AU2012255711A1 - Deuterated derivatives of ivacaftor - Google Patents

Deuterated derivatives of ivacaftor

Info

Publication number
AU2012255711A1
AU2012255711A1 AU2012255711A AU2012255711A AU2012255711A1 AU 2012255711 A1 AU2012255711 A1 AU 2012255711A1 AU 2012255711 A AU2012255711 A AU 2012255711A AU 2012255711 A AU2012255711 A AU 2012255711A AU 2012255711 A1 AU2012255711 A1 AU 2012255711A1
Authority
AU
Australia
Prior art keywords
compound
deuterium
acid
compounds
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2012255711A
Other versions
AU2012255711B2 (en
Inventor
Adam J. Morgan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vertex Pharmaceuticals Europe Ltd
Original Assignee
Vertex Pharmaceuticals Europe Ltd
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 Vertex Pharmaceuticals Europe Ltd filed Critical Vertex Pharmaceuticals Europe Ltd
Priority claimed from PCT/US2012/038297 external-priority patent/WO2012158885A1/en
Publication of AU2012255711A1 publication Critical patent/AU2012255711A1/en
Application granted granted Critical
Publication of AU2012255711B2 publication Critical patent/AU2012255711B2/en
Priority to AU2017208313A priority Critical patent/AU2017208313A1/en
Assigned to VERTEX PHARMACEUTICALS (EUROPE) LIMITED reassignment VERTEX PHARMACEUTICALS (EUROPE) LIMITED Request for Assignment Assignors: CONCERT PHARMACEUTICALS INC.
Priority to AU2019222862A priority patent/AU2019222862B2/en
Priority to AU2021200970A priority patent/AU2021200970B2/en
Priority to AU2021203786A priority patent/AU2021203786B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Description

