, L et al., Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette, L, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof. [0071] Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. Compositions [0072] 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. The carrier(s) are "acceptable" in the sense of being compatible with WO 2012/119006 PCT/US2012/027341 19 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. [0073] In some embodiments, the present invention provides 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. [0074] 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. [0075] If required, 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. [0076] 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 LUTROL 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. [0077] The pharmaceutical 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. In WO 2012/119006 PCT/US2012/027341 20 certain embodiments, 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). [0078] 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. In general, 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. [0079] In certain embodiments, the compound is administered orally. 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. [0080] In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When 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. [0081] 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. [0082] Compositions suitable for parenteral administration include aqueous and non aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended WO 2012/119006 PCT/US2012/027341 21 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 lyophilizedd) 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. [0083] 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. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, 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. [0084] The pharmaceutical 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. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols. [0085] The pharmaceutical 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 WO 2012/119006 PCT/US2012/027341 22 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. [0086] 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. For topical application topically to the skin, 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. Alternatively, 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. [0087] 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. [0088] Thus, according to yet another embodiment, 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. 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 WO 2012/119006 PCT/US2012/027341 23 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. [0089] According to another embodiment, 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. [0090] According to another embodiment, 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. [0091] According to another embodiment, 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. [0092] According to another embodiment, 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. [0093] Where 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. [0094] In another embodiment, 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.
WO 2012/119006 PCT/US2012/027341 24 [0095] Preferably, 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. [0096] In one embodiment, the second therapeutic agent is selected from kinase inhibitors. In one aspect of this embodiment, 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. PCT/US2009/053595), PF 00299804 and N-{2-[4-({3-chloro-4-[3-(trifluoromethyl)phenoxy]phenyl}amino)-5H pyrrolo [3,2-d]pyrimidin-5 -yl] ethyl} -3 -hydroxy-3 -methylbutaneamide (See US Patent Publication 2011/0003805). In a more specific embodiment, the deuterated form of D D 0 HN D 0 N D 0 N D D erlotinib is compound A, D D (A). In another more specific embodiment, the deuterated form of erlotinib is compound D D>j D O HN D 0 D D O N O I DD B, D D (B). In one more specific embodiment, the deuterated form of sorafenib is compound C, WO 2012/119006 PCT/US2012/027341 25 H D D 0 CI D D O IIN H F3C# N N D D
F
3 C H H D D D (C). In one aspect the kinase inhibitor is erlotinib or sorafenib. In one aspect 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). [0097] In one embodiment, a composition of this invention comprises a combination of the compound of Formula I with two second therapeutic agents selected from kinase inhibitors. In one aspect of this embodiment 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. 12/413,5 10, and sorafenib or a deuterated form of sorafenib as disclosed in PCT Patent Application No. PCT/US2009/053595). In a more specific aspect of this embodiment the combination is with erlotinib or compound A, and sorafenib or compound C. In another more specific aspect of this embodiment the combination is with erlotinib or compound B, and sorafenib or compound C. In one aspect of this embodiment the combination is erlotinib and sorafenib. In one aspect of this embodiment the combination is a deuterated form of erlotinib and a deuterated form of sorafenib. In one aspect of this embodiment the combination is a deuterated form of erlotinib and sorafenib. In one aspect of this embodiment the combination is erlotinib and a deuterated form of sorafenib. [0098] In another embodiment, 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. The term "associated with one another" as used herein 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). [0099] In the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term "effective WO 2012/119006 PCT/US2012/027341 26 amount" refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder. [00100] The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., Cancer Chemother. Rep, 1966, 50: 219. Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537. [00101] In one embodiment, 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: 300 mg, preferably administered orally twice a day; 250 mg, preferably administered orally twice a day; 200 mg, preferably administered orally twice a day, or once a day; 100 mg, preferably administered orally once a day; 50 mg, preferably administered orally once a day; from 200 to 300 mg, preferably administered orally twice a day; or 50-200 mg preferably administered orally once a day. [00102] 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. [00103] For pharmaceutical compositions that comprise a second therapeutic agent, 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. Preferably, 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 WO 2012/119006 PCT/US2012/027341 27 Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif (2000), each of which references are incorporated herein by reference in their entirety. [00104] It is expected that some of the second therapeutic agents referenced above will act synergistically with the compounds of this invention. When this occurs, it will allow the effective dosage of the second therapeutic agent and/or the compound of this invention to be reduced from that required in a monotherapy. This has the advantage of minimizing toxic side effects of either the second therapeutic agent of a compound of this invention, synergistic improvements in efficacy, improved ease of administration or use and/or reduced overall expense of compound preparation or formulation. Methods of Treatment [00105] In another embodiment, 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. [00106] According to another embodiment, 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. In one embodiment the subject is a patient in need of such treatment. Such 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 WO 2012/119006 PCT/US2012/027341 28 bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, glioblastoma, brain stem glioma, neuroblastoma, pituitary adenoma, solid tumors or a combination of one or more of the foregoing cancers. Such diseases include also abnormal cell growth disorders in which the disease is a benign proliferative disease, including, but not limited to psoriasis, benign prostatic hyperplasia and restinosis. [00107] According to another embodiment, the invention provides a method of treating abnormal cell growth in a mammal. [00108] In one particular embodiment, 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. [00109] 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). [00110] In another embodiment, 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. [00111] In particular, the combination therapies of this invention include co administering 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). [00112] The term "co-administered" as used herein 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 WO 2012/119006 PCT/US2012/027341 29 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. The administration of a 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. [00113] 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. [00114] In one embodiment of the invention, where a second therapeutic agent is administered to a subject, 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. [00115] In yet another aspect, 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 WO 2012/119006 PCT/US2012/027341 30 subject of a disease, disorder or symptom thereof delineated herein. Example. Evaluation of Metabolic Stability [00116] Microsomal Assay: Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). P-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCl 2 ), and dimethyl sulfoxide (DMSO) are purchased from Sigma-Aldrich. [00117] Determination of Metabolic Stability: 7.5 mM stock solutions of test compounds are prepared in DMSO. The 7.5 mM stock solutions are diluted to 12.5-50 tM in acetonitrile (ACN). 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 tL aliquot of the 12.5-50 pM 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 tM 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 tL aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 tL of ice-cold ACN with internal standard to stop the reactions. The plates are stored at 4 'C for 20 minutes after which 100 tL of water is added to the wells of the plate before centrifugation to pellet precipitated proteins. Supernatants are transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Bio-systems API 4000 mass spectrometer. The same procedure is followed for the non-deuterated counterpart of the compound of Formula I and the positive control, 7-ethoxycoumarin (1 gM). Testing is done in triplicate. [00118] Data analysis: The in vitro t 1
/
2 s for test compounds are calculated from the slopes of the linear regression of % parent remaining (ln) vs incubation time relationship. in vitro t / = 0.693/k k = -[slope of linear regression of % parent remaining(ln) vs incubation time] [00119] Data analysis is performed using Microsoft Excel Software.
WO 2012/119006 PCT/US2012/027341 31 [00120] Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. It should be understood that the foregoing discussion and examples merely present a detailed description of certain preferred embodiments. It will be apparent to those of ordinary skill in the art that various modifications and equivalents can be made without departing from the spirit and scope of the invention.

