WO2011038207A1 - Phosphorus-containing thyroid hormone receptor agonists and methods of use - Google Patents
Phosphorus-containing thyroid hormone receptor agonists and methods of use Download PDFInfo
- Publication number
- WO2011038207A1 WO2011038207A1 PCT/US2010/050169 US2010050169W WO2011038207A1 WO 2011038207 A1 WO2011038207 A1 WO 2011038207A1 US 2010050169 W US2010050169 W US 2010050169W WO 2011038207 A1 WO2011038207 A1 WO 2011038207A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- och
- compound
- group
- ocr
- atoms
- Prior art date
Links
- 0 **c1c(*)c(*)c(*c2c(*)c(*)c(*)c(*)c2*)c(*)c1* Chemical compound **c1c(*)c(*)c(*c2c(*)c(*)c(*)c(*)c2*)c(*)c1* 0.000 description 3
- GKGCQZJSVAKXST-UHFFFAOYSA-N CC(C)C(CC(O)=O)NC(c(cc(cc1)Oc(c(Br)cc(CP(C)=O)c2)c2Br)c1O)=O Chemical compound CC(C)C(CC(O)=O)NC(c(cc(cc1)Oc(c(Br)cc(CP(C)=O)c2)c2Br)c1O)=O GKGCQZJSVAKXST-UHFFFAOYSA-N 0.000 description 1
- SVXLLCKJKRYATC-UHFFFAOYSA-N CC(C)c(cc(Cc(c(C)c1)c(C)cc1OCP(O)(O)=O)cc1)c1O Chemical compound CC(C)c(cc(Cc(c(C)c1)c(C)cc1OCP(O)(O)=O)cc1)c1O SVXLLCKJKRYATC-UHFFFAOYSA-N 0.000 description 1
- QJMLAAOOHOSGIN-UHFFFAOYSA-N CCOC(CCc1cc(Cc(c(C)c2)c(C)cc2OCP(O)(O)=O)ccc1O)=O Chemical compound CCOC(CCc1cc(Cc(c(C)c2)c(C)cc2OCP(O)(O)=O)ccc1O)=O QJMLAAOOHOSGIN-UHFFFAOYSA-N 0.000 description 1
- ICAVAMJRWYYOOE-UHFFFAOYSA-N CCOC(CNC(c(cc(cc1)Oc(c(Br)cc(CP(C)(O)=O)c2)c2Br)c1O)=O)=O Chemical compound CCOC(CNC(c(cc(cc1)Oc(c(Br)cc(CP(C)(O)=O)c2)c2Br)c1O)=O)=O ICAVAMJRWYYOOE-UHFFFAOYSA-N 0.000 description 1
- CRXKDZHMXRPRDN-UHFFFAOYSA-N COC(c1ccc(CCNC(c(cc(cc2)Oc(c(Br)cc(CP(C)(O)=O)c3)c3[Br]=C)c2O)=O)cc1)=O Chemical compound COC(c1ccc(CCNC(c(cc(cc2)Oc(c(Br)cc(CP(C)(O)=O)c3)c3[Br]=C)c2O)=O)cc1)=O CRXKDZHMXRPRDN-UHFFFAOYSA-N 0.000 description 1
- CTLYEQZYOXJLDD-UHFFFAOYSA-N CP(Cc(cc1Br)cc(Br)c1Oc(cc1)cc(C(NCC(O)=O)=O)c1O)=O Chemical compound CP(Cc(cc1Br)cc(Br)c1Oc(cc1)cc(C(NCC(O)=O)=O)c1O)=O CTLYEQZYOXJLDD-UHFFFAOYSA-N 0.000 description 1
- DEQHOLODPWWCPD-SFHVURJKSA-N CP(Cc(cc1[Br]=C)cc([Br]=C)c1Oc(cc1C(N[C@@H](CCSC)C(O)=O)=O)ccc1O)(O)=O Chemical compound CP(Cc(cc1[Br]=C)cc([Br]=C)c1Oc(cc1C(N[C@@H](CCSC)C(O)=O)=O)ccc1O)(O)=O DEQHOLODPWWCPD-SFHVURJKSA-N 0.000 description 1
- MKWSQGNLZPNLKT-UHFFFAOYSA-N Cc1cc(CP(C)(O)=O)cc(C)c1Cc(cc1)cc(CC(O)=O)c1O Chemical compound Cc1cc(CP(C)(O)=O)cc(C)c1Cc(cc1)cc(CC(O)=O)c1O MKWSQGNLZPNLKT-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F9/00—Compounds containing elements of Groups 5 or 15 of the Periodic Table
- C07F9/02—Phosphorus compounds
- C07F9/28—Phosphorus compounds with one or more P—C bonds
- C07F9/30—Phosphinic acids [R2P(=O)(OH)]; Thiophosphinic acids ; [R2P(=X1)(X2H) (X1, X2 are each independently O, S or Se)]
- C07F9/306—Arylalkanephosphinic acids, e.g. Ar-(CH2)n-P(=X)(R)(XH), (X = O,S, Se; n>=1)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/66—Phosphorus compounds
- A61K31/662—Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/14—Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F9/00—Compounds containing elements of Groups 5 or 15 of the Periodic Table
- C07F9/02—Phosphorus compounds
- C07F9/28—Phosphorus compounds with one or more P—C bonds
- C07F9/38—Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)]
- C07F9/40—Esters thereof
- C07F9/4003—Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
- C07F9/4006—Esters of acyclic acids which can have further substituents on alkyl
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F9/00—Compounds containing elements of Groups 5 or 15 of the Periodic Table
- C07F9/02—Phosphorus compounds
- C07F9/28—Phosphorus compounds with one or more P—C bonds
- C07F9/38—Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)]
- C07F9/40—Esters thereof
- C07F9/4003—Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
- C07F9/4056—Esters of arylalkanephosphonic acids
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F9/00—Compounds containing elements of Groups 5 or 15 of the Periodic Table
- C07F9/02—Phosphorus compounds
- C07F9/28—Phosphorus compounds with one or more P—C bonds
- C07F9/38—Phosphonic acids [RP(=O)(OH)2]; Thiophosphonic acids ; [RP(=X1)(X2H)2(X1, X2 are each independently O, S or Se)]
- C07F9/40—Esters thereof
- C07F9/4071—Esters thereof the ester moiety containing a substituent or a structure which is considered as characteristic
- C07F9/4075—Esters with hydroxyalkyl compounds
Definitions
- the present invention is directed toward phosphorus-containing thyroid hormone receptor agonists ("THA”) and methods of their use.
- TAA thyroid hormone receptor agonists
- Thyroid hormones i.e., T4, T3, rT3, and/or T2 have profound effects on lipids and lipoprotein levels.
- THs lower plasma cholesterol levels in part by enhancing cholesterol conversion to bile acids through increased expression of the rate-limiting enzyme cholesterol 7a-hydroxylase CYP7a and by increasing hepatic uptake of LDL- cholesterol via LDL-receptors.
- THs increase free fatty acid oxidation and can as a result lower plasma and hepatic triglyceride levels.
- THs stimulate apoA-I expression and the secretion of apoA-I in HDL while reducing apoA-II.
- THs may also induce expression of SPvBl receptors which may in turn increase hepatic HDL uptake and possibly enhance reverse cholesterol transport.
- THs lower the atherogenic lipoprotein Lp(a). The effects of THs on lipids and lipoproteins are largely through effects on the liver and genes expressed in the liver controlling lipid and lipoprotein production, uptake and metabolism.
- T3 and T3 mimetics are thought to inhibit atherosclerosis by modulating the levels of certain lipoproteins known to be independent risk factors or potential risk factors of atherosclerosis, including low density lipoprotein (LDL)-cholesterol, high density lipoprotein (HDL)-cholesterol, apoA-I, which is a major apoprotein constituent of high density lipoprotein (HDL) particles and lipoprotein (a) or Lp(a).
- Lp(a) is an important risk factor, elevated in many patients with premature atherosclerosis. Lp(a) is considered highly atherogenic (de Bruin et ah, J. Clin. Endo. Metab., 76, 121-126 (1993)).
- Lp(a) is a hepatic acute phase protein that promotes the binding of LDL to cell surfaces independent of LDL receptors. Accordingly, Lp(a) is thought to provide supplementary cholesterol to certain cells, e.g., cells involved in inflammation or repair. Lp(a) is an independent risk factor for premature atherosclerosis.
- THs may also be utilized for lowering triglycerides, which occurs inconsistently with hyperthyroidism. Elevated triglycerides represents a condition that can result in increased risk for coronary heart disease, pancreatitis and/or NASH.
- THs are associated with a variety of adverse effects in animals and humans that largely arise from action of THs on non-hepatic tissues.
- Hyperthyroidism is associated with increased body temperature, general nervousness, weight loss despite increased appetite, muscle weakness and fatigue, increased bone resorption and enhanced calcification, and a variety of cardiovascular changes, including increased heart rate, increased stroke volume, increased cardiac index, cardiac hypertrophy, decreased peripheral vascular resistance, and increased pulse pressure.
- Hypothyroidism is generally associated with the opposite effects.
- TRs thyroid hormone receptors
- nuclear receptors which, along with its common partner, the retinoid X receptor, form heterodimers that act as ligand-inducible transcription factors.
- TRs have a ligand binding domain and a DNA binding domain and regulate gene expression through ligand- dependent interactions with DNA response elements (thyroid response elements, TREs).
- TRa and TRP are encoded by two distinct genes (TRa and TRP), which produce several isoforms through alternative splicing (Williams, Mol Cell Biol.
- TRa-1 is ubiquitously expressed in the rat with highest expression in skeletal muscle and brown fat.
- TR -l is also ubiquitously expressed with highest expression in the liver, brain and kidney.
- TR -2 is expressed in the anterior pituitary gland and specific regions of the hypothalamus as well as the developing brain and inner ear. In the rat and mouse liver, ⁇ ⁇ -l is the predominant isoform (80%).
- the TR isoforms found in human and rat are highly homologous with respect to their amino acid sequences which suggest that each serves a specialized function.
- THs brown adipose tissue
- WAT brown adipose tissue
- Hyperthyroidism is associated with increased food intake, an overall increase in the basal metabolic rate (BMR) and decreased body weight (ca. 15%) whereas hypothyroidism is associated with a 25-30%) increase in body weight.
- BMR basal metabolic rate
- hypothyroidism is associated with a 25-30%) increase in body weight.
- Treating hypothyroidism patients with T3 leads to a decrease in body weight for most patients but not all (17% of the patients maintain weight). Consequently, THs and TH mimetics have been of interest for decades as antiobesity drugs.
- THs are tightly regulated through feedback loops that affect production of hormones such as thyrotropin releasing factor (TRF) from the hypothalamus and thyroid stimulating hormone (TSH) from the pituitary which in turn affect the production of THs from the thyroid.
- TRF thyrotropin releasing factor
- TSH thyroid stimulating hormone
- THs also tightly regulate tissue levels of T3 through effects on enzymes (e.g. deiodinases) that govern TH metabolism.
- TSH is an anterior pituitary hormone that regulates thyroid hormone production. TSH formation and secretion is in turn regulated by the hypothalamic TRF. TSH controls the uptake of iodide by the thyroid, the subsequent release of iodinated thyronines from thyroglobulin ⁇ e.g., T3, T4) as well as possibly the intrapituitary conversion of circulating T4 to T3. Compounds that mimic T3 and T4 can negatively regulate both TSH and TRF secretion resulting in suppression of TSH levels and decreased levels of THs such as T3 and/or T4. Negative regulation of TSH is postulated based on co-transfection and knockout studies (Abel et al., J. Clin. Invest., 104, 291-300, (1999)) to arise through activation of the thyroid receptor ⁇ , possibly the isoform ⁇ - 2, which is highly expressed in the pituitary.
- THs also affect the expression of deiodinases that convert T4 to the most biologically active TH, T3.
- THs and TH mimetics increase deiodinase Dl which increases T4 conversion to T3 and rT3.
- Increased T4 metabolism results in lower T4 levels and/or in higher TSH levels.
- T4 levels induce expression of the deiodinase D2 which helps ensure that certain tissues remain euthyroid through production of adequate levels of T3. Some tissues may not be able to compensate for the low T4 levels and as a consequence there may be some tissue-specific hypothyroidism.
- TH mimetics are reported to significantly reduce T4 levels in animals and humans. Thus, there remains a need to develop characterize and optimize drugs from this class that lower lipids effectively with minimal effects on thyroid hormone levels.
- the present invention relates to phosphorus-containing thyroid hormone receptor agonists ("TELA”) having a short plasma half-life and exhibiting reduced effects on endogenous thyroid hormone levels, and methods of their use.
- TELA phosphorus-containing thyroid hormone receptor agonists
- TR thyroid hormone receptor
- the phosphorus-containing TR agonists of this invention and prodrugs thereof exhibit a short plasma half-life, particularly in mammalian and more particularly human subjects.
- TR agonists of this invention are at least 2 to 10-fold more selective for the TRP-l receptor over the TRa-1 receptor. Preferably the TR agonists are at least 20-fold to 50-fold, more selective for TRP-l over TRa-1.
- TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects on cardiovascular function. Reduced or minimal effects may include clinically insignificant impact on cardiac function.
- TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on muscle and bone function, as compared to baseline or normal levels.
- TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on thyroid hormones, and combinations thereof.
- TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit minimal effects on oxygen consumption.
- TR agonists of this invention and prodrugs thereof achieve the desired therapeutic index (TI) by using a dosing frequency wherein drug levels are reduced from their Cmax prior to the next dose.
- TR agonists of the invention are dosed at a frequency that sustains cholesterol lowering over time with minimal effects on T4 or while maintaining lipid lowering effects compared to baseline.
- the compound is a liver targeted compound that does not substantially impact other tissues. Demonstration of liver targeting and lack of impact on other tissues can be evaluated by measusing changes in mRNAs in liver as opposed to muscle, heart, pituitary, etc.
- Compounds of the invention may show changes in liver transcripts of genes encoding, for example, Dl, m-GPDH, CYP7a, malic enzyme, sterol regulating element binding protein lc (SREBPlc), LDL-cholesterol receptor in the liver, TSH , or Dl in the pituitary, Dl and m-GPDH in the heart, uncoupling protein 3 (UCP3) in the muscle.
- SREBPlc sterol regulating element binding protein lc
- UCP3 uncoupling protein 3
- TR agonists of this invention and prodrugs thereof are combined with one or more lipid lowering agents such as statins or cholesterol absorption inhibitors to treat patients with hyperlipidemia.
- lipid lowering agents such as statins or cholesterol absorption inhibitors
- such combination allows therapeutic effects at a reduced dose of one or more of the agents, improves lipid profile, or improves safety/therapeutic index of the therapy of one or more of the agents.
- thyroid hormone receptor ligands compounds that are thyroid hormone receptor ligands, pharmaceutically acceptable salts, and prodrugs of these compounds as well as their preparation and uses for preventing effects on the thyroid hormone axis (THA) while retaining the beneficial effects of activation of TR in the liver for preventing and/or treating hypercholesterolemia, hyperlipidemia, elevated Lp(a), and associated diseases such as atherosclerosis, coronary heart disease, and pancreatitis.
- TR -l- selective agonists would lower lipids with an improved cardiac therapeutic index relative to less TR selective agonists. Further improvement in the cardiac therapeutic index was realized with the discovery of liver-targeted phosphorus-containing TR agonists (Erion et al. PNAS, 2007, 104 (39), 15490). No therapeutic window was found for TR -l- selective agonists relative to effects on THs in animals and humans, possibly because these compounds affected mechanisms controlling TH metabolism in the liver (e.g.
- phosphorus-containing TR agonists particularly those with a short plasma half-life, could be combined with statins without narrowing the therapeutic index since TR agonists have been reported to adversely affect the same target organs of toxicity as statins, i.e., liver and muscle. It was also unknown that phosphorus-containing TR agonists with a short plasma half- life selective for TRP-l would be as useful as a longer-acting TR agonist in treating one or more of the following conditions and/or diseases: dyslipidemia, hypercholesterolemia, hypertriglyceridemia, elevated Lp(a) levels, atherosclerosis, coronary heart disease, pancreatitis.
- suitable functional groups to shorten the half life of said compound include: esters, iodines, aldehydes, carbamates, carbonates, thioethers, disulfides, or ⁇ -substituted cysteines, and phosphates that would be substrates for ubiquitous, highly expressed and highly active enzymes such as but not limited to esterases, deiodinases, carboxylesterases, aldehyde oxidases, glutathione transferases cysteine ⁇ -lyases and phosphatases (Rooseboom et al. Pharmacol. Rev. 56:53 (2004)).
- the compounds of the invention are designed to be rapidly metabolized by incorporation of metabolically unstable functional groups, these compounds are also designed with highly charged functional groups such as phospho(i)nic acids and phosphates, so as to target the liver and limit distribution in order to avoid activation of TR in tissues such as heart, muscle and pituitary. Activation of TR in extrahepatic tissues leads to side effects and a decrease in the therapeutic index (TI).
- TI therapeutic index
- the compounds of the invention are designed to be liver targeted and rapidly metabolized by incorporation of metabolically unstable functional groups, these compounds are also designed so as to bind and activate the TR in the liver to modulate gene expression of genes regulated by thyroid hormones.
- the compounds of the invention are designed as such that once metabolized, the metabolite produced does not bind to the TR or, if it does bind to TR, it does not activate TR.
- the present invention relates to phosphorus-containing TR agonists that have a short plasma half- life, are selective for the TRP-l receptor, target the liver, and lower lipids and lipoproteins without affecting THs
- the present invention relates to phospho(i)nic acid or phosphates containing compounds that bind to thyroid receptors in the liver and have a short half life. Activation of these receptors results in modulation of gene expression of genes regulated by TH.
- the present invention relates to phospho(i)nic acid or phosphates containing compounds designed with metabolic liabilities to shorten their half life and that bind to thyroid receptors in the liver and wherein administration of the compounds or prodrugs thereof results in lipid-lowering with minimal effects on T4.
- the present invention relates to phospho(i)nic acid or phosphates containing compounds described herein that bind to thyroid receptors in the liver, designed with metabolic liabilities to shorten their half life and whose metabolite do not activate TR receptors and wherein administration of the compounds or prodrugs thereof results in lipid-lowering with minimal effects on T4.
- the present invention relates to compounds designed with metabolic liabilities to shorten their half lives.
- the compounds may be thyroid hormone agonists of Formula I:
- Ar and Ar are substituted aryl or heteroaryl groups
- G is an atom or group of atoms that links Ar and Ar through 1-2 contiguous atoms;
- T is an atom or group of atoms linking Ar to E through 1 -4 contiguous atoms or is absent;
- E is a functional group with a pKa ⁇ 7.4, and more preferably pKa ⁇ 4.0, containing a phosphorus atom, and prodrug thereof.
- the present invention relates to methods of preventing or treating metabolic diseases with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
- the present invention relates to methods of reducing levels of Lp(a) with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
- the present invention relates to methods of reducing LDL- cholesterol levels with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
- the present invention also relates to pharmaceutically acceptable salts and co- crystals, prodrugs, and pharmaceutically acceptable salts and co-crystals of these prodrugs of these compounds.
- the present invention relates to methods of improving the therapeutic index by treating with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
- prodrugs of the compounds of the invention are also covered.
- the prodrugs of the compounds of the invention that have a short pharmacokinetic half life.
- the prodrug Upon oral absorption, the prodrug enters the portal vein and a fraction is uptaken by the liver. The remaining fraction not absorbed by the liver enters then the general circulation where, if the half life is not short, it has the potential to enter other organs, such as the heart and the brain.
- Prodrug breakdown and release of the parent TR agonist in these extra hepatic tissues could lead to activation of TR and a decrease of the TI.
- distribution of the intact prodrug into the whole body of an animal would lead to a significant increase in the volume of distribution of the prodrug.
- Prodrugs of the short acting TR compounds are useful for increasing oral bioavailability. A. Definitions
- phosphorus-containing thyroid hormone receptor agonist refers to TR agonists which bear a functional group with a pKa ⁇ 7.4, and preferably with a pKa ⁇ 4.0 containing a phosphorus atom such as a phospho(i)nic acid, phosphonic acid mono- ester, phosphoramidic acid or phosphate group.
- non-rodent mammalian animals refers to a mammal that is not from a rodent species.
- Preferred non-rodent mammalians are dogs, monkeys and humans.
- short plasma half-life refers to the pharmacokinetic half-life of a compound in plasma that is less than 4 h, less than 2 h, less than 1 h, less than 30 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes in non-rodent mammalian species.
- minimal effects on TH levels or endogenous TH levels refers to the effects of a drug on the levels of total T4, free T4, total T3 and free T3 in an animal. Such effects mean a change of less than 50%, preferably less than 20%, 10%>, or 5%>,.
- thyroid hormones refers to tyrosine-based hormones produced by the thyroid gland such as thyroxine (T4), 3',3,5-triiodothyronine (T3), 3',5',3- triiodothyronine (rT3) and 3,5-diodothyronine (T2).
- lipids refers to the group of naturally-occurring molecules which includes fats (fatty acids, mono, di and triglycerides), waxes, sterols (cholesterol), fat- soluble vitamins (such as vitamins A, D, E and K), phospholipids, and others.
- fats fatty acids, mono, di and triglycerides
- sterols cholesterol
- fat- soluble vitamins such as vitamins A, D, E and K
- phospholipids and others.
- the main biological functions of lipids include energy storage, as structural components of cell membranes, and as important signaling molecules.
- lipoproteins refers to biochemical assemblies containing both proteins and lipids and are classified by their relative density such as chylomicron, very low density lipoprotein (VLDL), low density lipoprotein (LDL) and high density lipoprotein (HDL).
- VLDL very low density lipoprotein
- LDL low density lipoprotein
- HDL high density lipoprotein
- T3 mimetic or TH mimetic refers to compounds that mimic the pharmacodynamic effects of T3.
- metabolic rate refers to resting metabolic rate (RMR) which is the amount of energy expanded by an animal while at rest in a neutrally temperate environment.
- RMR resting metabolic rate
- the RMR is measured by gas analysis through direct or indirect calorimetry.
- RMR can be determined by calculating the respiratory quotient which measures the inherent composition and utilization of carbohydrates, fats and proteins as they are converted to energy substrate units that can be used by the body as energy.
- oxygen consumption refers to the rate of oxygen consumed by an animal while at rest in a neutrally temperate environment. Oxygen consumption is used to calculate the respiratory quotient which is directly related to RMR. An elevation in oxygen consumption is associated with an increase in RMR.
- cardiac parameters refers to the measurable parameters used to evaluate cardiac function such as gene expression changes (e.g., Dl, m-GPDH), heart rate, pulse rate, rhythm, hypertrophy, or contractility.
- the term “elevated Lp(a)” refers to levels of the lipoprotein Lp(a) that are >10 mg/dL.
- prodrug with a short plasma half-life refers to the pharmacokinetic half-life of a compound in plasma that is less than 4 h, less than 2 h, less than 1 h, less than 30 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes.
- prodrug refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each.
- Standard prodrugs are formed using groups attached to functionality, e.g., the phenol group of the compounds described herein, that cleave in vivo. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound.
- the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, and/or pharmacodynamic half-life, etc.
- Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site-specific delivery of the compound.
- Prodrugs are described in The Organic Chemistry of Drug Design and Drug Action, by Richard B. Silverman, Academic Press, San Diego, 1992. Chapter 8: “Prodrugs and Drug delivery Systems” pp.352-401; Design of Prodrugs, edited by H. Bundgaard, Elsevier Science, Amsterdam, 1985; Design of Biopharmaceutical Properties through Prodrugs and Analogs, Ed. by E. B. Roche, American Pharmaceutical Association, Washington, 1977; and Drug Delivery Systems, ed. by R. L. Juliano, Oxford Univ. Press, Oxford, 1980.
- alkyl generally refers to saturated hydrocarbyl radicals of straight, branched or cyclic configuration including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, cyclohexyl, n-heptyl, octyl, n-octyl, and the like.
- alkyl substituents may be Ci to C 2 o , Ci to C 12j Ci to C 8 , Ci to C 6 , or Ci to C 4 alkyl groups.
- the alkyl group may be optionally substituted.
- the alkyl group may be a haloalkyl, including monohaloalkyl, dihaloalkyl, and trihaloalkyl.
- alkylene generally refers to linear, branched or cyclic alkene radicals having one or more carbon-carbon double bonds, such as C 2 to C 6 alkylene groups including 3-propenyl. Again, in certain embodiments, the alkyl group may be optionally substituted.
- alkenyl refers to unsaturated groups which have, e.g., 2 to 12 atoms and contain at least one carbon-carbon double bond and includes straight-chain, branched-chain and cyclic groups. Alkenyl groups may be optionally substituted. Suitable alkenyl groups include allyl.
- alkynyl refers to unsaturated groups which have, e.g., 2 to 12 atoms and contain at least one carbon-carbon triple bond and includes straight-chain, branched-chain and cyclic groups. Alkynyl groups may be optionally substituted. Suitable alkynyl groups include ethynyl.
- aryl refers to a carbocyclic aromatic ring structure. Included in the scope of aryl groups are aromatic rings having from five to twenty ring atoms.
- Aryl ring structures include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds, and includes carbocyclic aryl and heterocyclic aryl and biaryl groups. Examples of aryl groups that include phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, phenanthrenyl (i.e., phenanthrene), and napthyl (i.e., napthalene) ring structures. Again, in certain embodiments, the alkyl group may be optionally substituted.
- heterocycle refers to cyclic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heterocycle, and independently selectable, are O, N, and S heterocycle ring structures.
- the ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds, and may be aromatic, i.e., the ring structure may be a heteroaryl.
- heterocyclo groups include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl or tetrahydrothiopyranyl and the like.
- the alkyl group may be optionally substituted.
- heteroaryl refers to cyclic aromatic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heteroaryl, and independently selectable, are O, N, and S heteroaryl ring structures.
- the ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds.
- the heteroaryl groups may be selected from heteroaryl groups that contain two or more heteroatoms, three or more heteroatoms, or four or more heteroatoms.
- Heteroaryl ring structures may be selected from those that contain five or more atoms, six or more atoms, or eight or more atoms. In a preferred embodiment, the heteroaryl including five to ten atoms.
- heteroaryl ring structures include: acridine, benzimidazole, benzoxazole, benzodioxole, benzofuran, 1,3- diazine, 1,2-diazine, 1,2-diazole, 1 ,4-diazanaphthalene, furan, furazan, imidazole, indole, isoxazole, isoquinoline, isothiazole, oxazole, purine, pyridazine, pyrazole, pyridine, pyrazine, pyrimidine, pyrrole, quinoline, quinoxaline, thiazole, thiophene, 1,3,5-triazine, 1,2,4-triazine, 1,2,3-
- alkoxy generally refers to a group with the structure -O-R.
- R may be an alkyl group, such as a Ci to C 8 , Ci to C 6 alkyl group, or C ⁇ to C 4 alkyl group.
- the R group of the alkoxy may optionally be substituted, e.g., with at least one halogen.
- the R group of the alkoxy may be a haloalkyl, i.e., haloalkoxy.
- Halogen substituents may be independently selected from the halogens such as fluorine, chlorine, bromine, iodine, and astatine.
- the term "optionally substituted” or “substituted” includes groups substituted by one, two, three, four, five, or six substituents, independently selected from lower alkyl, lower aryl, lower aralkyl, lower cyclic alkyl, lower heterocycloalkyl, hydroxy, lower alkoxy, lower aryloxy, perhaloalkoxy, aralkoxy, lower heteroaryl, lower heteroaryloxy, lower heteroarylalkyl, lower heteroaralkoxy, azido, amino, halo, lower alkylthio, oxo, lower acylalkyl, lower carboxy esters, carboxyl, - carboxamido, nitro, lower acyloxy, lower aminoalkyl, lower alkylaminoaryl, lower alkylaryl, lower alkylaminoalkyl, lower alkoxyaryl, lower arylamino, lower aralkylamino, sulfonyl, lower-car
- terapéuticaally effective amount means an amount of a compound or a combination of compounds that modifies, ameliorates, attenuates or eliminates one or more of the symptoms of a particular disease or condition or prevents, modifies, or delays the onset of one or more of the symptoms of a particular disease or condition.
- the term "pharmaceutically acceptable salt” includes salts of compounds of Formula I and its prodrugs derived from the combination of a compound of this invention and an organic or inorganic acid or base.
- Suitable acids include acetic acid, adipic acid, benzenesulfonic acid, (+)-7,7-dimethyl-2-oxobicyclo[2.2.1 ]heptane- 1 -methanesulfonic acid, citric acid, 1 ,2-ethanedisulfonic acid, dodecyl sulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glucuronic acid, hippuric acid, hydrochloride hemiethanolic acid, HBr, HC1, HI, 2-hydroxyethanesulfonic acid, lactic acid, lactobionic acid, maleic acid, methanesulfonic acid, methylbromide acid, methyl sulfuric acid, 2- naphthalenesulfonic acid,
- patient and “subject” are used interchangably, and may include in vitro and in vivo subjects such as cells, tissues, and animals.
- animal includes birds and mammals.
- a mammal includes a rat, mouse, monkey, dog, cat, cow, horse, goat, sheep, pig or human of either gender.
- the term "increased or enhanced liver specificity” refers to an increase in the liver specificity ratio in animals treated with a compound of the present invention and a control compound.
- the term “enhanced oral bioavailability” refers to an increase of at least 50% of the absorption of the dose of the parent drug, unless otherwise specified. In an additional aspect the increase in oral bioavailability of the prodrug (compared to the parent drug) is at least 100%, that is a doubling of the absorption.
- Measurement of oral bioavailability usually refers to measurements of the prodrug, drug, or drug metabolite in blood, plasma, tissues, or urine following oral administration compared to measurements following systemic administration of the compound administered orally.
- treating includes a slowing of the progress or development of a disease after onset or actually reversing some or all of the disease affects. Treatment also includes palliative treatment.
- the term "preventing” includes a slowing of the progress or development of a disease before onset or precluding onset of a disease.
- prodrug refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each.
- Standard prodrugs are formed using groups attached to functionality, e.g., HO-, HS-, HOOC-, R 2 N-, associated with the drug, that cleave in vivo.
- Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate.
- the groups illustrated are exemplary, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Such prodrugs of the compounds of the present invention fall within this scope. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound.
- the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, and/or pharmacodynamic half-life, etc.
- Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site-specific delivery of the compound.
- Prodrugs are described in The Organic Chemistry of Drug Design and Drug Action, by Richard B. Silverman, Academic Press, San Diego, 1992.
- Prodrugs of carboxylic acid-containing thyromimetics are convertible by solvolysis or under physiological conditions to the free carboxylic acids.
- Examples of prodrugs include carboxylic acid esters, and are preferably lower alkyl esters, cycloalkyl esters, lower alkenyl esters, benzyl esters, aryl esters, mono- or di-substituted lower alkyl esters, e.g., the ro-(amino, mono- or di-lower alkylamino, carboxy, lower alkoxycarbonyl)-lower alkyl esters, and the a-(lower alkanoyloxy, lower alkoxycarbonyl or di-lower alkylaminocarbonyl)-lower alkyl esters, such as the pivaloyloxy-methyl ester.
- Prodrugs of phosphorus-containing thyromimetics breakdown chemically or enzymatically to a phospho(i)nic acid or phosphate group thereof in vivo.
- the term includes, but is not limited to, the following groups and combinations of these groups:
- esters are possible in which a cyclic alkyl ring is formed. These esters have been shown to generate phosphorus-containing nucleotides inside cells through a postulated sequence of reactions beginning with deesterification and followed by a series of elimination reactions ⁇ e.g., Freed et al, Biochem. Pharm, 38:3193-3198 (1989)).
- alkyloxycarbonyloxymethyl esters as shown in formula A, where R is alkoxy, aryloxy, alkylthio, arylthio, alkylamino, and arylamino; R', and R" are independently -H, alkyl, aryl, alkylaryl, and heterocycloalkyl have been studied in the area of ⁇ -lactam antibiotics (Nishimura et al., J. Antibiotics 40(l):$l-90 (1987); for a review see Ferres, H., Drugs of Today, 19:499 (1983)). More recently Cathy, M. S. et al.
- acyloxyalkyl esters are possible in which a cyclic alkyl ring is formed such as shown in formula B. These esters have been shown to generate phosphorus- containing nucleotides inside cells through a postulated sequence of reactions beginning with deesterification and followed by a series of elimination reactions (e.g., Freed et al, Biochem. Pharm. 38:3193-3198 (1989)).
- R is -H, alkyl, aryl, alkylaryl, alkoxy, aryloxy, alkylthio, arylthio, alkylamino, arylamino, or cyclo alkyl.
- Aryl esters have also been used as phosphonate prodrugs (e.g., DeLombaert et al, J. Med. Chem. 57 ⁇ :498-511 (1994); Serafmowska et al, J. Med. Chem. 38(8): ⁇ 3 ⁇ 2- 9 (1995). Phenyl as well as mono and poly-substituted phenyl proesters have generated the parent phosphonic acid in studies conducted in animals and in man (Formula C). Another approach has been described where Y is a carboxylic ester ortho to the phosphate (Khamnei et al, J. Med. Chem. J :4109-15 (1996)). wherein Y is -H, alkyl, aryl, alkylaryl, alkoxy, acyloxy, halogen, amino, alkoxycarbonyl, hydroxy, cyano, and heterocycloalkyl.
- Benzyl esters have also been reported to generate the parent phosphonic acid. In some cases, using substituents at the /?ara-position can accelerate the hydrolysis.
- X and Y are independently -H, alkyl, aryl, alkylaryl, alkoxy, acyloxy, hydroxy, cyano, nitro, perhaloalkyl, halo, or alkyloxycarbonyl;
- R and R are independently -H, alkyl, aryl, alkylaryl, halogen, and cyclic alkyl.
- Thio-containing phosphonate proesters may also be useful in the delivery of drugs to hepatocytes. These proesters contain a protected thioethyl moiety as shown in formula E. One or more of the oxygens of the phosphonate can be esterified. Since the mechanism that results in de-esterification requires the generation of a free thiolate, a variety of thiol protecting groups are possible. For example, the disulfide is reduced by a reductase-mediated process (Puech et al, Antiviral Res. 22: 155-174 (1993)). Thioesters will also generate free thiolates after esterase-mediated hydrolysis Benzaria, et al, J. Med. Chem.
- Cyclic analogs are also possible and were shown to liberate phosphonate in isolated rat hepatocytes.
- the cyclic disulfide shown below has not been previously described and is novel. wherein Z is alkylcarbonyl, alkoxycarbonyl, arylcarbonyl, aryloxycarbonyl, or alkylthio.
- prodrugs include proester classes exemplified by Biller and Magnin (U.S. Patent No. 5,157,027); Serafinowska et al., J. Med. Chem. 55 ⁇ :1372-9 (1995); Starrett et al, J. Med. Chem. 57:1857 (1994); Martin et al. J. Pharm. Sci. 7(5: 180 (1987); Alexander et al., Collect. Czech. Chem. Commun. 5 :1853 (1994); and EP 0 632 048 Al .
- R is -H, alkyl, cycloalkyl, or heterocycloalkyl
- Y is -H, alkyl, aryl, alkylaryl, cyano, alkoxy, acyloxy, halogen, amino, heterocycloalkyl, and alkoxycarbonyl.
- prodrugs of Formula E-3 are an example of "optionally substituted heterocycloalkyl where the cyclic moiety contains a carbonate or thiocarbonate.”
- Propyl phosphonate proesters can also be used to deliver drugs into hepatocytes. These proesters may contain a hydroxyl and hydroxyl group derivatives at the 3 -position of the propyl group as shown in formula F.
- the R and X groups can form a cyclic ring system as shown in formula F.
- One or more of the oxygens of the phosphonate can be esterified.
- R is alkyl, aryl, heteroaryl
- X is hydrogen, alkylcarbonyloxy, alkyloxycarbonyloxy
- Y is alkyl, aryl, heteroaryl, alkoxy, alkylamino, alkylthio, halogen,
- Phosphoramidate derivatives have been explored as phosphate prodrugs (e.g., McGuigan et al., J. Med. Chem. 42:393 (1999) and references cited therein) as shown in Formula G and H.
- Cyclic phosphoramidates have also been studied as phosphonate prodrugs because of their speculated higher stability compared to non-cyclic phosphoramidates (e.g., Starrett et al, J. Med. Chem. 57: 1857 (1994)).
- prodrugs are possible based on literature reports such as substituted ethyls, for example, bis(trichloroethyl)esters as disclosed by McGuigan, et ah, Bioorg Med. Chem. Lett. 5: 1207-1210 (1993), and the phenyl and benzyl combined nucleotide esters reported by Meier, C. et al, Bioorg. Med. Chem. Lett. 7:99-104 (1997).
- substituted ethyls for example, bis(trichloroethyl)esters as disclosed by McGuigan, et ah, Bioorg Med. Chem. Lett. 5: 1207-1210 (1993), and the phenyl and benzyl combined nucleotide esters reported by Meier, C. et al, Bioorg. Med. Chem. Lett. 7:99-104 (1997).
- cyclic phosphonate ester of 1,3-propane diol refers to the following:
- the structure above has an acyloxy substituent that is three carbon atoms from a Y, and an optional substituent, -CH 3 , on the new 6-membered ring.
- V aryl
- a spiro-fused cyclopropyl group for W and W spiro-fused cyclopropyl group for W and W.
- R 5 and R 3 be a substituent on the ring bearing the R 1 and R2 groups.
- the naming of the prodrugs is done by having the diaryl system with its linker T (Formula I, II, III, V, VI, and VIII) or D (Formula IV) be a substituent on the phosphorus atom contained in X.
- T Formula I, II, III, V, VI, and VIII
- D Formulamula IV
- N-[3-R -5-R -4-(4'-R -3'-R -phenoxy)phenyl]carbamoylphosphonic acid represents the formula:
- cis stereochemistry refers to the spatial relationship of the V group and the carbon attached to the phosphorus atom on the six-membered ring.
- the formula below shows a cis stereochemistry.
- trans stereochemistry refers to the spatial relationship of the V group and the carbon, attached to the phosphorus atom, on the six-membered ring.
- the formula below shows a trans-stereochemistry.
- ⁇ -configuration refers to the absolute configuration S of carbon C.
- ⁇ -isomer refers to the absolute configuration S of carbon C.
- ⁇ -prodrug refers to the absolute configuration S of carbon C.
- the formula below shows the ⁇ -stereochemistry.
- enantioenriched or “enantiomerically enriched” refers to a sample of a chiral compound that consists of more of one enantiomer than the other. The extent to which a sample is enantiomerically enriched is quantitated by the enantiomeric ratio or the enantiomeric excess.
- compounds of the invention that have a short pharmacokinetic half-life and are thyroid receptor ligands are provided, pharmaceutically acceptable salts thereof, and prodrugs of these compounds as well as their preparation and uses for preventing and/or treating metabolic diseases such as NASH, hypercholesterolemia and hyperlipidemia as well as associated conditions such as atherosclerosis, and coronary heart disease.
- the present invention is also related to the use of the compounds of the invention to treat the above-mentioned diseases and improve the therapeutic index by decreasing effects on the thyroid hormone axis.
- the invention is also related to the use of these compounds for the prevention and treatment of diseases responsive to modulation of T3 -responsive genes in the liver.
- Preferred compounds of the present invention include those described herein.
- the compounds may be thyroid hormone agonists of Formula I:
- Ar and Ar are substituted aryl or heteroaryl groups
- G is an atom or group of atoms that links Ar and Ar through 1-2 contiguous atoms;
- T is an atom or group of atoms linking Ar to E through 1 -4 contiguous atoms or is absent;
- E is a functional group with a pKa ⁇ 4.0 containing a phosphorus atom, and prodrug thereof.
- the thyroid hormone agonists of Formula I are a compound of Formula II:
- G is selected from:
- E is a functional group with a pKa ⁇ 4.0 containing a phosphorus atom
- thyroid hormone agonists of Formula I include compounds of Formula III:
- n is an integer from 0-2;
- p is an integer from 0-1;
- Each R a is independently selected from:
- Y" is -Ci-Ce-alkyl
- Y and Y' are each independently selected from the group consisting of -0-, and
- R 11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH 2 -heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(R z ) 2 OC(0)NR z 2 , -NR z -C(0)-R y , -C(R z ) 2 -OC(0)R y , -C(R z ) 2 -0-C(0)OR y , -C(R z ) 2 OC(0)SR y , -alkyl-S-C(0)R y , -alkyl-S-S-alkylhydroxy, and --
- R 11 attached to -NR V - is independently selected from the group consisting of -H, -[C(R z ) 2 ] q -COOR y , -C(R x ) 2 COOR y ,
- R 11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH 2 -heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl,
- R 11 attached to -NR V - is independently selected from the group consisting of -H, -[C(R z ) 2 ] q -COOR y , -C(R x ) 2 COOR y , -[C(R z ) 2 ] q -C(0)SR y , and
- R 11 and R 11 together form a cyclic group comprising -alkyl-S-S-alkyl-, or together R 11 and R 11 are the group:
- V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or
- V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
- V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or
- V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or
- Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or
- W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
- Z is selected from the group consisting of -CHR z OH, -CHR z OC(0)R y ,
- Each R z is selected from the group consisting of R y and -H;
- Each R y is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
- Each R x is independently selected from the group consisting of -H, and alkyl, or together R x and R x form a cycloalkyl group;
- Each R v is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl;
- V, Z, W, W are not all -H;
- R may be selected from:
- R f and R g may together form:
- E may be selected from:
- T may be selected from:
- E may be selected from: -P0 3 H 2 , -P(0)[-OCR z 2 OC(0)R y ] 2 ,
- G may be selected from:
- R may be OH
- E may be selected from:-P0 3 H 2 , -P(0)[-OCH 2 OC(0)-i-butyl] 2 ,
- the compounds of the invention exhibit metabolic instability which may be mediated by the deactivating enzyme selected from the group consisting of glutathione transferase, and cysteine ⁇ -lyase.
- the metabolically unstable substituent is contained within the T substituent of a compound of Formula III.
- T may be selected from:
- T may be selected from:
- E may be se ected from: -P0 3 H 2 ,
- G may be selected from:
- E may be selected from:-P0 3 H 2 , -P(0)[-OCR z 2 OC(0)R y ] 2 ,
- G may be selected from:
- R is OH
- E is selected from:-P0 3 H 2 , -P(0)[-OCH 2 OC(0)-t-butyl] 2 , -P(0)[-OCH 2 OC(0)0-z-propyl] 2 , -P(0)[-N(H)CH 2 C(0)OCH 2 CH 3 ] 2 , -P(0)[-N(H)CH(CH 3 )C(0)OCH 2 CH 3 ] 2 , -P(0)[-N(H)C(CH 3 ) 2 C(0)OCH 2 CH 3 ] 2 , -P(0)[-N(H)CH(CH 3 )C(0)OCH 2 CH 3 ][3,4-methylenedioxyphenyl], -P(0)[-N(H)C(CH 3 )2C(0)OCH 2 CH 3 ][3,4-methylenedioxyphenyl], -P(0)[-OCH (3-chlorophenyl)CH 2 CH 2 0-], -P(0)[-OCH(pyrid-4-yl)
- the compounds of the invention exhibit metabolic instability which is mediated by the deactivating enzyme deiodinase.
- the compound of Formula III contains at least one metabolically unstable iodo substituent.
- T may be selected from:
- R f and R g may together form:
- E may be selected from
- G may be selected from:
- R g may be selected from:
- E may be selected from: -F -P(0)[-OCR z 2 OC(0)R y ] 2 ,
- G may be selected from:
- each R e may independently be selected from:
- R 5 may be OH
- E may be selected from:-P0 3 H 2 , -P(0)[-OCH 2 OC(0)-t-butyl] 2 ,
- the compounds of the invention may exhibit metabolic instability which is mediated by the deactivating enzyme phosphatase.
- the metabolically unstable substituent may be contained within the E substituent of a compound of Formula III.
- T may preferably be selected from:
- n is an integer from 0-2;
- Each R is independently selected from:
- E is PCC OR OR 11 ;
- R attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(R z ) 2 OC(0)NR z 2 , -NR z -C(0)-R y , -C(R z ) 2 -OC(0)R y , -C(R z ) 2 -0-C(0)OR y , -C(R z ) 2 OC(0)SR y , -alkyl-S-C(0)R y ,
- R 11 and R 11 together form a cyclic group comprising -alkyl-S-S-alkyl-, or together R 11 and R 11 are the group:
- V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or
- V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
- V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or
- V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or
- Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or
- W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
- Z is selected from the group consisting of -CHR z OH, -CHR z OC(0)R y ,
- q is an integer 2 or 3;
- Each R z is selected from the group consisting of R y and -H;
- Each R y is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
- Each R x is independently selected from the group consisting of -H, and alkyl, or together R x and R x form a cycloalkyl group;
- Each R v is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl;
- V, Z, W, W are not all -H;
- T may be selected from:
- E may be se ected from: -P0 3 H 2 ,
- V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
- G may be selected from:
- R may be selected from:
- E may be selected from:-P0 3 H 2 , -P(0)[-OCR z 2 OC(0)R y ] 2 ,
- V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
- G may be selected from:
- T may be selected from:
- R may be OH
- E may be selected from:-P0 3 H 2 , -P(0)[-OCH 2 OC(0)-t-butyl] 2 ,
- the compounds are selected from the group consisting of and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
- the compounds of the invention may exhibit metabolic instability which is mediated by the deactivating enzyme selected from the group consisting of esterase, deiodinase, carboxylesterase, aldehyde oxidase, glutathione transferase, cysteine ⁇ -lyase, and phosphatase.
- the deactivating enzyme selected from the group consisting of esterase, deiodinase, carboxylesterase, aldehyde oxidase, glutathione transferase, cysteine ⁇ -lyase, and phosphatase.
- the compounds of the invention may exhibit a metabolic instability which is mediated by the deactivating enzyme selected from the group consisting of the deactivating enzyme is selected from esterase, deiodinase, carboxylesterase, and aldehyde oxidase.
- All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form.
- the compounds of the present invention can have stereogenic centers. Consequently, the compounds can exist in enantiomeric or diastereomeric forms or in mixture thereof.
- the processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When enantiomeric or diastereomeric products are prepared, they can be separated by conventional methods for example, chromatographic or fractional crystallization.
- synthesis of the scaffolds such as biaryl ethers and phenyl benzyl can be accomplished by processes known to one skilled in the art using the numerous methods described in the prior art for the synthesis of thyromimetics. More specifically, thyromimetics with phosphonic and phosphinic acids were described in WO06128055 and WO06128056 (the contents of which are herein incorporated by reference in their entirety) modified as described herein and as recognized by those in the art.
- the group is introduced by treating the appropriate scaffold with a strong base such as BuLi and trapping the anion with a symmetrical disulfide.
- a strong base such as BuLi
- the anion can be trapped with Ss to give the ortho thiol.
- the thiol is then alkylated using procedures known to those skilled in the art such as treatment with Mel and triethylamine.
- an ortho hydroxyl group is introduced on an anisole scaffold by treating the anisole with BuLi and trapping the anion with trimethyl borate.
- the borate ester is then oxidized with hydrogen peroxide to the corresponding guaiacol which is then acylated with an isocyanate and deprotected.
- a phenolic scaffold can be hydroxylated with one of the numerous methods known to those skilled in the art using hydrogen peroxide and an oxidizing catalyst such as iron salt (ferrocene, Fe(acac) 3 ) and selectively acylated with an isocyanate.
- thiophenols are synthesized from the corresponding phenol via palladium coupling of the respective aryl trifluoromethylsulfonate with trialkylsilyl thiolate (Tetrahedron Lett. 37:4523 (1996)) followed by removal of the silyl group with TBAF.
- the thiophenol is then alkylated using an activated phosphono-serine derivative (Angew. Chem. 98:836 (1986)) and deprotected.
- Compounds of the invention where the metabolically unstable group is a phosphate or a phosphoramidate are prepared by reaction of the corresponding thyromimetic scaffold bearing a phenol, alcohol or amine with a phosphoramidite, such as di-t-butyl-N,N-diisopropyl phosphoramidite, followed by oxidation of the phosphite or imidite with t-BuOOH, and t-butyl removal with TFA.
- a phosphoramidite such as di-t-butyl-N,N-diisopropyl phosphoramidite
- prodrugs of phosphonic and phosphinic acids can be accomplished by processes known to one skilled in the art using the numerous methods described in WO06128055 and WO06128056 (the contents of which are herein incorporated by reference in their entirety) modified as described herein and as recognized by those in the art.
- Prodrugs of phosphates and phosphoramidates are made by preparing the phosphorylating reagents bearing the prodrug moiety, such as trans 2-(4-nitrophenyl)-4- aryl-2-oxo-l,3,2-dioxaphosphorinanes and bis-(pivaloyloxymethyl)-phosphoryl chloride, and phosphorylating phenols, alcohols or amines using processes known to those skilled in the art.
- compounds of the invention may be resolved to enantiomerically pure compositions or synthesized as enantiomerically pure compositions using any method known in art.
- compounds of the invention may be resolved by direct crystallization of enantiomer mixtures, by diastereomer salt formation of enantiomers, by the formation and separation of diasteriomers or by enzymatic resolution of a racemic mixture.
- the methods of the invention comprise administering a therapeutically effective amount of at least one compound of the invention, e.g., a compound of Formula I, II, or III.
- Relative activity of the compounds of the invention may be determined by any method known in the art, including the assay described herein.
- the compounds of the invention and their prodrugs and salts are useful in preventing or treating arteriosclerosis by modulating levels of atherogenic proteins, e.g., Lp(a), apoAI, apoAII, LDL, HDL.
- atherogenic proteins e.g., Lp(a), apoAI, apoAII, LDL, HDL.
- Clinically overt hypothyroidism is associated with accelerated and premature coronary atherosclerosis and subclinical hypothyroidism is considered a condition with an increased risk for these diseases (Vanhaelst et al. and Bastenie et al., Lancet 2 (1967)).
- T3 and T3 mimetics modulate atherogenic proteins in a manner that could prove beneficial for patients at risk to develop atherosclerosis or patients with atherosclerosis or diseases associated with atherosclerosis.
- T3 and T3 mimetics are known to decrease Lp(a) levels, e.g., in the monkey, with 3,5-dichloro-4-[4-hydroxy-3-(l- methylethyl)phenoxy]benzeneacetic acid (Graver et al, Proc. Natl. Acad. Sci. U.S.A. 700: 10067-10072 (2003)).
- the T3 mimetic CGS23425 [[4-[4- hydroxy-3 -( 1 -methyl ethyl)phenoxy] -3 ,5 -dimethylphenyl] amino]oxo acetic acid) increased apoAI expression via thyroid hormone receptor activation (Taylor et al., Mol. Pharm. 52:542-547 (1997)).
- the compounds of the invention can be used to treat or prevent atherosclerosis, coronary heart disease and heart failure because such compounds are expected to distribute to the liver and modulate the expression and production of atherogenic proteins.
- the compounds of the invention and their prodrugs and salts are useful for preventing and/or treating metabolic diseases such as hypercholesterolemia and hyperlipidemia and conditions such as atherosclerosis, coronary heart disease, heart failure, nephrotic syndrome, and chronic renal failure without affecting thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4.
- T3 mimetics in these methods would reduce or minimize effects on thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4.
- the compounds or the present invention that distribute more readily to the liver and have a short half life result in pharmacological effects at doses that do not adversely affect thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4.
- the compounds of the present invention have a therapeutic index "TI", defined as the difference between the dose at which a significant effect is observed for a use disclosed herein, e.g., lowering lipids, lowering cholesterol, etc., and the dose at which a significant decrease in T3 or significant decrease in T4, or significant change in the ratio of T3 to T4 is observed, is at least 50 fold, 100 fold, 200 fold, 300 fold, 400 fold, 500 fold, 600 fold, 700 fold, 800 fold, 900 fold, 1000 fold, 2000 fold, 3000 fold, 4000 fold, 5000 fold, 6000 fold, 7000 fold, 8000 fold, 9000 fold or at least 10000 fold.
- TI therapeutic index
- an observed desired TI may include a significant effect on lowering lipids, e.g., 15% lipid lowering over baseline, without a corresponding significant effect, e.g., no more than a 25% reduction, on T4 in a non-rodent mammalian species, by using a dosing frequency wherein drug levels are reduced from their Cmax at least 50%, preferably 80%, 90%, 95%, or 99%, prior to the next dose.
- the amount of change in T3 or T4 may be a decrease selected from at least 5%, 10%>, 15%, 20%>, 25% or at least 30% of circulating levels.
- the compounds of the invention may be at least 2 to 10-fold more selective for the ⁇ ⁇ -l receptor over the TRa-1 receptor, preferably at least 20-fold to 50-fold, more selective for TRP-l over TRa-1.
- the compounds of the invention exhibit a short plasma half-life, particularly in mammalian and more particularly human subjects.
- the compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects on cardiovascular function.
- Reduced or minimal effects may include clinically insignificant impact on cardiac function.
- cardiac function may be evaluated by measuring changes in the maximum resting heart rate in a subject.
- Reduced or minimal effects may include a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%.
- reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED 5 o for cholesterol lowering over 14 days.
- clinically insignificant effects may include a resting heart rate of less than 100 bpm or an increase in resting heart rate of less than 20 bpm as compared to baseline or normal levels.
- TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on oxygen consumption.
- reduced or minimal effects may be a change of less than 50% of baseline levels, more preferably less than 20% or less than 10%.
- Oxygen consumption may be measured in any suitable manner recognized in the art.
- reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED50 for cholesterol lowering over 14 days. Again, alternatively one could use the minimally efficacious dose for cholesterol lowering.
- compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on muscle and bone function, as compared to baseline or normal levels.
- reduced or minimal effects may be a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%.
- reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED 50 for cholesterol lowering over 14 days for muscle effects, or over 6 months for bone effects (or minimally efficacious dose for cholesterol lowering).
- Thyroid hormones can affect bone mineral density and increase the risk of bone fractures.
- Serum osteocalcin and bone-specific alkaline phosphatase are markers of bone formation
- urinary pyridinoline cross-link (Pyr) excretion is a marker of bone resorption.
- Impact of thyroid hormones on muscle may be measured using any suitable assay methodology, including for example measurement of gastrocnemius muscle mass.
- 3-Methylhistidine urinary excretion and net balances across the leg or forearm may also be used as markers of contractile protein breakdown in muscle tissue.
- the compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on thyroid hormones, and combinations thereof.
- reduced or minimal effects may be a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%.
- reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED 5 o for cholesterol lowering over 14 days (or minimally efficacious dose for cholesterol lowering).
- the compounds of the invention have reduced or minimal effects on thyroid hormone levels at lipid lowering doses as compared to baseline levels.
- the effect is a change of less than 50% of baseline T4 levels, more preferably less than 20% or less than 10%.
- the compounds of the invention may be used to significantly lower cholesterol levels without having a significant effect on TSH levels.
- the compounds of the present invention significantly lower cholesterol levels without lowering TSH levels by more than 30%, 25%, 20%, 15%, 10%, or 5%.
- the compounds of the invention may be dosed at a frequency that sustains cholesterol lowering over time with minimal effects on T4.
- the dosing frequency may be determined by maintaining lipid lowering effect compared to baseline of at least 5%, 10%>, 15%, or 20%>.
- dosing is once or twice daily, once every other day, every third day, 3, 2, or 1 times weekly, or once or twice monthly.
- the compounds of the invention may be liver targeted compounds that do not substantially impact other tissues. Demonstration of liver targeting and lack of impact on other tissues can be evaluated by measuring changes in mR As levels in liver as opposed to muscle, heart, pituitary, etc.
- Compounds of the invention may show changes in liver transcripts of genes encoding, for example, Dl, m- GPDH, CYP7a, malic enzyme, sterol regulating element binding protein lc (SREBPlc), LDL-cholesterol receptor in the liver, ⁇ , or Dl in the pituitary, Dl and m-GPDH in the heart, uncoupling protein 3 (UCP3) in the muscle.
- Liver changes are of a magnitude of preferably at least 10%, 20%, 30%, 50%, 100%, 200%, 500% or more, while systemic tissue changes are preferably less than 100%, more preferably less than 50%>, 30%>, 20%>, or 10%>.
- the compounds of the present invention are able to increase the therapeutic index as compared to T3 and T3 mimetics.
- the compounds of the present invention can therefore be dosed at levels that are effective in treating metabolic and other disorders where the liver is the drug target without significantly negatively affecting THA.
- the compounds of the present invention have a therapeutic index, defined as the difference between the dose at which a significant effect is observed for a use disclosed herein, e.g., lowering cholesterol, and the dose at which a significant effect on a property or function, as disclosed herein ⁇ e.g., heart rate), is observed, is at least 50 fold, 100 fold, 200 fold, 300 fold, 400 fold, 500 fold, 600 fold, 700 fold, 800 fold, 900 fold, 1000 fold, 2000 fold, 3000 fold, 4000 fold, 5000 fold, 6000 fold, 7000 fold, 8000 fold, 9000 fold or at least 10000 fold.
- Examples of said use disclosed herein includes but is not limited to reducing lipid levels, increasing the ratio of HDL to LDL or apoAI to LDL, increasing mitochondrial biogenesis, AMP activated protein kinase or nuclear respiratory factor, or for the treatment or prevention of a disease or disorder selected from the group consisting of atherosclerosis, hypercholesterolemia, hyperlipidemia, NASH, NAFLD, nephrotic syndrome, chronic renal failure, metabolic syndrome X, hyperlipidemia, coronary heart disease, thyroid disease, thyroid cancer, depression, glaucoma, cardiac arrhythmias, heart failure, and osteoporosis.
- a disease or disorder selected from the group consisting of atherosclerosis, hypercholesterolemia, hyperlipidemia, NASH, NAFLD, nephrotic syndrome, chronic renal failure, metabolic syndrome X, hyperlipidemia, coronary heart disease, thyroid disease, thyroid cancer, depression, glaucoma, cardiac arrhythmias, heart failure, and osteoporosis.
- Examples wherein the property or function is a cardiac property/function include but are not limited to cardiac hypertrophy (heart weight to body weight ratio), heart rate, various hemodynamic parameters, including systolic and diastolic arterial pressure, end-systolic left ventricular pressure and maximal speeds of contraction and relaxation.
- cardiac hypertrophy heart weight to body weight ratio
- various hemodynamic parameters including systolic and diastolic arterial pressure, end-systolic left ventricular pressure and maximal speeds of contraction and relaxation.
- Other methods are also available to assess the therapeutic index including effects on muscle wasting, bone density, TSH levels, levels of T3 and T4, and the ratio T3/T4.
- the therapeutic index is determined by administering to animals a wide range of doses and determining the minimal dose capable of inducing a response in the liver relative to the dose capable of inducing side effects in the heart or on the THA.
- In vivo assays include but are not limited to treating animals with compounds of the invention or a prodrug thereof and monitoring the expression of T3-responsive genes in the liver or the functional consequences of changes of T3 -responsive genes.
- compounds useful in the novel methods bind to thyroid receptors and produce changes in the expression of two or more hepatic genes.
- Animals used for testing compounds useful in the methods include normal rats and mice, animals made hypothyroid using methods well described in the literature, including thyroid hormone receptor knockout mice (e.g., TRa ⁇ ⁇ such as those used in Grover et al, 2003), or animals exhibiting high cholesterol (e.g., high cholesterol fed rat or hamster), obesity and/or diabetes (e.g., fa/fa rat, Zucker diabetic fatty rat, ob/ob mice, db/db mice, high fat fed rodent).
- TRa ⁇ ⁇ such as those used in Grover et al, 2003
- high cholesterol e.g., high cholesterol fed rat or hamster
- obesity and/or diabetes e.g., fa/fa rat, Zucker diabetic fatty rat, ob/ob mice, db/db mice, high fat fed rod
- the drug or prodrug may be administered by a variety of routes including by bolus injection, oral, and continuous infusion.
- animals may be treated for 1-28 days and the liver, heart and blood are isolated.
- Changes in gene transcription relative to vehicle treated animals and T3 -treated animals determined using northern blot analysis, RNAase protection or reverse-transcription and subsequent PCR. While methods are available for monitoring changes in thousands of hepatic genes, only a small number need to be monitored to demonstrate the biological effect of compounds in this invention.
- genes such as spot-14, FAS, mGPDH, CPT-1, and LDL receptor may be monitored.
- Changes of >1.5 fold in two or more genes may be considered proof that the compound modulates T3 -responsive genes in vivo.
- Alternative methods for measuring changes in gene transcription include monitoring the activity or expression level of the protein encoded by the gene. For instance, in cases where the genes encode enzyme activities (e.g., FAS, mGPDH), direct measurements of enzyme activity in appropriately extracted liver tissue can be made using standard enzymological techniques. In cases where the genes encode receptor functions (e.g., the LDL receptor), ligand binding studies or antibody-based assays (e.g., Western blots) can be performed to quantify the number of receptors expressed. Depending on the gene, TR agonists may either increase or decrease enzyme activity or increase or decrease receptor binding or number.
- Administering compounds of the invention that bind to a TR to animals can result in changes in lipids, including hepatic and/or plasma cholesterol levels; changes in lipoprotein levels including LDL-cholesterol, lipoprotein a (Lp(a)); and changes in energy expenditure as measured by changes in oxygen consumption and in some cases animal weight.
- the effect on cholesterol may be determined using cholesterol fed animals such as normal rats and hamsters, or TRa ⁇ ⁇ knockout mice.
- Cholesterol may be measured using standard tests. Changes in energy expenditure may be monitored by measuring changes in oxygen consumption (MVo ).