DEUTERATED DERIVATIVES OF IVACAFTOR
Cross-Reference to Related Application
[1] This application claims the benefit of priority under 35 USC § 119 to United States Provisional Patent Application Serial No. 61/487,497, filed May 18, 2011, the contents of which are incorporated herein by reference in their entirety.
Background of the Invention
[2] Many current medicines suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties that prevent their wider use or limit their use in certain indications. Poor ADME properties are also a major reason for the failure of drug candidates in clinical trials. While formulation technologies and prodrug strategies can be employed in some cases to improve certain ADME properties, these approaches often fail to address the underlying ADME problems that exist for many drugs and drug candidates. One such problem is rapid metabolism that causes a number of drugs, which otherwise would be highly effective in treating a disease, to be cleared too rapidly from the body. A possible solution to rapid drug clearance is frequent or high dosing to attain a sufficiently high plasma level of drug. This, however, introduces a number of potential treatment problems such as poor patient compliance with the dosing regimen, side effects that become more acute with higher doses, and increased cost of treatment. A rapidly metabolized drug may also expose patients to undesirable toxic or reactive metabolites.
[3] Another ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites. As a result, some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent. In certain cases, modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
[4] 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, the CYP2D6 inhibitor quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism of dextromethorphan in a treatment of pseudobulbar affect. Quinidine, however, has unwanted side effects that greatly limit its use in potential combination therapy (see Wang, L et al., Clinical Pharmacology and Therapeutics, 1994, 56(6 Pt 1): 659-67; and FDA label for quinidine at www.accessdata.fda.gov).
[5] 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.
[6] A potentially attractive strategy for improving a drug's metabolic properties is deuterium modification. In this approach, one attempts to slow the CYP-mediated metabolism of a drug or to reduce the formation of undesirable metabolites by replacing one or more hydrogen atoms with deuterium atoms. Deuterium is a safe, stable, nonradioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability. 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.
[7] 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).
[8] The effects of deuterium modification on a drug's metabolic properties are not predictable even when deuterium atoms are incorporated at known sites of metabolism. Only by actually preparing and testing a deuterated drug can one determine if and how the rate of metabolism will differ from that of its non-deuterated counterpart. See, for example, Fukuto et al. (J. Med. Chem., 1991, 34, 2871-76). Many drugs have multiple sites where metabolism is possible. The site(s) where deuterium substitution is required and the extent of deuteration necessary to see an effect on metabolism, if any, will be different for each drug.
[9] This invention relates to novel derivatives of ivacaftor, 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 CFTR (cystic fibrosis transmembrane conductance regulator) potentiator.
[10] Ivacaftor, also known as VX-770 and by the chemical name, N-(2,4-di-tert-butyl- 5-hydroxyphenyl)-4-oxo-l,4-dihydroquinoline-3-carboxamide, acts as a CFTR potentiator. Results from phase III trials of VX-770 in patients with cystic fibrosis carrying at least one copy of the G551D-CFTR mutation demonstrated marked levels of improvement in lung function and other key indicators of the disease including sweat chloride levels, likelihood of pulmonary exacerbations and body weight. VX-770 is also currently in phase II clinical trials in combination with VX-809 (a CFTR corrector) for the oral treatment of cystic fibrosis patients who carry the more common AF508-CFTR mutation. VX-770 was granted fast track designation and orphan drug designation by the FDA in 2006 and 2007, respectively.
[11] Despite the beneficial activities of VX-770, there is a continuing need for new compounds to treat the aforementioned diseases and conditions.
Brief Description of the Drawings
[12] Figure 1 depicts the percentage of compound remaining over time for
Compound 110 of the invention and for ivacaftor in human cytochrome P450-specific SUPERSOMES™. Definitions
[13] The term "treat" means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
[14] "Disease" means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
[15] 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 VX-770 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.
[16] In the compounds of this invention any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Also unless otherwise stated, when a position is designated specifically as "D" or "deuterium", the position is understood to have deuterium at an abundance that is at least 3000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 45% incorporation of deuterium).
[17] The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
[18] 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). [19] 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.
[20] 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.
[21] 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.
[22] The term "pharmaceutically acceptable," as used herein, refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention. A "pharmaceutically acceptable counterion" is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
[23] 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.
[24] The term "stable compounds," as used herein, refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
[25] "D" and "d" both refer to deuterium. "Stereoisomer" refers to both enantiomers and diastereomers. "Tert" and "t-" each refer to tertiary. "US" refers to the United States of America.
[26] "Substituted with deuterium" refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms. [27] 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
[28] The present invention provides a compound of Formula I:
Formula I
or a pharmaceutically acceptable salt thereof, wherein each of X 11, X 2, 3 4 5 0
X\ X X X°, and X' is independently hydro gen or deuterium:
each of Y1, Y2, Y3, Y4, Y5, and Y6 is independently CH3 or CD3;
provided that if each of Y1, Y2, Y3, Y4, Y5, and Y6 is CH3, then at least one of X1, X2, X3, X4, X5, X6, and X7 is deuterium.
[29] In one embodiment, X1, X2, X3, and X4 are the same. In one aspect of this embodiment, X6 and X7 are the same. In one aspect of this embodiment, Y1, Y2, and Y3 are the same. In one aspect of this embodiment, Y4, Y5, and Y6 are the same. In one example of this aspect, Y1, Y2, and Y3 are the same. In a more particular example, X6 and X7 are the same.
[30] In one embodiment, each of Y1, Y2, and Y3 is the same. In one aspect of this embodiment, each of Y4, Y5, and Y6 is the same. In one example of this aspect, X6 and X7 are the same.
[31] In one embodiment, Y1, Y2, and Y3 are CD3. In one aspect of this embodiment, Y4, Y5, and Y6 are CH3. In another embodiment, Y4, Y5, and Y6 are CD3. In one aspect 1 2 3 1 2 3 4 of this embodiment, Y , Y , and Y are C¾. In yet another embodiment, Y , Y , Y , Y , Y5, and Y6 are CD3. In yet another embodiment, Y1, Y2, Y3, Y4, Y5, and Y6 are CH3.
[32] In one embodiment, each of Y4, Y5, and Y6 is the same. In one aspect of this embodiment, X6 and X7 are the same.
[33] In one example of any of the foregoing embodiments, aspects or examples, X5 is hydrogen. In another example, X5 is deuterium.
[34] In one embodiment, the compound of Formula I is any one of the compounds of table 1,
Table 1
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
[35] In one embodiment, the compound of Formula I is any one of the compounds of table 2,
Table 2
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
[36] In one embodiment, the compound of Formula I is any one of the compounds of table 3,
Table 3
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
[37] In another set of embodiments, any atom not designated as deuterium in any of the embodiments, examples or aspects set forth above is present at its natural isotopic abundance.
[38] The synthesis of compounds of Formula I may be readily achieved by synthetic chemists of ordinary skill by reference to the Exemplary Synthesis and Examples disclosed herein. Relevant procedures analogous to those of use for the preparation of compounds of Formula I and intermediates thereof are disclosed, for instance in WO 2007075946, WO 201 1019413, WO 2010019239, WO 2007134279, WO 2007079139 and WO 2006002421, the teachings of which are incorporated herein by reference.
[39] Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure. Exemplary Synthesis
[40] A convenient method for synthesizing compounds of Formula I is depicted in Scheme 1.
Scheme 1:
(A) (B)
Formula I
[41] Compounds of Formula I may be prepared as shown in Scheme 1 via the coupling of A and B employing HATU (N,N,N',N'-tetramethyl-0-(7-azabenzotriazol-l- yl)uronium hexafluorophosphate) in the presence of DIEA (N,N'- diisopropylethylamine).
[42] Deuterated intermediates of type A (scheme 1) may be prepared as outlined in scheme 2, analogously to Singh, A.; Van Goor, F.; Worley, F. J. Ill; Knapp, T.
Compounds Useful in CFTR Assays and Methods Therewith WO 2007075946 Al July 5, 2007, the entire teachings of which are incorporated herein by reference.
(3) (2) [43] As shown in Scheme 2, heating a mixture of an aniline 1 in the presence of a malonate derivative 2 affords the appropriately deuterated
((phenylamino)methylene)malonate, subsequent exposure of which to polyphosphoric acid in the presence of POCI3 followed by ester hydrolysis provides carboxylic acid A. In Scheme 2, X is hydrogen or deuterium. In one embodiment, X, X1, X2, X3 and X4 are the same.
[44] Exemplary compounds for use in Scheme 2 include the embodiment of compound (1) where X, X1, X2, X3 and X4 are each deuterium, commercially available from Aldrich; and the embodiment of compound (2) where X5 is deuterium, prepared analogously to the procedure described in Scheme 2b (Parham, W. E.; Reed, L. J. Org. Syn. , 1948, 28, 60, the teachings of which are incorporated herein by reference) for the case where X5 is hydrogen by employing the embodiment of 3 where X5 is deuterium (available from CDN Isotopes). As shown in Scheme 2b, the appropriately deuterated (ethoxymethylene)malonate of type 2 may be prepared by reaction of diethylmalonate with the appropriately deuterated triethylorthoformate of type 3 in the presence of acetic anhydride and facilitated by ZnC¾.
[45] Deuterated intermediates of type B (Scheme 1) may be prepared as outlined in scheme 3, analogously to Singh, A. et al., supra.
Scheme 3
[46] As shown in Scheme 3, protection of ditertbutylphenols of type 4 with methyl chloro formate followed by exposure to nitric acid results in the formation of a nitro- methylcarbonate intermediate. Subsequent carbonate hydrolysis followed by palladium catalyzed reduction of the nitro group ultimately affords aminophenols of type B. In Scheme 3, X' is hydrogen or deuterium. In one embodiment, X', X6 and X7 are the same. [47] Deuterated intermediates of type 4 (Scheme 3) may be prepared as outlined in Schemes 4a-4d, analogously to Sun, Y.; Tang, N. Huaxue Shiji 2004, 26, 266-268, the entire teachings of which are incorporated herein by reference.
Scheme 4a:
Scheme 4b:
Scheme 4c:
Scheme 4d:
[48] As shown in Schemes 4a-4d, ditertbutylphenols of type 4 may be prepared via Friedel-Crafts alkylation of the appropriately deuterated phenol (phenol, 4-ieri-butyl phenol or 2-tert butylphenol) with d9-tert butylchloride. The embodiments of compound (4) that may be obtained as shown in scheme 4 are exemplary compounds for use in Scheme 3. In the embodiments 4a, 4b, 4c, and 4d in Scheme 4, any atom not designated as deuterium is present at its natural isotopic abundance. In Scheme 4, both Compound 5 and compound 6 are commercially available (CDN Isotopes). [49] 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.
[50] Additional methods of synthesizing compounds of Formula I and their synthetic precursors, including those within routes not explicitly shown in schemes herein, are within the means of chemists of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the applicable compounds are known in the art and include, for example, those described in Larock R, Comprehensive Organic Transformations, VCH Publishers
rd
(1989); Greene, TW et al., Protective Groups in Organic Synthesis, 3 Ed., John Wiley and Sons (1999); Fieser, L et al., Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette, L, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof.
[51] Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds.
Compositions
[52] The invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier. The carrier(s) are "acceptable" in the sense of being compatible with 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.
[53] 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.
[54] 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.
[55] 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.
[56] 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).
[57] Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
[58] In certain embodiments, the compound is administered orally. Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc. Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
[59] In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
[60] Compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
[61] Compositions suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit- dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