Claims (27)

1. A compound of Formula I: X2b2 x 3 a NHXla X3b x5 Xlb N-N X 4 b X 4 a R 1 R 3 y2 R2 NH 2 F (I) or a pharmaceutically acceptable salt thereof, wherein: RI and R 2 are each independently selected from Cl, CH 3 and CD 3 ; R 3 is CH 3 or CD 3 ; la lb 2a 2b 3a 3b 4a 4b5 X , X , Xa, X , Xa, X , Xa, X , and X are each independently selected from hydrogen and deuterium; Y' is hydrogen or deuterium; and Y 2 is hydrogen or deuterium; provided that when R 1 and R 2 are each independently selected from Cl and CH 3 , each or la lb 2a 2b 3a 3b 4a 4b X , X , Xa, X2, Xa, X , Xa, X4, and X 5 is hydrogen and each of Y' and Y 2 is hydrogen, then R 3 is CD 3 .
2. The compound of claim 1, wherein Xia and Xlb are the same; X 2 a and X 2 b are the same; X 3 a and X3b are the same; X 4 a and X4b are the same; and R 1 and R 2 are independently selected from Cl and CD 3 .
3. The compound of claim 2, wherein Xia, XI , X 2 a and X 2 b are the same; and X 3 a X 3, X4a and X 4 b are the same.
4. The compound of claim 3, wherein R1 and R 2 are the same. WO 2012/119006 PCT/US2012/027341 33
5. The compound of claim 4 wherein each X', each X 2 , each X 3 and each X 4 is hydrogen.
6. The compound of claim 4, wherein each X 1 , each X 2 , each X 3 and each X 4 is deuterium.
7. The compound of claim 4, wherein each X 1 and each X 2 are hydrogen; and each x 3 and each X 4 are deuterium.
8. The compound of claim 4, wherein each X 1 and each X 2 are deuterium; and each x 3 and each X 4 are hydrogen.
9. The compound of any one of claims 5-8 wherein X 5 is hydrogen, Y' is hydrogen and Y 2 is hydrogen.
10. The compound of any one of claims 5-8 wherein X 5 is hydrogen, Y' is deuterium and Y 2 is hydrogen.
11. The compound of any one of claims 5-8 wherein X 5 is hydrogen, Y' is hydrogen and Y 2 is deuterium
12. The compound of any one of claims 5-8 wherein X 5 is hydrogen, Y' is deuterium and Y 2 is deuterium.
13. The compound of any one of claims 5-8 wherein X 5 is deuterium , Y' is hydrogen and Y 2 is hydrogen.
14. The compound of any one of claims 5-8 wherein X 5 is deuterium , Y' is deuterium and Y 2 is hydrogen.
15. The compound of any one of claims 5-8 wherein X 5 is deuterium , Y' is hydrogen and Y 2 is deuterium. WO 2012/119006 PCT/US2012/027341 34
16. The compound of any one of claims 5-8 wherein X 5 is deuterium, Y is deuterium and Y 2 is deuterium.
17. The compound of claim 3 or 4 wherein R 3 is CH3.
18. The compound of claim 3 or 4 wherein R 3 is CD 3 .
19. The compound of claim 4 selected from any one of the compounds in the table below: Cmpd IXa/ X 2 a/ X 3 a/ X 4 a/ X 5 y 1 Y 2 R 1 and R 2 R 3 XIb X 2b X 3b X 4b 100 D D D D D D D Cl CD 3 101 D D D D D H D Cl CD 3 102 D D D D H D H Cl CH 3 103 D D H H D H D Cl CD 3 104 D D H H D H H Cl CH 3 105 D D H H D D H Cl CH 3 106 D D H H H D H Cl CH 3 107 D D H H H D D Cl CD 3 108 H H D D D D H Cl CD 3 109 H H D D D H H Cl CH 3 110 H H D D D H D Cl CD 3 111 H H D D D D H Cl CH 3 112 H H H H H H H Cl CD 3 113 H H H H H H H Cl CD 3 114 D D D D D D D CD 3 CD 3 115 D D D D D H D CD 3 CD 3 116 D D D D H D H CD 3 CH 3 117 D D H H D H D CD 3 CD 3 118 D D H H D H H CD 3 CH 3 119 D D H H D D H CD 3 CH 3 120 D D H H H D H CD 3 CH 3 121 D D H H H D D CD 3 CD 3 122 H H D D D D H CD 3 CD 3 123 H H D D D H H CD 3 CH 3 124 H H D D D H D CD 3 CD 3 125 H H D D D D H CD 3 CH 3 126 H H H H H H H CD 3 CD 3 127 H H H H H H H CD 3 CD 3 or a pharmaceutically acceptable salt thereof WO 2012/119006 PCT/US2012/027341 35
20. The compound of any one of claims 1-19 wherein any atom not designated as deuterium is present at its natural isotopic abundance.
21. A pyrogen-free pharmaceutical composition comprising a compound of claim 1 or 20 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
22. The composition of claim 21, further comprising a second therapeutic agent selected from kinase inhibitors.
23. The composition of claim 22, wherein the kinase inhibitor is selected from erlotinib, d-erlotinib, sorafenib, d-sorafenib, PF-00299804 and 454283.
24. The composition of claim 21, further comprising a combination of two second therapeutic agents selected from erlotinib or d-erlotinib and sorafenib or d-sorafenib.
25. A method of treating a disease or condition selected from 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 (CNS), primary CNS lymphoma, spinal axis tumors, glioblastoma, brain stem glioma, neuroblastoma, pituitary adenoma, solid tumors or a combination of one or more of the foregoing cancers; benign proliferative disease, including, but not limited to psoriasis, benign prostatic hyperplasia and restinosis in a subject comprising the step of administering to the subject in need thereof a composition of claim 21. WO 2012/119006 PCT/US2012/027341 36
26. The method of claim 25, wherein the disease or condition is selected from non small cell lung cancer (NSCLC), solid tumor cancer, neuroblastoma and lymphoma.
27. The method of claim 26, wherein the disease or condition is non-small cell lung cancer (NSCLC).
AU2012223281A 2011-03-03 2012-03-01 Derivatives of pyrazole-substituted amino-heteroaryl compounds Abandoned AU2012223281A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161448887P 2011-03-03 2011-03-03
US61/448,887 2011-03-03
PCT/US2012/027341 WO2012119006A2 (en) 2011-03-03 2012-03-01 Derivatives of pyrazole-substituted amino-heteroaryl compounds