- Compounds of the invention that bind to a TR modulate expression of certain genes in the liver resulting in effects on lipids (e.g., cholesterol), lipoproteins, and triglycerides.
- lipids e.g., cholesterol
- lipoproteins e.g., cholesterol
- lipoproteins e.g., Lp(a) or LDL
- Such compounds can raise levels of lipoproteins such as apoAI or HDL and are useful in preventing or treating atherosclerosis and heart disease in patients.
- said compound is an active form. In another embodiment said compound is a prodrug. In another embodiment said compound of the invention comprises a stereocenter, is enantiomerically enriched or diastereomerically enriched, or a stereoisomer covered later. In another embodiment said compound is administered as a racemic mixture. In another embodiment said compound is administered as an enantiomerically enriched mixture. In another embodiment said compound is administered as a diastereomerically enriched mixture. In still another embodiment said compound is administered as an individual stereoisomer.
- said methods of reducing cholesterol results in a lowering of total cholesterol.
- said methods of reducing cholesterol results in a reduction of high density lipoprotein (HDL).
- said methods of reducing cholesterol results in a reduction of low-density lipoprotein (LDL).
- said methods of reducing cholesterol results in a reduction of very low-density lipoprotein (VLDL).
- said LDL is reduced to a greater extent than said HDL.
- said VLDL is reduced to a greater extent than said HDL.
- said VLDL is reduced to a greater extent than said LDL.
- the lipid is triglycerides.
- said lipid is liver triglycerides.
- said lipid is in the form of a lipoprotein.
- said lipoprotein is Lp(a).
- said lipoprotein is apoAII. Also provided are methods of increasing the ratio of HDL to LDL, HDL to VLDL, LDL to VLDL, apoAI to LDL or apoAI to VLDL in an animal. Also provided are methods of treating hyperlipidemia or hypercholesterolemia in an animal,
- the methods generally comprise the step of administering to a patient in need thereof, such as an animal subject including a human subject, an effective amount of a compound of the invention.
- a patient in need thereof such as an animal subject including a human subject
- an effective amount of a compound of the invention in one embodiment said compound is an active form.
- said compound is a prodrug.
- said compound of the invention comprises a stereocenter.
- said compound is administered as a racemic mixture.
- said compound is administered as an enantiomerically enriched mixture.
- said compound is a administered as a diastereomeric mixture.
- said compound is administered as an individual stereoisomer.
- the liver is a major target organ of thyroid hormone with an estimated 8% of the hepatic genes regulated by thyroid hormone. Quantitative fluorescent-labeled cDNA microarray hybridization was used to identify thyroid-responsive genes in the liver as shown in Table 1 below (Feng et al, Mol. Endocrinol. 14:941-955 (2000)). Hepatic RNAs from T3-treated and hypothyroid mice were used in the study. Thyroid hormone treatment affected the expression of 55 genes from the 2225 different mouse genes sampled with 14 increasing >2-fold and 41 decreasing >60%.
- T3-responsive genes in the liver include genes affecting lipogenesis, including spot 14, fatty acid transport protein, malic enzyme, fatty acid synthase (Blennemann et al. 5 Mol. Cell. Endocrinol. 110(l-2): ⁇ -% (1995)) and CYP4A. HMG CoA reductase and LDL receptor genes have been identified as affecting cholesterol synthesis and as being responsive to T3.
- CPT-1 is a T3-responsive gene involved in fatty acid oxidation.
- Genes affecting energy expenditure including mitochondrial genes such as mitochondrial sn- glycerol 3-phosphate dehydrogenase (mGPDH), and/or enzymes associated with proton leakage such as the adenine nucleotide transporter (ANT), Na + /K + -ATPase, Ca 2+ -ATPase and ATP synthase are also T3 -responsive genes.
- T3 -responsive genes affecting glycogeno lysis and gluconeogenesis include glucose 6-phosphatase and PEPCK.
- Thyroid hormone-responsive genes in the heart are not as well described as the liver but could be determined using similar techniques as described by Feng et al. Many of the genes described to be affected in the heart are the same as described above for the liver. Common genes evaluated include mitochondrial sn-glycerol 3-phosphate dehydrogenase (mGPDH), and myosin heavy and light chains (Danzi et al., Thyroid 72 ⁇ :467-72 (2002)).
- high triglycerides may be reduced, which in turn may reduce risk/incidence of pancreatitis (prevention/treatment); reduce risk of major advanced cardiac event (MACE) (stroke, heart attack) via reduced triglycerides, Lp(a).
- MACE major advanced cardiac event
- Lp(a) stroke, heart attack
- a specific population with only increased Lp(a) may be identifed and preferentially treated.
- the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammalian tissue or a mammal for a period of time sufficient to yield a metabolic product thereof.
- Such products typically are identified by preparing a radio-labeled ⁇ e.g.
- C ⁇ or H ⁇ ) compound of the invention administering it in a detectable dose (e.g., greater than about 0.5 mg/kg) to a mammal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours), and isolating its conversion products from urine, blood or other biological samples.
- a detectable dose e.g., greater than about 0.5 mg/kg
- a mammal such as rat, mouse, guinea pig, monkey, or to man
- sufficient time for metabolism to occur typically about 30 seconds to 30 hours
- the metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites may be done in the same way as conventional drug metabolism studies well-known to those skilled in the art.
- the conversion products so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they
- compositions useful in the methods of the invention are provided.
- the pharmaceutical compositions of the invention may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form.
- the pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5.0 to about pH 8.0.
- the pharmaceutical compositions of the invention comprise a therapeutically or prophylactically effective amount of at least one compound of the present invention, together with one or more pharmaceutically acceptable excipients.
- the pharmaceutical compositions of the invention may comprise a combination of compounds of the present invention, or may include a second active ingredient useful in a method disclosed herein.
- Formulations of the present invention are most typically solids, liquid solutions, emulsions or suspensions, while inhaleable formulations for pulmonary administration are generally liquids or powders, with powder formulations being generally preferred.
- a preferred pharmaceutical composition of the invention may also be formulated as a lyophilized solid that is reconstituted with a physiologically compatible solvent prior to administration.
- Alternative pharmaceutical compositions of the invention may be formulated as syrups, creams, ointments, tablets, and the like.
- compositions of the invention can be administered to the subject via any drug delivery route known in the art.
- Specific exemplary administration routes include oral, ocular, rectal, buccal, topical, nasal, ophthalmic, subcutaneous, intramuscular, intraveneous (bolus and infusion), intracerebral, transdermal, and pulmonary.
- pharmaceutically acceptable excipient refers to an excipient for administration of a pharmaceutical agent, such as the compounds of the present invention.
- the term refers to any pharmaceutical excipient that may be administered without undue toxicity.
- Pharmaceutically acceptable excipients are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington's Pharmaceutical Sciences).
- Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles.
- Other exemplary excipients include antioxidants such as ascorbic acid; chelating agents such as EDTA; carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid; liquids such as oils, water, saline, glycerol and ethanol; wetting or emulsifying agents; pH buffering substances; and the like. Liposomes are also included within the definition of pharmaceutically acceptable excipients.
- compositions of the invention may be formulated in any form suitable for the intended method of administration.
- tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared.
- Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
- the therapeutically effective amount refers to an amount of a pharmaceutical composition of the invention to treat, ameliorate, or modulate an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect.
- the effect can be detected by, for example, assays of the present invention.
- the effect can also be the prevention of a disease or condition where the disease or condition is predicted for an individual or a high percentage of a population.
- the precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; the therapeutic or combination of therapeutics selected for administration, the protein half-life, the mRNA half-life and the protein localization. Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician.
- the therapeutically effective amount can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs.
- the animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
- Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED 5 o (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population).
- the dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, ED50/LD 50 .
- compositions that exhibit large therapeutic indices are preferred.
- the data obtained from cell culture assays and animal studies may be used in formulating a range of dosage for human use.
- the dosage contained in such compositions is preferably within a range of circulating concentrations that include an ED 5 o with little or no toxicity.
- the dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
- a prophylactic or therapeutic dose of a particular active ingredient of the invention in the acute or chronic management of a disease or condition will vary, however, with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
- the dose, and perhaps the dose frequency will also vary according to the age, body weight, and response of the individual patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art.
- the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response.
- the compounds of the invention are administered orally in a total daily dose of about 0.375 ⁇ g/kg/day to about 3.75 mg/kg/day. In another aspect the total daily dose is from about 3.75 ⁇ g/kg/day to about 0.375 mg/kg/day. In another aspect the total daily dose is from about 3.75 ⁇ g/kg/day to about 37.5 ⁇ g/kg/day. In another aspect the total daily dose is from about 3.75 ⁇ g/kg/day to about 60 ⁇ g/kg/day. In a further aspect the dose range is from 30 ⁇ g/kg/day to 3.0 mg/kg/day.
- the compounds of the invention are administered orally in a unit dose of about 0.375 ⁇ g/kg to about 3.75 mg/kg. In another aspect the unit dose is from about 3.75 ⁇ g/kg to about 0.375 mg/kg. In another aspect the unit dose is from about 3.75 ⁇ g/kg to about 37.5 ⁇ g/kg. In another aspect the unit dose is from about 3.75 ⁇ g/kg to about 60 ⁇ g/kg. In one aspect, the compounds of the invention are administered orally in a unit dose of about 0.188 ⁇ g/kg to about 1.88 mg/kg. In another aspect the unit dose is from about 1.88 ⁇ g/kg to about 0.188 mg/kg.
- the unit dose is from about 1.88 ⁇ g/kg to about 18.8 ⁇ g/kg. In another aspect the unit dose is from about 1.88 ⁇ g/kg to about 30 ⁇ g/kg. In one aspect, the compounds of the invention are administered orally in a unit dose of about 0.125 ⁇ g/kg to about 1.25 mg/kg. In another aspect the unit dose is from about 1.25 ⁇ g/kg to about 0.125 mg/kg. In another aspect the unit dose is from about 1.25 ⁇ g/kg to about 12.5 ⁇ g/kg. In another aspect the unit dose is from about 1.25 ⁇ g/kg to about 20 ⁇ g/kg. In one embodiment the unit dose is administered once a day. In another embodiment the unit dose is administered twice a day. In another embodiment the unit dose is administered three times a day. In another embodiment the unit dose is administered four times a day.
- Dose refers to the equivalent of the free acid.
- the use of controlled-release preparations to control the rate of release of the active ingredient may be preferred.
- the daily dose may be administered in multiple divided doses over the period of a day. Doses and dosing schedules may be adjusted to the form of the drug or form of delivery used. For example, different dosages and scheduling of doses may be used when the form of the drug is in a controlled release form or intravenous delivery is used with a liquid form.
- therapeutically effective amount encompasses the above described dosage amounts and dose frequency schedules. Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to treat or prevent such diseases, but insufficient to cause, or sufficient to reduce, adverse effects associated with conventional therapies are also encompassed by the above described dosage amounts and dose frequency schedules.
- the exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time, protein of interest half- life, R A of interest half-life, frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.
- any compound of the present invention with one or more other active ingredients useful in the methods described herein, including compounds in a unitary dosage form, or in separate dosage forms intended for simultaneous or sequential administration to a patient in need of treatment. When administered sequentially, the combination may be administered in two or more administrations. In an alternative embodiment, it is possible to administer one or more compounds of the present invention and one or more additional active ingredients by different routes.
- the compounds of the invention may be combined with one or more lipid lowering agents such as statins or cholesterol absorption inhibitors to treat patients with hyperlipidemia.
- lipid lowering agents such as statins or cholesterol absorption inhibitors
- such combination allows therapeutic effect at a reduced dose of one or more of the agents, improves lipid profile, or improves safety/therapeutic index of the therapy or one or more of the agents.
- the combination of active ingredients may be: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by any other combination therapy regimen known in the art.
- the methods of the invention may comprise administering or delivering the active ingredients sequentially, e.g., in separate solution, emulsion, suspension, tablets, pills or capsules, or by different injections in separate syringes.
- an effective dosage of each active ingredient is administered sequentially, i.e., serially
- simultaneous therapy effective dosages of two or more active ingredients are administered together.
- Various sequences of intermittent combination therapy may also be used.
- the compounds of the present invention can be administered in combination with other pharmaceutical agents that are used to lower serum cholesterol such as a cholesterol biosynthesis inhibitor or a cholesterol absorption inhibitor, especially a HMG-CoA reductase inhibitor, or a HMG-CoA synthase inhibitor, or a HMG-CoA reductase or synthase gene expression inhibitor, a cholesteryl ester transfer protein (CETP) inhibitor (e.g., torcetrapib), a bile acid sequesterant (e.g., cholestyramine (Questran®), colesevelam and colestipol (Colestid®)), or a bile acid reabsorption inhibitor (see, for example, U.S.
- a cholesterol biosynthesis inhibitor or a cholesterol absorption inhibitor especially a HMG-CoA reductase inhibitor, or a HMG-CoA synthase inhibitor, or a HMG-CoA reductase or synthase gene expression inhibitor
- CETP chol
- a cholesterol absorption inhibitor as described (e.g., ezetimibe, tiqueside, pamaqueside or see, e.g., in WO 0250027), a PPARalpha agonist, a mixed PPAR alpha/gamma agonist such as, for example, AZ 242 (Tesaglitazar, (S)-3-(4-[2-(4- methanesulfonyloxyphenyl)ethoxy]phenyl)-2-ethoxypropionic acid), BMS 298585 (N- [(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4- oxazolyl)ethoxy]phenyl]methyl]glycine) or as described in WO 99/628
- the HMG-CoA reductase inhibitor is from a class of therapeutics commonly called statins.
- statins include but are not limited to lovastatin (MEVACOR; see U.S. Pat. Nos. 4,231,938; 4,294,926; 4,319,039), simvastatin (ZOCOR; see U.S. Pat. Nos. 4,444,784; 4,450,171, 4,820,850; 4,916,239), pravastatin (PRAVACHOL; see U.S. Pat. Nos.
- Non-limiting examples of suitable bile acid sequestrants include cholestyramine (a styrene-divinylbenzene copolymer containing quaternary ammonium cationic groups capable of binding bile acids, such as QUESTRAN or QUESTRAN LIGHT cholestyramine which are available from Bristol-Myers Squibb), colestipol (a copolymer of diethyl enetriamine and l-chloro-2,3-epoxypropane, such as COLESTID tablets which are available from Pharmacia), colesevelam hydrochloride (such as WelChol Tablets (poly(allylamine hydrochloride) cross-linked with epichlorohydrin and alkylated with 1-bromodecane and (6-bromohexyl)-trimethylammonium bromide) which are available from Sankyo), water soluble derivatives such as 3,3-ioene, N- (cycloalkyl)al
- Suitable inorganic cholesterol sequestrants include bismuth salicylate plus montmorillonite clay, aluminum hydroxide and calcium carbonate antacids.
- a fibrate base compound is a medicament for inhibiting synthesis and secretion of triglycerides in the liver and activating lipoprotein lipase, thereby lowering the triglyceride level in the blood.
- examples include bezafibrate, beclofibrate, binifibrate, ciprofibrate, clinofibrate, clofibrate, clofibric acid, ethofibrate, fenofibrate, gemfibrozil, nicofibrate, pirifibrate, ronifibrate, simfibrate and theofibrate.
- Such an AC AT inhibitor includes, for example: a compound having the general formula (I) disclosed in WO 92/09561 [preferably FR-129169, of which the chemical name is N- (l,2-diphenylethyl)-2-(2-octyloxyphenyl)acetamide]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 8-510256 (WO 94/26702, U.S. Pat. No.
- CI-1011 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in EP 421441 (U.S. Pat. No.
- F-1394 ⁇ preferably F-1394, of which the chemical name is (lS,2S)-2-[3-(2,2-dimethylpropyl)-3-nonylureido]cyclohexan-l-yl 3-[(4R)-N-(2,2,5,5- tetramethyl-1,- 3-dioxane-4-carbonyl)amino]propionate, and in the present invention F- 1394 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof ⁇ ; a compound including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) 2000-500771 (WO 97/19918, U.S.
- T-2591 of which the chemical name is l-(3-t-butyl-2-hydroxy-5- methoxyphenyl)-3-(2-cyclohexylethyl)-3-(4-dimethylaminophenyl)urea, and in the present invention T-2591 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in WO 96/26948 ⁇ preferably FCE-28654, of which the chemical name is l-(2,6- diisopropylphenyl)-3-[(4R,5R)-4,5-dimethyl-2-(4-phosphonophenyl)-l,3-dioxolan-2- ylmethyl]urea, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof
- Pat. No. 5,475,130 [preferably HL-004, of which the chemical name is N-(2,6- diisopropylphenyl)-2-tetradecylthioacetamide]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 7-82232 (EP 718281) ⁇ preferably NTE-122, of which the chemical name is trans- l,4-bis[l -cyclohexyl-3-(4- dimethylaminophenyl)ureidomethyl]cyclohexane, and in the present invention NTE-122 includes pharmacologically acceptable salts of NTE-122 ⁇ ; a compound including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 10-510512 (WO 96/10559) ⁇ preferably FR-
- the ACAT inhibitor preferably is a compound selected from the group consisting of FR129169, CI-1011, F-1394, F-12511, T-2591, FCE-28654, K-10085, HL-004, NTE-122, FPv-186054, N-(l -octyl-5-carboxymethyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (hereinafter referred as compound A), and N-(l-pentyl-4,6- dimethylindolin-7-yl)-2,2-dimethylpropaneamide (hereinafter referred as compound B), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof.
- the ACAT inhibitor more preferably is a compound selected from the group consisting of CI- 1011, F-12511, N-(l-octyl-5-carboxymethyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (compound A), and N-(l-pentyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (compound B), including a pharmacologically acceptable salt/co- crystal, ester or prodrug thereof; most preferred is N-(l-octyl-5-carboxymethyl-4,6- dimethylindolin-7-yl)-2,2-dimethylpropaneamide (compound A).
- An angiotensin II receptor antagonist includes, for example, a biphenyl tetrazole compound or biphenylcarboxylic acid derivative such as: a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Sho 63-23868 (U.S. Pat. No.
- losartan of which the chemical name is 2-butyl-4-chloro- 1- [2 '-(lH-tetrazol-5-yl)biphenyl-4-ylmethyl]-lH-imidazol-5 -methanol, and in the present invention losartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof ⁇ ; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 4-506222 (WO 91/14679) ⁇ preferably irbesartan, of which the chemical name is 2-N-butyl-4-spirocyclopentane-l-[2'-(lH- tetrazol-5-yl)biphenyl-4-ylmethyl]-2-imidazoline-5-one, and in the present invention irbesart
- Pat. No. 5,196,444 ⁇ preferably candesartan, of which the chemical name is 1- (cyclohexyloxycarbonyloxy)ethyl 2-ethoxy- 1 - [2 ' -( 1 H-tetrazol-5 -yl)biphenyl-4-ylmethyl] - lH-benzimidazole-7-carboxylate, and in the present invention candesartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof (TCV-116 or the like), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof ⁇ ; a carboxylic acid derivative having the general formula (I), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 5-78328 (U.S.
- olmesartan includes carboxylic acid derivatives thereof, pharmacologically acceptable esters of the carboxylic acid derivatives (CS-866 or the like), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof ⁇ ; and a compound having the general formula (I), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 4-346978 (U.S.
- telmisartan of which the chemical name is 4'-[[2-n-propyl-4-methyl-6-(l- methylbenzimidazol-2-yl)-benzimidazol- 1 -yl]- methyl]biphenyl-2-carboxylate, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof ⁇ .
- the angiotensin II receptor antagonist preferably is losartan, irbesartan, valsartan, candesartan, olmesartan, or telmisartan; more preferred is losartan or olmesartan; and most preferred is olmesartan.
- combination therapy with compounds of this invention maybe useful in reducing the dosage of the second drug or agent (e.g., atorvastatin).
- the compounds of the present invention can be used in combination with an apolipoprotein B secretion inhibitor and/or microsomal triglyceride transfer protein (MTP) inhibitor.
- MTP microsomal triglyceride transfer protein
- HMG-CoA reductase inhibitor refers to a compound that inhibits the biotransformation of hydroxymethylglutaryl-coenzyme A to mevalonic acid as catalyzed by the enzyme HMG- CoA reductase. Such inhibition may be determined readily by one of skill in the art according to standard assays (e.g., Methods of Enzymology, 71 : 455-509 (1981); and the references cited therein). A variety of these compounds are described and referenced below. U.S.
- U.S. 4,231,938 discloses certain compounds isolated after cultivation of a microorganism belonging to the genus Aspergillus, such as lovastatin.
- U.S. 4,444,784 discloses synthetic derivatives of the aforementioned compounds, such as simvastatin.
- U.S. 4,739,073 discloses certain substituted indoles, such as fluvastatin.
- U.S. 4,346,227 discloses ML-236B derivatives, such as pravastatin.
- EP 491,226 teaches certain pyridyldihydroxyheptenoic acids, such as rivastatin.
- 4,647,576 discloses certain 6-[2-(substituted-pyrrol-l-yl)-alkyl]- pyran-2-ones such as atorvastatin.
- Other HMG-CoA reductase inhibitors will be known to those skilled in the art. Examples of currently or previously marketed products containing HMG-CoA reductase inhibitors include cerivastatin Na, rosuvastatin Ca, fluvastatin, atorvastatin, lovastatin, pravastatin Na and simvastatin.
- HMG-CoA synthase inhibitor refers to a compound that inhibits the biosynthesis of hydroxymethylglutaryl-coenzyme A from acetyl-coenzyme A and acetoacetyl-coenzyme A, catalyzed by the enzyme HMG- CoA synthase. Such inhibition may be determined readily by one of skill in the art according to standard assays (e.g., Methods of Enzymology 35: 155-160 (1975); and Methods of Enzymology, 110: 19-26 (1985); and the references cited therein). A variety of these compounds are described and referenced below.
- U.S. 5,120,729 discloses certain beta-lactam derivatives.
- U.S. 5,064,856 discloses certain spiro-lactone derivatives prepared by culturing the microorganism MF5253.
- U.S. 4,847,271 discloses certain oxetane compounds such as 1 l-(3-hydroxymethyl-4-oxo-2-oxetayl)- 3,5,7-trimethyl-2,4-undecadienoic acid derivatives.
- Other HMG-CoA synthase inhibitors useful in the methods, compositions and kits of the present invention will be known to those skilled in the art.
- Any compound that decreases HMG-CoA reductase gene expression may be used as an additional compound in the combination therapy aspect of this invention.
- These agents may be HMG-CoA reductase transcription inhibitors that block the transcription of DNA or translation inhibitors that prevent translation of mRNA coding for HMG-CoA reductase into protein.
- Such inhibitors may either affect transcription or translation directly, or may be biotransformed into compounds that have the aforementioned attributes by one or more enzymes in the cholesterol biosynthetic cascade or may lead to the accumulation of an isoprene metabolite that has the aforementioned activities.
- Such regulation is readily determined by those skilled in the art according to standard assays (Methods of Enzymology, 110: 9-19 (1985)).
- CETP inhibitor refers to compounds that inhibit the cholesteryl ester transfer protein (CETP) mediated transport of various cholesteryl esters and triglycerides from HDL to LDL and VLDL. A variety of these compounds are described and referenced below; however, other CETP inhibitors will be known to those skilled in the art.
- CETP inhibitors U.S. 5,512,548 discloses certain polypeptide derivatives having activity as CETP inhibitors, while certain CETP- inhibitory rosenonolactone derivatives and phosphate-containing analogs of cholesteryl ester are disclosed in J. Antibiot., 49(8): 815-816 (1996), and Bioorg. Med. Chem. Lett., 6: 1951-1954 (1996), respectively.
- ACAT inhibitor refers to a compound that inhibits the intracellular esterification of dietary cholesterol by the enzyme acyl CoA: cholesterol acyltransferase. Such inhibition may be determined readily by one of skill in the art according to standard assays, such as the method of Heider et al. described in Journal of Lipid Research, 24:1127 (1983). A variety of these compounds are described and referenced below; however, other ACAT inhibitors will be known to those skilled in the art.
- U.S. 5,510,379 discloses certain carboxysulfonates, while WO 96/26948 and WO 96/10559 both disclose urea derivatives having ACAT inhibitory activity.
- squalene synthetase inhibitor refers to compounds that inhibit the condensation of two molecules of farnesylpyrophosphate to form squalene, a reaction that is catalyzed by the enzyme squalene synthetase. Such inhibition is readily determined by those skilled in the art according to standard methodology (Methods of Enzymology 15:393-454 (1969); and Methods of Enzymology 110: 359-373 (1985); and references cited therein). A summary of squalene synthetase inhibitors has been compiled in Curr. Op.
- EP 0 567 026 Al discloses certain 4,1-benzoxazepine derivatives as squalene synthetase inhibitors and their use in the treatment of hypercholesterolemia and as fungicides.
- EP 0 645 378 Al discloses certain seven- or eight-membered heterocycles as squalene synthetase inhibitors and their use in the treatment and prevention of hypercholesterolemia and fungal infections.
- EP 0 645 377 Al discloses certain benzoxazepine derivatives as squalene synthetase inhibitors useful for the treatment of hypercholesterolemia or coronary sclerosis.
- EP 0 611 749 Al discloses certain substituted amic acid derivatives useful for the treatment of arteriosclerosis.
- EP 0 705 607 A2 discloses certain condensed seven- or eight-membered heterocyclic compounds useful as antihypertriglyceridemic agents.
- WO 96/09827 discloses certain combinations of cholesterol absorption inhibitors and cholesterol biosynthesis inhibitors including benzoxazepine derivatives and benzothiazepine derivatives.
- EP 0 701 725 Al discloses a process for preparing certain optically-active compounds, including benzoxazepine derivatives, having plasma cholesterol and triglyceride lowering activities.
- Other compounds that are currently or previously marketed for hyperlipidemia, including hypercholesterolemia, and which are intended to help prevent or treat atherosclerosis include bile acid sequestrants, such as colestipol HC1 and cholestyramine; and fibric acid derivatives, such as clofibrate, fenofibrate, and gemfibrozil. These compounds can also be used in combination with a compound of the present invention.
- the compounds of the present invention be administered with a lipase inhibitor and/or a glucosidase inhibitor, which are typically used in the treatment of conditions resulting from the presence of excess triglycerides, free fatty acids, cholesterol, cholesterol esters or glucose including, inter alia, obesity, hyperlipidemia, hyperlipoproteinemia, Syndrome X, and the like.
- any lipase inhibitor or glucosidase inhibitor may be employed.
- lipase inhibitors comprise gastric or pancreatic lipase inhibitors.
- glucosidase inhibitors comprise amylase inhibitors. Examples of glucosidase inhibitors are those inhibitors selected from the group consisting of acarbose, adiposine, voglibose, miglitol, emiglitate, camiglibose, tendamistate, trestatin, pradimicin-Q and salbostatin.
- amylase inhibitors include tendamistat and the various cyclic peptides related thereto disclosed in U.S. Pat. No. 4,451,455, AI-3688 and the various cyclic polypeptides related thereto disclosed in U.S. Pat. No. 4,623,714, and trestatin, consisting of a mixture of trestatin A, trestatin B and trestatin C and the various trehalose-containing aminosugars related thereto disclosed in U.S. Pat. No. 4,273,765.
- a lipase inhibitor is a compound that inhibits the metabolic cleavage of dietary triglycerides into free fatty acids and monoglycerides.
- lipolysis occurs via a two-step process that involves acylation of an activated serine moiety of the lipase enzyme. This leads to the production of a fatty acid-lipase hemiacetal intermediate, which is then cleaved to release a diglyceride. Following further deacylation, the lipase-fatty acid intermediate is cleaved, resulting in free lipase, a monoglyceride and a fatty acid.
- bile acid phospholipid micelles which are subsequently absorbed at the level of the brush border of the small intestine.
- the micelles eventually enter the peripheral circulation as chylomicrons. Accordingly, compounds, including lipase inhibitors that selectively limit or inhibit the absorption of ingested fat precursors are useful in the treatment of conditions including obesity, hyperlipidemia, hyperlipoproteinemia, Syndrome X, and the like.
- Pancreatic lipase mediates the metabolic cleavage of fatty acids from triglycerides at the 1- and 3 -carbon positions.
- the primary site of the metabolism of ingested fats is in the duodenum and proximal jejunum by pancreatic lipase, which is usually secreted in vast excess of the amounts necessary for the breakdown of fats in the upper small intestine.
- pancreatic lipase is the primary enzyme required for the absorption of dietary triglycerides, inhibitors have utility in the treatment of obesity and the other related conditions.
- Gastric lipase is an immunologically distinct lipase that is responsible for approximately 10 to 40% of the digestion of dietary fats. Gastric lipase is secreted in response to mechanical stimulation, ingestion of food, the presence of a fatty meal or by sympathetic agents. Gastric lipolysis of ingested fats is of physiological importance in the provision of fatty acids needed to trigger pancreatic lipase activity in the intestine and is also of importance for fat absorption in a variety of physiological and pathological conditions associated with pancreatic insufficiency. See, for example, C. K. Abrams, et al., Gastroenterology 92: 125 (1987).
- lipase inhibitors are known to one of ordinary skill in the art. However, in the practice of the methods, pharmaceutical compositions, and kits of the instant invention, generally lipase inhibitors are those inhibitors that are selected from the group consisting of lipstatin, tetrahydrolipstatin (orlistat), FL-386, WAY-121898, Bay-N-3176, valilactone, esterastin, ebelactone A, ebelactone B and RHC 80267.
- pancreatic lipase inhibitor FL-386 l-[4-(2-methylpropyl)cyclohexyl]-2- [(phenylsulfonyl)oxy]-ethanone, and the variously substituted sulfonate derivatives related thereto, are disclosed in U.S. 4,452,813.
- pancreatic lipase inhibitor WAY-121898 4-phenoxyphenyl-4- methylpiperidin-l-yl-carboxylate, and the various carbamate esters and pharmaceutically acceptable salts related thereto, are disclosed in U.S. 5,512,565; 5,391,571 and 5,602,151.
- the lipase inhibitor Bay-N-3176, N-3-trifluoromethylphenyl-N'-3-chloro- 4-trifluoromethylphenylurea, and the various urea derivatives related thereto, are disclosed in U.S. 4,405,644.
- the pancreatic lipase inhibitor valilactone, and a process for the preparation thereof by the microbial cultivation of Actinomycetes strain MG147— CF2, are disclosed in Kitahara, et al, J. Antibiotics, 40(11): 1647-50 (1987).
- pancreatic lipase inhibitors ebelactone A and ebelactone B and a process for the preparation thereof by the microbial cultivation of Actinomycetes strain MG7-G1, are disclosed in Umezawa, et al, J. Antibiotics, 33, 1594-1596 (1980).
- the use of ebelactones A and B in the suppression of monoglyceride formation is disclosed in Japanese Kokai 08-143457, published Jun. 4, 1996.
- the lipase inhibitor RHC 80267, cyclo-0,0'-[(l,6-hexanediyl)-bis- (iminocarbonyl)]dioxime, and the various bis(iminocarbonyl)dioximes related thereto may be prepared as described in Petersen et al, Liebig's Annalen, 562: 205-29 (1949).
- RHC 80267 to inhibit the activity of myocardial lipoprotein lipase is disclosed in Carroll et al, Lipids, 27 305-7 (1992) and Chuang et al, J. Mol Cell Cardiol, 22: 1009-16 (1990).
- the compounds of Formula I can be used in combination with an anti-obesity agent.
- the anti-obesity agent in one aspect is selected from the group consisting of a p 3 -adrenergic receptor agonist, a cholecystokinin-A agonist, a monoamine reuptake inhibitor, a sympathomimetic agent, a serotonergic agent, a dopamine agonist, a melanocyte-stimulating hormone receptor agonist or mimetic, a melanocyte-stimulating hormone receptor analog, a cannabinoid receptor antagonist, a melanin concentrating hormone antagonist, leptin, a leptin analog, a leptin receptor agonist, a galanin antagonist, a lipase inhibitor, a bombesin agonist, a neuropeptide-Y antagonist, a thyromimetic agent, dehydroepiandrosterone or an analog thereof, a gluco
- the anti-obesity agents comprise those compounds selected from the group consisting of sibutramine, fenfluramine, dexfenfluramine, bromocriptine, phentermine, ephedrine, leptin, phenylpropanolamine pseudoephedrine, ⁇ 4-[2-(2-[6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl ⁇ acetic acid, ⁇ 4 ⁇ 2-(2-[6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl ⁇ benzoic acid, ⁇ 4-[2-(2 ⁇ 6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl ⁇
- Representative agents that can be used to treat diabetes in combination with a compound of the present invention include insulin and insulin analogs (e.g., LysPro insulin); GLP-1 (7-37) (insulinotropin) and GLP-1 (7-36)— N3 ⁇ 4.
- insulin and insulin analogs e.g., LysPro insulin
- GLP-1 (7-37) insulinotropin
- Agents that enhance insulin secretion e.g., chlorpropamide, glibenclamide, tolbutamide, tolazamide, acetohexamide, glypizide, glimepiride, repaglinide, nateglinide, meglitinide; biguanides: metformin, phenformin, buformin; A2-antagonists and imidazolines: midaglizole, isaglidole, deriglidole, idazoxan, efaroxan, fluparoxan; other insulin secretagogues linogliride, A-4166; glitazones: ciglitazone, pioglitazone, englitazone, troglitazone, darglitazone, BRL49653; fatty acid oxidation inhibitors: clomoxir, etomoxir; a-glucosidase inhibitors: acarbose, miglitol,
- the compounds of the present invention can be used in combination with one or more aldose reductase inhibitors, DPP-IV inhibitor, glycogen phosphorylase inhibitors, sorbitol dehydrogenase inhibitors, NHE-1 inhibitors and/or glucocorticoid receptor antagonists.
- Any compound having activity as a fructose -1,6-bisphosphatase (FBPase) inhibitor can serve as the second compound in the combination therapy aspect of the instant invention (e.g., 2-Amino-5-isobutyl-4- ⁇ 2-[5-(N,N'-bis((S)-l- ethoxycarbonyl)ethyl)phosphonamido]furanyl ⁇ thiazoles).
- FBPase is a key regulatory enzyme in gluconeogenesis, the metabolic pathway by which the liver synthesizes glucose from 3 -carbon precursors.
- FBPase inhibitor refers to compounds that inhibit FBPase enzyme activity and thereby block the conversion of fructose -1,6- bisphosphate, the substrate of the enzyme, to fructose 6-phosphate. FBPase inhibition can be determined directly at the enzyme level by those skilled in the art according to standard methodology (e.g., Gidh-Jain M, Zhang Y, van Poelje PD et al., J Biol Chem. 1994, 269(44): 27732-8).
- FBPase inhibition can be assessed according to standard methodology by measuring the inhibition of glucose production by isolated hepatocytes or in a perfused liver, or by measuring blood glucose lowering in normal or diabetic animals (e.g., Vincent MF, Erion MD, Gruber HE, Van den Berghe, Diabetologia. 1996, 39(10): 1148-55.; Vincent MF, Marangos PJ, Gruber HE, Van den Berghe G, Diabetes 1991 40(10): 1259-66).
- in vivo metabolic activation of a compound may be required to generate the FBPase inhibitor.
- This class of compounds may be inactive in the enzyme inhibition screen, may or may not be active in hepatocytes, but is active in vivo as evidenced by glucose lowering in the normal, fasted rat and/or in animal models of diabetes.
- FBPase inhibitors A variety of FBPase inhibitors are described and referenced below; however, other FBPase inhibitors will be known to those skilled in the art. Gruber et al. U.S. Patent No. 5,658,889 described the use of inhibitors of the AMP site of FBPase to treat diabetes; WO 98/39344 and US 6,284,748 describe purine inhibitors; WO 98/39343 and US 6,110,903 describe benzothiazole inhibitors to treat diabetes; WO 98/39342 and US 6,054,587 describe indole inhibitors to treat diabetes; and WO 00/14095 and US 6,489476 describe heteroaromatic phosphonate inhibitors to treat diabetes. Other FBPase inhibitors are described in Wright SW, Carlo AA, Carty MD et al., J Med Chem. 2002 45(18):3865-77 and WO 99/47549.
- the compounds of the present invention can also be used in combination with sulfonylureas such as amaryl, alyburide, glucotrol, chlorpropamide, diabinese, tolazamide, tolinase, acetohexamide, glipizide, tolbutamide, orinase, glimepiride, DiaBeta, micronase, glibenclamide, and gliclazide.
- sulfonylureas such as amaryl, alyburide, glucotrol, chlorpropamide, diabinese, tolazamide, tolinase, acetohexamide, glipizide, tolbutamide, orinase, glimepiride, DiaBeta, micronase, glibenclamide, and gliclazide.
- the compounds of the present invention can also be used in combination with antihypertensive agents. Any anti-hypertensive agent can be used as the second agent in such combinations.
- antihypertensive agents include calcium channel blockers, such as Cardizem, Adalat, Calan, Cardene, Covera, Dilacor, DynaCirc, Procardia XL, Sular, Tiazac, Vascor, Verelan, Isoptin, Nimotop, Norvasc, and Plendil; angiotensin converting enzyme (ACE) inhibitors, such as Accupril, Altace, Captopril, Lotensin, Mavik, Monopril, Prinivil, Univasc, Vasotec and Zestril.
- ACE angiotensin converting enzyme
- Step a A solution of HBr in acetic acid (33%, 25.12 mL, 140.1 mmol) was added to a slightly heterogeneous solution of 4-benzyloxy-benzyl alcohol in acetic acid (50 mL) at 0 °C. After stirring at 0 °C for 1 h the thick slurry was poured into a mixture of ice and water. The precipitate that formed was collected by filtration, rinsed with water and dried to give crude benzyl bromide as a white solid (11.10 g).
- Step b Diethoxymethylphosphine (11.15 mL, 80.10 mmol) was added to a solution of crude benzyl bromide from step a (11.10 g) in DMF. The clear reaction mixture was heated at 110 °C for 4 h. The cooled solution was partitioned between EtOAc and water and the layers separated. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with water (3X), dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure to give crude ethyl phosphinate (12.1 g).
- Step c Pd/C (10%, 1.28 g) was added to a degassed solution of crude phosphinate in methanol. After stirring at rt under 1 atm of hydrogen for 16 hours, the catalyst was removed by filtration over Celite and the pad was rinsed with methanol. The combined filtrates were concentrated under reduced pressure and the residue was partitioned between water and EtOAc. The layers were separated and the aqueous phase was extracted with CH 2 C1 2 (5X). The combined organic extracts were dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure to give crude phenol (8.80 g).
- Step d Ca 2 C0 3 (11.21 g, 112.0 mmol) followed by benzyltrimethylammonium tribromide (30.03 g, 77.0 mmol) were added to a solution of phenol (7.5 g, 35.0 mmol) in methanol (60 mL) at rt. After stirring at rt for 5 h, the heterogeneous mixture was filtered through Celite and the pad rinsed with methanol. The combined filtrates were concentrated under reduced pressure. The residue was partitioned between CH 2 C1 2 and water. The layers were separated and the organics were washed with water (2X), dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure. The residue was purified by column
- Step a Fuming nitric acid (7.82 mL, 184.9 mmol) was added to neat acetic anhydride (20 mL) at -20 °C. Iodine (7.79 g, 30.69 mmol) was then added followed by TFA (14.25 mL). After stirring at -20 °C for 15 minutes, the cold bath was removed and the reaction mixture was stirred at rt. After 2 h at rt, all the iodine was consumed and the orange vapors were blown away under a stream of nitrogen. The black reaction mixture was concentrated under reduced pressure and the residue was taken up in acetic anhydride (60 mL) and the black solution cooled to -20 °C.
- Step b A solution of ethyl 3,5-dibromo-4-hydroxy-benzyl-methylphosphinate
- Step c A solution of 1 , 1-dichloromethyl-methyl ether (5.3 mL, 58.56 mmol) and tin tetrachloride (20.58 mL, 175.68 mmol) in CH 2 CI 2 (20 mL) was added to a solution of diphenyl ether (14.0 g, 29.28 mmol) in CH 2 C1 2 (60 mL) at -78 °C. Upon completion of the addition, the temperature was raised to 0 °C. After stirring at 0 °C for 4 h, 2 M HC1 (80 mL) was added and the reaction mixture was partitioned between EtOAc and water. The layers were separated and the organics were washed with a saturated solution of NaHC0 3 , brine, dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure to give a brown foam (13.20 g, 89%).
- Step d NaH 2 P0 4 » H 2 0 (47.75 g, 346.1 mmol) followed by sodium chlorite (80%, 57.18, 506 mmol) were added to a solution of aldehyde (13.10 g, 26.62 mmol) in t-butanol (120 mL), 2-methyl-2-butene (60 mL) and water (120 mL) at rt. The heterogeneous mixture becomes a clear solution after 20 minutes. After stirring at rt for 4 h, the reaction mixture was partitioned between CH 2 CI 2 and water, and the layers separated. The aqueous phase was extracted with CH 2 CI 2 and the combined organic extracts were washed with 0.3 N HC1, water (2X), dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure (15.5g).
- Step e Neat BBr (1 1.44 mL, 118.76 mmol) was added to a solution of anisole (15.5 g, 29.69 mmol) in CH 2 CI 2 (150 mL) at -50 °C. The amber reaction mixture was warmed to 0 °C and stirred for 4 h, then warmed to rt and stirred 4h. Water and ice were carefully added and most of the CH 2 CI 2 was removed under reduced pressure. The residual mixture was partitioned between EtOAc and water and the layers separated. The organic layer was washed with water (3X), dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure to give the desired carboxylic acid intermediate (15.10 g).
- Step a A 50 L, 4-neck flask was equipped with an overhead stirrer, temperature probe, and cooling bath. The flask was charged with 3,5-dimethylphenol (2497 g, 20.5 mol), water (14 L), and 50% (wt/wt) aqueous sodium hydroxide (1636 g, 20.5 mol). The mixture was stirred 1.5 h to complete dissolution. The mixture was cooled to 4 °C using an ice/water bath (note 2). Formaldehyde (37% aqueous solution, 1496 g, 18.5 mol) was added in one portion. The mixture was stirred cold throughout the daytime and allowed to warm slowly overnight.
- Step b A 12 L flask was equipped with an overhead stirrer, heating mantle, temperature probe, condenser with a nitrogen bubbler on the outlet, and an addition funnel.
- the flask was charged with paraformaldehyde (488 g, 16.3 mol), potassium carbonate (94 g, 0.7 mol), and 2-propanol (4.5 L). The mixture was heated to 50 °C then the heating mantle was turned off. Diisopropylphosphite (2260 g, 13.6 mol) was added from the addition funnel at a rate that would maintain the temperature at 50-60 °C. The mixture was cooled to 35 °C over 2 h then filtered through Celite. The pad was washed with 2-propanol (2 x 200 mL). The mixture was concentrated under reduced pressure. The colorless residual oil (2950 g) was dissolved in dichloromethane (9 L).
- Step c A 22 L, four-neck flask was equipped with an overhead stirrer, cooling bath, temperature probe, 2 L addition funnel, and a nitrogen bubbler. The flask was charged with diisopropyl hydroxymethylphosphonate (1408g, 6.67 mol), triethylamine (1350 g, 13.34 mol), and dichloromethane (4 L). The resulting solution was cooled to 5 °C using an ice/water bath. A solution of /?-toluenesulfonyl chloride (1335 g, 7.0 mol) in dichloromethane (10 L) was added from the addition funnel at a rate that would keep the temperature below 10 °C.
- the mixture was stirred in an ice bath for 1.75 h then at ambient temperature ( ⁇ 20 °C) for 15 h.
- the reaction mixture was washed with 1 M HC1 (6 L) followed by saturated aqueous NaHC0 3 (6 L).
- the organic layer was dried over MgS0 4 (400 g) and filtered.
- the filtrate was concentrated under reduced pressure to give the tosylate as a yellow oil weighing 2111 g (90% yield).
- Step d A 22 L, four-neck flask was equipped with an overhead stirrer, temperature probe, heating mantle, and condenser with a nitrogen bubbler on the outlet.
- the flask was charged with the tosylate (2054 g, 5.45 mol), DMSO (2 L), the benzyl alcohol from step a (928 g, 5.62 mol), cesium carbonate (2841 g, 8.72 mol), and DMSO (2.5 L).
- the mixture was heated to 55 °C over 2 h and maintained at 50-60 °C for 6 h.
- the reaction mixture was cooled to 20 °C overnight then cooled to 5 °C (ice/water bath).
- Step a TFA (58.6 mL, 0.789 mol) was added to an heterogeneous mixture of benzyl alcohol from intermediate 3/step a (20 g, 0.132 mol) and anisole (42.8 mL, 0.395 mol) in CH 2 CI 2 (200 mL) at -20 °C.
- the ice bath was removed and the reaction mixture was allowed to warm to rt over 1 h.
- the clear solution was poured into a mixture of ice (200 g) and a concentrated solution of NH 4 OH (60 mL). After stirring for 5 minutes, the mixture was extracted with CH 2 CI 2 (2X).
- Step b Trifluoromethanesulfonic anhydride was added to a solution of phenol (16.48 g, 68.1 mmol) and pyridine (10.95 mL), 136.2 mmol) in CH 2 C1 2 (680 mL) at 0 °C. After 15 minutes at 0 °C, the ice bath was removed and the reaction allowed to warm to rt. After stirring at rt for 1 h, the orange solution was washed with 10%> hydrochloric acid, water, dried (Na 2 S0 4 ), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (5% EtOAc in hexanes) to give the aryl triflate as a clear oil (19.76 g, 78%).
- Step c A mixture of aryl triflate (19.76 g, 52.8 mmol), Pd(OAc) 2 (1.18 g, 5.28 mmol), diphenylphosphinopropane (2.18 g, 5.28 mmol), triethylamine (14.7 mL, 105.7 mmol) in methanol (50 mL) and DMF (100 mL) was charged in a bomb. The bomb was sealed, evacuated and charged with 60 psi of CO. After heating the bomb at 90 °C for 16 h, the cooled bomb was opened. The black reaction mixture was filtered over Celite and rinsed with EtOAc. The combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (5% EtOAc in hexanes) to give the methyl benzoate as a yellow solid (6.65 g, 44%).
- Step d L1AIH 4 (1.87 g, 49.2 mmol) was added in small portion to a solution of methyl benzoate (6.65 g, 23.4 mmol) in THF (230 mL) at 0 °C. After stirring at 0 °C for 90 minutes, the grey slurry was quenched carefully with 1 N NaoH. The solids were filtered off over Celite and the pad was rinsed with EtOAc. The combined filtrates were extracted with EtOAc (2X). The combined organic extracts were washed with brine, dried (Na 2 S0 4 ), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (20 to 40% EtOAc in hexanes) to give the benzylic alcohol as a white solid (4.7 g, 78%).
- Step e A mixture of CBr 4 (9.13 g, 27.5 mmol) and PPh 3 (7.22 g, 27.5 mmol) in THF (160 mL) was stirred at rt for 5 minutes. A solution of the benzylic alcohol (4.7 g, 18.4 mmol) in THF (24 mL) was added to the stirring solution of CBr 4 and PPh 3 . The off white heterogeneous mixture was stirred at rt for 90 minutes. The solids were filtered off over Celite and rinsed with ether. The combined organic extracts were concentrated under reduced pressure and the residue was purified by column chromatography to give the benzyl bromide as a white solid (4.68 g, 83%).
- Step a Methanesulfonic acid (0.46 mL, 7.2 mmol) was added to a solution of intermediate 3 (990 mg, 3 mmol) and methyl 3-(2-hydroxyphenyl)-propanoate (810 mg, 4.5 mmol) in CH 2 CI 2 (30 mL) at -40 °C. The bath was removed and the reaction mixture was allowed to warm to 0 °C. Upon reaching 0 °C, water was added and the layers were separated. The aqueous phase was extracted with CH 2 CI 2 and the combined organics were washed with water, dried (Na 2 S0 4 ), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (10% to 25 %> EtOAc in CH 2 CI 2 ) to give the lactone (500 mg, 34%>) and the methyl carboxylate (350 mg, 24%>).
- Step b Bromotrimethylsilane (1 mL, 3.6 mmol) was added to a solution of methyl carboxylate (350 mg, 0.71 mmol) in CH 2 CI 2 (20 mL) at rt. After stirring at rt for 16 h, the reaction mixture was concentrated under reduced pressure. The residue was taken up in EtOAc and washed with water, brine, dried (Na 2 S0 4 ), filtered, and concentrated under reduced pressure to give the lactone phosphonic acid.
- Step c Asolution of HC1 in dioxane (4 N, 0.25 mL, 1 mmol) was added to a solution of lactone phosphonic acid (100 mg, 0.26 mmol) in ethanol (4 mL) at rt. After stirring at rt for 1 h, the reaction mixture was concentrated under reduced pressure and azeotrpoed with ethanol. The residue was taken up in ether and sonicated. The solid was collected by filtration, rinsed with ether and dried to give the ethyl carboxylate (90 mg, 82%); 1H
- Step a A solution of NaCN (288 mg, 5.88 mmol) in water (10 mL) was added to a solution of benzaldehyde intermediate (intermediate in the synthesis of intermediate 4, 1.10 g, 2.94 mmol), ethyl chloroformate (0.3 mL, 3.09 mmol) and tetrabutyl ammonium bromide (47 mg, 0.15 mmol) in CH 2 C1 2 (10 mL) at rt. After stirring at rt for 16 h, the biphasic reaction mixture was partitioned between CH 2 C1 2 and water, and the layers separated.
- Step b Pd/C (400 mg) was added to a solution of (ethoxycarbonyloxy)-benzyl-cyanide (1.10 g, 2.32 mmol) in ethanol (35 mL) in a pressure vessel.
- the vessel was sealed, degassed and pressurize with 50 psi of hydrogen.
- the sealed vessel was heated at 70 °C for 20 h.
- the cooled bomb was opened, and the black reaction mixture was filtered over Celite and rinsed with EtOAc.
- the combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (5% EtOAc in hexanes) to give the benzyl cyanide (1.10 g, 100%).
- Step c A solution of potassium hydroxide (567 mg, 10.1 mmol) in water (4 mL) was added to a solution of benzyl cyanide (391 mg, 1.01 mmol) in ethanol (8 mL) and the resulting solution was heated at reflux for 16 h. The cooled reaction mixture was concentrated under reduced pressure. The residue was taken up in water and washed with ether. The aqueous phase was acidified to pH 1 with cone hydrochloric acid. The white precipitate was collected by filtration, rinsed with water and dried to give the diacid (331 mg, 87%).
- Step d Neat boron tribromide (0.51 mL, 5.27 mmol) was added over 2 min to a suspension of diacid (331 mg, 0.88 mmol) in CH 2 CI 2 at rt. After stirring at rt for 18 h, the reaction mixture was poured into ice/water (50 mL) and the pH adjusted to 14 with solid NaOH. The layers were separated and the organic phase was extracted with 1 N NaOH. The combined aqueous extracts were acidified to pH 1 with cone HC1.
- Step a A solution of 2-methoxy-phenyl-magnesium bromide (1 M in THF, 32 mL, 32mmol) was added to a solution of methyl 3-carboxaldehyde-benzoate (5.0 g, 30.5 mmol) in CH 2 C1 2 (30 mL) at rt over 90 minutes. Upon completion of the addition, the reaction mixture was quenched with saturated aqueous solution of NH 4 C1 and diluted with EtOAc. The layers were separated and the organics were washed with water then brine, dried (Na 2 S0 4 ), filtered, concentrated to dryness and purified by column
- Step b TFA (4.53 mL, 61 mmol) was added to a solution of carbinol from step a (9.52 g, 30.5 mmol) and triethylsilane (9.85 mL, 61 mmol) in CH 2 C1 2 (300 mL) at rt. After stirring at rt for 4 h, water (100 mL) was added and the reaction mixture was stirred vigourously for 5 minutes. The layers were separated and the organics were dried
- Step c Fuming nitric acid (1.17 mL, 27.6 mmol) was added to neat acetic anhydride (2.97 mL) at -20 °C. Iodine (1.15 g, 4.59 mmol) was then added followed by TFA (2.13 mL). After stirring at -20 °C for 15 minutes, the cold bath was removed and the reaction mixture was stirred at rt. After 2 h at rt, all the iodine was consumed and the orange vapors were blown away under a stream of nitrogen. The black reaction mixture was concentrated under reduced pressure and the residue was taken up in acetic anhydride (8.95 mL) and the black solution cooled to -20 °C. A solution of methyl 3-(2- methoxybenzyl)benzoate (7.08 g, 27.6 mmol) in acetic anhydride (2.97 mL) and TFA
- the solvents were decanted and the residue was washed with hexanes.
- Step d A solution of ethyl 3,5-dibromo-4-hydroxy-benzyl-methylphosphinate (372 mg, 1 mmol) and triethylamine (0.15 mL, 1.1 mmol) in CH 2 CI 2 (4 mL) was added to a suspension of iodonium salt from step c (850 mg, 1.3 mmol) and copper powder (127 mg, 2 mmol) in CH 2 CI 2 (8 mL) at 0 °C. The ice bath was removed and the flask was covered with aluminum foil. After stirring at rt for 18 h, the reaction mixture was filtered over Celite and the pad rinsed with CH 2 CI 2 .
- Step e Neat BBr 3 (0.67 mL, 6.96 mmol) was added to a solution of compound from step d (436 mg, 0.696 mmol) in CH 2 CI 2 (15 mL) at 0 °C. The ice bath was removed and the amber reaction mixture was stirred at rt for 22 h. Water and ice were carefully added and the layers were separated. The aqueous layer was extracted with EtOAc. The combined organic extracts were dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure. The brown solid was taken up in EtOH (15 mL) and a solution of KOH (195 mg, 3.48 mmol) in water (15 mL) was added.
- Step a A mixture of palladium acetate and X-phos in THF (3 mL) was degassed and stirred at rt. After 30 minutes, this solution was added to an heterogeneous mixture of methyl 3-bromo-benzoate (6.85g, 31.85 mmol), 2-methoxyphenyl boronic acid (7.26 g, 47.78 mmol) and ⁇ 3 ⁇ 0 4 ⁇ 2 0 (21.98 g, 95.56 mmol) in THF (32 mL). After stirring at rt for 18 h, the insolubles were removed by filtration through Celite and rinsed with EtOAc.
- Step a EDCI (90 mg, 0.47 mmol) was added to an heterogeneous mixture of methyl glycine hydrochloride (99 mg, 0.79 mmol), carboxylic acid intermediate 2 (200 mg, 0.39 mmol), HOBT (72 mg, 0.47 mmol) and diisopropylethylamine (0.26 mL, 1.50 mmol) in CH 2 C1 2 (6 mL) at rt. After stirring at rt for 16 h, the clear reaction mixture was partitioned between CH 2 C1 2 and 0.5 N HCl. The layers were separated and the organics were washed with 0.5 N HCl, a saturated solution of NaHC0 3 (2X), dried (Na 2 S0 4 ), filtered, concentrated under reduced pressure and the residue was purified by column
- Step a Thionyl chloride (0.05 mL, 0.71 mmol) was added to a solution of carboxylic acid intermediate 2 (120 mg, 0.236 mmol) in chcloroform (4 mL). The reaction mixture was refluxed for 2 h, cooled to rt, concentrated under reduced pressure, azeotroped with CH 2 C1 2 (2X) and dried under high vacuum. The residue was taken up in CH 2 C1 2 (4 mL) and a solution of ethyl glycine hydrochloride (40 mg, 0.28 mmol) and triethylamine (0.13 mL, 0.94 mmol) in CH 2 C1 2 (4 mL) was added at rt.
- ethyl glycine hydrochloride 40 mg, 0.28 mmol
- triethylamine (0.13 mL, 0.94 mmol
- Ci 9 H 2 oBr 2 N07P Calcd: C:40.38, H:3.57, N:2.48; Found: C:40.09, H:3.45, N:2.30.
- Ci 8 Hi 8 Br 2 N0 7 P +1 Anal for Ci 8 Hi 8 Br 2 N0 7 P; Calcd: C:39.23, H:3.29, N:2.54; Found: C:38.95, H:3.19, N:2.41.
- Step a Diisopropylethylamine (0.35 mL, 2.02 mmol) was added dropwise to a suspension of ethyl alanine hydrochloride (163 mg, 1.06 mmol) in CH 2 CI 2 (5.3 mL) at 0 °C. After stirring at 0 °C for 10 minutes, the carboxylic acid intermediate 4 (200 mg, 0.53 mmol), HO AT (89 mg, 0.64 mmol) and EDCI (122 mg, 0.64 mmol) was added to the clear solution and the ice bath was removed. After stirring at rt for 16 h, the clear reaction mixture was partitioned between CH 2 CI 2 and 10% hydrochloric acid.
- Step b Bromotrimethylsilane (0.28 mL, 2.1 mmol) was added to a solution of
- Step a A mixture N-Boc-Valine (1.55 g, 7.13 mmol), iodomethyl pivalate (1.73 g, 7.13 mmol) and Cs 2 C0 3 (2.32 g, 7.13 mmol) in DMF (50 mL) was heated at 60 °C. After 3 h at 60 °C, the yellow heterogeneous mixture was partitioned between EtOAc and water. The layers were separated and the organics were washed with, water (2X), A saturated solution of NaHC0 3 , 10% hydrochloric acid, brine, dried (Na 2 S0 4 ), filtered and concentrated under reduced pressure to give a clear oil (1.685 g, 71%).
- Step b TFA ( 7.5 mL) was added to a solution of pivaloyloxymethyl N-Boc-valinate (500 mg) in CH 2 C1 2 (7.5 mL) at 0 °C. After stirring at 0 °C for 2 h, the reaction mixture was concentrated under reduced pressure and azeotroped with toluene to give the free amine.
- Methyl 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)benzoate was prepared from 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)phenol (Chiellini et al., Bioorg. Med. Chem. Lett. 10:2607 (2000)) according to the procedure described for the synthesis of intermediate 4/step b-c.
- Step a To a mixture of 3,5-dimethyl-4-(3-isopropyl-4-hydroxybenzyl)benzyl alcohol (0.80 g, 2.82 mmol) and di-tert-butyl diethylphosphoramidate (0.98 g, 3.94 mmol) in DMF (20.0 mL) at rt was added 5-methylthiotetrazole (0.46 g, 3.84 mmol). The reaction mixture was stirred at rt for 40 min and cooled to 0 °C. To it was added t-butyl hydrogen peroxide (1.16 mL, 8.46 mmol).
- Step b A mixture of di-tert-butyl 3,5-dimethyl-4-(4-hydroxy-3-isopropylbenzyl)benzyl phosphate (0.44 g, 0.92 mmol) and 1 N HCl-dioxane (8.0 mL, 8.0 mmol) was stirred at room temperature for 3 h. The solvent was removed under reduced pressure and the residue was dissolved in ether (10 mL). The organic solution was washed with water (10 mL), dried in MgS0 4 and filtered. The solvent was removed under reduced pressure and the residue was dissolved in THF (6 mL).
- Example A34 The title compound was prepared from 3,5-dimethyl-4-(3-isopropyl-4- hydroxybenzyl)phenol (Example A34) according to the procedure described for the synthesis of Example E33-1.
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Diabetes (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Endocrinology (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to sulfonic acid containing compounds that bind to thyroid receptors in the liver. Activation of these receptors results in modulation of gene expression of genes regulated by thyroid hormones. The compounds can be used to treat diseases and disorders including metabolic diseases such as NASH, hypercholesterolemia and hyperlipidemia, as well as associated conditions such as atherosclerosis, coronary heart disease, impaired glucose tolerance, and metabolic syndrome X.
Description
PHOSPHOROUS-CONTAINING THYROID HORMONE
RECEPTOR AGONISTS AND METHODS OF USE
INCORPORATED APPLICATIONS [0001] This application is related to US Provisional Application Nos. 61/246,092, filed September 25, 2009, and 61/290,812, filed December 29, 2009, the contents of which are herein incorporated by reference in their entireties. This application is also related to US Application Nos. 61/151,312, 10/580,134; 11/137,773; 11/816,774; 11/842,067; 11/814,819; 11/814,824; and the applications to which they claim the benefit of priority.
FIELD OF THE INVENTION
[0002] The present invention is directed toward phosphorus-containing thyroid hormone receptor agonists ("THA") and methods of their use.
BACKGROUND OF THE INVENTION [0003] Thyroid hormones (THs, i.e., T4, T3, rT3, and/or T2) have profound effects on lipids and lipoprotein levels. THs lower plasma cholesterol levels in part by enhancing cholesterol conversion to bile acids through increased expression of the rate-limiting enzyme cholesterol 7a-hydroxylase CYP7a and by increasing hepatic uptake of LDL- cholesterol via LDL-receptors. THs increase free fatty acid oxidation and can as a result lower plasma and hepatic triglyceride levels. THs stimulate apoA-I expression and the secretion of apoA-I in HDL while reducing apoA-II. THs may also induce expression of SPvBl receptors which may in turn increase hepatic HDL uptake and possibly enhance reverse cholesterol transport. Last, THs lower the atherogenic lipoprotein Lp(a). The effects of THs on lipids and lipoproteins are largely through effects on the liver and genes expressed in the liver controlling lipid and lipoprotein production, uptake and metabolism.
[0004] T3 and T3 mimetics are thought to inhibit atherosclerosis by modulating the levels of certain lipoproteins known to be independent risk factors or potential risk factors of atherosclerosis, including low density lipoprotein (LDL)-cholesterol, high density
lipoprotein (HDL)-cholesterol, apoA-I, which is a major apoprotein constituent of high density lipoprotein (HDL) particles and lipoprotein (a) or Lp(a). Lp(a) is an important risk factor, elevated in many patients with premature atherosclerosis. Lp(a) is considered highly atherogenic (de Bruin et ah, J. Clin. Endo. Metab., 76, 121-126 (1993)). In man, Lp(a) is a hepatic acute phase protein that promotes the binding of LDL to cell surfaces independent of LDL receptors. Accordingly, Lp(a) is thought to provide supplementary cholesterol to certain cells, e.g., cells involved in inflammation or repair. Lp(a) is an independent risk factor for premature atherosclerosis.
[0005] THs may also be utilized for lowering triglycerides, which occurs inconsistently with hyperthyroidism. Elevated triglycerides represents a condition that can result in increased risk for coronary heart disease, pancreatitis and/or NASH.
[0006] THs are associated with a variety of adverse effects in animals and humans that largely arise from action of THs on non-hepatic tissues. Hyperthyroidism is associated with increased body temperature, general nervousness, weight loss despite increased appetite, muscle weakness and fatigue, increased bone resorption and enhanced calcification, and a variety of cardiovascular changes, including increased heart rate, increased stroke volume, increased cardiac index, cardiac hypertrophy, decreased peripheral vascular resistance, and increased pulse pressure. Hypothyroidism is generally associated with the opposite effects.
[0007] The biological activity of THs is mediated largely through thyroid hormone receptors (TRs). TRs belong to the receptor superfamily known as nuclear receptors, which, along with its common partner, the retinoid X receptor, form heterodimers that act as ligand-inducible transcription factors. Like other nuclear receptors, TRs have a ligand binding domain and a DNA binding domain and regulate gene expression through ligand- dependent interactions with DNA response elements (thyroid response elements, TREs). Currently, the literature shows that TRs are encoded by two distinct genes (TRa and TRP), which produce several isoforms through alternative splicing (Williams, Mol Cell Biol. 20(22):8329-42 (2000); Nagaya, et al, Biochem Biophys Res Commun 226(2):426- 30 (1996)). The major isoforms that have so far been identified are TRa-1, TRa-2, TR - 1 and TR -2. TRa-1 is ubiquitously expressed in the rat with highest expression in skeletal muscle and brown fat. TR -l is also ubiquitously expressed with highest expression in the liver, brain and kidney. TR -2 is expressed in the anterior pituitary gland and specific regions of the hypothalamus as well as the developing brain and inner
ear. In the rat and mouse liver, ΤΙ β-l is the predominant isoform (80%). The TR isoforms found in human and rat are highly homologous with respect to their amino acid sequences which suggest that each serves a specialized function.
[0008] The beneficial effects of THs on lipids and lipoproteins are attributed to ΤΙ β- 1 based in part on findings from ΤΙ β-l -deficient mice. In contrast, the adverse cardiac effects of THs are largely attributed to TRa-1. Accordingly, the search for T3 mimetics that are selective for TRP-l has been a major goal for pharmaceutical research as a means of obtaining the desired lipid lowering properties while minimizing effects on the heart.
[0009] The most widely recognized effects of THs are an increase in metabolic rate, oxygen consumption and heat production. Much of these effects are attributed to TRa-1 and action of THs on muscle and fat. Both TRa-1 and TR -l are expressed in brown adipose tissue (BAT). THs induce differentiation of white adipose tissue (WAT) as well as a variety of lipogenic genes, including ACC, FAS, glucose-6-phosphate dehydrogenase and spot- 14. Overall THs play an important role in regulating basal oxygen consumption, fat stores, lipogenesis and lipolysis (Oppenheimer, et ah, J. Clin. Invest. 87(1): 125-32 (1991)). Hyperthyroidism is associated with increased food intake, an overall increase in the basal metabolic rate (BMR) and decreased body weight (ca. 15%) whereas hypothyroidism is associated with a 25-30%) increase in body weight. Treating hypothyroidism patients with T3 leads to a decrease in body weight for most patients but not all (17% of the patients maintain weight). Consequently, THs and TH mimetics have been of interest for decades as antiobesity drugs.
[0010] THs are tightly regulated through feedback loops that affect production of hormones such as thyrotropin releasing factor (TRF) from the hypothalamus and thyroid stimulating hormone (TSH) from the pituitary which in turn affect the production of THs from the thyroid. THs also tightly regulate tissue levels of T3 through effects on enzymes (e.g. deiodinases) that govern TH metabolism.
[0011] TSH is an anterior pituitary hormone that regulates thyroid hormone production. TSH formation and secretion is in turn regulated by the hypothalamic TRF. TSH controls the uptake of iodide by the thyroid, the subsequent release of iodinated thyronines from thyroglobulin {e.g., T3, T4) as well as possibly the intrapituitary conversion of circulating T4 to T3. Compounds that mimic T3 and T4 can negatively regulate both TSH and TRF secretion resulting in suppression of TSH levels and decreased levels of THs such as T3 and/or T4. Negative regulation of TSH is postulated
based on co-transfection and knockout studies (Abel et al., J. Clin. Invest., 104, 291-300, (1999)) to arise through activation of the thyroid receptor ΤΡνβ, possibly the isoform ΤΡνβ- 2, which is highly expressed in the pituitary.
[0012] THs also affect the expression of deiodinases that convert T4 to the most biologically active TH, T3. In the liver, THs and TH mimetics increase deiodinase Dl which increases T4 conversion to T3 and rT3. Increased T4 metabolism results in lower T4 levels and/or in higher TSH levels.
[0013] Low T4 levels induce expression of the deiodinase D2 which helps ensure that certain tissues remain euthyroid through production of adequate levels of T3. Some tissues may not be able to compensate for the low T4 levels and as a consequence there may be some tissue-specific hypothyroidism.
[0014] TH mimetics are reported to significantly reduce T4 levels in animals and humans. Thus, there remains a need to develop characterize and optimize drugs from this class that lower lipids effectively with minimal effects on thyroid hormone levels.
[0015] All documents referred to herein are incorporated by reference into the present application as though fully set forth herein.
SUMMARY OF THE INVENTION
[0016] In certain aspects, the present invention relates to phosphorus-containing thyroid hormone receptor agonists ("TELA") having a short plasma half-life and exhibiting reduced effects on endogenous thyroid hormone levels, and methods of their use.
[0017] In accordance with the present invention, novel phosphorus-containing thyroid hormone receptor (TR) agonists that have reduced or minimal effects on thyroid hormone levels at lipid lowering doses as compared to baseline levels, have been identified, and methods for their use provided. Preferably, the effect is a change of less than 50% of baseline T4 levels, more preferably less than 20% or less than 10%.
[0018] In certain aspects, the phosphorus-containing TR agonists of this invention and prodrugs thereof exhibit a short plasma half-life, particularly in mammalian and more particularly human subjects.
[0019] In certain aspects, TR agonists of this invention are at least 2 to 10-fold more selective for the TRP-l receptor over the TRa-1 receptor. Preferably the TR agonists are at least 20-fold to 50-fold, more selective for TRP-l over TRa-1.
[0020] In certain aspects, TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects on cardiovascular function. Reduced or minimal effects may include clinically insignificant impact on cardiac function.
[0021] In certain aspects, TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on muscle and bone function, as compared to baseline or normal levels.
[0022] In certain aspects, TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on thyroid hormones, and combinations thereof.
[0023] In certain aspects, TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit minimal effects on oxygen consumption.
[0024] In certain aspects, TR agonists of this invention and prodrugs thereof achieve the desired therapeutic index (TI) by using a dosing frequency wherein drug levels are reduced from their Cmax prior to the next dose.
[0025] In certain aspects, TR agonists of the invention are dosed at a frequency that sustains cholesterol lowering over time with minimal effects on T4 or while maintaining lipid lowering effects compared to baseline.
[0026] In certain aspects, the compound is a liver targeted compound that does not substantially impact other tissues. Demonstration of liver targeting and lack of impact on other tissues can be evaluated by measusing changes in mRNAs in liver as opposed to muscle, heart, pituitary, etc. Compounds of the invention may show changes in liver transcripts of genes encoding, for example, Dl, m-GPDH, CYP7a, malic enzyme, sterol regulating element binding protein lc (SREBPlc), LDL-cholesterol receptor in the liver, TSH , or Dl in the pituitary, Dl and m-GPDH in the heart, uncoupling protein 3 (UCP3) in the muscle. Such changes would be expected in liver, but no significant corresponding change in heart, muscle, pituitary, or other systemic tissues would be expected.
[0027] In certain aspects, TR agonists of this invention and prodrugs thereof are combined with one or more lipid lowering agents such as statins or cholesterol absorption inhibitors to treat patients with hyperlipidemia. Preferably, such combination allows therapeutic effects at a reduced dose of one or more of the agents, improves lipid profile, or improves safety/therapeutic index of the therapy of one or more of the agents.
[0028] In certain aspects, compounds that are thyroid hormone receptor ligands, pharmaceutically acceptable salts, and prodrugs, preferably prodrugs that are short lived in vivo, of these compounds, as well as their preparation and uses for preventing and/or treating dyslipidemia, hypercholesterolemia, hyperlipidemia, elevated Lp(a), and associated diseases such as atherosclerosis, coronary heart disease, and pancreatitis.
[0029] In certain aspects, compounds that are thyroid hormone receptor ligands, pharmaceutically acceptable salts, and prodrugs of these compounds as well as their preparation and uses for preventing effects on the thyroid hormone axis (THA) while retaining the beneficial effects of activation of TR in the liver for preventing and/or treating hypercholesterolemia, hyperlipidemia, elevated Lp(a), and associated diseases such as atherosclerosis, coronary heart disease, and pancreatitis.
[0030] These and other aspects of the invention will be more clearly understood with reference to the following preferred embodiments and detailed description.
DETAILED DESCRIPTION OF THE INVENTION
[0031] Prior to the discoveries of the present invention, it was thought that TR -l- selective agonists would lower lipids with an improved cardiac therapeutic index relative to less TR selective agonists. Further improvement in the cardiac therapeutic index was realized with the discovery of liver-targeted phosphorus-containing TR agonists (Erion et al. PNAS, 2007, 104 (39), 15490). No therapeutic window was found for TR -l- selective agonists relative to effects on THs in animals and humans, possibly because these compounds affected mechanisms controlling TH metabolism in the liver (e.g. Dl) or because these compounds affected TSH and/or TRF production in the pituitary and hypothalamus as a result of both being regulated by TR or because of an indirect effect of activation of TR-sensitive genes such as Dl in the liver which in turn modulated THs in a manner that affected central mechanisms controlling TH homeostasis. Some improvement in the therapeutic window was reported for phospho(i)nic acid TR agonists, possibly because of improved liver targeting but even these compounds produced a significant reduction in total T4 levels in animals and humans at minimally effective lipid lowering doses. Consequently it was not known that liver-targeted TR agonists could reduce lipids without affecting systemic T4 levels.
[0032] Prior to the discoveries of the present invention it was not known whether the effects of TR agonists on lipids lasted longer, shorter or the same amount of time as the
effects of these compounds on THs. Further, it was not known that phosphorus- containing TR agonists with a short plasma half-life would result in reduced effects on THs at lipid lowering doses. And it was not known that phosphorus-containing TR agonists with a short plasma half- life would have an improved therapeutic index. It was also unknown whether phosphorus-containing TR agonists with a short plasma half-life would result in reduced effects on liver toxicity and/or oxygen consumption and whether these reduced effects would result in an improved therapeutic index.
[0033] Prior to the discoveries of the invention, it was unknown whether phosphorus- containing TR agonists, particularly those with a short plasma half-life, could be combined with statins without narrowing the therapeutic index since TR agonists have been reported to adversely affect the same target organs of toxicity as statins, i.e., liver and muscle. It was also unknown that phosphorus-containing TR agonists with a short plasma half- life selective for TRP-l would be as useful as a longer-acting TR agonist in treating one or more of the following conditions and/or diseases: dyslipidemia, hypercholesterolemia, hypertriglyceridemia, elevated Lp(a) levels, atherosclerosis, coronary heart disease, pancreatitis.
[0034] Prior to the discoveries of the invention, it was not known that phosphorus- containing TR agonists could be dosed at a frequency that involved prolonged periods of low drug levels and that this dosing regimen would result in sustained lipid lowering with an improved therapeutic index, and that such a regimen would still be useful in preventing and/or treating the following conditions and/or diseases: dyslipidemia, hypercholesterolemia, hypertriglyceridemia, elevated Lp(a) levels, atherosclerosis, coronary heart disease, pancreatitis, while reducing adverse effects on TH levels and/or extra-hepatic tissues.
[0035] Prior to the discoveries of the invention, it was unknown whether phosphorus- containing TR agonists with the desired plasma half-life could be identified by selecting prodrugs that resulted in rapid conversion to the biologically-active TR agonist and by selecting TR agonists that undergo rapid clearance via transporters that exist in the liver and/or kidney and transport negatively-charged compounds; or by selecting a TR agonist that is subject to rapid metabolism resulting in metabolites devoid of significant TR agonist activity.
[0036] It was discovered that the compounds of the invention are able to solve the above problems; that short-acting phosphorous containing TR agonists can specifically
target the liver and activate lipid lowering gene transcription such as LDL receptor expression, without the expected increase in systemic T4 clearance or other adverse systemic THA effects.
[0037] Alternatively and contrary to traditional medicinal chemistry teachings, compounds of the inventions are designed with metabolic liabilities to take advantage of deactivating metabolic pathways in order to shorten the half life of said compounds (Bodor et al. Med. Res. Rev. 58 (2000). In accordance with embodiments of the invention, suitable functional groups to shorten the half life of said compound include: esters, iodines, aldehydes, carbamates, carbonates, thioethers, disulfides, or ^-substituted cysteines, and phosphates that would be substrates for ubiquitous, highly expressed and highly active enzymes such as but not limited to esterases, deiodinases, carboxylesterases, aldehyde oxidases, glutathione transferases cysteine β -lyases and phosphatases (Rooseboom et al. Pharmacol. Rev. 56:53 (2004)).
[0038] While the compounds of the invention are designed to be rapidly metabolized by incorporation of metabolically unstable functional groups, these compounds are also designed with highly charged functional groups such as phospho(i)nic acids and phosphates, so as to target the liver and limit distribution in order to avoid activation of TR in tissues such as heart, muscle and pituitary. Activation of TR in extrahepatic tissues leads to side effects and a decrease in the therapeutic index (TI).
[0039] While the compounds of the invention are designed to be liver targeted and rapidly metabolized by incorporation of metabolically unstable functional groups, these compounds are also designed so as to bind and activate the TR in the liver to modulate gene expression of genes regulated by thyroid hormones. In addition, the compounds of
the invention are designed as such that once metabolized, the metabolite produced does not bind to the TR or, if it does bind to TR, it does not activate TR.
[0040] In certain aspects, the present invention relates to phosphorus-containing TR agonists that have a short plasma half- life, are selective for the TRP-l receptor, target the liver, and lower lipids and lipoproteins without affecting THs
[0041] In other aspects, the present invention relates to phospho(i)nic acid or phosphates containing compounds that bind to thyroid receptors in the liver and have a short half life. Activation of these receptors results in modulation of gene expression of genes regulated by TH.
[0042] In other aspects, the present invention relates to phospho(i)nic acid or phosphates containing compounds designed with metabolic liabilities to shorten their half life and that bind to thyroid receptors in the liver and wherein administration of the compounds or prodrugs thereof results in lipid-lowering with minimal effects on T4.
[0043] In other aspects, the present invention relates to phospho(i)nic acid or phosphates containing compounds described herein that bind to thyroid receptors in the liver, designed with metabolic liabilities to shorten their half life and whose metabolite do not activate TR receptors and wherein administration of the compounds or prodrugs thereof results in lipid-lowering with minimal effects on T4.
[0044] In other aspect, the present invention relates to compounds designed with metabolic liabilities to shorten their half lives.
[0045] In certain embodiments, the compounds may be thyroid hormone agonists of Formula I:
(Ar1)-G-(Ar2)-T-E
wherein:
1 2
Ar and Ar are substituted aryl or heteroaryl groups;
1 2
G is an atom or group of atoms that links Ar and Ar through 1-2 contiguous atoms;
T is an atom or group of atoms linking Ar to E through 1 -4 contiguous atoms or is absent; and
E is a functional group with a pKa < 7.4, and more preferably pKa < 4.0, containing a phosphorus atom, and prodrug thereof.
[0046] In certain aspects, the present invention relates to methods of preventing or treating metabolic diseases with compounds of the invention, pharmaceutically acceptable
salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
[0047] In certain aspects, the present invention relates to methods of reducing levels of Lp(a) with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
[0048] In certain aspects, the present invention relates to methods of reducing LDL- cholesterol levels with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
[0049] The present invention also relates to pharmaceutically acceptable salts and co- crystals, prodrugs, and pharmaceutically acceptable salts and co-crystals of these prodrugs of these compounds.
[0050] In certain aspects, the present invention relates to methods of improving the therapeutic index by treating with compounds of the invention, pharmaceutically acceptable salts and prodrugs thereof, and pharmaceutically acceptable salts of the prodrugs, where the said compounds bind to a thyroid hormone receptor.
[0051] Also covered are the prodrugs of the compounds of the invention. Especially preferred are the prodrugs of the compounds of the invention that have a short pharmacokinetic half life. Upon oral absorption, the prodrug enters the portal vein and a fraction is uptaken by the liver. The remaining fraction not absorbed by the liver enters then the general circulation where, if the half life is not short, it has the potential to enter other organs, such as the heart and the brain. Prodrug breakdown and release of the parent TR agonist in these extra hepatic tissues could lead to activation of TR and a decrease of the TI. In addition, distribution of the intact prodrug into the whole body of an animal would lead to a significant increase in the volume of distribution of the prodrug. This increase in volume of distribution in turn would lead to a slow release of the prodrug from the extra hepatic tissues, as the parent TR agonist is cleared by the liver, which would lead to a lengthening of the pharmacokinetic half-life of the parent drug.
[0052] Prodrugs of the short acting TR compounds are useful for increasing oral bioavailability.
A. Definitions
[0053] The term "phosphorus-containing thyroid hormone receptor agonist" refers to TR agonists which bear a functional group with a pKa < 7.4, and preferably with a pKa < 4.0 containing a phosphorus atom such as a phospho(i)nic acid, phosphonic acid mono- ester, phosphoramidic acid or phosphate group.
[0054] The term "non-rodent mammalian" animals refers to a mammal that is not from a rodent species. Preferred non-rodent mammalians are dogs, monkeys and humans.
[0055] The term "short plasma half-life" refers to the pharmacokinetic half-life of a compound in plasma that is less than 4 h, less than 2 h, less than 1 h, less than 30 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes in non-rodent mammalian species.
[0056] The term "minimal effects on TH levels or endogenous TH levels" refers to the effects of a drug on the levels of total T4, free T4, total T3 and free T3 in an animal. Such effects mean a change of less than 50%, preferably less than 20%, 10%>, or 5%>,.
[0057] The term "thyroid hormones" refers to tyrosine-based hormones produced by the thyroid gland such as thyroxine (T4), 3',3,5-triiodothyronine (T3), 3',5',3- triiodothyronine (rT3) and 3,5-diodothyronine (T2).
[0058] The term "lipids" refers to the group of naturally-occurring molecules which includes fats (fatty acids, mono, di and triglycerides), waxes, sterols (cholesterol), fat- soluble vitamins (such as vitamins A, D, E and K), phospholipids, and others. The main biological functions of lipids include energy storage, as structural components of cell membranes, and as important signaling molecules.
[0059] The term "lipoproteins" refers to biochemical assemblies containing both proteins and lipids and are classified by their relative density such as chylomicron, very low density lipoprotein (VLDL), low density lipoprotein (LDL) and high density lipoprotein (HDL).
[0060] The term "T3 mimetic or TH mimetic" refers to compounds that mimic the pharmacodynamic effects of T3.
[0061] The term "metabolic rate" refers to resting metabolic rate (RMR) which is the amount of energy expanded by an animal while at rest in a neutrally temperate environment. The RMR is measured by gas analysis through direct or indirect calorimetry. Alternatively RMR can be determined by calculating the respiratory quotient which measures the inherent composition and utilization of carbohydrates, fats and
proteins as they are converted to energy substrate units that can be used by the body as energy.
[0062] The term "oxygen consumption" refers to the rate of oxygen consumed by an animal while at rest in a neutrally temperate environment. Oxygen consumption is used to calculate the respiratory quotient which is directly related to RMR. An elevation in oxygen consumption is associated with an increase in RMR.
[0063] The term "cardiac parameters" refers to the measurable parameters used to evaluate cardiac function such as gene expression changes (e.g., Dl, m-GPDH), heart rate, pulse rate, rhythm, hypertrophy, or contractility.
[0064] The term "elevated Lp(a)" refers to levels of the lipoprotein Lp(a) that are >10 mg/dL.
[0065] The term "prodrug with a short plasma half-life" refers to the pharmacokinetic half-life of a compound in plasma that is less than 4 h, less than 2 h, less than 1 h, less than 30 minutes, less than 20 minutes, less than 15 minutes, less than 10 minutes, less than 5 minutes.
[0066] The term "prodrug" as used herein refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each. Standard prodrugs are formed using groups attached to functionality, e.g., the phenol group of the compounds described herein, that cleave in vivo. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, and/or pharmacodynamic half-life, etc.
[0067] Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site-specific delivery of the compound. Prodrugs are described in The Organic Chemistry of Drug Design and Drug Action, by Richard B. Silverman, Academic Press, San Diego, 1992. Chapter 8: "Prodrugs and Drug delivery Systems" pp.352-401; Design of Prodrugs, edited by H. Bundgaard, Elsevier Science, Amsterdam,
1985; Design of Biopharmaceutical Properties through Prodrugs and Analogs, Ed. by E. B. Roche, American Pharmaceutical Association, Washington, 1977; and Drug Delivery Systems, ed. by R. L. Juliano, Oxford Univ. Press, Oxford, 1980.
[0068] As used herein, the term "alkyl" generally refers to saturated hydrocarbyl radicals of straight, branched or cyclic configuration including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, cyclohexyl, n-heptyl, octyl, n-octyl, and the like. In some embodiments, alkyl substituents may be Ci to C2o, Ci to C12j Ci to C8, Ci to C6, or Ci to C4 alkyl groups. In certain embodiments, the alkyl group may be optionally substituted. For instance, the alkyl group may be a haloalkyl, including monohaloalkyl, dihaloalkyl, and trihaloalkyl.
[0069] As used herein, "alkylene" generally refers to linear, branched or cyclic alkene radicals having one or more carbon-carbon double bonds, such as C2 to C6 alkylene groups including 3-propenyl. Again, in certain embodiments, the alkyl group may be optionally substituted.
[0070] The term "alkenyl" refers to unsaturated groups which have, e.g., 2 to 12 atoms and contain at least one carbon-carbon double bond and includes straight-chain, branched-chain and cyclic groups. Alkenyl groups may be optionally substituted. Suitable alkenyl groups include allyl.
[0071] The term "alkynyl" refers to unsaturated groups which have, e.g., 2 to 12 atoms and contain at least one carbon-carbon triple bond and includes straight-chain, branched-chain and cyclic groups. Alkynyl groups may be optionally substituted. Suitable alkynyl groups include ethynyl.
[0072] As used herein, "aryl" refers to a carbocyclic aromatic ring structure. Included in the scope of aryl groups are aromatic rings having from five to twenty ring atoms. Aryl ring structures include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds, and includes carbocyclic aryl and heterocyclic aryl and biaryl groups. Examples of aryl groups that include phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, phenanthrenyl (i.e., phenanthrene), and napthyl (i.e., napthalene) ring structures. Again, in certain embodiments, the alkyl group may be optionally substituted.
[0073] As used herein, "heterocycle" refers to cyclic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heterocycle, and
independently selectable, are O, N, and S heterocycle ring structures. The ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds, and may be aromatic, i.e., the ring structure may be a heteroaryl. Example of heterocyclo groups include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl or tetrahydrothiopyranyl and the like. Again, in certain embodiments, the alkyl group may be optionally substituted.
[0074] As used herein, "heteroaryl" refers to cyclic aromatic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heteroaryl, and independently selectable, are O, N, and S heteroaryl ring structures. The ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds. In some embodiments, the heteroaryl groups may be selected from heteroaryl groups that contain two or more heteroatoms, three or more heteroatoms, or four or more heteroatoms. Heteroaryl ring structures may be selected from those that contain five or more atoms, six or more atoms, or eight or more atoms. In a preferred embodiment, the heteroaryl including five to ten atoms. Examples of heteroaryl ring structures include: acridine, benzimidazole, benzoxazole, benzodioxole, benzofuran, 1,3- diazine, 1,2-diazine, 1,2-diazole, 1 ,4-diazanaphthalene, furan, furazan, imidazole, indole, isoxazole, isoquinoline, isothiazole, oxazole, purine, pyridazine, pyrazole, pyridine, pyrazine, pyrimidine, pyrrole, quinoline, quinoxaline, thiazole, thiophene, 1,3,5-triazine, 1,2,4-triazine, 1,2,3-triazine, tetrazole and quinazoline. Again, in certain embodiments, the alkyl group may be optionally substituted.
[0075] As used herein, "alkoxy" generally refers to a group with the structure -O-R. In certain embodiments, R may be an alkyl group, such as a Ci to C8, Ci to C6 alkyl group, or C\ to C4 alkyl group. In certain embodiments, the R group of the alkoxy may optionally be substituted, e.g., with at least one halogen. For example, the R group of the alkoxy may be a haloalkyl, i.e., haloalkoxy.
[0076] Halogen substituents may be independently selected from the halogens such as fluorine, chlorine, bromine, iodine, and astatine.
[0077] More specifically, the term "optionally substituted" or "substituted" includes groups substituted by one, two, three, four, five, or six substituents, independently
selected from lower alkyl, lower aryl, lower aralkyl, lower cyclic alkyl, lower heterocycloalkyl, hydroxy, lower alkoxy, lower aryloxy, perhaloalkoxy, aralkoxy, lower heteroaryl, lower heteroaryloxy, lower heteroarylalkyl, lower heteroaralkoxy, azido, amino, halo, lower alkylthio, oxo, lower acylalkyl, lower carboxy esters, carboxyl, - carboxamido, nitro, lower acyloxy, lower aminoalkyl, lower alkylaminoaryl, lower alkylaryl, lower alkylaminoalkyl, lower alkoxyaryl, lower arylamino, lower aralkylamino, sulfonyl, lower-carboxamidoalkylaryl, lower -carboxamidoaryl, lower hydroxyalkyl, lower haloalkyl, lower alkylaminoalkylcarboxy-, lower aminocarboxamidoalkyl-, cyano, lower alkoxyalkyl, lower perhaloalkyl, and lower arylalkyloxyalkyl.
[0078] The phrase "therapeutically effective amount" means an amount of a compound or a combination of compounds that modifies, ameliorates, attenuates or eliminates one or more of the symptoms of a particular disease or condition or prevents, modifies, or delays the onset of one or more of the symptoms of a particular disease or condition.
[0079] The term "pharmaceutically acceptable salt" includes salts of compounds of Formula I and its prodrugs derived from the combination of a compound of this invention and an organic or inorganic acid or base. Suitable acids include acetic acid, adipic acid, benzenesulfonic acid, (+)-7,7-dimethyl-2-oxobicyclo[2.2.1 ]heptane- 1 -methanesulfonic acid, citric acid, 1 ,2-ethanedisulfonic acid, dodecyl sulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glucuronic acid, hippuric acid, hydrochloride hemiethanolic acid, HBr, HC1, HI, 2-hydroxyethanesulfonic acid, lactic acid, lactobionic acid, maleic acid, methanesulfonic acid, methylbromide acid, methyl sulfuric acid, 2- naphthalenesulfonic acid, nitric acid, oleic acid, 4,4'-methylenebis [3-hydroxy-2- naphthalenecarboxylic acid], phosphoric acid, polygalacturonic acid, stearic acid, succinic acid, sulfuric acid, sulfosalicylic acid, tannic acid, tartaric acid, terphthalic acid, and p- toluenesulfonic acid.
[0080] The terms "patient" and "subject" are used interchangably, and may include in vitro and in vivo subjects such as cells, tissues, and animals. In this regard, the term "animal" includes birds and mammals. In one embodiment a mammal includes a rat, mouse, monkey, dog, cat, cow, horse, goat, sheep, pig or human of either gender.
[0081] The term "increased or enhanced liver specificity" refers to an increase in the liver specificity ratio in animals treated with a compound of the present invention and a control compound.
[0082] The term "enhanced oral bioavailability" refers to an increase of at least 50% of the absorption of the dose of the parent drug, unless otherwise specified. In an additional aspect the increase in oral bioavailability of the prodrug (compared to the parent drug) is at least 100%, that is a doubling of the absorption. Measurement of oral bioavailability usually refers to measurements of the prodrug, drug, or drug metabolite in blood, plasma, tissues, or urine following oral administration compared to measurements following systemic administration of the compound administered orally.
[0083] The terms "treating" or "treatment" of a disease includes a slowing of the progress or development of a disease after onset or actually reversing some or all of the disease affects. Treatment also includes palliative treatment.
[0084] The term "preventing" includes a slowing of the progress or development of a disease before onset or precluding onset of a disease.
[0085] The term "prodrug" as used herein refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each. Standard prodrugs are formed using groups attached to functionality, e.g., HO-, HS-, HOOC-, R2N-, associated with the drug, that cleave in vivo. Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate. The groups illustrated are exemplary, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Such prodrugs of the compounds of the present invention fall within this scope. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, and/or pharmacodynamic half-life, etc. Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site-specific delivery of the compound. Prodrugs are described in The Organic Chemistry of Drug Design and Drug Action, by
Richard B. Silverman, Academic Press, San Diego, 1992. Chapter 8: "Prodrugs and Drug delivery Systems" pp.352-401; Design of Prodrugs, edited by H. Bundgaard, Elsevier Science, Amsterdam, 1985; Design of Biopharmaceutical Properties through Prodrugs and Analogs, Ed. by E. B. Roche, American Pharmaceutical Association, Washington, 1977; and Drug Delivery Systems, ed. by R. L. Juliano, Oxford Univ. Press, Oxford, 1980.
[0086] Prodrugs of carboxylic acid-containing thyromimetics are convertible by solvolysis or under physiological conditions to the free carboxylic acids. Examples of prodrugs include carboxylic acid esters, and are preferably lower alkyl esters, cycloalkyl esters, lower alkenyl esters, benzyl esters, aryl esters, mono- or di-substituted lower alkyl esters, e.g., the ro-(amino, mono- or di-lower alkylamino, carboxy, lower alkoxycarbonyl)-lower alkyl esters, and the a-(lower alkanoyloxy, lower alkoxycarbonyl or di-lower alkylaminocarbonyl)-lower alkyl esters, such as the pivaloyloxy-methyl ester.
[0087] Prodrugs of phosphorus-containing thyromimetics breakdown chemically or enzymatically to a phospho(i)nic acid or phosphate group thereof in vivo. As employed herein the term includes, but is not limited to, the following groups and combinations of these groups:
[0088] Acyloxyalkyl esters which are well described in the literature (Farquhar et al., J. Pharm. Sci. 72:324-325 (1983)).
[0089] Other acyloxyalkyl esters are possible in which a cyclic alkyl ring is formed. These esters have been shown to generate phosphorus-containing nucleotides inside cells through a postulated sequence of reactions beginning with deesterification and followed by a series of elimination reactions {e.g., Freed et al, Biochem. Pharm, 38:3193-3198 (1989)).
[0090] Another class of these double esters known as alkyloxycarbonyloxymethyl esters, as shown in formula A, where R is alkoxy, aryloxy, alkylthio, arylthio, alkylamino, and arylamino; R', and R" are independently -H, alkyl, aryl, alkylaryl, and heterocycloalkyl have been studied in the area of β-lactam antibiotics (Nishimura et al., J. Antibiotics 40(l):$l-90 (1987); for a review see Ferres, H., Drugs of Today, 19:499 (1983)). More recently Cathy, M. S. et al. (Abstract from AAPS Western Regional Meeting, April, 1997) showed that these alkyloxycarbonyloxymethyl ester prodrugs on (9-[(R)-2-phosphonomethoxy)propyl]adenine (PMPA) are bioavailable up to 30% in dogs.
wherein R, R', and R" are independently H, alkyl, aryl, alkylaryl, and alicyclic (see WO 90/08155; WO 90/10636).
[0091] Other acyloxyalkyl esters are possible in which a cyclic alkyl ring is formed such as shown in formula B. These esters have been shown to generate phosphorus- containing nucleotides inside cells through a postulated sequence of reactions beginning with deesterification and followed by a series of elimination reactions (e.g., Freed et al, Biochem. Pharm. 38:3193-3198 (1989)).
wherein R is -H, alkyl, aryl, alkylaryl, alkoxy, aryloxy, alkylthio, arylthio, alkylamino, arylamino, or cyclo alkyl.
[0092] Aryl esters have also been used as phosphonate prodrugs (e.g., DeLombaert et al, J. Med. Chem. 57^:498-511 (1994); Serafmowska et al, J. Med. Chem. 38(8):\3Ί2- 9 (1995). Phenyl as well as mono and poly-substituted phenyl proesters have generated the parent phosphonic acid in studies conducted in animals and in man (Formula C). Another approach has been described where Y is a carboxylic ester ortho to the phosphate (Khamnei et al, J. Med. Chem. J :4109-15 (1996)).
wherein Y is -H, alkyl, aryl, alkylaryl, alkoxy, acyloxy, halogen, amino, alkoxycarbonyl, hydroxy, cyano, and heterocycloalkyl.
[0093] Benzyl esters have also been reported to generate the parent phosphonic acid. In some cases, using substituents at the /?ara-position can accelerate the hydrolysis. Benzyl analogs with 4-acyloxy or 4-alkyloxy group [Formula D, X = -H, OR or 0(CO)R or 0(CO)OR] can generate the 4-hydroxy compound more readily through the action of enzymes, e.g., oxidases, esterases, etc. Examples of this class of prodrugs are described in Mitchell et al, J. Chem. Soc. Perkin Trans. 12345 (1992); WO 91/19721.
wherein X and Y are independently -H, alkyl, aryl, alkylaryl, alkoxy, acyloxy, hydroxy, cyano, nitro, perhaloalkyl, halo, or alkyloxycarbonyl; and
R and R are independently -H, alkyl, aryl, alkylaryl, halogen, and cyclic alkyl.
[0094] Thio-containing phosphonate proesters may also be useful in the delivery of drugs to hepatocytes. These proesters contain a protected thioethyl moiety as shown in formula E. One or more of the oxygens of the phosphonate can be esterified. Since the mechanism that results in de-esterification requires the generation of a free thiolate, a variety of thiol protecting groups are possible. For example, the disulfide is reduced by a reductase-mediated process (Puech et al, Antiviral Res. 22: 155-174 (1993)). Thioesters will also generate free thiolates after esterase-mediated hydrolysis Benzaria, et al, J. Med. Chem. 3 (25,):4958-65 (1996)). Cyclic analogs are also possible and were shown to liberate phosphonate in isolated rat hepatocytes. The cyclic disulfide shown below has not been previously described and is novel.
wherein Z is alkylcarbonyl, alkoxycarbonyl, arylcarbonyl, aryloxycarbonyl, or alkylthio.
[0095] Other examples of suitable prodrugs include proester classes exemplified by Biller and Magnin (U.S. Patent No. 5,157,027); Serafinowska et al., J. Med. Chem. 55^:1372-9 (1995); Starrett et al, J. Med. Chem. 57:1857 (1994); Martin et al. J. Pharm. Sci. 7(5: 180 (1987); Alexander et al., Collect. Czech. Chem. Commun. 5 :1853 (1994); and EP 0 632 048 Al . Some of the structural classes described are optionally substituted, including fused lactones attached at the omega position (formulae E-1 and E- 2) and optionally substituted 2-oxo-l,3-dioxolenes attached through a methylene to the phosphorus oxygen (formula E-3) such as:
wherein Y is -H, alkyl, aryl, alkylaryl, cyano, alkoxy, acyloxy, halogen, amino, heterocycloalkyl, and alkoxycarbonyl.
[0096] The prodrugs of Formula E-3 are an example of "optionally substituted heterocycloalkyl where the cyclic moiety contains a carbonate or thiocarbonate."
[0097] Propyl phosphonate proesters can also be used to deliver drugs into hepatocytes. These proesters may contain a hydroxyl and hydroxyl group derivatives at the 3 -position of the propyl group as shown in formula F. The R and X groups can form a cyclic ring system as shown in formula F. One or more of the oxygens of the phosphonate can be esterified.
X is hydrogen, alkylcarbonyloxy, alkyloxycarbonyloxy; and
Y is alkyl, aryl, heteroaryl, alkoxy, alkylamino, alkylthio, halogen,
hydrogen, hydroxy, acyloxy, amino.
[0098] Phosphoramidate derivatives have been explored as phosphate prodrugs (e.g., McGuigan et al., J. Med. Chem. 42:393 (1999) and references cited therein) as shown in Formula G and H.
[0099] Cyclic phosphoramidates have also been studied as phosphonate prodrugs because of their speculated higher stability compared to non-cyclic phosphoramidates (e.g., Starrett et al, J. Med. Chem. 57: 1857 (1994)).
[00100] Another type of phosphoramidate prodrug was reported as the combination of S-acyl-2-thioethyl ester and phosphoramidate (Egron et al., Nucleosides Nucleotides 75:981 (1999)) as shown in Formula J:
[00101] Other prodrugs are possible based on literature reports such as substituted ethyls, for example, bis(trichloroethyl)esters as disclosed by McGuigan, et ah, Bioorg Med. Chem. Lett. 5: 1207-1210 (1993), and the phenyl and benzyl combined nucleotide esters reported by Meier, C. et al, Bioorg. Med. Chem. Lett. 7:99-104 (1997).
[00102] The structure
has a plane of symmetry running through the phosphorus-oxygen double bond when R6=R6, V=W, and V and W are either both pointing up or both pointing down. The same is true of structures where each -NR6 is replaced with -0-.
[00103] The term "cyclic phosphonate ester of 1,3-propane diol", "cyclic phosphonate diester of 1,3-propane diol", "2 oxo 2λ5 [1,3,2] dioxaphosphonane", "2 oxo [1,3,2] dioxaphosphonane", "dioxaphosphonane" refers to the following:
[00104] The phrase "together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkylthiocarbonyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy
attached to a carbon atom that is three atoms from both Y groups attached to the phosphorus" includes the following:
[00105] The structure shown above (left) has an additional 3 carbon atoms that forms a five member cyclic group. Such cyclic groups must possess the listed substitution to be oxidized.
[00106] The phrase "together V and Z are connected via an additional 3-5 atoms to form a cyclic group, optionally containing one heteroatom, that is fused to an aryl group attached at the beta and gamma position to the Y attached to the phosphorus" includes the following:
[00107] The phrase "together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said additional carbon atoms that is three atoms from a Y attached to the phosphorus" includes the following:
[00108] The structure above has an acyloxy substituent that is three carbon atoms from a Y, and an optional substituent, -CH3, on the new 6-membered ring. There has to be at least one hydrogen at each of the following positions: the carbon attached to Z; both carbons alpha to the carbon labeled "3"; and the carbon attached to "OC(0)CH3" above.
[00109] The phrase "together W and W are connected via an additional 2-5 atoms to form a cyclic group, optionally containing 0-2 heteroatoms, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl" includes the following:
[00110] The structure above has V=aryl, and a spiro-fused cyclopropyl group for W and W.
[00111] The term "cyclic phosphon(amid)ate" refers to
where Y is independently -O- or -NR -. The carbon attached to V must have a C-H bond. The carbon attached to Z must also have a C-H bond.
[00112] The naming of the compounds is done by having the ring bearing the groups
R 5 and R 3 be a substituent on the ring bearing the R 1 and R2 groups. The naming of the prodrugs is done by having the diaryl system with its linker T (Formula I, II, III, V, VI, and VIII) or D (Formula IV) be a substituent on the phosphorus atom contained in X. For example:
1 2 5 3
[3-R -5-R -4-(4'-R -3'-R -benzyl)phenoxy]methylphosphonic acid represents the formula:
1 2 5 3
[3-R -5-R -4-(4'-R -3'-R -phenoxy)phenoxy]methylphosphonic acid represents formula:
1 2 5 3
2-[(3-R1-5-R2-4-(4'-R5-3'-R3-benzyl)phenoxy)methyl]-4-aryl-2-oxo-2 5-[l,3,2]-dioxapho sphonane:
[00113] The term "cis" stereochemistry refers to the spatial relationship of the V group and the carbon attached to the phosphorus atom on the six-membered ring. The formula below shows a cis stereochemistry.
[00114] The term "trans" stereochemistry refers to the spatial relationship of the V group and the carbon, attached to the phosphorus atom, on the six-membered ring. The formula below shows a trans-stereochemistry.
[00115] The formula below shows another trans- stereochemistry.
[00116] The terms "^-configuration," "^-isomer" and "^-prodrug" refers to the absolute configuration S of carbon C. The formula below shows the ^-stereochemistry.
[00117] The terms "^-configuration," "R -isomer" and "R -prodrug" refers to the absolute configuration R of carbon C. The formula below shows the i?-stereochemistry.
[00118] The term "percent enantiomeric excess (% ee)" refers to optical purity. It is obtained by using the following formula:
where [R] is the amount of the R isomer and [S] is the amount of the S isomer. This formula provides the % ee when R is the dominant isomer.
[00119] The term "enantioenriched" or "enantiomerically enriched" refers to a sample of a chiral compound that consists of more of one enantiomer than the other. The extent
to which a sample is enantiomerically enriched is quantitated by the enantiomeric ratio or the enantiomeric excess.
B. Compounds of the Invention
[00120] In one aspect of the invention, compounds of the invention that have a short pharmacokinetic half-life and are thyroid receptor ligands are provided, pharmaceutically acceptable salts thereof, and prodrugs of these compounds as well as their preparation and uses for preventing and/or treating metabolic diseases such as NASH, hypercholesterolemia and hyperlipidemia as well as associated conditions such as atherosclerosis, and coronary heart disease. The present invention is also related to the use of the compounds of the invention to treat the above-mentioned diseases and improve the therapeutic index by decreasing effects on the thyroid hormone axis. The invention is also related to the use of these compounds for the prevention and treatment of diseases responsive to modulation of T3 -responsive genes in the liver.
[00121] Preferred compounds of the present invention include those described herein. In certain embodiments, the compounds may be thyroid hormone agonists of Formula I:
(Ar1)-G-(Ar2)-T-E
wherein:
1 2
Ar and Ar are substituted aryl or heteroaryl groups;
1 2
G is an atom or group of atoms that links Ar and Ar through 1-2 contiguous atoms;
T is an atom or group of atoms linking Ar to E through 1 -4 contiguous atoms or is absent; and
E is a functional group with a pKa < 4.0 containing a phosphorus atom, and prodrug thereof.
[00122] In particular embodiments, the thyroid hormone agonists of Formula I are a compound of Formula II:
wherein:
k is an integer from 0-4; m is an integer from 0-3; n is an integer from 0-2; p is an integer from 0-1;
E is a functional group with a pKa < 4.0 containing a phosphorus atom;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00123] In another embodiment, thyroid hormone agonists of Formula I include compounds of Formula III:
wherein:
n is an integer from 0-2;
p is an integer from 0-1;
Each Ra is independently selected from:
Y and Y' are each independently selected from the group consisting of -0-, and
-NRV-;
when Y is -O- and Y" is -Ci-C6-alkyl, or when Y and Y' are both -0-, R11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy, -C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry, -alkyl-S-S-alkylhydroxy, and -alkyl-S-S-S-alkylhydroxy;
when Y and Y' are both -NRV-, then R11 attached to -NRV- is independently selected from the group consisting of -H, -[C(Rz)2]q-COORy, -C(Rx)2COORy,
-[C(Rz)2]q-C(0)SRy, and -cycloalkylene-COORy;
when Y is -O- and Y' is NRV, then R11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl,
-C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy,
-C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry, -alkyl-S-S-alkylhydroxy, and -alkyl-S-S-S- alkylhydroxy; and R11 attached to -NRV- is independently selected from the group consisting of -H, -[C(Rz)2]q-COORy, -C(Rx)2COORy, -[C(Rz)2]q-C(0)SRy, and
-cycloalkylene-COORy;
or when Y and Y' are independently selected from -O- and -NRV-, then R11 and R11 together form a cyclic group comprising -alkyl-S-S-alkyl-, or together R11 and R11 are the group:
wherein:
V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or
together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both Y groups attached to the phosphorus; or
together V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or
together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or
together Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or
together W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
Z is selected from the group consisting of -CHRzOH, -CHRzOC(0)Ry,
-CHRzOC(S)Ry, -CHRzOC(S)ORy, -CHRzOC(0)SRy, -CHRzOC02Ry, -ORz,
-SRZ, -CHRZN3, -CH2aryl, -CH(aryl)OH, -CH(CH=CRz 2)OH,
-CH(C≡CRz)OH, -Rz, -NR -OCORy, -OC02Ry, -SCORy, -SC02Ry,
-NHCOR2, -NHC02Ry, -CH2NHaryl, -(CH2)q-ORz, and -(CH2)q-SRz;
q is an integer 2 or 3;
Each Rz is selected from the group consisting of Ry and -H;
Each Ry is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
Each Rx is independently selected from the group consisting of -H, and alkyl, or together Rx and Rx form a cycloalkyl group;
Each Rv is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl;
with the provisos that:
a) V, Z, W, W are not all -H; and
b) when Z is -Rz, then at least one of V, W, and W is not -H, alkyl, aralkyl, or heterocycloalkyl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00124] In certain preferred embodiments, R may be selected from:
Rf and Rg may together form:
[00129] In yet other embodiments, E may be selected from:
-P(0)[-OCRz 2OC(0)Ry] -P(0)[-OCRz 2OC(0)ORy] -P(0)[-N(H)CRz 2C(0)ORy]2
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected
from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
[00130] In yet other embodiments, T may be selected from:
E may be selected from: -P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2 -P(0)[-N(H)CRz 2C(0)ORy]2
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00131] In yet other embodiments, G may be selected from:
E may be selected from:-P03H2, -P(0)[-OCH2OC(0)-i-butyl]2,
-P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2, -P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl],
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH
(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3),
-P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3),
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00132] In yet other embodiments, the compounds of the invention exhibit metabolic instability which may be mediated by the deactivating enzyme selected from the group consisting of glutathione transferase, and cysteine β-lyase. In certain embodiments, the metabolically unstable substituent is contained within the T substituent of a compound of Formula III.
[00133] In such embodiments, T may be selected from:
[00134] More particularly, in certain preferred embodiments, T may be selected from:
[00137] In other embodiments, E may be se ected from: -P03H2,
-P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
[00138] and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
E may be selected from:-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2 -P(0)[-N(H)CRz 2C(0)ORy]
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected
from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00140] In yet other embodiments, G may be selected from:
R is OH;
E is selected from:-P03H2, -P(0)[-OCH2OC(0)-t-butyl]2, -P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2, -P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2, -P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2, -P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl],
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH (3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3), -P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3);
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00141] In yet other embodiments, the compounds of the invention exhibit metabolic instability which is mediated by the deactivating enzyme deiodinase. In certain embodiments, the compound of Formula III contains at least one metabolically unstable iodo substituent.
[00142] In such embodiments, T may be selected from:
[00145] In yet other embodiments, E may be selected from
-P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy] -P(0)[-N(H)CRz 2C(0)ORy]2
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
[00146] In yet other embodiments, G may be selected from:
Rg may be selected from:
with the proviso that a least R 1 , or R 3 is iodo;
E may be selected from: -F -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2 -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11 ] , -P(O) [-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00147] In certain preferred embodiments, G may be selected from:
each Re may independently be selected from:
with the proviso that a least R 1 , or R 3 is iodo;
R5 may be OH
E may be selected from:-P03H2, -P(0)[-OCH2OC(0)-t-butyl]2,
-P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2, -P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl],
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH
(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3),
-P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3);
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00148] In yet other embodiments, the compounds of the invention may exhibit metabolic instability which is mediated by the deactivating enzyme phosphatase. In certain embodiments, the metabolically unstable substituent may be contained within the E substituent of a compound of Formula III.
n is an integer from 0-2;
E is PCC OR OR11;
R attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy, -C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry,
-alkyl-S-S-alkylhydroxy, and -alkyl-S-S-S-alkylhydroxy;
or when Y and Y' are independently selected from -O- and -NRV-, then R11 and R11 together form a cyclic group comprising -alkyl-S-S-alkyl-, or together R11 and R11 are the group:
V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or
together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both Y groups attached to the phosphorus; or
together V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon
substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or
together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or
together Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or
together W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
Z is selected from the group consisting of -CHRzOH, -CHRzOC(0)Ry,
-CHRzOC(S)Ry, -CHRzOC(S)ORy, -CHRzOC(0)SRy, -CHRzOC02Ry, -ORz,
-SRZ, -CHRZN3, -CH2aryl, -CH(aryl)OH, -CH(CH=CRz 2)OH,
-CH(C≡CRz)OH, -Rz, -NR -OCORy, -OC02Ry, -SCORy, -SC02Ry,
-NHCOR2, -NHC02Ry, -CH2NHaryl, -(CH2)q-ORz, and -(CH2)q-SRz;
q is an integer 2 or 3;
Each Rz is selected from the group consisting of Ry and -H;
Each Ry is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
Each Rx is independently selected from the group consisting of -H, and alkyl, or together Rx and Rx form a cycloalkyl group;
Each Rv is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl;
with the provisos that:
a) V, Z, W, W are not all -H; and
b) when Z is -Rz, then at least one of V, W, and W is not -H, alkyl, aralkyl, or heterocycloalkyl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00150] In certain embodiments, T may be selected from:
[00153] In other embodiments, E may be se ected from: -P03H2,
-P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy]2 -P(0)[-OCH(V)CH2CH20-], wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
[00154] In yet other embodiments, G may be selected from:
R may be selected from:
E may be selected from:-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-OCH(V)CH2CH20-], wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
T may be selected from:
R may be OH
E may be selected from:-P03H2, -P(0)[-OCH2OC(0)-t-butyl]2,
-P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-OCH(3-chlorophenyl)CH2CH20-],
-P(0)[-OCH(pyrid-4-yl)CH2CH20-] ;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00156] In yet another embodiment the compounds are selected from the group consisting of
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
[00157] In other embodiments, the compounds of the invention may exhibit metabolic instability which is mediated by the deactivating enzyme selected from the group consisting of esterase, deiodinase, carboxylesterase, aldehyde oxidase, glutathione transferase, cysteine β-lyase, and phosphatase.
[00158] In yet other embodiments, the compounds of the invention may exhibit a metabolic instability which is mediated by the deactivating enzyme selected from the group consisting of the deactivating enzyme is selected from esterase, deiodinase, carboxylesterase, and aldehyde oxidase.
C. Preparation of Compounds of the Invention
[00159] The compounds in this invention may be prepared by the processes described herein in the following general schemes and examples, as well as relevant published literature procedures that are used by those skilled in the art. It should be understood that the following schemes are provided solely for the purpose of illustration and do not limit the invention which is defined by the claims.
[00160] All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. The compounds of the present invention can have stereogenic centers. Consequently, the compounds can exist in enantiomeric or diastereomeric forms or in mixture thereof. The processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When enantiomeric or diastereomeric products are prepared, they can be separated by conventional methods for example, chromatographic or fractional crystallization.
[00161] The synthesis of the scaffolds such as biaryl ethers and phenyl benzyl can be accomplished by processes known to one skilled in the art using the numerous methods
described in the prior art for the synthesis of thyromimetics. More specifically, thyromimetics with phosphonic and phosphinic acids were described in WO06128055 and WO06128056 (the contents of which are herein incorporated by reference in their entirety) modified as described herein and as recognized by those in the art.
[00162] For compounds of the invention with a metabolically unstable group at the R position such as esters or amides of amino acids, the group is introduced by functionalizing the position ortho to the phenol on the already built scaffold. Formylation followed by oxidation of the aldehyde results in the carboxylic acid which can be homologated and converted to esters by methods known to one skilled in the art. Amide coupling using one of the numerous coupling reagents known in the art, such as EDCI or making the acyl chloride with SOCl2, affords the desired amides (Bioorg Med. Chem. Lett. 14:3549 (2004)).
[00163] For compounds of the invention with a metabolically unstable group at the R position such as a sulfide, the group is introduced by treating the appropriate scaffold with a strong base such as BuLi and trapping the anion with a symmetrical disulfide. Alternatively, the anion can be trapped with Ss to give the ortho thiol. The thiol is then alkylated using procedures known to those skilled in the art such as treatment with Mel and triethylamine.
[00164] For compounds of the invention with a metabolically unstable group at the R position such as a disulfide, the ortho thiol described above is treated with a symmetrical disulfide.
[00165] For compounds of the invention with a metabolically unstable group at the R position such as a carbamate, an ortho hydroxyl group is introduced on an anisole scaffold by treating the anisole with BuLi and trapping the anion with trimethyl borate. The borate ester is then oxidized with hydrogen peroxide to the corresponding guaiacol which is then acylated with an isocyanate and deprotected. Alternatively, a phenolic scaffold can be hydroxylated with one of the numerous methods known to those skilled in the art using hydrogen peroxide and an oxidizing catalyst such as iron salt (ferrocene, Fe(acac)3) and selectively acylated with an isocyanate.
[00166] For compounds of the invention where the metabolically unstable group at the T position such as S-aryl phosphono-cysteine, thiophenols are synthesized from the corresponding phenol via palladium coupling of the respective aryl trifluoromethylsulfonate with trialkylsilyl thiolate (Tetrahedron Lett. 37:4523 (1996)) followed by removal of the silyl group with TBAF. The thiophenol is then alkylated using an activated phosphono-serine derivative (Angew. Chem. 98:836 (1986)) and deprotected.
[00167] Compounds of the invention where the metabolically unstable group is a phosphate or a phosphoramidate, are prepared by reaction of the corresponding thyromimetic scaffold bearing a phenol, alcohol or amine with a phosphoramidite, such as di-t-butyl-N,N-diisopropyl phosphoramidite, followed by oxidation of the phosphite or imidite with t-BuOOH, and t-butyl removal with TFA.
[00168] For compounds of the invention where the metabolically unstable group at the T position is a disulfide, the homo disulfide of the thiophenol described above is reacted with the desired thiol bearing a phosphonate group and deprotected
[00169] The synthesis of prodrugs of phosphonic and phosphinic acids can be accomplished by processes known to one skilled in the art using the numerous methods described in WO06128055 and WO06128056 (the contents of which are herein incorporated by reference in their entirety) modified as described herein and as recognized by those in the art.
[00170] Prodrugs of phosphates and phosphoramidates are made by preparing the phosphorylating reagents bearing the prodrug moiety, such as trans 2-(4-nitrophenyl)-4- aryl-2-oxo-l,3,2-dioxaphosphorinanes and bis-(pivaloyloxymethyl)-phosphoryl chloride, and phosphorylating phenols, alcohols or amines using processes known to those skilled in the art.
[00171] In certain preferred embodiments, compounds of the invention may be resolved to enantiomerically pure compositions or synthesized as enantiomerically pure
compositions using any method known in art. By way of example, compounds of the invention may be resolved by direct crystallization of enantiomer mixtures, by diastereomer salt formation of enantiomers, by the formation and separation of diasteriomers or by enzymatic resolution of a racemic mixture.
[00172] These and other reaction methodologies may be useful in preparing the compounds of the invention, as recognized by one of skill in the art. Various modifications to the above schemes and procedures will be apparent to one of skill in the art, and the invention is not limited specifically by the method of preparing the compounds of the invention.
D. Methods of the Invention
[00173] In another aspect of the invention, methods are provided. In preferred embodiments, the methods of the invention comprise administering a therapeutically effective amount of at least one compound of the invention, e.g., a compound of Formula I, II, or III. Relative activity of the compounds of the invention may be determined by any method known in the art, including the assay described herein.
[00174] In one aspect, the compounds of the invention and their prodrugs and salts are useful in preventing or treating arteriosclerosis by modulating levels of atherogenic proteins, e.g., Lp(a), apoAI, apoAII, LDL, HDL. Clinically overt hypothyroidism is associated with accelerated and premature coronary atherosclerosis and subclinical hypothyroidism is considered a condition with an increased risk for these diseases (Vanhaelst et al. and Bastenie et al., Lancet 2 (1967)).
[00175] T3 and T3 mimetics modulate atherogenic proteins in a manner that could prove beneficial for patients at risk to develop atherosclerosis or patients with atherosclerosis or diseases associated with atherosclerosis. T3 and T3 mimetics are known to decrease Lp(a) levels, e.g., in the monkey, with 3,5-dichloro-4-[4-hydroxy-3-(l- methylethyl)phenoxy]benzeneacetic acid (Graver et al, Proc. Natl. Acad. Sci. U.S.A. 700: 10067-10072 (2003)). In human hepatoma cells, the T3 mimetic CGS23425 ([[4-[4- hydroxy-3 -( 1 -methyl ethyl)phenoxy] -3 ,5 -dimethylphenyl] amino]oxo acetic acid) increased apoAI expression via thyroid hormone receptor activation (Taylor et al., Mol. Pharm. 52:542-547 (1997)).
[00176] Thus in one aspect, the compounds of the invention, their salts and prodrugs can be used to treat or prevent atherosclerosis, coronary heart disease and heart failure
because such compounds are expected to distribute to the liver and modulate the expression and production of atherogenic proteins.
[00177] In another aspect, the compounds of the invention and their prodrugs and salts are useful for preventing and/or treating metabolic diseases such as hypercholesterolemia and hyperlipidemia and conditions such as atherosclerosis, coronary heart disease, heart failure, nephrotic syndrome, and chronic renal failure without affecting thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4. Compounds previously reported that contain a carboxylic acid moiety and are not liver targeted, e.g., GC-1 ([4-[[4-hydroxy-3-(l-methylethyl)phenyl]methyl]-3,5- dimethylphenoxy] acetic acid)(Trost et al., Endocrinology 747:3057-3064 (2000)) and 3,5-Dichloro-4-[4-hydroxy-3-(l-methylethyl)phenoxy] benzeneacetic acid (Grover et al., Proc. Natl. Acad. Sci. U.S.A. 700: 10067-10072 (2003)) report that these ΤΡνβ-selective compounds dose-dependently lower cholesterol and TSH levels. Effects on cholesterol and TSH occur at the same dose or at doses stated to be not pharmacologically different (e.g., 2-fold).
[00178] Particularly useful T3 mimetics in these methods would reduce or minimize effects on thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4. Unlike prior T3 mimetics, the compounds or the present invention that distribute more readily to the liver and have a short half life result in pharmacological effects at doses that do not adversely affect thyroid function, thyroid production of circulating iodinated thyronines such as T3 and T4, and/or the ratio of T3 to T4.
[00179] In one embodiment the compounds of the present invention have a therapeutic index "TI", defined as the difference between the dose at which a significant effect is observed for a use disclosed herein, e.g., lowering lipids, lowering cholesterol, etc., and the dose at which a significant decrease in T3 or significant decrease in T4, or significant change in the ratio of T3 to T4 is observed, is at least 50 fold, 100 fold, 200 fold, 300 fold, 400 fold, 500 fold, 600 fold, 700 fold, 800 fold, 900 fold, 1000 fold, 2000 fold, 3000 fold, 4000 fold, 5000 fold, 6000 fold, 7000 fold, 8000 fold, 9000 fold or at least 10000 fold.
[00180] By way of example, an observed desired TI may include a significant effect on lowering lipids, e.g., 15% lipid lowering over baseline, without a corresponding significant effect, e.g., no more than a 25% reduction, on T4 in a non-rodent mammalian
species, by using a dosing frequency wherein drug levels are reduced from their Cmax at least 50%, preferably 80%, 90%, 95%, or 99%, prior to the next dose. In another example, rather than a significant amount, the amount of change in T3 or T4 may be a decrease selected from at least 5%, 10%>, 15%, 20%>, 25% or at least 30% of circulating levels.
[00181] In certain aspects, the compounds of the invention may be at least 2 to 10-fold more selective for the ΤΙ β-l receptor over the TRa-1 receptor, preferably at least 20-fold to 50-fold, more selective for TRP-l over TRa-1. In other aspects, the compounds of the invention exhibit a short plasma half-life, particularly in mammalian and more particularly human subjects.
[00182] In other embodiments, the compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects on cardiovascular function. Reduced or minimal effects may include clinically insignificant impact on cardiac function. By way of example, cardiac function may be evaluated by measuring changes in the maximum resting heart rate in a subject. Reduced or minimal effects may include a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%. In other embodiments, reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED5o for cholesterol lowering over 14 days. Alternatively one could use the minimally efficacious dose for cholesterol lowering. More particularly, clinically insignificant effects may include a resting heart rate of less than 100 bpm or an increase in resting heart rate of less than 20 bpm as compared to baseline or normal levels.
[00183] In certain aspects, TR agonists of this invention and prodrugs thereof lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on oxygen consumption. Again, reduced or minimal effects may be a change of less than 50% of baseline levels, more preferably less than 20% or less than 10%. Oxygen consumption may be measured in any suitable manner recognized in the art. In other embodiments, reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED50 for cholesterol lowering over 14 days. Again, alternatively one could use the minimally efficacious dose for cholesterol lowering.
[00184] In another embodiment, compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on muscle and bone function, as compared to baseline or normal levels. As
above, reduced or minimal effects may be a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%. In other embodiments, reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED50 for cholesterol lowering over 14 days for muscle effects, or over 6 months for bone effects (or minimally efficacious dose for cholesterol lowering). Thyroid hormones can affect bone mineral density and increase the risk of bone fractures. The following markers of bone turnover may be used to predict long-term changes in bone density: Serum osteocalcin and bone-specific alkaline phosphatase (B-ALP) are markers of bone formation, and urinary pyridinoline cross-link (Pyr) excretion is a marker of bone resorption.
[00185] Impact of thyroid hormones on muscle may be measured using any suitable assay methodology, including for example measurement of gastrocnemius muscle mass. 3-Methylhistidine urinary excretion and net balances across the leg or forearm may also be used as markers of contractile protein breakdown in muscle tissue.
[00186] In another embodiment, the compounds of the invention may be used to lower lipids at doses that exhibit reduced or minimal effects (including clinically insignificant effects) on thyroid hormones, and combinations thereof. As above, reduced or minimal effects may be a change of less than 50% of baseline or normal levels, more preferably less than 20%, less than 10%, or less than 3%. In other embodiments, reduced or minimal effects may be observed at 3 times, preferably 10 times, or 30 times the ED5o for cholesterol lowering over 14 days (or minimally efficacious dose for cholesterol lowering). In other aspects, the compounds of the invention have reduced or minimal effects on thyroid hormone levels at lipid lowering doses as compared to baseline levels.
Preferably, the effect is a change of less than 50% of baseline T4 levels, more preferably less than 20% or less than 10%.
[00187] In a particular embodiment, the compounds of the invention may be used to significantly lower cholesterol levels without having a significant effect on TSH levels. In another embodiment, the compounds of the present invention significantly lower cholesterol levels without lowering TSH levels by more than 30%, 25%, 20%, 15%, 10%, or 5%.
[00188] In another embodiment, the compounds of the invention may be dosed at a frequency that sustains cholesterol lowering over time with minimal effects on T4. By way of example, the dosing frequency may be determined by maintaining lipid lowering effect compared to baseline of at least 5%, 10%>, 15%, or 20%>. In preferred embodiments, dosing is once or twice daily, once every other day, every third day, 3, 2, or 1 times weekly, or once or twice monthly.
[00189] In certain aspects, the compounds of the invention may be liver targeted compounds that do not substantially impact other tissues. Demonstration of liver targeting and lack of impact on other tissues can be evaluated by measuring changes in mR As levels in liver as opposed to muscle, heart, pituitary, etc. Compounds of the invention may show changes in liver transcripts of genes encoding, for example, Dl, m- GPDH, CYP7a, malic enzyme, sterol regulating element binding protein lc (SREBPlc), LDL-cholesterol receptor in the liver, ΤΞΗβ, or Dl in the pituitary, Dl and m-GPDH in the heart, uncoupling protein 3 (UCP3) in the muscle. Such changes would be expected in liver, but no significant corresponding change in heart, muscle, pituitary, or other systemic tissues would be expected. Liver changes are of a magnitude of preferably at least 10%, 20%, 30%, 50%, 100%, 200%, 500% or more, while systemic tissue changes are preferably less than 100%, more preferably less than 50%>, 30%>, 20%>, or 10%>.
[00190] As discussed above, the previous use of T3 and T3 mimetics to treat metabolic diseases have been limited by the deleterious side-effects on the heart and THA. Previous attempts to overcome this limitation have first focused on ΤΡνβ-selective compounds to reduce cardiac side-effects, then liver-targeted compounds to reduce effects on the THA while eliminating cardiac side-effects. However, small decreases in total T4 were still observed over prolonged treatment with liver-targeted T3 mimetics. It was therefore unexpected when the present Inventors discovered that shortening the pharmacokinetic half life of liver-targeted thyroid hormone agonists led to a widening of the TI respective
to the THA. Thus the compounds of the present invention are able to increase the therapeutic index as compared to T3 and T3 mimetics. The compounds of the present invention can therefore be dosed at levels that are effective in treating metabolic and other disorders where the liver is the drug target without significantly negatively affecting THA.
[00191] Changes in the therapeutic index are readily determined using assays and methods well described in the literature. Genes in extrahepatic tissues are monitored using methods well understood by those skilled in the art. Assays include using cDNA microarray analysis of tissues isolated from treated animals. The sensitivity of the heart to T3 makes analysis of T3 -responsive genes in the heart as well as the functional consequences of these changes on cardiac properties one further strategy for evaluating the therapeutic index of the compounds of the present invention. Cardiac genes measured include mGPDH and myosin heavy and light chain. One method of measuring the effects of T3 mimetics on the heart is by the use of assays that measure T3 mediated myosin heavy chain gene transcription in the heart.
[00192] In one embodiment the compounds of the present invention have a therapeutic index, defined as the difference between the dose at which a significant effect is observed for a use disclosed herein, e.g., lowering cholesterol, and the dose at which a significant effect on a property or function, as disclosed herein {e.g., heart rate), is observed, is at least 50 fold, 100 fold, 200 fold, 300 fold, 400 fold, 500 fold, 600 fold, 700 fold, 800 fold, 900 fold, 1000 fold, 2000 fold, 3000 fold, 4000 fold, 5000 fold, 6000 fold, 7000 fold, 8000 fold, 9000 fold or at least 10000 fold. Examples of said use disclosed herein includes but is not limited to reducing lipid levels, increasing the ratio of HDL to LDL or apoAI to LDL, increasing mitochondrial biogenesis, AMP activated protein kinase or nuclear respiratory factor, or for the treatment or prevention of a disease or disorder selected from the group consisting of atherosclerosis, hypercholesterolemia, hyperlipidemia, NASH, NAFLD, nephrotic syndrome, chronic renal failure, metabolic syndrome X, hyperlipidemia, coronary heart disease, thyroid disease, thyroid cancer, depression, glaucoma, cardiac arrhythmias, heart failure, and osteoporosis. Examples wherein the property or function is a cardiac property/function include but are not limited to cardiac hypertrophy (heart weight to body weight ratio), heart rate, various hemodynamic parameters, including systolic and diastolic arterial pressure, end-systolic left ventricular pressure and maximal speeds of contraction and relaxation.
[00193] A variety of methods are described that provide a means for evaluating the functional consequences of T3 -cardiac action, including measurement of cardiac hypertrophy (heart weight to body weight ratio), heart rate, and various hemodynamic parameters, including systolic and diastolic arterial pressure, end-systolic left ventricular pressure and maximal speeds of contraction and relaxation using methods described by Trost et al, (Endocrinology 141:3057-64 (2000)). Other methods are also available to assess the therapeutic index including effects on muscle wasting, bone density, TSH levels, levels of T3 and T4, and the ratio T3/T4.
[00194] The therapeutic index is determined by administering to animals a wide range of doses and determining the minimal dose capable of inducing a response in the liver relative to the dose capable of inducing side effects in the heart or on the THA.
[00195] In vivo assays include but are not limited to treating animals with compounds of the invention or a prodrug thereof and monitoring the expression of T3-responsive genes in the liver or the functional consequences of changes of T3 -responsive genes.
[00196] In one aspect, compounds useful in the novel methods bind to thyroid receptors and produce changes in the expression of two or more hepatic genes. Animals used for testing compounds useful in the methods include normal rats and mice, animals made hypothyroid using methods well described in the literature, including thyroid hormone receptor knockout mice (e.g., TRa~ ~ such as those used in Grover et al, 2003), or animals exhibiting high cholesterol (e.g., high cholesterol fed rat or hamster), obesity and/or diabetes (e.g., fa/fa rat, Zucker diabetic fatty rat, ob/ob mice, db/db mice, high fat fed rodent). (Liureau et al, Biochem. Pharmacol. 35(70): 1691-6 (1986); Trost et al., Endocrinology 141 (9) :3057-64 (2000); and Grover et al, 2003).
[00197] The drug or prodrug may be administered by a variety of routes including by bolus injection, oral, and continuous infusion. By way of example, animals may be treated for 1-28 days and the liver, heart and blood are isolated. Changes in gene transcription relative to vehicle treated animals and T3 -treated animals determined using northern blot analysis, RNAase protection or reverse-transcription and subsequent PCR. While methods are available for monitoring changes in thousands of hepatic genes, only a small number need to be monitored to demonstrate the biological effect of compounds in this invention. Typically, genes such as spot-14, FAS, mGPDH, CPT-1, and LDL receptor may be monitored. Changes of >1.5 fold in two or more genes may be considered proof that the compound modulates T3 -responsive genes in vivo. Alternative
methods for measuring changes in gene transcription include monitoring the activity or expression level of the protein encoded by the gene. For instance, in cases where the genes encode enzyme activities (e.g., FAS, mGPDH), direct measurements of enzyme activity in appropriately extracted liver tissue can be made using standard enzymological techniques. In cases where the genes encode receptor functions (e.g., the LDL receptor), ligand binding studies or antibody-based assays (e.g., Western blots) can be performed to quantify the number of receptors expressed. Depending on the gene, TR agonists may either increase or decrease enzyme activity or increase or decrease receptor binding or number.
[00198] The functional consequences of changing the expression levels of hepatic genes responsive to T3 are many-fold and readily demonstrated using assays well described in the literature. Administering compounds of the invention that bind to a TR to animals can result in changes in lipids, including hepatic and/or plasma cholesterol levels; changes in lipoprotein levels including LDL-cholesterol, lipoprotein a (Lp(a)); and changes in energy expenditure as measured by changes in oxygen consumption and in some cases animal weight. For example, the effect on cholesterol may be determined using cholesterol fed animals such as normal rats and hamsters, or TRa ~ ~ knockout mice. Cholesterol may be measured using standard tests. Changes in energy expenditure may be monitored by measuring changes in oxygen consumption (MVo ). A variety of methods are well described in the literature and include measurement in the whole animal using Oxymax chambers (U.S. Patent No. 6,441,015). Livers from treated rats can also be evaluated (Fernandez et al., Toxicol. Lett. 69(2):205-l0 (1993)) as well as isolated mitochondria from liver (Carreras et al., Am. J. Physiol. Heart Circ. Physiol. 2Si(¾):H2282-8 (2001)). Hepatocytes from treated rats can also be evaluated (Ismail- Beigi F et al, J Gen Physiol. 75^:369-83 (1979)). .
[00199] Compounds of the invention that bind to a TR modulate expression of certain genes in the liver resulting in effects on lipids (e.g., cholesterol), lipoproteins, and triglycerides. Such compounds can lower cholesterol levels which is useful in the treatment of patients with hypercholesterolemia. Such compounds can lower levels of lipoproteins such as Lp(a) or LDL and are useful in preventing or treating atherosclerosis and heart disease in patients. Such compounds can raise levels of lipoproteins such as apoAI or HDL and are useful in preventing or treating atherosclerosis and heart disease in patients.
[00200] Also provided are methods of reducing plasma lipid levels in an animal, the method comprising the step of administering to a patient an amount of a compound of the invention. In one embodiment said compound is an active form. In another embodiment said compound is a prodrug. In another embodiment said compound of the invention comprises a stereocenter, is enantiomerically enriched or diastereomerically enriched, or a stereoisomer covered later. In another embodiment said compound is administered as a racemic mixture. In another embodiment said compound is administered as an enantiomerically enriched mixture. In another embodiment said compound is administered as a diastereomerically enriched mixture. In still another embodiment said compound is administered as an individual stereoisomer.
[00201] Also provided are methods of reducing plasma lipid levels in an animal wherein the lipid is cholesterol. In one embodiment said methods of reducing cholesterol results in a lowering of total cholesterol. In one embodiment said methods of reducing cholesterol results in a reduction of high density lipoprotein (HDL). In one embodiment said methods of reducing cholesterol results in a reduction of low-density lipoprotein (LDL). In one embodiment said methods of reducing cholesterol results in a reduction of very low-density lipoprotein (VLDL). In another embodiment said LDL is reduced to a greater extent than said HDL. In another embodiment said VLDL is reduced to a greater extent than said HDL. In another embodiment said VLDL is reduced to a greater extent than said LDL.
[00202] In one embodiment of the method of reducing lipids, the lipid is triglycerides. In one embodiment said lipid is liver triglycerides. In another embodiment said lipid is in the form of a lipoprotein. In another embodiment said lipoprotein is Lp(a). In another embodiment said lipoprotein is apoAII. Also provided are methods of increasing the ratio of HDL to LDL, HDL to VLDL, LDL to VLDL, apoAI to LDL or apoAI to VLDL in an animal. Also provided are methods of treating hyperlipidemia or hypercholesterolemia in an animal,
[00203] Also provided are methods of preventing or treating atherosclerosis in an animal. Also provided are methods of reducing fat content in the liver or of preventing or treating fatty liver/steatosis, NASH or NAFLD in an animal. Also provided are methods of preventing or treating nephrotic syndrome or chronic renal failure in an animal. Also provided are methods of preventing or treating coronary heart disease in an animal.
[00204] Also provided are methods of preventing or treating a liver disease responsive to modulation of T3-responsive genes in an animal. Also provided are methods of preventing or treating thyroid disease, thyroid cancer, depression, glaucoma, cardiac arrhythmias, heart failure, or osteoporosis in an animal. Also provided are methods of increasing mitochondrial biogenesis in an animal. Also provided are methods of increasing expression of AMP activated protein kinase or nuclear respiratory factor in an animal
[00205] In all methods described above, the methods generally comprise the step of administering to a patient in need thereof, such as an animal subject including a human subject, an effective amount of a compound of the invention. In one embodiment said compound is an active form. In another embodiment said compound is a prodrug. In another embodiment said compound of the invention comprises a stereocenter. In another embodiment said compound is administered as a racemic mixture. In another embodiment said compound is administered as an enantiomerically enriched mixture. In another embodiment said compound is a administered as a diastereomeric mixture. In still another embodiment said compound is administered as an individual stereoisomer.
[00206] Without intending to be limited by theory, it is believed that the methods of the present invention act through a combination of mechanisms. The liver is a major target organ of thyroid hormone with an estimated 8% of the hepatic genes regulated by thyroid hormone. Quantitative fluorescent-labeled cDNA microarray hybridization was used to identify thyroid-responsive genes in the liver as shown in Table 1 below (Feng et al, Mol. Endocrinol. 14:941-955 (2000)). Hepatic RNAs from T3-treated and hypothyroid mice were used in the study. Thyroid hormone treatment affected the expression of 55 genes from the 2225 different mouse genes sampled with 14 increasing >2-fold and 41 decreasing >60%.
TABLE 1
[00207] Genes reported to be affected by thyroid hormone are identified using a variety of techniques include microarray analysis. Studies have identified genes that are affected by T3 and T3 mimetics that are important in metabolic diseases.
[00208] T3-responsive genes in the liver include genes affecting lipogenesis, including spot 14, fatty acid transport protein, malic enzyme, fatty acid synthase (Blennemann et al.5 Mol. Cell. Endocrinol. 110(l-2): \-% (1995)) and CYP4A. HMG CoA reductase and LDL receptor genes have been identified as affecting cholesterol synthesis and as being responsive to T3. CPT-1 is a T3-responsive gene involved in fatty acid oxidation. Genes affecting energy expenditure, including mitochondrial genes such as mitochondrial sn- glycerol 3-phosphate dehydrogenase (mGPDH), and/or enzymes associated with proton leakage such as the adenine nucleotide transporter (ANT), Na+/K+-ATPase, Ca2+-ATPase and ATP synthase are also T3 -responsive genes. T3 -responsive genes affecting glycogeno lysis and gluconeogenesis include glucose 6-phosphatase and PEPCK.
[00209] Thyroid hormone-responsive genes in the heart are not as well described as the liver but could be determined using similar techniques as described by Feng et al. Many of the genes described to be affected in the heart are the same as described above for the liver. Common genes evaluated include mitochondrial sn-glycerol 3-phosphate dehydrogenase (mGPDH), and myosin heavy and light chains (Danzi et al., Thyroid 72^:467-72 (2002)).
[00210] Compounds used in the methods bind to thyroid receptors and produce a change in some hepatic gene expression. Evidence for agonist activity may be obtained using standard assays described in the literature.
[00211] In other embodiments, high triglycerides may be reduced, which in turn may reduce risk/incidence of pancreatitis (prevention/treatment); reduce risk of major advanced cardiac event (MACE) (stroke, heart attack) via reduced triglycerides, Lp(a). In certain preferred embodiments, a specific population with only increased Lp(a) may be identifed and preferentially treated.
E. Metabolites of the Compounds of the Invention
[00212] Also falling within the scope of the present invention are the in vivo metabolic products of the compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammalian tissue or a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radio-labeled {e.g. C^ or H^) compound of the invention, administering it in a detectable
dose (e.g., greater than about 0.5 mg/kg) to a mammal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours), and isolating its conversion products from urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites may be done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no biological activity of their own.
F. Pharmaceutical Compositions of the Invention
[00213] While it is possible for the compounds of the present invention to be administered neat, it may be preferable to formulate the compounds as pharmaceutical compositions. As such, in yet another aspect of the invention, pharmaceutical compositions useful in the methods of the invention are provided. The pharmaceutical compositions of the invention may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form. The pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5.0 to about pH 8.0.
[00214] More particularly, the pharmaceutical compositions of the invention comprise a therapeutically or prophylactically effective amount of at least one compound of the present invention, together with one or more pharmaceutically acceptable excipients. Optionally, the pharmaceutical compositions of the invention may comprise a combination of compounds of the present invention, or may include a second active ingredient useful in a method disclosed herein.
[00215] Formulations of the present invention, e.g., for parenteral or oral administration, are most typically solids, liquid solutions, emulsions or suspensions, while inhaleable formulations for pulmonary administration are generally liquids or powders, with powder formulations being generally preferred. A preferred pharmaceutical
composition of the invention may also be formulated as a lyophilized solid that is reconstituted with a physiologically compatible solvent prior to administration. Alternative pharmaceutical compositions of the invention may be formulated as syrups, creams, ointments, tablets, and the like.
[00216] The pharmaceutical compositions of the invention can be administered to the subject via any drug delivery route known in the art. Specific exemplary administration routes include oral, ocular, rectal, buccal, topical, nasal, ophthalmic, subcutaneous, intramuscular, intraveneous (bolus and infusion), intracerebral, transdermal, and pulmonary.
[00217] The term "pharmaceutically acceptable excipient" refers to an excipient for administration of a pharmaceutical agent, such as the compounds of the present invention. The term refers to any pharmaceutical excipient that may be administered without undue toxicity. Pharmaceutically acceptable excipients are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington's Pharmaceutical Sciences).
[00218] Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Other exemplary excipients include antioxidants such as ascorbic acid; chelating agents such as EDTA; carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid; liquids such as oils, water, saline, glycerol and ethanol; wetting or emulsifying agents; pH buffering substances; and the like. Liposomes are also included within the definition of pharmaceutically acceptable excipients.
[00219] The pharmaceutical compositions of the invention may be formulated in any form suitable for the intended method of administration. When intended for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or
more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
[00220] The therapeutically effective amount, as used herein, refers to an amount of a pharmaceutical composition of the invention to treat, ameliorate, or modulate an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect. The effect can be detected by, for example, assays of the present invention. The effect can also be the prevention of a disease or condition where the disease or condition is predicted for an individual or a high percentage of a population.
[00221] The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; the therapeutic or combination of therapeutics selected for administration, the protein half-life, the mRNA half-life and the protein localization. Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician.
[00222] For any compound, the therapeutically effective amount can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED5o (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, ED50/LD50. Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies may be used in formulating a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that include an ED5o with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
[00223] The magnitude of a prophylactic or therapeutic dose of a particular active ingredient of the invention in the acute or chronic management of a disease or condition will vary, however, with the nature and severity of the disease or condition, and the route
by which the active ingredient is administered. The dose, and perhaps the dose frequency, will also vary according to the age, body weight, and response of the individual patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response.
[00224] In one aspect, the compounds of the invention are administered orally in a total daily dose of about 0.375 μg/kg/day to about 3.75 mg/kg/day. In another aspect the total daily dose is from about 3.75 μg/kg/day to about 0.375 mg/kg/day. In another aspect the total daily dose is from about 3.75 μg/kg/day to about 37.5 μg/kg/day. In another aspect the total daily dose is from about 3.75 μg/kg/day to about 60 μg/kg/day. In a further aspect the dose range is from 30 μg/kg/day to 3.0 mg/kg/day. In one aspect, the compounds of the invention are administered orally in a unit dose of about 0.375 μg/kg to about 3.75 mg/kg. In another aspect the unit dose is from about 3.75 μg/kg to about 0.375 mg/kg. In another aspect the unit dose is from about 3.75 μg/kg to about 37.5 μg/kg. In another aspect the unit dose is from about 3.75 μg/kg to about 60 μg/kg. In one aspect, the compounds of the invention are administered orally in a unit dose of about 0.188 μg/kg to about 1.88 mg/kg. In another aspect the unit dose is from about 1.88 μg/kg to about 0.188 mg/kg. In another aspect the unit dose is from about 1.88 μg/kg to about 18.8 μg/kg. In another aspect the unit dose is from about 1.88 μg/kg to about 30 μg/kg. In one aspect, the compounds of the invention are administered orally in a unit dose of about 0.125 μg/kg to about 1.25 mg/kg. In another aspect the unit dose is from about 1.25 μg/kg to about 0.125 mg/kg. In another aspect the unit dose is from about 1.25 μg/kg to about 12.5 μg/kg. In another aspect the unit dose is from about 1.25 μg/kg to about 20 μg/kg. In one embodiment the unit dose is administered once a day. In another embodiment the unit dose is administered twice a day. In another embodiment the unit dose is administered three times a day. In another embodiment the unit dose is administered four times a day.
[00225] Dose refers to the equivalent of the free acid. The use of controlled-release preparations to control the rate of release of the active ingredient may be preferred. The daily dose may be administered in multiple divided doses over the period of a day. Doses
and dosing schedules may be adjusted to the form of the drug or form of delivery used. For example, different dosages and scheduling of doses may be used when the form of the drug is in a controlled release form or intravenous delivery is used with a liquid form.
[00226] The phrases "therapeutically effective amount", "prophylactically effective amount" and "therapeutically or prophylactically effective amount," as used herein encompass the above described dosage amounts and dose frequency schedules. Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to treat or prevent such diseases, but insufficient to cause, or sufficient to reduce, adverse effects associated with conventional therapies are also encompassed by the above described dosage amounts and dose frequency schedules.
[00227] The exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time, protein of interest half- life, R A of interest half-life, frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.
G. Combination Therapy
[00228] It is also possible to combine any compound of the present invention with one or more other active ingredients useful in the methods described herein, including compounds in a unitary dosage form, or in separate dosage forms intended for simultaneous or sequential administration to a patient in need of treatment. When administered sequentially, the combination may be administered in two or more administrations. In an alternative embodiment, it is possible to administer one or more compounds of the present invention and one or more additional active ingredients by different routes.
[00229] The skilled artisan will recognize that a variety of active ingredients may be administered in combination with the compounds of the present invention that may act to augment or synergistically enhance the activity of the compounds of the invention.
[00230] In certain aspects, the compounds of the invention may be combined with one or more lipid lowering agents such as statins or cholesterol absorption inhibitors to treat patients with hyperlipidemia. Preferably, such combination allows therapeutic effect at a reduced dose of one or more of the agents, improves lipid profile, or improves safety/therapeutic index of the therapy or one or more of the agents.
[00231] According to the methods of the invention, the combination of active ingredients may be: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by any other combination therapy regimen known in the art. When delivered in alternation therapy, the methods of the invention may comprise administering or delivering the active ingredients sequentially, e.g., in separate solution, emulsion, suspension, tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in simultaneous therapy, effective dosages of two or more active ingredients are administered together. Various sequences of intermittent combination therapy may also be used.
[00232] By way of example, the compounds of the present invention can be administered in combination with other pharmaceutical agents that are used to lower serum cholesterol such as a cholesterol biosynthesis inhibitor or a cholesterol absorption inhibitor, especially a HMG-CoA reductase inhibitor, or a HMG-CoA synthase inhibitor, or a HMG-CoA reductase or synthase gene expression inhibitor, a cholesteryl ester transfer protein (CETP) inhibitor (e.g., torcetrapib), a bile acid sequesterant (e.g., cholestyramine (Questran®), colesevelam and colestipol (Colestid®)), or a bile acid reabsorption inhibitor (see, for example, U.S. Pat. No. 6,245,744, U.S. Pat. No. 6,221,897, U.S. Pat. No. 6,277,831, EP 0683 773, EP 0683 774), a cholesterol absorption inhibitor as described (e.g., ezetimibe, tiqueside, pamaqueside or see, e.g., in WO 0250027), a PPARalpha agonist, a mixed PPAR alpha/gamma agonist such as, for example, AZ 242 (Tesaglitazar, (S)-3-(4-[2-(4- methanesulfonyloxyphenyl)ethoxy]phenyl)-2-ethoxypropionic acid), BMS 298585 (N- [(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4- oxazolyl)ethoxy]phenyl]methyl]glycine) or as described in WO 99/62872, WO 99/62871, WO 01/40171, WO 01/40169, W096/38428, WO 01/81327, WO 01/21602, WO 03/020269, WO 00/64888 or WO 00/64876, a MTP inhibitor such as, for example,
implitapide, a fibrate, an ACAT inhibitor (e.g., avasimibe), an angiotensin II receptor antagonist, a squalene synthetase inhibitor, a squalene epoxidase inhibitor, a squalene cyclase inhibitor, combined squalene epoxidase/squalene cyclase inhibitor, a lipoprotein lipase inhibitor, an ATP citrate lyase inhibitor, lipoprotein(a) antagonist, an antioxidant or niacin (e.g., slow release niacin). The compounds of the present invention may also be administered in combination with a naturally occurring compound that acts to lower plasma cholesterol levels. Such naturally occurring compounds are commonly called nutraceuticals and include, for example, garlic extract and niacin.
[00233] In one aspect, the HMG-CoA reductase inhibitor is from a class of therapeutics commonly called statins. Examples of HMG-CoA reductase inhibitors that may be used include but are not limited to lovastatin (MEVACOR; see U.S. Pat. Nos. 4,231,938; 4,294,926; 4,319,039), simvastatin (ZOCOR; see U.S. Pat. Nos. 4,444,784; 4,450,171, 4,820,850; 4,916,239), pravastatin (PRAVACHOL; see U.S. Pat. Nos. 4,346,227; 4,537,859; 4,410,629; 5,030,447 and 5,180,589), lactones of pravastatin (see U.S. Pat. No. 4,448,979), fluvastatin (LESCOL; see U.S. Pat. Nos. 5,354,772; 4,911,165; 4,739,073; 4,929,437; 5,189,164; 5,118,853; 5,290,946; 5,356,896), lactones of fluvastatin, atorvastatin (LIPITOR; see U.S. Pat. Nos. 5,273,995; 4,681,893; 5,489,691; 5,342,952), lactones of atorvastatin, cerivastatin (also known as rivastatin and BAYCHOL; see U.S. Pat. No. 5,177,080, and European Application No. EP-491226A), lactones of cerivastatin, rosuvastatin (CRESTOR; see U.S. Pat. Nos. 5,260,440 and RE37314, and European Patent No. EP521471), lactones of rosuvastatin, itavastatin, nisvastatin, visastatin, atavastatin, bervastatin, compactin, dihydrocompactin, dalvastatin, fluindostatin, pitivastatin, mevastatin (see U.S. Pat. No. 3,983,140), and velostatin (also referred to as synvinolin). Other examples of HMG-CoA reductase inhibitors are described in U.S. Pat. Nos. 5,217,992; 5,196,440; 5,189,180; 5,166,364; 5,157,134; 5,110,940; 5,106,992; 5,099,035; 5,081,136; 5,049,696; 5,049,577; 5,025,017; 5,011,947; 5,010,105; 4,970,221; 4,940,800; 4,866,058; 4,686,237; 4,647,576; European Application Nos. 0142146A2 and 0221025A1; and PCT Application Nos. WO 86/03488 and WO 86/07054. Also included are pharmaceutically acceptable forms of the above. All of the above references are incorporated herein by reference.
[00234] Non-limiting examples of suitable bile acid sequestrants include cholestyramine (a styrene-divinylbenzene copolymer containing quaternary ammonium cationic groups capable of binding bile acids, such as QUESTRAN or QUESTRAN
LIGHT cholestyramine which are available from Bristol-Myers Squibb), colestipol (a copolymer of diethyl enetriamine and l-chloro-2,3-epoxypropane, such as COLESTID tablets which are available from Pharmacia), colesevelam hydrochloride (such as WelChol Tablets (poly(allylamine hydrochloride) cross-linked with epichlorohydrin and alkylated with 1-bromodecane and (6-bromohexyl)-trimethylammonium bromide) which are available from Sankyo), water soluble derivatives such as 3,3-ioene, N- (cycloalkyl)alkylamines and poliglusam, insoluble quaternized polystyrenes, saponins and mixtures thereof. Other useful bile acid sequestrants are disclosed in PCT Patent Applications Nos. WO 97/11345 and WO 98/57652, and U.S. Pat. Nos. 3,692,895 and 5,703,188 which are incorporated herein by reference. Suitable inorganic cholesterol sequestrants include bismuth salicylate plus montmorillonite clay, aluminum hydroxide and calcium carbonate antacids.