[62] Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long- chain alcohol diluent or dispersant.
[63] The pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
[64] The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
[65] Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For topical application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol, and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
[66] Application of the subject therapeutics may be local, so as to be administered at the site of interest. Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
[67] 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 having the same mechanism of action as VX-770.
[68] Preferably, the second therapeutic agent is an agent useful in the treatment of a variety of conditions, including cystic fibrosis, Hereditary emphysema, Hereditary hemochromatosis, Coagulation-Fibrinolysis deficiencies, such as Protein C deficiency, Type 1 hereditary angioedema, Lipid processing deficiencies, such as Familial hypercholesterolemia, Type 1 chylomicronemia, Abetalipoproteinemia, Lysosomal storage diseases, such as I-cell disease/Pseudo-Hurler, Mucopolysaccharidoses, Sandhof/T ay-Sachs, Crigler-Najjar type II, Polyendocrinopathy/Hyperinsulemia, Diabetes mellitus, Laron dwarfism, Myleoperoxidase deficiency, Primary
hypoparathyroidism, Melanoma, Glycanosis CDG type 1, Hereditary emphysema, Congenital hyperthyroidism, Osteogenesis imperfecta, Hereditary hypofibrinogenemia, ACT deficiency, Diabetes insipidus (DI), Neurophyseal DI, Neprogenic DI, Charcot- Marie Tooth syndrome, Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Progressive supranuclear palsy, Pick's disease, several polyglutamine neurological disorders asuch as Huntington, Spinocerebullar ataxia type I, Spinal and bulbar muscular atrophy,
Dentatorubal pallidoluysian, and Myotonic dystrophy, as well as Spongiform
encephalopathies, such as Hereditary Creutzfeldt- Jakob disease, Fabry disease,
Straussler- Scheinker syndrome, COPD, dry-eye disease, and Sjogren's disease.
[69] In one embodiment, the second therapeutic agent is an agent useful in the treatment of cystic fibrosis.
[70] In one embodiment, the second therapeutic agent is VX-809. [71] In another embodiment, the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another. The term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
[72] In the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term "effective amount" refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
[73] The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., Cancer Chemother. Rep, 1966, 50: 219. Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
[74] In one embodiment, an effective amount of a compound of this invention can range from about 0.02 to 2500 mg per treatment. In more specific embodiments the range is from about 0.2 to 1250 mg or from about 0.4 to 500 mg or most specifically from 2 to 250 mg per treatment. Treatment typically is administered one to two times daily.
[75] Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
[76] 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.
[77] It is expected that some of the second therapeutic agents referenced above will act synergistically with the compounds of this invention. When this occurs, it will allow the effective dosage of the second therapeutic agent and/or the compound of this invention to be reduced from that required in a monotherapy. This has the advantage of minimizing toxic side effects of either the second therapeutic agent of a compound of this invention, synergistic improvements in efficacy, improved ease of administration or use and/or reduced overall expense of compound preparation or formulation.
Methods of Treatment
[78] In another embodiment, the invention provides a method of potentiating the activity of CFTR in an infected cell, comprising contacting such a cell with a compound of Formula I herein, or a pharmaceutically acceptable salt thereof.
[79] According to another embodiment, the invention provides a method of treating a disease that is beneficially treated by VX-770 in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound or a composition of this invention. In one embodiment the subject is a patient in need of such treatment. Such diseases include cystic fibrosis, Hereditary emphysema,
Hereditary hemochromatosis, Coagulation-Fibrinolysis deficiencies, such as Protein C deficiency, Type 1 hereditary angioedema, Lipid processing deficiencies, such as Familial hypercholesterolemia, Type 1 chylomicronemia, Abetalipoproteinemia, Lysosomal storage diseases, such as I-cell disease/Pseudo-Hurler,
Mucopolysaccharidoses, Sandhof/T ay-Sachs, Crigler-Najjar type II,
Polyendocrinopathy/Hyperinsulemia, Diabetes mellitus, Laron dwarfism,
Myleoperoxidase deficiency, Primary hypoparathyroidism, Melanoma, Glycanosis CDG type 1, Hereditary emphysema, Congenital hyperthyroidism, Osteogenesis imperfecta, Hereditary hypofibrinogenemia, ACT deficiency, Diabetes insipidus (DI), Neurophyseal DI, Neprogenic DI, Charcot-Marie Tooth syndrome, Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease,
Amyotrophic lateral sclerosis, Progressive supranuclear palsy, Pick's disease, several polyglutamine neurological disorders such as Huntington, Spinocerebullar ataxia type I, Spinal and bulbar muscular atrophy, Dentatorubal pallidoluysian, and Myotonic dystrophy, as well as Spongiform encephalopathies, such as Hereditary Creutzfeldt- Jakob disease, Fabry disease, Straussler- Scheinker syndrome, COPD, dry-eye disease, and Sjogren's disease.
[80] In one embodiment, the method of this invention is used to treat cystic fibrosis in a subject such as a patient in need thereof.
[81] In another embodiment, any of the above methods of treatment comprises the further step of co-administering to the subject in need thereof one or more second therapeutic agents. The choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for co-administration with VX-770. 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.
[82] In particular, the combination therapies of this invention include coadministering a compound of Formula I or a pharmaceutically acceptable salt thereof and a second therapeutic agent to a subject in need thereof for treatment of the following conditions (with the particular second therapeutic agent indicated in parentheses following the indication):.
[83] The term "co-administered" as used herein means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods. The
administration of a composition of this invention, comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
[84] 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.
[85] 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.
[86] In yet another aspect, the invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment or prevention in a subject of a disease, disorder or symptom set forth above. Another aspect of the invention is a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention in a subject of a disease, disorder or symptom thereof delineated herein.
Examples
[87] Example 1. Synthesis of N-(2.4-Di-(tert-butyl-dQ)-3.6-dz-5-hvdroxyphenyl)-4- oxo-1.4-dihvdroquinoline-3-carboxamide (Compound 110).
[88] Compound 110 was prepared as outlined in Scheme 5 below.
Scheme 5. Preparation of Compound 110
Compound 110
[89] Step 1. 2,4-Di-(½rt-butyl- do )-3,5,6- d phenol (4a). Intermediate 4a was prepared according to the procedure described for the synthesis for 2,4-di-teri-butyl-3,5- d2-phenol employing ieri-butyl chloride-dg in place of ieri-butylchloride (Kurahashi, T.; Hada, M.; Fujii, H. /. Am. Chem. Soc. 2009, 131, 12394-12405): To a solution of phenol-d6 (459 mg, 4.59 mmol, 99 atom %D, Sigma Aldrich) and ieri-butyl chloride-dg (2.50 mL, 23.0 mmol, 98 atom%D, Cambridge Isotope Laboratories, Inc.) in 1,2- dichloroethane (10.0 mL) was added ReBr(CO)s (19.0 mg, 0.0459 mmol). The reaction mixture was stirred at 85 °C for 15 hours at which time additional ieri-butyl chloride-d9 (2.50 mL, 23.0 mmol, 98 atom%D, Cambridge Isotope Laboratories, Inc.) and
ReBr(CO)5 (19.0 mg, 0.0459 mmol) was added. Stirring was continued at 85 °C for 2 hours, the mixture was cooled to room temperature, concentrated in vacuo and purified by column chromatography (Si02, 30% CH2Cl2/heptanes) to afford 4a (0.789 g, 76% yield) as a light yellow oil. MS (ESI) 228.1 [(M+H)+].
[90] Step 2. 2.4-Di-(tert-butyl-dQ)-3.5.6-d^-phenyl methyl carbonate (20). To a solution of 4a (2.72 g, 12.0 mmol), triethylamine (3.33 mL, 23.9 mmol) and N,N- dimethylaminopyridine (73.0 mg, 0.598 mmol) in CH2C12 (30.0 mL) at 0 °C was added methyl chloroformate (1.38 mL, 17.9 mmol). The reaction mixture was stirred at room temperature for 15 hours then was diluted with 10% ethyl acetate/heptanes and filtered through a silica plug. The silica plug was then rinsed with additional 10% ethyl acetate/heptanes. The filtrate was combined, and concentrated in vacuo to provide 20 (2.40 g, 70% yield) as a light yellow oil which was carried forward without purification.
[91] Step 3. 2.4-Di-(tert-butyl-dQ)-3.6-d9-5-nitrophenol (21). To a solution of 20 (2.40 g, 8.41 mmol) in sulfuric acid (1.00 mL) at 0 °C was added a 1 : 1 mixture of sulfuric acid and nitric acid (2.00 mL) dropwise. The reaction mixture was then stirred at room temperature for 2 hours then slowly added to ice water with vigorous stirring. The resulting slurry was extracted with ethyl acetate (3 x lOOmL) and the combined organic layers were dried ( a2S04), filtered and concentrated to afford an amber oil containing a mixture of regioisomers. This crude oil was then taken up in MeOH (50 mL) and KOH (1.54 g, 27.5 mmol) was added. The reaction mixture was stirred at room temperature for 2 hours then was acidified to pH = 2 with concentrated HC1. The resulting solution was extracted with diethyl ether (3 x 100 mL), dried (MgS04), filtered and concentrated. The residue was then purified via column chromatography (S1O2, 0-5% ethyl acetate/heptanes) to afford 21 (526 mg, 23%) as a light yellow solid. MS (ESI) 270.3 [(M-H)-].
[92] Step 4. 5-Amino-2.4-di-(tert-butyl-dQ)-3.6-d9-phenol (22). A solution of 21 (526 mg, 1.94 mmol) and ammonium formate (489 mg, 7.75 mmol) in ethanol (25.0 mL) was heated to the point of reflux. At this time, 10% Pd/C (250 mg, 50% wet) was added in small portions and the reaction mixture was stirred at reflux for 2 hours. The mixture was then cooled to room temperature, diluted with THF, filtered through Celite® and concentrated in vacuo to afford 22 (473 mg, 100%) as a tan solid. MS (ESI) 242.4
[(M+H)+].
[93] Step 5. N-(2.4-Di-(tert-butyl-do )-3.6-dz-5-hvdroxyphenyl)-4-oxo- 1.4- dihydroquinoline-3-carboxamide (Compound 110). To a solution of 22 (250 mg, 1.04 mmol), 4-oxo- l,4-dihydroquinoline-3-carboxylic acid (23, purchased from Matrix Scientific, 98.0 mg, 0.518 mmol) and N,N-diisopropylethylamine (181 xL, 1.04 mmol) in DMF (5.00 mL) was added HATU (197 mg, 0.518 mmol). The reaction mixture was stirred at room temperature for 3 hours then was diluted with saturated aHC03 and extracted with ethyl acetate (3 x 50 mL). The combined organic extracts were washed with water (3 x 20 mL), dried (Na2S04), filtered and concentrated in vacuo. The resulting residue was purified via column chromatography (S1O2, 0-70% ethyl acetate/heptanes) to afford Compound 110 (77.0 mg, 36% Yield) as a white solid. JH NMR (d6-DMSO, 400 MHz): δ 12.87 (br s, 1H), 1 1.80 (s, 1H), 9.18 (s, 1H), 8.86 (s, 1H), 8.32 (d, / = 8.2 Hz, 1H), 7.81 (t, J = 7.9 Hz, 1H), 7.76 (t, 7 = 8.2 Hz, 1H), 7.51 (t, / = 7.4 Hz, 1H), 7.10 (s, 0.2H)*; MS (ESI) 413.5 [(M+H)+]. *The ¾ NMR signal at 7.10 ppm indicates approximately 80% deuterium incorporation at one of the two deuterated aryl positions. The absence of signals at 7.20 ppm and 1.37 ppm indicate high levels of incorporation (>95%) at the remaining deuterated positions.
[94] Example 2. Evaluation of Metabolic Stability - Human CYP3A4
Supersomes™.
[95] SUPERSOMES™ Assay. 7.5 mM stock solutions of test compounds,
Compound 110 and ivacaftor, were prepared in DMSO. The 7.5 mM stock solutions were diluted to 50 mM in acetonitrile (ACN). Human CYP3A4 supersomes™ (1000 pmol/mL, purchased from BD Gentest™ Products and Services) were diluted to 62.5 pmol/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCi2. The diluted supersomes were added to wells of a 96-well polypropylene plate in triplicate. A 10 mL aliquot of the 50 mM test compound was added to the supersomes and the mixture was pre-warmed for 10 minutes. Reactions were initiated by addition of pre- warmed NADPH solution. The final reaction volume was 0.5 mL and contained 50 pmol/mL CYP3A4 supersomes™, 1.0 mM test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl2. The reaction mixtures were incubated at 37 °C, and 50 mL aliquots were removed at 0, 5, 10, 20, and 30 minutes and added to 96-well plates which contained 50 mL of ice-cold ACN with internal standard to stop the reactions. The plates were stored at 4 °C for 20 minutes after which 100 mL of water was added to the wells of the plate before centrifugation to pellet precipitated proteins. Supernatants were 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.
[96] Data analysis: The in vitro half-lives (ti/2 values) for test compounds were calculated from the slopes of the linear regression of LN(% parent remaining) vs incubation time relationship: [97] in vitro t ½ = 0.693/k, where k = -[slope of linear regression of % parent remaining (In) vs incubation time]
[98] Figure 1 shows a plot of the percentage of parent compound remaining over time for Compound 110 and for ivacaftor in human cytochrome P450-specific
SUPERSOMES™.The ti/2 values, and the percentage increase ( _ ) in average ti/2, are shown in Table 4 below.
Table 4. Results of fa Vitro Human Cytochrome P450-Specific SUPERSOMES™
[99] Table 4 shows that Compound 110 has a 55% longer half life in the assay than ivacaftor.
[100] 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 (14)