Publications (1)

Publication Number Publication Date
AU2012223281A1 true AU2012223281A1 (en) 2013-09-19

Family

ID=46758504

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012223281A Abandoned AU2012223281A1 (en) 2011-03-03 2012-03-01 Derivatives of pyrazole-substituted amino-heteroaryl compounds

Country Status (5)

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

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103304552B (en) * 2012-03-09 2016-12-28 广东东阳光药业有限公司 Substituted pyridine compounds and using method thereof and purposes
MX2015008187A (en) 2012-12-20 2016-02-05 Concert Pharmaceuticals Inc Deuterated alk inhibitors.
CN104650049B (en) * 2013-08-28 2018-06-08 广东东阳光药业有限公司 Substituted pyridine compounds and its application method and purposes
WO2018213426A1 (en) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006021884A2 (en) * 2004-08-26 2006-03-02 Pfizer Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
KR101026676B1 (en) * 2005-12-05 2011-04-04 화이자 프로덕츠 인코포레이티드 Method of treating abnormal cell growth
EP2001870A2 (en) * 2006-03-31 2008-12-17 Schering Corporation Kinase inhibitors
CN101967140A (en) * 2010-09-14 2011-02-09 郑州泰基鸿诺药物科技有限公司 Deuterated crizotinib as well as derivant, preparation method and application thereof

Also Published As

Publication number Publication date
WO2012119006A3 (en) 2014-04-17
EP2680843A2 (en) 2014-01-08
CA2829025A1 (en) 2012-09-07
WO2012119006A2 (en) 2012-09-07
EP2680843A4 (en) 2015-05-06
MX2013010067A (en) 2014-01-31

Similar Documents

Publication Publication Date Title
EP2970209B1 (en) Deuterated palbociclib with improved metabolic stability
US9776973B2 (en) Deuterated momelotinib
BR122023027277A2 (en) DEUTERATE DERIVATIVES OF RUXOLITINIB AND PHARMACEUTICAL COMPOSITION
US20150166601A1 (en) Deuterated carfilzomib
US20120208837A1 (en) Substituted azaindoles
AU2012223281A1 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
JP2013521289A (en) Fluorouracil derivatives
JP2014005265A (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
US8410082B2 (en) Fluorinated diaryl urea derivatives
US9707218B2 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
WO2012129381A1 (en) Deuterated preladenant
WO2015009889A1 (en) Deuterated intedanib derivatives and their use for the treatment of proliferative disorders
AU2014240478A1 (en) Deuterated pacritinib
WO2013192512A1 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
US20150299170A1 (en) Fluoro-derivatives of pyrazole-substituted amino-heteroaryl compounds
WO2010068480A1 (en) Deuterated derivatives of dimeboline

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted