WO2011060216A1 - Substituted azaindoles - Google Patents

Substituted azaindoles Download PDF

Info

Publication number
WO2011060216A1
WO2011060216A1 PCT/US2010/056447 US2010056447W WO2011060216A1 WO 2011060216 A1 WO2011060216 A1 WO 2011060216A1 US 2010056447 W US2010056447 W US 2010056447W WO 2011060216 A1 WO2011060216 A1 WO 2011060216A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
deuterium
hydrogen
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/US2010/056447
Other languages
French (fr)
Inventor
Roger Tung
Craig E. Masse
I. Robert Silverman
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 WO2011060216A1 publication Critical patent/WO2011060216A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • ADME absorption, distribution, metabolism and/or excretion
  • 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.
  • quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism in a treatment of pseudobulbar affect.
  • Quinidine is a CYP2D6 inhibitor that 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).
  • 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.
  • a potentially attractive strategy for improving a drug's metabolic properties is deuterium modification.
  • 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.
  • This invention relates to novel deuterated azaindoles 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 a B-Raf V600E protein kinase inhibitor.
  • N-[3 - [(5 -chloro- 1 H-pyrrolo [2,3 -b]pyridine-3 -yl)carbonyl]-2,4-difluorophenyl] - 1 - propanesulfonamide is known to selectively inhibit oncogenic B-Raf V600E protein kinase (Puzanov, I. et al, J. Clin. Oncol, 2009, 27(15s): suppl; abstr 9021 ; Flaherty, K. et al, J. Clin. Oncol, 2009, 27(15s): suppl; abstr 9000).
  • Oncogenic mutations in the B-Raf gene have been linked to a variety of cancers.
  • PLX4032 is currently undergoing clinical evaluation for treatment of malignant melanoma and for colorectal cancer (see http://clinicaltrials.gov).
  • treat means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of 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.
  • alkyl refers to a monovalent saturated hydrocarbon group.
  • Ci-C alkyl is an alkyl having from 1 to 6 carbon atoms.
  • An alkyl may be linear or branched.
  • alkyl groups include methyl; ethyl; propyl, including w-propyl and isopropyl; butyl, including M-butyl, isobutyl, sec-butyl, and ?-butyl; pentyl, including, for example, w-pentyl, isopentyl, and neopentyl; and hexyl, including, for example, w-hexyl and 2-methylpentyl.
  • heteroaryl refers to a monovalent aromatic ring system having from 5 to 14 ring atoms, wherein from 1 to 4 ring atoms are heteroatoms independently selected from the group consisting of O, N and S.
  • the ring system may be a monocyclic, fused bicyclic, or fused tricyclic ring system. Examples of 5 to 14-membered heteroaryl include 5 to
  • heteroaryl groups include furanyl, furazanyl, imidazolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrimidinyl, phenanthridinyl, pyrazinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolinyl, thiadiazinyl, thiadiazolyl, thienyl, thienothiazolyl, thienooxazolyl,
  • halo or halogen refers to -CI, -Br, -F, and -I.
  • 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 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1% incorporation of deuterium).
  • isotopic enrichment factor 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.
  • 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.
  • 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 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.
  • the 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 salt that is non-toxic upon administration to a recipient at a therapeutically effective dose level, and 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,
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate,
  • 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. 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 of another possible stereoisomer.
  • substantially free of other stereoisomers 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 specified 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).
  • D refers to deuterium.
  • Stepoisomer refers to both enantiomers and
  • substituted with deuterium means that one or more positions in the indicated moiety are substituted with a deuterium atom.
  • 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 provides a compound of Formula II:
  • R 1 is C 1 -C4 alkyl optionally substituted with deuterium
  • each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is independently selected from hydrogen and deuterium;
  • R 2 is halo, C 1 -C4 alkyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein any hydrogen atom in R 2 is optionally replaced with deuterium;
  • At least one of R 1 and R 2 comprises deuterium.
  • R 1 is isopropyl optionally substituted with deuterium. In one aspect of this embodiment, R 1 is -CH(CH 3 ) 2 , -CD(CH 3 ) 2 , -CH(CD 3 ) 2 , or -CD(CD 3 ) 2 . [33] In one embodiment, R 1 is n-propyl optionally substituted with deuterium.
  • R 1 is -CH 2 CH 2 CH 3 , -CD 2 CH 2 CH 3 , -CH 2 CD 2 CH 3 , - CH 2 CH 2 CD 3 , -CD 2 CD 2 CH 3 , -CD 2 CH 2 CD 3 , -CH 2 CD 2 CD 3 , or -CD 2 CD 2 CD 3 .
  • R 2 is -CI, -Br, -CH 3 , or -CD 3 . In one aspect of this embodiment, R 2 is -CI. In one aspect of this embodiment, R 2 is -Br. In one aspect of this embodiment, R 2 is -CH 3 . In one aspect of this embodiment, R 2 is -CD 3 . In one example of each of the above aspects, R 1 is -CD 2 CD 2 CD 3 .
  • R 2 is -CH(CH 3 ) 2 , -CD(CH 3 ) 2 , -CH(CD 3 ) 2 , or -CD(CD 3 ) 2 .
  • R 2 is -C(CH 3 ) 3 or -C(CD 3 ) 3 .
  • R 2 is a 6-membered heteroaryl. In one aspect of this
  • R 2 is pyridinyl. In one aspect R 2 is pyridin-3-yl. In one example of each of the above aspects, R 1 is -CD 2 CD 2 CD 3 .
  • each of Y 1 , Y 2 , and Y 3 is deuterium. In another embodiment, each of Y 1 , Y 2 , and Y 3 is hydrogen.
  • each of Y 4 and Y 5 is deuterium. In one aspect of this
  • each of Y , Y , and Y is deuterium. In another aspect, each of Y , Y , and Y is hydrogen.
  • each of Y 4 and Y 5 is hydrogen. In one aspect of this
  • each of Y , Y , and Y is hydrogen. In another aspect, each of Y , Y , and Y is deuterium.
  • the compound of Formula II is a compound of Formula Ila:
  • the compound of Formula II is a compound of Formula lib:
  • Z 2 is isopropyl optionally substituted with deuterium and Z 3 is pyridin-3-yl optionally substituted with deuterium,
  • Z 3 is selected from
  • Z 3 may be selected from
  • any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
  • a compound of Formula Ila include a compound selected from the group consisting of the following, wherein (a) in 100a, 101a, 102a, 103a, 104a, 105a and 106a, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is deuterium; and (b) in 100b, 101b, 102b, 103b, 104b, 105b and 106b, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is hydrogen:
  • a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 200a, 201a, 202a, 203a, 204a, 205a and 206a, each of Y 1 , Y 2 , Y ? , Y 4 , and Y 5 is deuterium; and (b) in 200b, 201b, 202b, 203b 204b, 205b and 206b, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is hydrogen:
  • a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 300a, 301a, 302a, 303a, 304a, 305a and 306a, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is deuterium; and (b) in 300b, 301b, 302b, 303b, 304b, 305b and 306b, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is hydrogen:
  • a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 400a, 401a, 402a, 403a, 404a, 405a and 406a, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is deuterium; and (b) in 400b, 401b, 402b, 403b 404b, 405b and 406b, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is hydrogen:
  • a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 500a, 501a, 502a, 503a, 504a, 505a and 506a, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is deuterium; and (b) in 500b, 501b, 502b, 503b, 504b, 505b and 506b, each of Y 1 , Y 2 , Y 3 , Y 4 , and Y 5 is hydrogen: and in both (a) andb), all hydrogens on the 3-pyridyl substituent are at their naturally occurring abundance:
  • the compound is a compound of Formula I:
  • R 1 is C1-C4 alkyl optionally substituted with deuterium
  • R 2 is halo, C1-C4 alkyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein any hydrogen atom in R 2 is optionally replaced with deuterium;
  • R 1 and R 2 comprises deuterium
  • R 1 is isopropyl optionally substituted with deuterium. In one aspect of this embodiment, R 1 is -CH(CH 3 ) 2 , -CD(CH 3 ) 2 , -CH(CD 3 ) 2 , or -CD(CD 3 ) 2 .
  • R 1 is propyl optionally substituted with deuterium.
  • R 1 is -CH 2 CH 2 CH 3 , -CD 2 CH 2 CH 3 , -CH 2 CD 2 CH 3 , -CH 2 CH 2 CD 3 , -CD 2 CD 2 CH 3 , -CD 2 CH 2 CD 3 , -CH 2 CD 2 CD 3 , or -CD 2 CD 2 CD 3 .
  • R 2 is -CI, -Br, -CH 3 , or -CD 3 . In one aspect of this embodiment, R 2 is -CI.
  • R 2 is -CH(CH 3 ) 2 , -CD(CH 3 ) 2 , -CH(CD 3 ) 2 , or -CD(CD 3 ) 2 .
  • R 2 is -C(CH 3 ) 3 or -C(CD 3 ) 3 .
  • R 2 is a 6-membered heteroaryl. In one aspect of this
  • R 2 is pyridinyl. In one aspect R 2 is pyridin-3-yl.
  • the compound of the invention is a compound of Formula la:
  • the compound of Formula I is a compound of Formula lb:
  • Z 2 is isopropyl optionally substituted with deuterium and Z 3 is pyridin-3-yl optionally substituted with deuterium,
  • Z 3 is selected from
  • Z 3 may be selected from
  • a compound of Formula lb include a compound selected from the group consisting of:
  • any atom not designated as deuterium in any of the embodiments 107-112 set forth above is present at its natural isotopic abundance.
  • CD 3 CD 2 CD 2 S0 2 C1
  • CD 3 CD 2 CH 2 S0 2 C1
  • Scheme 1 depicts a route for preparing compounds of Formula II in a manner analogous to that presented in WO2007002325.
  • the route is useful, for example, for compounds where R 1 is -CD 2 CH 2 CH 3 or -CD(CH 3 ) 2 and R 2 is -CI, -Br, -CH 3 or -CD 3 .
  • Aniline 1, wherein Y 4 and Y 5 are each H, is prepared as described in patent publication WO 2007002325.
  • Aniline 1, wherein Y 4 and Y 5 are each D may be prepared from perdeuterated 2,4-difluoroaniline.
  • the latter compound is available from commercially available perdeuterated 1,2,4-trichlorobenzene following a procedure analogous to that of Mason et al., Syn. Comm., 1994, 24(4): 529-32.
  • the appropriately deuterated aniline 1 is treated with appropriately deuterated sulfonylchloride 2 to provide sulfonamide 3.
  • the benzyl group of 3 is removed via hydrogenation in the presence of palladium hydroxide on carbon to give carboxylic acid 4 which, upon treatment with thionyl chloride, affords acyl chloride 5.
  • Intermediate 2 may be one of 2a - 2g.
  • Intermediates 2a-2g may be prepared from the appropriately deuterated n-propyl thiol in a manner analogous to that described by Prakash, GKS et al, JOC, 2007, 72(15): 5847-50.
  • Starting material for the preparation of 2a CD 3 CD 2 CD 2 SH, is commercially available.
  • Other appropriately deuterated n-propyl thiols may be prepared from
  • deuterated n-propyl bromides in a manner analogous to that described by Cossar, BC, et al., JOC, 1962, 27:93-95.
  • deuterated n-propyl bromides include CH 3 CD 2 CD 2 Br, CD 3 CH 2 CD 2 Br, CD 3 CD 2 CH 2 Br, CD 3 CH 2 CH 2 Br, CH 3 CD 2 CH 2 Br, CH 3 CH 2 CD 2 Br. Additional intermediates 2 may be prepared as shown in Scheme 4 below. Intermediates 6a(i), 6b(i), and 6c(i) are commercially available.
  • Intermediate 6d(i) may be prepared from commercially available 2,3-dihydro-7-azaindole in a manner analogous to that described in patent publication WO 2004078757 using commercially available CD 3 I.
  • Intermediate 6a(ii) or 6a wherein Y 1 and Y 2 are each D and Y 3 is H may be prepared from commercially available perdeutero-5-chloro-2-pyridinamine in a manner analogous to that described in WO 2005095400.
  • Intermediate 6b(ii) may be prepared from commercially available perdeutero-7-azaindole in a manner analogous to that described by Wu, P.W. et al, JACS, 2006, 128(45): 14426-27.
  • Intermediate 6c(ii) may be prepared from commercially available perdeutero-7-azaindole in a manner analogous to that described in WO 2004078757.
  • Intermediate 6d(ii) may be prepared from commercially available perdeutero-7-azaindole and commercially available CD 3 I in a manner analogous to that described in WO 2004078757.
  • deuterated 3-bromopyridine examples include (a) commercially available cU-3- bromopyridine, (b) 3-bromo-6-deuteropyridine, prepared as described in Organic Letters, vol. 6, p. 4905-07 (2004); and (c) 3-bromo-5-deuteropyridine, prepared as described in Tetrahedron, vol. 56, p. 1349-60 (2000).
  • Scheme 3 depicts an example of a route to preparing compounds of Formula II.
  • the route is useful, for example, for compounds where R 1 is -CD 2 CH 2 CH 3 or -CD(CH 3 ) 2 and R 2 is -CI or 3-pyridyl.
  • Aniline 8a wherein Y 4 and Y 5 are each H, is commercially available.
  • Aniline 8b, wherein Y 4 and Y 5 are each D may be prepared from commercially available perdeutero-l,3-benzenediamine in a manner analogous to that described by Wang et al, Nongyao, 2005, 44(1): 13-15.
  • Appropriately deuterated aniline 8 is treated with
  • Oxidation of 11 with either Dess-Martin periodinane (WO 2008079906) or DDQ (Tsai, J. et al. Proc. Natl. Acad. Sci. USA 2008, 105(8): 3041-3046) provides compounds of Formula II wherein R 2 is -CI or 3-pyridyl. It would be readily apparent to one of skill in the art that compounds of Formula I, la, and lb may be prepared as described in schemes 1, 2 and/or 3 above.
  • deuterated intermediates 2 may be prepared as depicted in Scheme 4 in a manner analogous to that described in patent publication WO 2007140317A2. Conversion of appropriately deuterated alkyl halide 12a or 12b to the corresponding sulfonic acid in the presence of aqueous sodium sulfite followed by treatment with thionyl chloride results in the formation of sulfonylchloride 2.
  • alkyl bromides (or chlorides), 12a and 12b, of use in Scheme 4 are shown below.
  • CD 3 CDBr(CI) CD 3 (CH 3 )CHBr(CI) (CH 3 ) 2 CDBr(CI) (CD 3 ) 2 CHBr(CI)
  • the invention also provides pyrogen-free compositions comprising a compound of Formula I or II (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and an acceptable carrier.
  • the composition comprises an effective amount of the compound of Formula I or II.
  • a compound of Formula I or II e.g., including any of the formulae herein
  • a pharmaceutically acceptable salt of said compound e.g., including any of the formulae herein
  • composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is a pharmaceutically acceptable carrier.
  • 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.
  • 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 phosphat
  • 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,
  • a composition of this invention further comprises a second therapeutic agent.
  • the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound that selectively inhibits oncogenic B-Raf V600E protein kinase.
  • the second therapeutic agent is an agent useful in the treatment or prevention of a disease or condition selected from neoplastic diseases and associated complications, including chemotherapy -induced hypoxia.
  • a disease or condition selected from neoplastic diseases and associated complications, including chemotherapy -induced hypoxia.
  • gastrointestinal stromal tumors GISTs
  • prostate tumors including canine mast ceil tumors
  • mast cell tumors including canine mast ceil tumors
  • acute myeloid leukemia acute lymphocytic leukemia
  • chronic myeloid leukemia multiple myeloma
  • melanoma melanoma.
  • mastocytosis gliomas, glioblastoma, astrocytoma, neuroblastoma
  • sarcomas e.g. sarcomas of neuroectodermal origin
  • carcinomas e.g.
  • lung breast, pancreatic, renal, female genital tract, carcinoma in situ
  • lymphoma e.g. histiocytic lymphoma
  • neurofibromatosis including Schwann cell neoplasia
  • myelodysplasia syndrome leukemia, tumor angiogenesis, and cancers of the thyroid, liver, bone, skin, brain, pancreas, lung (e.g. small cell lung cancer), breast, colon, prostate, testes and ovary.
  • the disease or condition is selected from melanoma, thyroid cancer, ovarian cancer, and colorectal cancer.
  • 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 a disease or disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
  • an effective amount of a compound of this invention can range from about 0.01 to about 5000 mg per treatment. In more specific embodiments the range is from about 0.1 to 2500 mg, or from 0.2 to 1000 mg, or most specifically from about 1 to 500 mg. Treatment typically is administered one to three times daily.
  • 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 patient, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
  • 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, 2 nd 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 treating a patient suffering from, or susceptible to, a disease that is beneficially treated by an inhibitor of oncogenic B-Raf protein kinase comprising the step of administering to said patient an effective amount of a compound of this invention or a pharmaceutically acceptable salt of said compound or a composition of this invention.
  • diseases include diseases that are treated by selectively inhibiting oncogenic B-Raf V600E protein kinase.
  • any of the above methods of treatment comprises the further step of co-administering to the patient 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 a compound that selectively inhibits oncogenic B-Raf V600E protein kinase.
  • 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.
  • co-administered means that the second therapeutic agent may be administered (i) 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; or (ii) prior to, consecutively with, or following the administration of a compound of this invention.
  • 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
  • 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 patient does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said patient 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, 2 nd 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 or II, or a pharmaceutically acceptable salt of said compound, 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 patient of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula I or II or a pharmaceutically acceptable salt thereof for use in the treatment or prevention in a patient of a disease, disorder or symptom thereof delineated herein.
  • Step 2. 1.1.2.2.3.3.3-d7-Propane-l-sulfonyl chloride (2a): A solution of 21 (824 mg, 7.61 mmol) in AcOD (2.20 mL, 38.1 mmol, Sigma-Aldrich, 99 atom % D) and D 2 0 (0.230 mL, 1 1.4 mmol, Cambridge Isotope Laboratories, 99.8 atom % D) was stirred at 50 °C for 30 minutes. Sulfuryl chloride (6.17 mL, 76.1 mmol) was then added dropwise and the reaction continued to stir for an additional 30 minutes.
  • Step 4 N-(2.4-Difluoro-3 -formylphenyl)- 1.1.2.2.3.3.3-d7-propane- 1 -sulfonamide (23): To a solution of N,N-diisopropylamine (0.775 mL, 5.48 mmol) in THF (10 mL) at -78 °C was added dropwise w-butyllithium (2.95 mL, 5.48 mmol, 1.86M solution in hexane). The resulting solution stirred at 0 °C for 1 hour then was cooled to -78 °C and a solution of 7 (604 mg, 2.49 mmol) in THF (5 mL) was added dropwise.
  • N-formylmorpholine (0.302 mL, 2.99 mmol) was added and the reaction stirred at room temperature for 15 hours and subsequently quenched with IN HC1 and extracted with EtOAc (3 x 50 mL). The organic layers were combined, washed with water, dried ( a 2 S04), filtered and concentrated under reduced pressure. The resulting brown solid was
  • Step 6 N-(3-(5-Chloro-lH-pyrrolor2.3-b1pyridine-3-carbonyl)-2.4-difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (106b): 2,3-Dichloro-5,6-dicyano-l,4-benzoquinone (DDQ) (42.0 mg, 0.184 mmol) was added to a solution of azaindole 24 (60.0 mg, 0.142 mmol) in 1,4-dioxane (1.00 mL) and water (0.100 mL). The resulting solution stirred at room temperature for 2 hours then was quenched with sat.
  • DDQ 2,3-Dichloro-5,6-dicyano-l,4-benzoquinone
  • Step 2 N-(3 -(5 -Bromo- 1 H-pyrrolo ⁇ 2.3 -blpyridine-3 -carbonyl)-2.4-difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (206b) : The preparation was carried out according to the procedure for compound 106b to afford ketone 206b. MS (ESI) 463.0 [(M - H) ].
  • Human liver microsomes (20 mg/mL) are available from Xenotech, LLC (Lenexa, KS).
  • ⁇ -nicotinamide adenine dinucleotide phosphate, reduced form ( ADPH), magnesium chloride (MgC ⁇ ), and dimethyl sulfoxide (DMSO) are available 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 MgCl2.
  • 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.

Abstract

This invention relates to novel substituted azaindoles 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 a compound showing selective inhibitory activity of oncogenic B-RafV600E protein kinase.

Description

SUBSTITUTED AZAINDOLES
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 61/260,752, filed November 12, 2009, and U.S. Provisional Application No. 61/360,043, filed June 30, 2010, both of which are incorporated by reference herein in their entirety.
Background of the Invention
[1] Many current medicines suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties that prevent their wider use. 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 maintain 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.
[2] 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 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, quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism in a treatment of pseudobulbar affect. Quinidine, however, is a CYP2D6 inhibitor that 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). [3] 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.
[4] 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 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.
[5] 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 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).
[6] 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 overall metabolism will differ from that of its undeuterated 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.
[7] This invention relates to novel deuterated azaindoles 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 a B-RafV600E protein kinase inhibitor.
[8] N-[3 - [(5 -chloro- 1 H-pyrrolo [2,3 -b]pyridine-3 -yl)carbonyl]-2,4-difluorophenyl] - 1 - propanesulfonamide is known to selectively inhibit oncogenic B-RafV600E protein kinase (Puzanov, I. et al, J. Clin. Oncol, 2009, 27(15s): suppl; abstr 9021 ; Flaherty, K. et al, J. Clin. Oncol, 2009, 27(15s): suppl; abstr 9000). Oncogenic mutations in the B-Raf gene have been linked to a variety of cancers. Reportedly, approximately 70% of melanoma cases, 30- 70% of thyroid cancer cases, 15-30% of ovarian cancer cases and 5-20% of colorectal cancer cases have been associated with B-Raf-activating mutations (Sala, E. et al, Mol Cancer Res., 2008 May, 6(5): 751-9). A single-base mutation to B-Raf at nucleotide 1799 in codon 600 of exon 15 leading to a valine-to-glutamate substitution (V600E) has been found in more than 85% of melanomas with a B-Raf mutation (Langland, R. et al, EP2036990A.1).
[9] PLX4032 is currently undergoing clinical evaluation for treatment of malignant melanoma and for colorectal cancer (see http://clinicaltrials.gov).
[10] Despite the purported beneficial activities of N-[3-[(5-chloro-lH-pyrrolo[2,3- b]pyridine-3-yl)carbonyl]-2,4-difluorophenyl]-l-propanesulfonamide, there is a continuing need for new compounds that have beneficial effects for treatment of malignant melanoma and for colorectal cancer.
Definitions
[11] 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).
[12] "Disease" means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
[13] The term "alkyl" refers to a monovalent saturated hydrocarbon group. Ci-C alkyl is an alkyl having from 1 to 6 carbon atoms. An alkyl may be linear or branched. Examples of alkyl groups include methyl; ethyl; propyl, including w-propyl and isopropyl; butyl, including M-butyl, isobutyl, sec-butyl, and ?-butyl; pentyl, including, for example, w-pentyl, isopentyl, and neopentyl; and hexyl, including, for example, w-hexyl and 2-methylpentyl.
[14] The term "heteroaryl" refers to a monovalent aromatic ring system having from 5 to 14 ring atoms, wherein from 1 to 4 ring atoms are heteroatoms independently selected from the group consisting of O, N and S. The ring system may be a monocyclic, fused bicyclic, or fused tricyclic ring system. Examples of 5 to 14-membered heteroaryl include 5 to
10-membered heteroaryl and 5 to 6-membered heteroaryl. More particular examples of heteroaryl groups include furanyl, furazanyl, imidazolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrimidinyl, phenanthridinyl, pyrazinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolinyl, thiadiazinyl, thiadiazolyl, thienyl, thienothiazolyl, thienooxazolyl,
thienoimidazolyl, thiophenyl, triazinyl, and triazolyl.
[15] The term "halo" or "halogen" refers to -CI, -Br, -F, and -I.
[16] 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 N-[3-[(5-chloro-lH-pyrrolo[2,3-b]pyridine-3-yl)carbonyl]-2,4- difluorophenyl]-l-propanesulfonamide 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.
[17] 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 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1% incorporation of deuterium).
[18] The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
[19] 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).
[20] 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. [21] 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 17.5%, less than 10%, less than 5%, less than 3%, less than 1%, or less than 0.5% of the compound.
[22] 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. According to another embodiment, the compound is a pharmaceutically acceptable acid addition salt.
[23] 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 salt that is non-toxic upon administration to a recipient at a therapeutically effective dose level, and 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.
[24] 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-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, β-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.
[25] The compounds of the present invention (e.g., compounds of Formula I and II), 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 of 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 enantiomer for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates.
[26] 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.
[27] 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 specified 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). [28] "D" refers to deuterium. "Stereoisomer" refers to both enantiomers and
diastereomers. "Tert" and "t-" each refer to tertiary. "US" refers to the United States of America.
[29] The phrase "substituted with deuterium" means that one or more positions in the indicated moiety are substituted with a deuterium atom.
[30] Throughout this specification, a variable may be referred to generally (e.g., "each R") or may be referred to specifically (e.g., R1, R2, R3, 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
The present invention provides a compound of Formula II:
Figure imgf000008_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is C1-C4 alkyl optionally substituted with deuterium;
each of Y1, Y2, Y3, Y4, and Y5 is independently selected from hydrogen and deuterium;
and
R2 is halo, C1-C4 alkyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein any hydrogen atom in R2 is optionally replaced with deuterium;
with the proviso that if each of Y1, Y2, Y3, Y4, and Y5 is hydrogen, then
at least one of R1 and R2 comprises deuterium.
[32] In one embodiment, R1 is isopropyl optionally substituted with deuterium. In one aspect of this embodiment, R1 is -CH(CH3)2, -CD(CH3)2, -CH(CD3)2, or -CD(CD3)2. [33] In one embodiment, R1 is n-propyl optionally substituted with deuterium. In one aspect of this embodiment, R1 is -CH2CH2CH3, -CD2CH2CH3, -CH2CD2CH3, - CH2CH2CD3, -CD2CD2CH3, -CD2CH2CD3, -CH2CD2CD3, or -CD2CD2CD3.
[34] In one embodiment, R2 is -CI, -Br, -CH3, or -CD3. In one aspect of this embodiment, R2 is -CI. In one aspect of this embodiment, R2 is -Br. In one aspect of this embodiment, R2 is -CH3. In one aspect of this embodiment, R2 is -CD3. In one example of each of the above aspects, R1 is -CD2CD2CD3.
[35] In another embodiment, R2 is -CH(CH3)2, -CD(CH3)2, -CH(CD3)2, or -CD(CD3)2. In another embodiment, R2 is -C(CH3)3 or -C(CD3)3.
[36] In one embodiment, R2 is a 6-membered heteroaryl. In one aspect of this
embodiment, R2 is pyridinyl. In one aspect R2 is pyridin-3-yl. In one example of each of the above aspects, R1 is -CD2CD2CD3.
[37] In one embodiment, each of Y1, Y2, and Y3 is deuterium. In another embodiment, each of Y1, Y2, and Y3 is hydrogen.
[38] In one embodiment, each of Y4 and Y5 is deuterium. In one aspect of this
1 2 3 1 2 3 embodiment, each of Y , Y , and Y is deuterium. In another aspect, each of Y , Y , and Y is hydrogen.
[39] In one embodiment, each of Y4 and Y5 is hydrogen. In one aspect of this
embodiment, each of Y , Y , and Y is hydrogen. In another aspect, each of Y , Y , and Y is deuterium.
[40] In one embodiment of this invention the compound of Formula II is a compound of Formula Ila:
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein Z1 is n-propyl substituted with deuterium. [41] In one embodiment of this invention the compound of Formula II is a compound of Formula lib:
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein Z2 is isopropyl optionally substituted with deuterium and Z3 is pyridin-3-yl optionally substituted with deuterium,
with the proviso that at least one of Z2 and Z3 comprises deuterium. In one aspect of this embodiment, Z3 is selected from
Figure imgf000010_0002
For example, Z3 may be selected from
Figure imgf000010_0003
[42] In another set of embodiments, any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
[43] Specific examples of a compound of Formula Ila include a compound selected from the group consisting of the following, wherein (a) in 100a, 101a, 102a, 103a, 104a, 105a and 106a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 100b, 101b, 102b, 103b, 104b, 105b and 106b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
Figure imgf000011_0001
Figure imgf000012_0001
or a pharmaceutically acceptable salt of any of the foregoing.
[44] Specific examples of a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 200a, 201a, 202a, 203a, 204a, 205a and 206a, each of Y1, Y2, Y?, Y4, and Y5 is deuterium; and (b) in 200b, 201b, 202b, 203b 204b, 205b and 206b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
Figure imgf000012_0002
Figure imgf000013_0001
or a pharmaceutically acceptable salt of any of the foregoing. [45] Specific examples of a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 300a, 301a, 302a, 303a, 304a, 305a and 306a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 300b, 301b, 302b, 303b, 304b, 305b and 306b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
Figure imgf000014_0001
Figure imgf000015_0001
or a pharmaceutically acceptable salt of any of the foregoing.
[46] Specific examples of a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 400a, 401a, 402a, 403a, 404a, 405a and 406a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 400b, 401b, 402b, 403b 404b, 405b and 406b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
Figure imgf000015_0002
Figure imgf000016_0001

Figure imgf000017_0001
or a pharmaceutically acceptable salt of any of the foregoing.
[47] Specific examples of a compound of Formula Ila also include a compound selected from the group consisting of the following, wherein (a) in 500a, 501a, 502a, 503a, 504a, 505a and 506a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 500b, 501b, 502b, 503b, 504b, 505b and 506b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen: and in both (a) andb), all hydrogens on the 3-pyridyl substituent are at their naturally occurring abundance:
Figure imgf000017_0002
Figure imgf000018_0001
or a pharmaceutically acceptable salt of any of the foregoing.
[48] In one embodiment of this invention, the compound is a compound of Formula I:
Figure imgf000019_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is C1-C4 alkyl optionally substituted with deuterium; and
R2 is halo, C1-C4 alkyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein any hydrogen atom in R2 is optionally replaced with deuterium;
with the proviso that at least one of R1 and R2 comprises deuterium.
[49] In one embodiment, R1 is isopropyl optionally substituted with deuterium. In one aspect of this embodiment, R1 is -CH(CH3)2, -CD(CH3)2, -CH(CD3)2, or -CD(CD3)2.
[50] In one embodiment, R1 is propyl optionally substituted with deuterium. In one aspect of this embodiment, R1 is -CH2CH2CH3, -CD2CH2CH3, -CH2CD2CH3, -CH2CH2CD3, -CD2CD2CH3, -CD2CH2CD3, -CH2CD2CD3, or -CD2CD2CD3.
[51] In one embodiment, R2 is -CI, -Br, -CH3, or -CD3. In one aspect of this embodiment, R2 is -CI.
[52] In another embodiment, R2 is -CH(CH3)2, -CD(CH3)2, -CH(CD3)2, or -CD(CD3)2. In another embodiment, R2 is -C(CH3)3 or -C(CD3)3.
[53] In one embodiment, R2 is a 6-membered heteroaryl. In one aspect of this
embodiment, R2 is pyridinyl. In one aspect R2 is pyridin-3-yl.
[54] In one embodiment the compound of the invention is a compound of Formula la:
Figure imgf000019_0002
or a pharmaceutically acceptable salt thereof, wherein Z1 is n-propyl substituted with deuterium.
[55] In one embodiment of this invention the compound of Formula I is a compound of Formula lb:
Figure imgf000020_0001
or a pharmaceutically acceptable salt thereof, wherein Z2 is isopropyl optionally substituted with deuterium and Z3 is pyridin-3-yl optionally substituted with deuterium,
with the proviso that at least one of Z2 and Z3 comprises deuterium. In one aspect of this embodiment, Z3 is selected from
Figure imgf000020_0002
For example, Z3 may be selected from
Figure imgf000020_0003
[56] Specific examples of a compound of Formula lb include a compound selected from the group consisting of:
Figure imgf000021_0001
or a pharmaceutically acceptable salt of any of the foregoing. [57] In another set of embodiments, any atom not designated as deuterium in any of the embodiments 107-112 set forth above is present at its natural isotopic abundance.
[58] The synthesis of compounds of Formula I, la, lb, II, Ila and lib may be readily achieved by synthetic chemists of ordinary skill by reference to the Exemplary Synthesis disclosed herein. Relevant procedures analogous to those of use for the preparation of compounds of Formula I, la, lb, II, Ila and lib and intermediates thereof are disclosed, for instance in patent publication WO 2007002325.
Exemplary Synthesis
[59] Compounds of Formula I, la, lb, II, Ila and lib may be prepared according to the schemes shown below.
[60] Scheme 1 : Preparation of Compounds of Formula II.
Figure imgf000022_0001
Formula II
(R2 = CI, Br, CH3 01- CD3) Intermediates 2a-2g
CD3CD2CD2S02C1 CD3CD2CH2S02C1
CD3CH2CD2S02C1 CH3CD2CD2S02C1
CD3CH2CH2S02C1 CH3CD2CH2S02C1
CH3CH2CD2S02C1
Intermediates 6
Figure imgf000023_0001
6a(i) - 6d(i) 6a(ii) - 6d(ii)
6a: R2 = CI 6a: R2 = CI
6b: R2 = Br 6b: R2 = Br
6c: R2 = CH3 6c: R2 = CH3
6d: R2 = CD3 6d: R2 = CD3
[61] Scheme 1 depicts a route for preparing compounds of Formula II in a manner analogous to that presented in WO2007002325. The route is useful, for example, for compounds where R1 is -CD2CH2CH3 or -CD(CH3)2 and R2 is -CI, -Br, -CH3 or -CD3.
Aniline 1, wherein Y4 and Y5 are each H, is prepared as described in patent publication WO 2007002325. Aniline 1, wherein Y4 and Y5 are each D, may be prepared from perdeuterated 2,4-difluoroaniline. The latter compound is available from commercially available perdeuterated 1,2,4-trichlorobenzene following a procedure analogous to that of Mason et al., Syn. Comm., 1994, 24(4): 529-32. The appropriately deuterated aniline 1 is treated with appropriately deuterated sulfonylchloride 2 to provide sulfonamide 3. The benzyl group of 3 is removed via hydrogenation in the presence of palladium hydroxide on carbon to give carboxylic acid 4 which, upon treatment with thionyl chloride, affords acyl chloride 5.
Treatment of 5 with 7-azaindole 6a - 6d in the presence of aluminum trichloride gives compounds of Formula II wherein R2 is -CI, -Br, -CH3 or -CD3. Intermediate 2 may be one of 2a - 2g. Intermediates 2a-2g may be prepared from the appropriately deuterated n-propyl thiol in a manner analogous to that described by Prakash, GKS et al, JOC, 2007, 72(15): 5847-50. Starting material for the preparation of 2a, CD3CD2CD2SH, is commercially available. Other appropriately deuterated n-propyl thiols may be prepared from
commercially available deuterated n-propyl bromides in a manner analogous to that described by Cossar, BC, et al., JOC, 1962, 27:93-95. Examples of commercially available deuterated n-propyl bromides include CH3CD2CD2Br, CD3CH2CD2Br, CD3CD2CH2Br, CD3CH2CH2Br, CH3CD2CH2Br, CH3CH2CD2Br. Additional intermediates 2 may be prepared as shown in Scheme 4 below. Intermediates 6a(i), 6b(i), and 6c(i) are commercially available.
Intermediate 6d(i) may be prepared from commercially available 2,3-dihydro-7-azaindole in a manner analogous to that described in patent publication WO 2004078757 using commercially available CD3I. Intermediate 6a(ii) or 6a wherein Y1 and Y2 are each D and Y3 is H, may be prepared from commercially available perdeutero-5-chloro-2-pyridinamine in a manner analogous to that described in WO 2005095400. Intermediate 6b(ii) may be prepared from commercially available perdeutero-7-azaindole in a manner analogous to that described by Wu, P.W. et al, JACS, 2006, 128(45): 14426-27. Intermediate 6c(ii) may be prepared from commercially available perdeutero-7-azaindole in a manner analogous to that described in WO 2004078757. Intermediate 6d(ii) may be prepared from commercially available perdeutero-7-azaindole and commercially available CD3I in a manner analogous to that described in WO 2004078757.
[62] Scheme 2: Preparation of Compounds of Formula II wherein R2 is 3-pyridyl.
Figure imgf000024_0001
[63] Compounds of Formula II wherein R2 is 3-pyridyl may be prepared as depicted in Scheme 2 in a manner analogous to that presented in WO 2007002325. A compound of Formula II wherein R2 = Br is treated with palladium tetrakis(triphenylphosphine), potassium carbonate, and pyridin-3-ylboronic acid 7 to provide a compound of Formula II wherein R2 = 3-pyridyl. Analogously, a deuterated pyridin-3-ylboronic acid, in which one or more pyridinyl ring hydrogens are replaced with deuterium, may be used instead of 7 to provide a compound of Formula II where the pyridinyl ring is deuterated. Such a deuterated pyridin-3- ylboronic acid may be prepared from the corresponding deuterated 3-bromopyridine.
Examples of deuterated 3-bromopyridine include (a) commercially available cU-3- bromopyridine, (b) 3-bromo-6-deuteropyridine, prepared as described in Organic Letters, vol. 6, p. 4905-07 (2004); and (c) 3-bromo-5-deuteropyridine, prepared as described in Tetrahedron, vol. 56, p. 1349-60 (2000).
[64] Scheme 3 : Alternate Approach to the Preparation of Compounds of Formula II.
Figure imgf000025_0001
Formula II (R2 = CI or 3-pyridyl)
Intermediates 6
Figure imgf000025_0002
6a(i) or 6e(i) 6a(ii) or 6e(ii)
6a: R2 = CI 6a: R2 = CI
6e: R2 = 3-pyridyl 6e: R2 = 3-pyridyl