We claim:
1. A compound of Formula I:
Formula I
or a pharmaceutically acceptable salt thereof, wherein
1 2 3 4 5 0
each of X1, X , X\ X X X°, and X' is independently hydro gen or deuterium:
each of Y1, Y2, Y3, Y4, Y5, and Y6 is independently CH3 or CD3;
provided that if each of Y1, Y2, Y3, Y4, Y5, and Y6 is CH3, then at least one of X1, X2, X3, X4, X5, X6, and X7 is deuterium.
2. The compound of claim 1, wherein X1, X2, X3, and X4 are the same.
3. The compound of claim 2, wherein X6 and X7 are the same.
4. The compound of claim 2 or 3, wherein Y1, Y2, and Y3 are the same.
5. The compound of any one of claims 2-4, wherein Y4, Y5, and Y6 are the same.
6. The compound of any one of claims 2-5, wherein X6 and X7 are the same.
7. The compound any one of the preceding claims, wherein X5 is deuterium.
8. The compound of any one of the preceding claims wherein Y1, Y2, and Y3 are CD3.
9. The compound of any one of the preceding claims wherein Y4, Y5, and Y6 are CD,.
The compound of any one of the preceding claims, wherein any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
The compound of claim 1, wherein the compound of Formula I is any one of the compounds of the table below,
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
12. The compound of claim 1, wherein the compound of Formula I is any one of the compounds of the table below,
or a pharmaceutical y acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
13. A pharmaceutical composition comprising a compound of any one of the preceding claims or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
14. A method of treating cystic fibrosis in a subject comprising administering to the subject the pharmaceutical composition of claim 13 or a compound of any one of claims 1-12.
AU2012255711A 2011-05-18 2012-05-17 Deuterated derivatives of ivacaftor Active AU2012255711B2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2017208313A AU2017208313A1 (en) 2011-05-18 2017-07-27 Deuterated derivatives of ivacftor
AU2019222862A AU2019222862B2 (en) 2011-05-18 2019-08-28 Deuterated derivatives of ivacaftor
AU2021200970A AU2021200970B2 (en) 2011-05-18 2021-02-15 Deuterated derivatives of ivacaftor
AU2021203786A AU2021203786B2 (en) 2011-05-18 2021-06-08 Deuterated derivatives of ivacaftor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161487497P 2011-05-18 2011-05-18
US61/487,497 2011-05-18
PCT/US2012/038297 WO2012158885A1 (en) 2011-05-18 2012-05-17 Deuterated derivatives of ivacaftor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2017208313A Division AU2017208313A1 (en) 2011-05-18 2017-07-27 Deuterated derivatives of ivacftor