[65] Scheme 3 depicts an example of a route to preparing compounds of Formula II. The route is useful, for example, for compounds where R1 is -CD2CH2CH3 or -CD(CH3)2 and R2 is -CI or 3-pyridyl. Aniline 8a, wherein Y4 and Y5 are each H, is commercially available. Aniline 8b, wherein Y4 and Y5 are each D may be prepared from commercially available perdeutero-l,3-benzenediamine in a manner analogous to that described by Wang et al, Nongyao, 2005, 44(1): 13-15. Appropriately deuterated aniline 8 is treated with
appropriately deuterated sulfonyl chloride 2 to provide sulfonamide 9. Deprotonation with n- butyllithium followed by reaction with dimethylformamide gives aldehyde 10 (in a manner analogous to what is described in WO 2009012283 and WO 2008079906). Treatment of 10 with potassium hydroxide and commercially available 6a(i) or 6e(i) (prepared as described in patent publication WO 2007002325) gives appropriately deuterated 11. Similarly, treatment with commercially-available potassium deuteroxide and 6a(ii) (prepared as described above for Scheme 1) or 6e(ii) (prepared from 6b(ii) in a manner analogous to that described in WO 2007002325) gives appropriately deuterated 11. Oxidation of 11 with either Dess-Martin periodinane (WO 2008079906) or DDQ (Tsai, J. et al. Proc. Natl. Acad. Sci. USA 2008, 105(8): 3041-3046) provides compounds of Formula II wherein R2 is -CI or 3-pyridyl. It would be readily apparent to one of skill in the art that compounds of Formula I, la, and lb may be prepared as described in schemes 1, 2 and/or 3 above.
[66] Scheme 4. General Preparation of Sulfonylchloride (2).
1. aq Na2S03
R1-Br or R1-CI R1S02CI
2. SOCI2
12a 12b 2
[67] Appropriately deuterated intermediates 2 may be prepared as depicted in Scheme 4 in a manner analogous to that described in patent publication WO 2007140317A2. Conversion of appropriately deuterated alkyl halide 12a or 12b to the corresponding sulfonic acid in the presence of aqueous sodium sulfite followed by treatment with thionyl chloride results in the formation of sulfonylchloride 2. Commercially available alkyl bromides (or chlorides), 12a and 12b, of use in Scheme 4 are shown below.
CD3CD2CD2Br(CI) CH3CH2CD2Br(CI) CH3CD2CD2Br CD3CH2CD2Br
CD3CH2CH2Br CD3CD2CH2Br CH3CD2CH2Br
(CD3)2CDBr(CI) CD3(CH3)CHBr(CI) (CH3)2CDBr(CI) (CD3)2CHBr(CI)
[68] 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. Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds.
Compositions
[69] The invention also provides pyrogen-free compositions comprising a compound of Formula I or II (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and an acceptable carrier. In one embodiment, the composition comprises an effective amount of the compound of Formula I or II. Preferably, a
composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is a pharmaceutically acceptable carrier. 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.
[70] 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.
[71] 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. [72] 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 PLURONIC™ (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.
[73] 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 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).
[74] In another embodiment, a composition of this invention further comprises a second therapeutic agent. The second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound that selectively inhibits oncogenic B-RafV600E protein kinase.
[75] Preferably, the second therapeutic agent is an agent useful in the treatment or prevention of a disease or condition selected from neoplastic diseases and associated complications, including chemotherapy -induced hypoxia., gastrointestinal stromal tumors (GISTs), prostate tumors, mast cell tumors (including canine mast ceil tumors), acute myeloid leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, multiple myeloma, melanoma., mastocytosis, gliomas, glioblastoma, astrocytoma, neuroblastoma, sarcomas (e.g. sarcomas of neuroectodermal origin), carcinomas (e.g. lung, breast, pancreatic, renal, female genital tract, carcinoma in situ), lymphoma (e.g. histiocytic lymphoma), neurofibromatosis (including Schwann cell neoplasia), myelodysplasia syndrome, leukemia, tumor angiogenesis, and cancers of the thyroid, liver, bone, skin, brain, pancreas, lung (e.g. small cell lung cancer), breast, colon, prostate, testes and ovary. As an example, the disease or condition is selected from melanoma, thyroid cancer, ovarian cancer, and colorectal cancer.
[76] In one 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).
[77] In one embodiment of the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term "effective amount" refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat a disease or disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
[78] The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al, (1966) Cancer Chemother. Rep 50: 219. Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
[79] In one embodiment, an effective amount of a compound of this invention can range from about 0.01 to about 5000 mg per treatment. In more specific embodiments the range is from about 0.1 to 2500 mg, or from 0.2 to 1000 mg, or most specifically from about 1 to 500 mg. Treatment typically is administered one to three times daily.
[80] 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 patient, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
[81] 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 Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
Methods of Treatment
[82] According to another embodiment, the invention provides a method of treating a patient suffering from, or susceptible to, a disease that is beneficially treated by an inhibitor of oncogenic B-Raf protein kinase comprising the step of administering to said patient an effective amount of a compound of this invention or a pharmaceutically acceptable salt of said compound or a composition of this invention. Such diseases include diseases that are treated by selectively inhibiting oncogenic B-RafV600E protein kinase.
[83] In another embodiment, any of the above methods of treatment comprises the further step of co-administering to the patient 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 a compound that selectively inhibits oncogenic B-RafV600E protein kinase. 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.
[84] The term "co-administered" as used herein means that the second therapeutic agent may be administered (i) 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; or (ii) 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 patient does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said patient at another time during a course of treatment.
[85] 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.
[86] 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.
[87] In yet another aspect, the invention provides the use of a compound of Formula I or II, or a pharmaceutically acceptable salt of said compound, 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 patient of a disease, disorder or symptom set forth above. Another aspect of the invention is a compound of Formula I or II or a pharmaceutically acceptable salt thereof for use in the treatment or prevention in a patient of a disease, disorder or symptom thereof delineated herein.
Examples
[88] Example 1. Synthesis of N-(3-(5-Chloro-lH-pyrrolor2,3-b1pyridine-3-carbonyl -2,4- difluorophenyl)- 1, 1,2,2,3,3,3 -d7-propane- 1 -sulfonamide (106b).
[89] Scheme 5. Preparation of Compound 106b.
Figure imgf000031_0001
[90] Step 1. 3-Thiocyanatol .1.2.2.3.3.3-d7-propane (21): To a solution of άη- bromopropane 20 (0.740 mL, 7.69 mmol, CDN, 98 atom % D) in EtOD (1 1 mL, Sigma- Aldrich, 99.5+ atom % D) was added potassium thiocyanate (1.12 g, 11.5 mmol). The reaction was heated to reflux and stirred for 3 hours. Upon completion, the reaction was cooled to room temperature, diluted with D20 (Cambridge Isotope Laboratories, 99.8 atom % D), and extracted with EtOAc (3 x 50 mL). The organic layers were combined, washed with D20, dried (Na2S04), filtered and concentrated under reduced pressure to afford thiocyanate 21 (824 mg, 99%) which was used in the next step without further purification.
[91] Step 2. 1.1.2.2.3.3.3-d7-Propane-l-sulfonyl chloride (2a): A solution of 21 (824 mg, 7.61 mmol) in AcOD (2.20 mL, 38.1 mmol, Sigma-Aldrich, 99 atom % D) and D20 (0.230 mL, 1 1.4 mmol, Cambridge Isotope Laboratories, 99.8 atom % D) was stirred at 50 °C for 30 minutes. Sulfuryl chloride (6.17 mL, 76.1 mmol) was then added dropwise and the reaction continued to stir for an additional 30 minutes. Upon cooling to room temperature the reaction was quenched by the dropwise addition of water (10 mL) and the resulting solution was extracted with EtOAc (3 x 50 mL). The organic layers were combined, washed with water, dried (Na2S04), filtered and concentrated under reduced pressure to afford 2a (1.14 g, 99%) as a clear oil which was used in the next step without further purification. MS (ESI) ) sulfonic acid ion observed: 130.2 [(M - H)"].
[92] Step 3. N-(2.4-Difluorophenyl)-l .1.2.2.3.3.3 -d7-propane-l -sulfonamide (22):
Sulfonyl chloride 2a (1.14 g, 7.61 mmol) was added dropwise to a solution of 2,4- difluoroaniline (0.740 mL, 7.35 mmol), pyridine (0.615 mL, 7.61 mmol) and DMAP (45.0 mg, 0.370 mmol) in dichloromethane (10 mL) at room temperature. The reaction was stirred at 45 °C for 15 hours then cooled to room temperature, diluted with water and extracted with EtOAc (3 x 50 mL). The organic layers were combined, washed with brine, dried (Na2S04), filtered and concentrated under reduced pressure. The resulting brown solid was
recrystallized with EtO Ac/Heptanes to afford sulfonamide 22 (604 mg, 34%) as a tan solid. MS (ESI) 241.1 [(M - H)"].
[93] Step 4. N-(2.4-Difluoro-3 -formylphenyl)- 1.1.2.2.3.3.3-d7-propane- 1 -sulfonamide (23): To a solution of N,N-diisopropylamine (0.775 mL, 5.48 mmol) in THF (10 mL) at -78 °C was added dropwise w-butyllithium (2.95 mL, 5.48 mmol, 1.86M solution in hexane). The resulting solution stirred at 0 °C for 1 hour then was cooled to -78 °C and a solution of 7 (604 mg, 2.49 mmol) in THF (5 mL) was added dropwise. After stirring at -78 °C for 4 hours, N-formylmorpholine (0.302 mL, 2.99 mmol) was added and the reaction stirred at room temperature for 15 hours and subsequently quenched with IN HC1 and extracted with EtOAc (3 x 50 mL). The organic layers were combined, washed with water, dried ( a2S04), filtered and concentrated under reduced pressure. The resulting brown solid was
recrystallized from EtO Ac/heptanes to afford formylsulfonamide 23 (265 mg, 40%) as a tan solid. MS (ESI) 269.1 [(M - H)"].
[94] Step 5. N-(3 -((5-Chloro- 1 Η-ρνηχ)1οΓ2.3 -blpyridin-3 -yl)(hvdroxy)-methyl)-2.4- difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (24): To a solution of
formylsulfonamide 23 (50.0 mg, 0.185 mmol) in MeOH/Water 1 : 1 (1 mL) was added 5- chloroazaindole 6a(i) (30.0 mg, 0.196 mmol, Adesis, Inc.) followed by K2CO3 (171 mg, 1.24 mmol). The resulting solution stirred at room temperature for 3 days then was neutralized to pH 7 with 4N HC1 and extracted with EtO Ac (3 x 50 mL). The organic layers were combined, washed with water, dried (Na2S04), filtered and concentrated under reduced pressure to afford azaindole 24 (60 mg, 77%) as a tan solid which was used without further purification. MS (ESI) 421.0 [(M - H) "].
[95] Step 6. N-(3-(5-Chloro-lH-pyrrolor2.3-b1pyridine-3-carbonyl)-2.4-difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (106b): 2,3-Dichloro-5,6-dicyano-l,4-benzoquinone (DDQ) (42.0 mg, 0.184 mmol) was added to a solution of azaindole 24 (60.0 mg, 0.142 mmol) in 1,4-dioxane (1.00 mL) and water (0.100 mL). The resulting solution stirred at room temperature for 2 hours then was quenched with sat. aHC03 and extracted with 1 : 1 THF/EtOAc (3 x 15 mL). The organic layers were combined, diluted with heptanes (10 mL), dried (Na2S04), filtered through a pad of silica gel and concentrated under reduced pressure to afford ketone 106b as a tan solid. MS (ESI) 419.1 [(M - H) "].
[96] Example 2. Synthesis of N-(3-(5-Bromo-lH-pyrrolor2.3-b1pyridine-3-carbonyl)-2.4- difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (206b).
[97] Scheme 6. Preparation of Compound 206b.
Figure imgf000033_0001
Figure imgf000034_0001
[98] Step 1. N-G-(('5-Bromo-lH-pyrrolor2.3-blpyridin-3-vn(hvdroxy)-methvn-2.4- difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (25): The preparation was carried out according to the procedure for compound 24 employing 5-bromoazaindole 6b(i) (Adesis, Inc.) in lieu of 5-chloroazaindole 6a(i) to afford azaindole 25 (142 mg, 82%) as a tan solid which was used without further purification. MS (ESI) 465.0 [(M - H)"].
[99] Step 2. N-(3 -(5 -Bromo- 1 H-pyrrolo Γ2.3 -blpyridine-3 -carbonyl)-2.4-difluorophenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (206b) : The preparation was carried out according to the procedure for compound 106b to afford ketone 206b. MS (ESI) 463.0 [(M - H) ].
[100] Example 3. Synthesis of N-(2.4-Difluoro-3-(5-(pyridin-3-yl)-lH-pyrrolor2.3- blpyridine-3 -carbonvDphenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (506b)
Scheme 7. Preparation of Compound 506b.
Figure imgf000034_0002
[102] N-(2.4-Difluoro-3-(5-(pyridin-3-yl)-lH-pyrrolor2.3-b1pyridine-3-carbonyl)phenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (506b) : To a solution of ketone 206b (30.0 mg, 0.0645 mmol), in water (0.300 mL) and acetonitrile (0.600 mL) was added pyridine-3- boronic acid (40.0 mg, 0.323 mmol), K2C03 (54.0 mg, 0.387 mmol) and
tetrakis(triphenylphoshine)palladium(0) (1.00 mg, 8.6xl0"7 mol). The resulting solution stirred in a sealed pressure flask at 170 °C for 15 hours then was cooled to room temperature, diluted with water and extracted with EtOAc (3 x 5 mL). The organic layers were combined, dried (Na2S04), filtered through a pad of silica gel and concentrated under reduced pressure to afford ketone 506b. MS (ESI) 464.2 [(M + H)+].
[103] Example 4. Synthesis of N-(2.4-Difluoro-3-(5-methyl-lH-pyrrolor2.3-b1pyridine-3- carbonyDphenyl)- 1.1.2.2.3.3.3-d7-propane- 1 -sulfonamide (306b).
[104] Scheme 8. Preparation of Compound 306b.
Figure imgf000035_0001
206b 306b
[105] N-(2.4-Difluoro-3 -(5-methyl- 1 H-pyrrolo \2.3 -blpyridine-3 -carbonvDphenyl)- 1.1.2.2.3.3.3 -d7-propane- 1 -sulfonamide (306b) : The preparation was carried out according to the procedure for compound 506b employing trimethylboroxine in lieu of 3-pyridineboronic acid to afford ketone 306b. MS (ESI) 401.2 [(M + H)+].
[106] Example 5. Synthesis of N-(2.4-Difluoro-3-(5-d^-methyl-lH-pyrrolor2.3-blpyridine- 3-carbonyl)phenvD- 1.1.2.2.3.3.3-d7-propane-l -sulfonamide (406b).
[107] Scheme 9. Preparation of Compound 406b.
Figure imgf000035_0002
206b 406b
N-(2.4-Difluoro-3 -(5-d^-methyl- 1 H-pyrrolo \2.3 -blpyridine-3 -carbonvDphenyl)- 1.1.2.2.3.3.3 - d7-propane-l -sulfonamide (406b): The preparation is carried out according to the procedure for compound 506b employing d3-methylboronic acid in lieu of 3-pyridineboronic acid to afford ketone 406b. [108] Example 6. Evaluation of Metabolic Stability in Human Liver Microsomes.
Human liver microsomes (20 mg/mL) are available from Xenotech, LLC (Lenexa, KS). β-nicotinamide adenine dinucleotide phosphate, reduced form ( ADPH), magnesium chloride (MgC^), and dimethyl sulfoxide (DMSO) are available from Sigma-Aldrich.
[109] 7.5 mM stock solutions of test compounds are prepared in DMSO. The 7.5 mM stock solutions are diluted to 12.5 - 50 μΜ 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 MgCl2. 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 μΜ 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 μΜ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl2. The reaction mixtures are incubated at 37 °C, and 50 μΐ^ aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 μΐ^ of ice-cold ACN with internal standard to stop the reactions. The plates are stored at 4 °C for 20 minutes after which 100 μΐ^ 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 PLX4032 and the positive control, 7-ethoxycoumarin (1 μΜ). Testing is done in triplicate.
[110] The in vitro ti/2s for test compounds are calculated from the slopes of the linear regression of % parent remaining (In) vs incubation time relationship:
in vitro t ½ = 0.693/k
k = -[slope of linear regression of % parent remaining(ln) vs incubation time].
Data analysis is performed using Microsoft Excel Software.
[Ill] 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