Publications (2)

Publication Number Publication Date
AU2012255711A1 true AU2012255711A1 (en) 2013-12-12
AU2012255711B2 AU2012255711B2 (en) 2017-05-04

Family

ID=46201810

Family Applications (5)

Application Number Title Priority Date Filing Date
AU2012255711A Active AU2012255711B2 (en) 2011-05-18 2012-05-17 Deuterated derivatives of ivacaftor
AU2017208313A Abandoned AU2017208313A1 (en) 2011-05-18 2017-07-27 Deuterated derivatives of ivacftor
AU2019222862A Active AU2019222862B2 (en) 2011-05-18 2019-08-28 Deuterated derivatives of ivacaftor
AU2021200970A Active AU2021200970B2 (en) 2011-05-18 2021-02-15 Deuterated derivatives of ivacaftor
AU2021203786A Active AU2021203786B2 (en) 2011-05-18 2021-06-08 Deuterated derivatives of ivacaftor

Family Applications After (4)

Application Number Title Priority Date Filing Date
AU2017208313A Abandoned AU2017208313A1 (en) 2011-05-18 2017-07-27 Deuterated derivatives of ivacftor
AU2019222862A Active AU2019222862B2 (en) 2011-05-18 2019-08-28 Deuterated derivatives of ivacaftor
AU2021200970A Active AU2021200970B2 (en) 2011-05-18 2021-02-15 Deuterated derivatives of ivacaftor
AU2021203786A Active AU2021203786B2 (en) 2011-05-18 2021-06-08 Deuterated derivatives of ivacaftor

Country Status (20)

Country Link
EP (2) EP3235812B1 (en)
JP (2) JP6063455B2 (en)
AU (5) AU2012255711B2 (en)
BR (1) BR112013029240B1 (en)
CA (1) CA2834574C (en)
CY (2) CY1119090T1 (en)
DK (2) DK3235812T3 (en)
EA (1) EA028378B1 (en)
ES (2) ES2758028T3 (en)
HK (1) HK1244183A1 (en)
HR (2) HRP20170754T1 (en)
HU (1) HUE032771T2 (en)
LT (1) LT2709986T (en)
ME (2) ME03652B (en)
MX (1) MX349159B (en)
PL (2) PL3235812T3 (en)
PT (2) PT3235812T (en)
RS (2) RS56096B1 (en)
SI (2) SI3235812T1 (en)
WO (1) WO2012158885A1 (en)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0620960A2 (en) 2005-12-28 2011-11-29 Vertex Pharmaceuticals Incorporated n- [2,4-bis (1,1-dimethylethyl) -5-hydroxyphenyl] -1,4-didro-4-oxyquinoline-3-carboxamide solid forms
CA2834574C (en) * 2011-05-18 2019-09-17 Concert Pharmaceuticals Inc. Deuterated derivatives of ivacaftor
HUE047354T2 (en) 2011-05-18 2020-04-28 Vertex Pharmaceuticals Europe Ltd Deuterated derivatives of ivacaftor
WO2013151758A2 (en) 2012-04-06 2013-10-10 The Uab Research Foundation Methods for increasing cftr activity
JP6146990B2 (en) * 2012-11-16 2017-06-14 コンサート ファーマシューティカルズ インコーポレイテッド Deuterated CFTR enhancer
WO2014078842A1 (en) * 2012-11-19 2014-05-22 Concert Pharmaceuticals, Inc. Deuterated cftr potentiators
JP6896619B2 (en) 2014-10-06 2021-06-30 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Cystic fibrosis transmembrane conductance regulator modulator
RU2749213C2 (en) 2014-10-07 2021-06-07 Вертекс Фармасьютикалз Инкорпорейтед Co-crystals of transmembrane conduction regulator modulators in cystic fibrosis
AU2016243171B2 (en) 2015-03-31 2020-10-08 Concert Pharmaceuticals, Inc. Deuterated VX-661
AU2016297886B2 (en) 2015-07-24 2020-12-10 Proteostasis Therapeutics, Inc. Compounds, compositions and methods of increasing CFTR activity
CA2998911C (en) * 2015-09-21 2023-05-23 Vertex Pharmaceuticals (Europe) Limited Administration of deuterated cftr potentiators
AU2016327603B2 (en) * 2015-09-25 2021-04-22 Vertex Pharmaceuticals (Europe) Limited Deuterated CFTR potentiators
US10662207B2 (en) * 2016-04-07 2020-05-26 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
ES2954658T3 (en) 2016-06-21 2023-11-23 Proteostasis Therapeutics Inc Compounds, compositions and methods for increasing CFTR activity
JP2017105824A (en) * 2017-02-10 2017-06-15 コンサート ファーマシューティカルズ インコーポレイテッド Deuterated cftr enhancement material
US11708331B2 (en) * 2017-12-01 2023-07-25 Vertex Pharmaceuticals Incorporated Processes for making modulators of cystic fibrosis transmembrane conductance regulator
JP6654223B2 (en) * 2018-08-13 2020-02-26 バーテックス ファーマシューティカルズ (ヨーロッパ) リミテッド Deuterated CFTR enhancer
WO2020214921A1 (en) 2019-04-17 2020-10-22 Vertex Pharmaceuticals Incorporated Solid forms of modulators of cftr
KR20220064366A (en) 2019-08-14 2022-05-18 버텍스 파마슈티칼스 인코포레이티드 Crystalline Forms of CFTR Modulators
TW202115092A (en) 2019-08-14 2021-04-16 美商維泰克斯製藥公司 Modulators of cystic fibrosis transmembrane conductance regulator
US20220313698A1 (en) 2019-08-14 2022-10-06 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
LV15575A (en) 2019-12-20 2021-06-20 Latvijas Organiskās Sintēzes Institūts Deuterated analogues of selenophenochromenes, synthesis thereof, and methods of using same agents
CR20230120A (en) 2020-08-07 2023-09-01 Vertex Pharma Modulators of cystic fibrosis transmembrane conductance regulator
IL300413A (en) 2020-08-13 2023-04-01 Vertex Pharma Crystalline forms of cftr modulators
EP3970718A1 (en) 2020-09-18 2022-03-23 Charité - Universitätsmedizin Berlin New medical use of cystic fibrosis transmembrane conductance regulator (cftr) modulators
EP4225748A1 (en) 2020-10-07 2023-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
WO2022076620A1 (en) 2020-10-07 2022-04-14 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US20230373974A1 (en) 2020-10-07 2023-11-23 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US20230416275A1 (en) 2020-10-07 2023-12-28 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
CR20230197A (en) 2020-10-07 2023-07-06 Vertex Pharma Modulators of cystic fibrosis transmembrane conductance regulator
CA3197857A1 (en) 2020-10-07 2022-04-14 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
EP4225737A1 (en) 2020-10-07 2023-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
EP4225762A1 (en) 2020-10-07 2023-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US20230382924A1 (en) 2020-10-07 2023-11-30 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
IL301757A (en) 2020-10-07 2023-05-01 Vertex Pharma Modulators of cystic fibrosis transmembrane conductance regulator
WO2022125826A1 (en) 2020-12-10 2022-06-16 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
WO2023150236A1 (en) 2022-02-03 2023-08-10 Vertex Pharmaceuticals Incorporated Methods of preparing and crystalline forms of (6a,12a)-17-amino-12-methyl-6,15-bis(trifluoromethyl)-13,19-dioxa-3,4,18-triazatricyclo[ 12.3.1.12,5]nonadeca-1(18),2,4,14,16-pentaen-6-ol
WO2023150237A1 (en) 2022-02-03 2023-08-10 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
WO2023154291A1 (en) 2022-02-08 2023-08-17 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
WO2023196429A1 (en) 2022-04-06 2023-10-12 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
WO2023224931A1 (en) 2022-05-16 2023-11-23 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
WO2023224924A1 (en) 2022-05-16 2023-11-23 Vertex Pharmaceuticals Incorporated Solid forms of a macrocyclic compounds as cftr modulators and their preparation