What is claimed is
1. A compound of Formula II
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is C1-C4 alkyl optionally substituted with deuterium;
each of Y1, Y2, Y3, Y4, and Y5 is independently selected from hydrogen and deuterium;
and
R2 is halo, C1-C4 alkyl, 5-membered heteroaryl or 6-membered heteroaryl, wherein any hydrogen atom in R2 is optionally replaced with deuterium;
with the proviso that if each of Y1, Y2, Y3, Y4, and Y5 is hydrogen, then
at least one of R1 and R2 comprises deuterium.
2. The compound of claim 1, wherein R1 is isopropyl optionally substituted with deuterium.
3. The compound of claim 2, wherein R1 is -CH(CH3)2, -CD(CH3)2, - CH(CD3)2, or -CD(CD3)2.
4. The compound of claim 1 , wherein R1 is n-propyl optionally substituted with deuterium.
5. The compound of claim 4, wherein R1 is -CH2CH2CH3, -CD2CH2CH3, -CH2CD2CH3, -CH2CH2CD3, -CD2CD2CH3, -CD2CH2CD3, -CH2CD2CD3, or -CD2CD2CD3. 6. The compound of any one of the preceding claims, wherein R2 is -CI, -Br, - CH3, or -CD3.
7. The compound of claim 6, wherein R2 is -CI.
8. The compound of claim 6, wherein R2 is -Br.
9. The compound of claim 6, wherein R2 is -CH3 or -CD3.
10. The compound of any one of claims 1-5, wherein R2 is -CH(CH3)2, - CD(CH3)2, -CH(CD3)2, or -CD(CD3)2.
1 1. The compound of any one of claims 1 -5, wherein R2 is -C(CH3)3 or -C(CD3)3.
12. The compound of any one of claims 1 to 5, wherein R2 is a 6-membered heteroaryl.
13. The compound of claim 12, wherein R2 is pyridyl.
14. The compound of claim 13, wherein R2 is pyridin-3-yl.
15. The compound of any one of claims 1- 14, wherein each of Y1, Y2, and Y~ is deuterium.
16. The compound of any one of claims 1-14, wherein each of Y1, Y2, and Y3 is hydrogen.
17. The compound of any one of claims 1-16, wherein each of Y4 and Y5 is deuterium.
18. The compound of any one of claims 1-16, wherein each of Y4 and Y5 is hydrogen. compound of claim 1 , wherein the compound is a compound of Formula
Figure imgf000039_0001
or a pharmaceutically acceptable salt thereof, wherein Z1 is n-propyl substituted with deuterium.
20. The compound of claim 19, wherein the compound is selected from the group consisting of the following compounds, wherein (a) in 100a, 101a, 102a, 103a, 104a, 105a and 106a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 100b, 101b, 102b, 103b, 104b 105b and 106b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
a pharmaceutically acceptable salt of any of the foregoing. 21. The compound of claim 1 or 8 wherein the compound is selected from the group consisting of the following compounds, wherein (a) in 200a, 201a, 202a, 203a, 204a, 205a and 206a, each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 200b, 201b, 202b, 203b 204b, 205b and 206b, each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
22. The compound of claim 1 or 9 wherein the compound is selected from the group consisting of the following compounds, wherein (a) in 300a, 301a, 302a, 303a, 304a, 305a, 306a, 400a, 401a, 402a, 403a, 404a, 405a and 406a each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 300b, 301b, 302b, 303b, 304b, 305b, 306b, 400b, 401b, 402b, 403b, 404b 405b and 406b each of Y1, Y2, Y3, Y4, and Y5 is hydrogen:
Figure imgf000042_0001
or a pharmaceutically acceptable salt of any of the foregoing.
23. The compound of claim 1 or 14 wherein the compound is selected from the group consisting of the following compounds, wherein (a) in 500a, 501a, 502a, 503a, 504a, 405a and 406a each of Y1, Y2, Y3, Y4, and Y5 is deuterium; and (b) in 500b, 501b, 502b, 503b, 504b, 505b and 506b each of Y1, Y2, Y?, Y4, and Y5 is hydrogen:
506a, 506b or a pharmaceutically acceptable salt of any of the foregoing.
24. The compound of any one of claims 1 to 23, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
25. A pyrogen-free pharmaceutical composition comprising a compound of any one of claims 1 to 24 or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier.
26. The composition of claim 25 additionally comprising a second therapeutic agent useful in the treatment or prevention of a disease or condition selected from melanoma, thyroid cancer, ovarian cancer, and colorectal cancer.
27. A method of treating a patient suffering from, or susceptible to, a disease or condition selected from chemotherapy-induced hypoxia, gastrointestinal stromal tumors (GISTs), prostate tumors, mast cell tumors, acute myeloid leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, multiple myeloma, melanoma, mastocytosis, gliomas, glioblastoma, astrocytoma, neuroblastoma, sarcomas, carcinomas, lymphoma,
neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, and cancers of the thyroid, liver, bone, skin, brain, pancreas, lung, breast, colon, prostate, testes and ovary, comprising the step of administering to the patient in need thereof an effective amount of a composition of claim 25.
28. The method of claim 27, wherein the disease or condition is selected from melanoma, thyroid cancer, ovarian cancer, and colorectal cancer.
PCT/US2010/056447 2009-11-12 2010-11-12 Substituted azaindoles WO2011060216A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US26075209P 2009-11-12 2009-11-12
US61/260,752 2009-11-12
US36004310P 2010-06-30 2010-06-30
US61/360,043 2010-06-30

Publications (1)

Publication Number Publication Date
WO2011060216A1 true WO2011060216A1 (en) 2011-05-19

Family

ID=43415196

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/056447 WO2011060216A1 (en) 2009-11-12 2010-11-12 Substituted azaindoles

Country Status (1)

Country Link
WO (1) WO2011060216A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012037060A1 (en) * 2010-09-13 2012-03-22 Concert Pharmaceuticals Inc. Substituted azaindoles
EP2672967A1 (en) * 2011-02-07 2013-12-18 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078757A2 (en) 2003-03-06 2004-09-16 Eisai Co., Ltd. Synthesis of 5-substituted 7-azaindoles and 7-azaidonines
WO2005095400A1 (en) 2004-03-30 2005-10-13 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of jak and other protein kinases
US7014866B2 (en) 2001-05-03 2006-03-21 Hoffmann-La Roche Inc. High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
US20060079502A1 (en) 1999-11-02 2006-04-13 Steffen Lang Pharmaceutical compositions
US20060094744A1 (en) 2004-09-29 2006-05-04 Maryanoff Cynthia A Pharmaceutical dosage forms of stable amorphous rapamycin like compounds
WO2007002325A1 (en) 2005-06-22 2007-01-04 Plexxikon, Inc. Pyrrolo[2,3-b] pyridine derivatives as protein kinase inhibitors
WO2007140317A2 (en) 2006-05-26 2007-12-06 Wyeth Use of inhibitors of cytosolic ph0sph0lipase a2 in the treatment of thrombosis
WO2008079903A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives as kinase modulators
WO2009012283A1 (en) 2007-07-17 2009-01-22 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2036990A1 (en) 2007-09-11 2009-03-18 Roche Diagnostics GmbH Diagnostic test for susceptibility to B-Raf kinase inhibitors

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060079502A1 (en) 1999-11-02 2006-04-13 Steffen Lang Pharmaceutical compositions
US7014866B2 (en) 2001-05-03 2006-03-21 Hoffmann-La Roche Inc. High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
WO2004078757A2 (en) 2003-03-06 2004-09-16 Eisai Co., Ltd. Synthesis of 5-substituted 7-azaindoles and 7-azaidonines
WO2005095400A1 (en) 2004-03-30 2005-10-13 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of jak and other protein kinases
US20060094744A1 (en) 2004-09-29 2006-05-04 Maryanoff Cynthia A Pharmaceutical dosage forms of stable amorphous rapamycin like compounds
WO2007002325A1 (en) 2005-06-22 2007-01-04 Plexxikon, Inc. Pyrrolo[2,3-b] pyridine derivatives as protein kinase inhibitors
WO2007140317A2 (en) 2006-05-26 2007-12-06 Wyeth Use of inhibitors of cytosolic ph0sph0lipase a2 in the treatment of thrombosis
WO2008079903A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives as kinase modulators
WO2008079906A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives as kinase modulators
WO2009012283A1 (en) 2007-07-17 2009-01-22 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2036990A1 (en) 2007-09-11 2009-03-18 Roche Diagnostics GmbH Diagnostic test for susceptibility to B-Raf kinase inhibitors

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
"Informa Healthcare", 2007, article "Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences)"
"PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia", 2000, TARASCON PUBLISHING
"Pharmacotherapy Handbook", 2000, APPLETON AND LANGE
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
"Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples", 2006, WILEY-INTERSCIENCE
BLAKE, MI ET AL., J PHARM SCI, vol. 64, 1975, pages 367 - 91
COSSAR, BC ET AL., JOC, vol. 27, 1962, pages 93 - 95
FISHER, MB ET AL., CURR OPIN DRUG DISCOV DEVEL, vol. 9, 2006, pages 101 - 09
FLAHERTY, K. ET AL., J. CLIN. ONCOL., vol. 27, no. 15S, 2009, pages 9000
FOSTER, AB, ADV DRUG RES, vol. 14, 1985, pages 1 - 40
FREIREICH ET AL., CANCER CHEMOTHER. REP, vol. 50, 1966, pages 219
FUKUTO ET AL., J. MED. CHEM., vol. 34, 1991, pages 2871 - 76
GANNES LZ ET AL., COMP BIOCHEM PHYSIOL MOL INTEGR PHYSIOL, vol. 119, 1998, pages 725
KEMPF, D.J. ET AL., ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 41, no. 3, 1997, pages 654 - 60
KUSHNER, DJ ET AL., CAN J PHYSIOL PHARMACOL, 1999, pages 79 - 88
MASON ET AL., SYN. COMM., vol. 24, no. 4, 1994, pages 529 - 32
ORGANIC LETTERS, vol. 6, 2004, pages 4905 - 07
PRAKASH, GKS ET AL., JOC, vol. 72, no. 15, 2007, pages 5847 - 50
PUZANOV, I. ET AL., J. CLIN. ONCOL., vol. 27, 2009, pages 9021
SALA, E. ET AL., MOL CANCER RES., vol. 6, no. 5, May 2008 (2008-05-01), pages 751 - 9
TETRAHEDRON, vol. 56, 2000, pages 1349 - 60
TSAI, J. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 105, no. 8, 2008, pages 3041 - 3046
WADA E ET AL., SEIKAGAKU, vol. 66, 1994, pages 15
WANG ET AL., NONGYAO, vol. 44, no. 1, 2005, pages 13 - 15
WANG, L ET AL., CLINICAL PHARMACOLOGY AND THERAPEUTICS, vol. 56, 1994, pages 659 - 67
WU, P.W. ET AL., JACS, vol. 128, no. 45, 2006, pages 14426 - 27

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9487515B2 (en) 2006-11-22 2016-11-08 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9663517B2 (en) 2009-04-03 2017-05-30 Plexxikon Inc. Compositions and uses thereof
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2012037060A1 (en) * 2010-09-13 2012-03-22 Concert Pharmaceuticals Inc. Substituted azaindoles
EP2672967A1 (en) * 2011-02-07 2013-12-18 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US11337976B2 (en) 2011-02-07 2022-05-24 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2672967A4 (en) * 2011-02-07 2014-08-20 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
JP2016138119A (en) * 2011-02-07 2016-08-04 プレキシコン インコーポレーテッドPlexxikon Inc. Compound and method for kinase modulation, and indications therefor
JP2015518051A (en) * 2012-05-31 2015-06-25 プレキシコン インコーポレーテッドPlexxikon Inc. Synthesis of pyrrolo [2,3-b] pyridine
US9150570B2 (en) * 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
US9695169B2 (en) 2012-05-31 2017-07-04 Plexxikon Inc. Synthesis of heterocyclic compounds
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Similar Documents

Publication Publication Date Title
WO2011060216A1 (en) Substituted azaindoles
AU2009314568B2 (en) Substituted dioxopiperidinyl phthalimide derivaties
JP5852565B2 (en) Deuterated isoindoline-1,3-dione derivatives as PDE4 and TNF-α inhibitors
AU2013296627B2 (en) Deuterated ibrutinib
US20130150408A1 (en) Substituted dioxopiperidinyl phthalimide derivatives
US8575221B2 (en) Derivatives of dimethylcurcumin
AU2018271227A1 (en) Deuterated derivatives of ruxolitinib
EP2970209B1 (en) Deuterated palbociclib with improved metabolic stability
US20120208837A1 (en) Substituted azaindoles
US20110288126A1 (en) Substituted dioxopiperidinyl phthalimide derivatives
US20110082147A1 (en) Substituted imidazotriazines
WO2018106916A1 (en) Deuterated quinoxaline compounds
CA2897814A1 (en) Deuterated momelotinib
WO2014110322A2 (en) Substituted dioxopiperidinyl phthalimide derivatives
AU2011223895A1 (en) Fluorouracil derivatives
CN110229159B (en) Deuterated derivative of ruxotinib
AU2012223281A1 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
US20150366853A1 (en) Derivatives of Pyrazole-Substituted Amino-Heteroaryl Compounds
WO2014159511A1 (en) Deuterated pacritinib
WO2012129381A1 (en) Deuterated preladenant
WO2010068480A1 (en) Deuterated derivatives of dimeboline
WO2013192512A1 (en) Derivatives of pyrazole-substituted amino-heteroaryl compounds
WO2014081816A1 (en) Fluoro-derivatives of pyrazole-substituted amino-heteroaryl compounds
EP2678337A1 (en) 2-amino-naphthyridine derivatives
CA2824626A1 (en) 2-amino-naphthyridine derivatives

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10779645

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10779645

Country of ref document: EP

Kind code of ref document: A1