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20040068613A (en) * 1994-03-25 2004-07-31 이소테크니카 인코포레이티드 Deuterated compound and composition for treating hypertension
GB9925962D0 (en) 1999-11-02 1999-12-29 Novartis Ag Organic compounds
US20100074949A1 (en) 2008-08-13 2010-03-25 William Rowe Pharmaceutical composition and administration thereof
NZ528689A (en) 2001-05-03 2005-03-24 F Pharmaceutical dosage form of amorphous nelfinavir mesylate
CA2446904A1 (en) 2001-05-24 2003-04-03 Alexza Molecular Delivery Corporation Delivery of drug esters through an inhalation route
PT2489659T (en) * 2004-06-24 2018-03-07 Vertex Pharma Modulators of atp-binding cassette transporters
CA2581169A1 (en) 2004-09-29 2006-04-13 Cordis Corporation Pharmaceutical dosage forms of stable amorphous rapamycin like compounds
CA2635214A1 (en) 2005-12-27 2007-07-05 Vertex Pharmaceuticals Incorporated Compounds useful in cftr assays and methods therewith
BRPI0620960A2 (en) 2005-12-28 2011-11-29 Vertex Pharmaceuticals Incorporated n- [2,4-bis (1,1-dimethylethyl) -5-hydroxyphenyl] -1,4-didro-4-oxyquinoline-3-carboxamide solid forms
ATE534383T1 (en) 2006-05-12 2011-12-15 Vertex Pharma COMPOSITIONS OF N-Ä2,4-BIS(1,1-DIMETHYLETHYL)-5-HYDROXYPHENYLÜ-1,4-DIHYDRO-4-OXOCINOLIN-3-CARBOXAMIDE
EP2727915B1 (en) * 2007-09-13 2016-04-13 Concert Pharmaceuticals Inc. Synthesis of deuterated catechols and benzo[d][1,3]dioxoles and derivatives thereof
NO2328618T3 (en) 2008-08-13 2018-04-28
UA104005C2 (en) * 2008-09-03 2013-12-25 Тева Фармасьютикл Индастриз, Лтд. 2-oxo-1,2-dihydro-quinoline modulators of immune function
CA2834574C (en) * 2011-05-18 2019-09-17 Concert Pharmaceuticals Inc. Deuterated derivatives of ivacaftor

Similar Documents

Publication Publication Date Title
AU2021200970B2 (en) Deuterated derivatives of ivacaftor
US20220041557A1 (en) Deuterated cftr potentiators
AU2012255711A1 (en) Deuterated derivatives of ivacaftor
WO2014078842A1 (en) Deuterated cftr potentiators
JP6146990B2 (en) Deuterated CFTR enhancer
AU2016327603B2 (en) Deuterated CFTR potentiators
JP6654223B2 (en) Deuterated CFTR enhancer
KR102070361B1 (en) Deuterated cftr potentiators
JP2017105824A (en) Deuterated cftr enhancement material