[00235] In the above description, a fibrate base compound is a medicament for inhibiting synthesis and secretion of triglycerides in the liver and activating lipoprotein lipase, thereby lowering the triglyceride level in the blood. Examples include bezafibrate, beclofibrate, binifibrate, ciprofibrate, clinofibrate, clofibrate, clofibric acid, ethofibrate, fenofibrate, gemfibrozil, nicofibrate, pirifibrate, ronifibrate, simfibrate and theofibrate. Such an AC AT inhibitor includes, for example: a compound having the general formula (I) disclosed in WO 92/09561 [preferably FR-129169, of which the chemical name is N- (l,2-diphenylethyl)-2-(2-octyloxyphenyl)acetamide]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 8-510256 (WO 94/26702, U.S. Pat. No. 5,491,172) {preferably CI-1011, of which the chemical name is 2,6-diisopropylphenyl-N-[(2,4,6-triisopropylphenyl)acetyl]sulfamate, and in the present invention CI-1011 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in EP 421441 (U.S. Pat. No. 5,120,738) {preferably F-1394, of which the chemical name is (lS,2S)-2-[3-(2,2-dimethylpropyl)-3-nonylureido]cyclohexan-l-yl 3-[(4R)-N-(2,2,5,5- tetramethyl-1,- 3-dioxane-4-carbonyl)amino]propionate, and in the present invention F- 1394 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a compound including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) 2000-500771 (WO
97/19918, U.S. Pat. No. 5,990,173) [preferably F-12511, of which the chemical name is (S)-2',3',5'-trimethyl-4'-hydroxy-a-dodecylthio-.alpha.-phenylacetanilide, and in the present invention F-1251 1 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 10-195037 (EP 790240, U.S. Pat. No. 5,849,732) [preferably T-2591, of which the chemical name is l-(3-t-butyl-2-hydroxy-5- methoxyphenyl)-3-(2-cyclohexylethyl)-3-(4-dimethylaminophenyl)urea, and in the present invention T-2591 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in WO 96/26948 {preferably FCE-28654, of which the chemical name is l-(2,6- diisopropylphenyl)-3-[(4R,5R)-4,5-dimethyl-2-(4-phosphonophenyl)-l,3-dioxolan-2- ylmethyl]urea, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a compound having the general formula (I) or a pharmacologically acceptable salt thereof disclosed in the specification of WO 98/54153 (EP 987254) {preferably K- 10085, of which the chemical name is N-[2,4-bis(methylthio)-6-methyl-3-pyridyl]-2-[4- [2-(oxazolo[4,5-b]pyridine-2-ylthio)ethyl]piperazin- 1 -yl]acetamide, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a compound having the general formula (I) disclosed in WO 92/09572 (EP 559898, U.S. Pat. No. 5,475,130) [preferably HL-004, of which the chemical name is N-(2,6- diisopropylphenyl)-2-tetradecylthioacetamide]; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 7-82232 (EP 718281) {preferably NTE-122, of which the chemical name is trans- l,4-bis[l -cyclohexyl-3-(4- dimethylaminophenyl)ureidomethyl]cyclohexane, and in the present invention NTE-122 includes pharmacologically acceptable salts of NTE-122}; a compound including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 10-510512 (WO 96/10559) {preferably FR- 186054, of which the chemical name is 1 -benzyl- l-[3-(pyrazol-3-yl)benzyl]-3-[2,4- bis(methylthio)-6-methylpyridi- n-3-yl]urea, and in the present invention FR-186054 including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a compound having the general Formula I including a pharmacologically acceptable
salt/co-crystal, ester or prodrug thereof disclosed in WO 96/09287 (EP 0782986, U.S. Pat. No. 5,990,150) [preferably N-(l-pentyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide, and in the present invention including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof]; and a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in WO 97/12860 (EP 0866059, U.S. Pat. No. 6,063,806) [preferably N- (l-octyl-5-carboxymethyl-4,6-dimethylindolin-7-yl)-2,2-dimethylpropaneamide, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof]. The ACAT inhibitor preferably is a compound selected from the group consisting of FR129169, CI-1011, F-1394, F-12511, T-2591, FCE-28654, K-10085, HL-004, NTE-122, FPv-186054, N-(l -octyl-5-carboxymethyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (hereinafter referred as compound A), and N-(l-pentyl-4,6- dimethylindolin-7-yl)-2,2-dimethylpropaneamide (hereinafter referred as compound B), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof. The ACAT inhibitor more preferably is a compound selected from the group consisting of CI- 1011, F-12511, N-(l-octyl-5-carboxymethyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (compound A), and N-(l-pentyl-4,6-dimethylindolin-7-yl)-2,2- dimethylpropaneamide (compound B), including a pharmacologically acceptable salt/co- crystal, ester or prodrug thereof; most preferred is N-(l-octyl-5-carboxymethyl-4,6- dimethylindolin-7-yl)-2,2-dimethylpropaneamide (compound A).
[00236] An angiotensin II receptor antagonist includes, for example, a biphenyl tetrazole compound or biphenylcarboxylic acid derivative such as: a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Sho 63-23868 (U.S. Pat. No. 5,138,069) {preferably losartan, of which the chemical name is 2-butyl-4-chloro- 1- [2 '-(lH-tetrazol-5-yl)biphenyl-4-ylmethyl]-lH-imidazol-5 -methanol, and in the present invention losartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof }; a compound having the general formula (I) including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kohyo) Hei 4-506222 (WO 91/14679) {preferably irbesartan, of which the chemical name is 2-N-butyl-4-spirocyclopentane-l-[2'-(lH- tetrazol-5-yl)biphenyl-4-ylmethyl]-2-imidazoline-5-one, and in the present invention irbesartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug
thereof}; a compound having the general formula (I), an ester thereof, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 4-235149 (EP 433983) {preferably valsartan, of which the chemical name is (S)-N-valeryl-N- [2 '-(1 H-tetrazol-5 -yl)biphenyl-4- ylmethyljvaline, and in the present invention valsartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a carboxylic acid derivative having the general formula (I), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 4-364171 (U.S. Pat. No. 5,196,444) {preferably candesartan, of which the chemical name is 1- (cyclohexyloxycarbonyloxy)ethyl 2-ethoxy- 1 - [2 ' -( 1 H-tetrazol-5 -yl)biphenyl-4-ylmethyl] - lH-benzimidazole-7-carboxylate, and in the present invention candesartan including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof (TCV-116 or the like), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof}; a carboxylic acid derivative having the general formula (I), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 5-78328 (U.S. Pat. No. 5,616,599) {preferably olmesartan, of which the chemical name is (5-methyl-2-oxo-l,3-dioxolen-4-yl)methyl 4-( 1 -hydroxy- 1- methylethyl)-2-pr- opyl- 1 - [2 ' -( 1 H-tetrazol-5 -yl)biphenyl-4-ylmethyl]imidazole-5 - carboxylate, and in the present invention olmesartan includes carboxylic acid derivatives thereof, pharmacologically acceptable esters of the carboxylic acid derivatives (CS-866 or the like), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof } ; and a compound having the general formula (I), including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof disclosed in the Japanese Patent Publication (Kokai) Hei 4-346978 (U.S. Pat. No. 5,591,762, EP 502,314) {preferably telmisartan, of which the chemical name is 4'-[[2-n-propyl-4-methyl-6-(l- methylbenzimidazol-2-yl)-benzimidazol- 1 -yl]- methyl]biphenyl-2-carboxylate, including a pharmacologically acceptable salt/co-crystal, ester or prodrug thereof} . The angiotensin II receptor antagonist preferably is losartan, irbesartan, valsartan, candesartan, olmesartan, or telmisartan; more preferred is losartan or olmesartan; and most preferred is olmesartan.
[00237] In addition to being useful in treating or preventing certain diseases and disorders, combination therapy with compounds of this invention maybe useful in reducing the dosage of the second drug or agent (e.g., atorvastatin).
[00238] In addition, the compounds of the present invention can be used in combination with an apolipoprotein B secretion inhibitor and/or microsomal triglyceride transfer protein (MTP) inhibitor. Some apolipoprotein B secretion inhibitors and/or MTP inhibitors are disclosed in U.S. 5,919,795.
[00239] Any HMG-CoA reductase inhibitor may be employed as an additional compound in the combination therapy aspect of the present invention. The term HMG- CoA reductase inhibitor refers to a compound that inhibits the biotransformation of hydroxymethylglutaryl-coenzyme A to mevalonic acid as catalyzed by the enzyme HMG- CoA reductase. Such inhibition may be determined readily by one of skill in the art according to standard assays (e.g., Methods of Enzymology, 71 : 455-509 (1981); and the references cited therein). A variety of these compounds are described and referenced below. U.S. 4,231,938 discloses certain compounds isolated after cultivation of a microorganism belonging to the genus Aspergillus, such as lovastatin. Also U.S. 4,444,784 discloses synthetic derivatives of the aforementioned compounds, such as simvastatin. Additionally, U.S. 4,739,073 discloses certain substituted indoles, such as fluvastatin. Further, U.S. 4,346,227 discloses ML-236B derivatives, such as pravastatin. In addition, EP 491,226 teaches certain pyridyldihydroxyheptenoic acids, such as rivastatin. Also, U.S. 4,647,576 discloses certain 6-[2-(substituted-pyrrol-l-yl)-alkyl]- pyran-2-ones such as atorvastatin. Other HMG-CoA reductase inhibitors will be known to those skilled in the art. Examples of currently or previously marketed products containing HMG-CoA reductase inhibitors include cerivastatin Na, rosuvastatin Ca, fluvastatin, atorvastatin, lovastatin, pravastatin Na and simvastatin.
[00240] Any HMG-CoA synthase inhibitor may be used as an additional compound in the combination therapy aspect of this invention. The term HMG-CoA synthase inhibitor refers to a compound that inhibits the biosynthesis of hydroxymethylglutaryl-coenzyme A from acetyl-coenzyme A and acetoacetyl-coenzyme A, catalyzed by the enzyme HMG- CoA synthase. Such inhibition may be determined readily by one of skill in the art according to standard assays (e.g., Methods of Enzymology 35: 155-160 (1975); and Methods of Enzymology, 110: 19-26 (1985); and the references cited therein). A variety of these compounds are described and referenced below. U.S. 5,120,729 discloses certain beta-lactam derivatives. U.S. 5,064,856 discloses certain spiro-lactone derivatives prepared by culturing the microorganism MF5253. U.S. 4,847,271 discloses certain oxetane compounds such as 1 l-(3-hydroxymethyl-4-oxo-2-oxetayl)-
3,5,7-trimethyl-2,4-undecadienoic acid derivatives. Other HMG-CoA synthase inhibitors useful in the methods, compositions and kits of the present invention will be known to those skilled in the art.
[00241] Any compound that decreases HMG-CoA reductase gene expression may be used as an additional compound in the combination therapy aspect of this invention. These agents may be HMG-CoA reductase transcription inhibitors that block the transcription of DNA or translation inhibitors that prevent translation of mRNA coding for HMG-CoA reductase into protein. Such inhibitors may either affect transcription or translation directly, or may be biotransformed into compounds that have the aforementioned attributes by one or more enzymes in the cholesterol biosynthetic cascade or may lead to the accumulation of an isoprene metabolite that has the aforementioned activities. Such regulation is readily determined by those skilled in the art according to standard assays (Methods of Enzymology, 110: 9-19 (1985)). Several such compounds are described and referenced below; however, other inhibitors of HMG-CoA reductase gene expression will be known to those skilled in the art, for example, U.S. 5,041,432 discloses certain 15 -substituted lanosterol derivatives that are inhibitors of HMG-CoA reductase gene expression. Other oxygenated sterols that suppress the biosynthesis of HMG-CoA reductase are discussed by E. I. Mercer (Prog. Lip. Res., 32:357-416 (1993)).
[00242] Any compound having activity as a CETP inhibitor can serve as the second compound in the combination therapy aspect of the instant invention. The term CETP inhibitor refers to compounds that inhibit the cholesteryl ester transfer protein (CETP) mediated transport of various cholesteryl esters and triglycerides from HDL to LDL and VLDL. A variety of these compounds are described and referenced below; however, other CETP inhibitors will be known to those skilled in the art. U.S. 5,512,548 discloses certain polypeptide derivatives having activity as CETP inhibitors, while certain CETP- inhibitory rosenonolactone derivatives and phosphate-containing analogs of cholesteryl ester are disclosed in J. Antibiot., 49(8): 815-816 (1996), and Bioorg. Med. Chem. Lett., 6: 1951-1954 (1996), respectively.
[00243] Any ACAT inhibitor can serve as an additional compound in the combination therapy aspect of this invention. The term ACAT inhibitor refers to a compound that inhibits the intracellular esterification of dietary cholesterol by the enzyme acyl CoA: cholesterol acyltransferase. Such inhibition may be determined readily by one of skill in the art according to standard assays, such as the method of Heider et al. described in
Journal of Lipid Research, 24:1127 (1983). A variety of these compounds are described and referenced below; however, other ACAT inhibitors will be known to those skilled in the art. U.S. 5,510,379 discloses certain carboxysulfonates, while WO 96/26948 and WO 96/10559 both disclose urea derivatives having ACAT inhibitory activity.
[00244] Any compound having activity as a squalene synthetase inhibitor can serve as an additional compound in the combination therapy aspect of the instant invention. The term squalene synthetase inhibitor refers to compounds that inhibit the condensation of two molecules of farnesylpyrophosphate to form squalene, a reaction that is catalyzed by the enzyme squalene synthetase. Such inhibition is readily determined by those skilled in the art according to standard methodology (Methods of Enzymology 15:393-454 (1969); and Methods of Enzymology 110: 359-373 (1985); and references cited therein). A summary of squalene synthetase inhibitors has been compiled in Curr. Op. Ther Patents, 861-4, (1993). EP 0 567 026 Al discloses certain 4,1-benzoxazepine derivatives as squalene synthetase inhibitors and their use in the treatment of hypercholesterolemia and as fungicides. EP 0 645 378 Al discloses certain seven- or eight-membered heterocycles as squalene synthetase inhibitors and their use in the treatment and prevention of hypercholesterolemia and fungal infections. EP 0 645 377 Al discloses certain benzoxazepine derivatives as squalene synthetase inhibitors useful for the treatment of hypercholesterolemia or coronary sclerosis. EP 0 611 749 Al discloses certain substituted amic acid derivatives useful for the treatment of arteriosclerosis. EP 0 705 607 A2 discloses certain condensed seven- or eight-membered heterocyclic compounds useful as antihypertriglyceridemic agents. WO 96/09827 discloses certain combinations of cholesterol absorption inhibitors and cholesterol biosynthesis inhibitors including benzoxazepine derivatives and benzothiazepine derivatives. EP 0 701 725 Al discloses a process for preparing certain optically-active compounds, including benzoxazepine derivatives, having plasma cholesterol and triglyceride lowering activities.
[00245] Other compounds that are currently or previously marketed for hyperlipidemia, including hypercholesterolemia, and which are intended to help prevent or treat atherosclerosis, include bile acid sequestrants, such as colestipol HC1 and cholestyramine; and fibric acid derivatives, such as clofibrate, fenofibrate, and gemfibrozil. These compounds can also be used in combination with a compound of the present invention.
[00246] It is also contemplated that the compounds of the present invention be administered with a lipase inhibitor and/or a glucosidase inhibitor, which are typically used in the treatment of conditions resulting from the presence of excess triglycerides, free fatty acids, cholesterol, cholesterol esters or glucose including, inter alia, obesity, hyperlipidemia, hyperlipoproteinemia, Syndrome X, and the like.
[00247] In a combination with a compound of the present invention, any lipase inhibitor or glucosidase inhibitor may be employed. In one aspect lipase inhibitors comprise gastric or pancreatic lipase inhibitors. In a further aspect glucosidase inhibitors comprise amylase inhibitors. Examples of glucosidase inhibitors are those inhibitors selected from the group consisting of acarbose, adiposine, voglibose, miglitol, emiglitate, camiglibose, tendamistate, trestatin, pradimicin-Q and salbostatin. Examples of amylase inhibitors include tendamistat and the various cyclic peptides related thereto disclosed in U.S. Pat. No. 4,451,455, AI-3688 and the various cyclic polypeptides related thereto disclosed in U.S. Pat. No. 4,623,714, and trestatin, consisting of a mixture of trestatin A, trestatin B and trestatin C and the various trehalose-containing aminosugars related thereto disclosed in U.S. Pat. No. 4,273,765.
[00248] A lipase inhibitor is a compound that inhibits the metabolic cleavage of dietary triglycerides into free fatty acids and monoglycerides. Under normal physiological conditions, lipolysis occurs via a two-step process that involves acylation of an activated serine moiety of the lipase enzyme. This leads to the production of a fatty acid-lipase hemiacetal intermediate, which is then cleaved to release a diglyceride. Following further deacylation, the lipase-fatty acid intermediate is cleaved, resulting in free lipase, a monoglyceride and a fatty acid. The resultant free fatty acids and monoglycerides are incorporated into bile acid phospholipid micelles, which are subsequently absorbed at the level of the brush border of the small intestine. The micelles eventually enter the peripheral circulation as chylomicrons. Accordingly, compounds, including lipase inhibitors that selectively limit or inhibit the absorption of ingested fat precursors are useful in the treatment of conditions including obesity, hyperlipidemia, hyperlipoproteinemia, Syndrome X, and the like.
[00249] Pancreatic lipase mediates the metabolic cleavage of fatty acids from triglycerides at the 1- and 3 -carbon positions. The primary site of the metabolism of ingested fats is in the duodenum and proximal jejunum by pancreatic lipase, which is usually secreted in vast excess of the amounts necessary for the breakdown of fats in the
upper small intestine. Because pancreatic lipase is the primary enzyme required for the absorption of dietary triglycerides, inhibitors have utility in the treatment of obesity and the other related conditions.
[00250] Gastric lipase is an immunologically distinct lipase that is responsible for approximately 10 to 40% of the digestion of dietary fats. Gastric lipase is secreted in response to mechanical stimulation, ingestion of food, the presence of a fatty meal or by sympathetic agents. Gastric lipolysis of ingested fats is of physiological importance in the provision of fatty acids needed to trigger pancreatic lipase activity in the intestine and is also of importance for fat absorption in a variety of physiological and pathological conditions associated with pancreatic insufficiency. See, for example, C. K. Abrams, et al., Gastroenterology 92: 125 (1987).
[00251] A variety of lipase inhibitors are known to one of ordinary skill in the art. However, in the practice of the methods, pharmaceutical compositions, and kits of the instant invention, generally lipase inhibitors are those inhibitors that are selected from the group consisting of lipstatin, tetrahydrolipstatin (orlistat), FL-386, WAY-121898, Bay-N-3176, valilactone, esterastin, ebelactone A, ebelactone B and RHC 80267.
[00252] The pancreatic lipase inhibitors lipstatin, 2S, 3S, SS, 7Z, 1 OZ)-5-[(5)-2-formamido-4-methyl-valeryloxy]-2-hexyl-3-hydroxy-7, 1 (t-hexadecanoi c acid lactone, and tetrahydrolipostatin (orlistat), 2S, 3S, 55)-5-[(5)-2- formamido-4-methyl-valeryloxy]-2-hexyl-3-hydroxy-hexadecanoic acid lactone, and the variously substituted N-formylleucine derivatives and stereoisomers thereof, are disclosed in U.S. 4,598,089.
[00253] The pancreatic lipase inhibitor FL-386, l-[4-(2-methylpropyl)cyclohexyl]-2- [(phenylsulfonyl)oxy]-ethanone, and the variously substituted sulfonate derivatives related thereto, are disclosed in U.S. 4,452,813.
[00254] The pancreatic lipase inhibitor WAY-121898, 4-phenoxyphenyl-4- methylpiperidin-l-yl-carboxylate, and the various carbamate esters and pharmaceutically acceptable salts related thereto, are disclosed in U.S. 5,512,565; 5,391,571 and 5,602,151.
[00255] The lipase inhibitor Bay-N-3176, N-3-trifluoromethylphenyl-N'-3-chloro- 4-trifluoromethylphenylurea, and the various urea derivatives related thereto, are disclosed in U.S. 4,405,644.
[00256] The pancreatic lipase inhibitor valilactone, and a process for the preparation thereof by the microbial cultivation of Actinomycetes strain MG147— CF2, are disclosed in Kitahara, et al, J. Antibiotics, 40(11): 1647-50 (1987).
[00257] The lipase inhibitor esteracin, and certain processes for the preparation thereof by the microbial cultivation of Streptomyces strain ATCC 31336, are disclosed in U.S. 4,189,438 and 4,242,453.
[00258] The pancreatic lipase inhibitors ebelactone A and ebelactone B, and a process for the preparation thereof by the microbial cultivation of Actinomycetes strain MG7-G1, are disclosed in Umezawa, et al, J. Antibiotics, 33, 1594-1596 (1980). The use of ebelactones A and B in the suppression of monoglyceride formation is disclosed in Japanese Kokai 08-143457, published Jun. 4, 1996.
[00259] The lipase inhibitor RHC 80267, cyclo-0,0'-[(l,6-hexanediyl)-bis- (iminocarbonyl)]dioxime, and the various bis(iminocarbonyl)dioximes related thereto may be prepared as described in Petersen et al, Liebig's Annalen, 562: 205-29 (1949).
[00260] The ability of RHC 80267 to inhibit the activity of myocardial lipoprotein lipase is disclosed in Carroll et al, Lipids, 27 305-7 (1992) and Chuang et al, J. Mol Cell Cardiol, 22: 1009-16 (1990).
[00261] In another aspect of the present invention, the compounds of Formula I can be used in combination with an anti-obesity agent. The anti-obesity agent in one aspect is selected from the group consisting of a p3-adrenergic receptor agonist, a cholecystokinin-A agonist, a monoamine reuptake inhibitor, a sympathomimetic agent, a serotonergic agent, a dopamine agonist, a melanocyte-stimulating hormone receptor agonist or mimetic, a melanocyte-stimulating hormone receptor analog, a cannabinoid receptor antagonist, a melanin concentrating hormone antagonist, leptin, a leptin analog, a leptin receptor agonist, a galanin antagonist, a lipase inhibitor, a bombesin agonist, a neuropeptide-Y antagonist, a thyromimetic agent, dehydroepiandrosterone or an analog thereof, a glucocorticoid receptor agonist or antagonist, an orexin receptor antagonist, a urocortin binding protein antagonist, a glucagon- like peptide- 1 receptor agonist, and a ciliary neurotrophic factor.
[00262] In an additional aspect the anti-obesity agents comprise those compounds selected from the group consisting of sibutramine, fenfluramine, dexfenfluramine, bromocriptine, phentermine, ephedrine, leptin, phenylpropanolamine pseudoephedrine, {4-[2-(2-[6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl} acetic acid,
{4{2-(2-[6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl}benzoic acid, {4-[2-(2{6-aminopyridin-3-yl]-2(R)-hydroxyethylamino)ethoxy]phenyl}
propionic acid, and {4-[2-(2-[6-aminopyridin-3-yl]-2(R)- hydroxyethylamino)ethoxy]phenoxy} acetic acid.
[00263] Representative agents that can be used to treat diabetes in combination with a compound of the present invention include insulin and insulin analogs (e.g., LysPro insulin); GLP-1 (7-37) (insulinotropin) and GLP-1 (7-36)— N¾. Agents that enhance insulin secretion, e.g., chlorpropamide, glibenclamide, tolbutamide, tolazamide, acetohexamide, glypizide, glimepiride, repaglinide, nateglinide, meglitinide; biguanides: metformin, phenformin, buformin; A2-antagonists and imidazolines: midaglizole, isaglidole, deriglidole, idazoxan, efaroxan, fluparoxan; other insulin secretagogues linogliride, A-4166; glitazones: ciglitazone, pioglitazone, englitazone, troglitazone, darglitazone, BRL49653; fatty acid oxidation inhibitors: clomoxir, etomoxir; a-glucosidase inhibitors: acarbose, miglitol, emiglitate, voglibose, MDL25,637, camiglibose, MDL-73,945; 3-agonists: BRL 35135, BRL 37344, RO 16-8714, ICI D7114, CL 316,243; phosphodiesterase inhibitors: L-386,398; lipid-lowering agents benfluorex; antiobesity agents: fenfluramine; vanadate and vanadium complexes (e.g., bis(cysteinamide N-octyl) oxovanadium) and peroxovanadium complexes; amylin antagonists; glucagon antagonists; gluconeo genesis inhibitors; somatostatin analogs; antilipolytic agents: nicotinic acid, acipimox, WAG 994. Also contemplated to be used in combination with a compound of the present invention are pramlintide (symlin™), AC 2993 and nateglinide. Any agent or combination of agents can be administered as described above.
[00264] In addition, the compounds of the present invention can be used in combination with one or more aldose reductase inhibitors, DPP-IV inhibitor, glycogen phosphorylase inhibitors, sorbitol dehydrogenase inhibitors, NHE-1 inhibitors and/or glucocorticoid receptor antagonists.
[00265] Any compound having activity as a fructose -1,6-bisphosphatase (FBPase) inhibitor can serve as the second compound in the combination therapy aspect of the instant invention (e.g., 2-Amino-5-isobutyl-4-{2-[5-(N,N'-bis((S)-l- ethoxycarbonyl)ethyl)phosphonamido]furanyl}thiazoles). FBPase is a key regulatory enzyme in gluconeogenesis, the metabolic pathway by which the liver synthesizes glucose from 3 -carbon precursors. The term FBPase inhibitor refers to compounds that
inhibit FBPase enzyme activity and thereby block the conversion of fructose -1,6- bisphosphate, the substrate of the enzyme, to fructose 6-phosphate. FBPase inhibition can be determined directly at the enzyme level by those skilled in the art according to standard methodology (e.g., Gidh-Jain M, Zhang Y, van Poelje PD et al., J Biol Chem. 1994, 269(44): 27732-8). Alternatively, FBPase inhibition can be assessed according to standard methodology by measuring the inhibition of glucose production by isolated hepatocytes or in a perfused liver, or by measuring blood glucose lowering in normal or diabetic animals (e.g., Vincent MF, Erion MD, Gruber HE, Van den Berghe, Diabetologia. 1996, 39(10): 1148-55.; Vincent MF, Marangos PJ, Gruber HE, Van den Berghe G, Diabetes 1991 40(10): 1259-66). In some cases, in vivo metabolic activation of a compound may be required to generate the FBPase inhibitor. This class of compounds may be inactive in the enzyme inhibition screen, may or may not be active in hepatocytes, but is active in vivo as evidenced by glucose lowering in the normal, fasted rat and/or in animal models of diabetes.
[00266] A variety of FBPase inhibitors are described and referenced below; however, other FBPase inhibitors will be known to those skilled in the art. Gruber et al. U.S. Patent No. 5,658,889 described the use of inhibitors of the AMP site of FBPase to treat diabetes; WO 98/39344 and US 6,284,748 describe purine inhibitors; WO 98/39343 and US 6,110,903 describe benzothiazole inhibitors to treat diabetes; WO 98/39342 and US 6,054,587 describe indole inhibitors to treat diabetes; and WO 00/14095 and US 6,489476 describe heteroaromatic phosphonate inhibitors to treat diabetes. Other FBPase inhibitors are described in Wright SW, Carlo AA, Carty MD et al., J Med Chem. 2002 45(18):3865-77 and WO 99/47549.
[00267] The compounds of the present invention can also be used in combination with sulfonylureas such as amaryl, alyburide, glucotrol, chlorpropamide, diabinese, tolazamide, tolinase, acetohexamide, glipizide, tolbutamide, orinase, glimepiride, DiaBeta, micronase, glibenclamide, and gliclazide.
[00268] The compounds of the present invention can also be used in combination with antihypertensive agents. Any anti-hypertensive agent can be used as the second agent in such combinations. Examples of presently marketed products containing antihypertensive agents include calcium channel blockers, such as Cardizem, Adalat, Calan, Cardene, Covera, Dilacor, DynaCirc, Procardia XL, Sular, Tiazac, Vascor, Verelan, Isoptin, Nimotop, Norvasc, and Plendil; angiotensin converting enzyme (ACE)
inhibitors, such as Accupril, Altace, Captopril, Lotensin, Mavik, Monopril, Prinivil, Univasc, Vasotec and Zestril.
[00269] To assist in understanding the present invention, the following Examples are included. The experiments relating to this invention should not, of course, be construed as specifically limiting the invention and such variations of the invention, now known or later developed, which would be within the purview of one skilled in the art are considered to fall within the scope of the invention as described herein and hereinafter claimed.
EXAMPLES [00270] The present invention is described in more detail with reference to the following non-limiting examples, which are offered to more fully illustrate the invention, but are not to be construed as limiting the scope thereof. The examples illustrate the preparation of certain compounds of the invention, and the testing of these compounds in vitro and/or in vivo. Those of skill in the art will understand that the techniques described in these examples represent techniques described by the inventors to function well in the practice of the invention, and as such constitute preferred modes for the practice thereof. However, it should be appreciated that those of skill in the art should in light of the present disclosure, appreciate that many changes can be made in the specific methods that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Intermediate 1 :
Step a: A solution of HBr in acetic acid (33%, 25.12 mL, 140.1 mmol) was added to a slightly heterogeneous solution of 4-benzyloxy-benzyl alcohol in acetic acid (50 mL) at 0 °C. After stirring at 0 °C for 1 h the thick slurry was poured into a mixture of ice and water. The precipitate that formed was collected by filtration, rinsed with water and dried to give crude benzyl bromide as a white solid (11.10 g).
Step b: Diethoxymethylphosphine (11.15 mL, 80.10 mmol) was added to a solution of crude benzyl bromide from step a (11.10 g) in DMF. The clear reaction mixture was heated at 110 °C for 4 h. The cooled solution was partitioned between EtOAc and water
and the layers separated. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with water (3X), dried (Na2S04), filtered and concentrated under reduced pressure to give crude ethyl phosphinate (12.1 g).
Step c: Pd/C (10%, 1.28 g) was added to a degassed solution of crude phosphinate in methanol. After stirring at rt under 1 atm of hydrogen for 16 hours, the catalyst was removed by filtration over Celite and the pad was rinsed with methanol. The combined filtrates were concentrated under reduced pressure and the residue was partitioned between water and EtOAc. The layers were separated and the aqueous phase was extracted with CH2C12 (5X). The combined organic extracts were dried (Na2S04), filtered and concentrated under reduced pressure to give crude phenol (8.80 g).
Step d: Ca2C03 (11.21 g, 112.0 mmol) followed by benzyltrimethylammonium tribromide (30.03 g, 77.0 mmol) were added to a solution of phenol (7.5 g, 35.0 mmol) in methanol (60 mL) at rt. After stirring at rt for 5 h, the heterogeneous mixture was filtered through Celite and the pad rinsed with methanol. The combined filtrates were concentrated under reduced pressure. The residue was partitioned between CH2C12 and water. The layers were separated and the organics were washed with water (2X), dried (Na2S04), filtered and concentrated under reduced pressure. The residue was purified by column
chromatography (2% to 5% methanol in CH2C12) to give intermediate 1 as an oil (10.6 g, 81%).
Step a: Fuming nitric acid (7.82 mL, 184.9 mmol) was added to neat acetic anhydride (20 mL) at -20 °C. Iodine (7.79 g, 30.69 mmol) was then added followed by TFA (14.25 mL). After stirring at -20 °C for 15 minutes, the cold bath was removed and the reaction mixture was stirred at rt. After 2 h at rt, all the iodine was consumed and the orange vapors were blown away under a stream of nitrogen. The black reaction mixture was concentrated under reduced pressure and the residue was taken up in acetic anhydride (60 mL) and the black solution cooled to -20 °C. A solution of anisole (20.0 g, 184.9 mmol) in acetic anhydride (20 mL) and TFA (15.67 mL) was added to the black solution at -20 °C and the resulting mixture was placed in a fridge at 5 °C overnight. The resulting
solution was then stirred at rt for 4 h and concentrated under reduced pressure. The residue was taken up in methanol (60 mL) and a solution of NaHS03 (10.0 g in 100 mL) followed by a solution of NaBF4 (109.8 g in 500 mL) were added. After stirring at rt for 3 h, the solvent was decanted and the brown sludge was washed with water. The solids were taken up in hexanes, filtered, rinsed with hexanes and dried to give a beige solid (13.14 g, 50% based on iodine).
Step b: A solution of ethyl 3,5-dibromo-4-hydroxy-benzyl-methylphosphinate
(intermediate 1 , 10.5 g, 28.22 mmol) and triethylamine (4.32 mL, 31.0 mmol) in CH2CI2 (30 mL) was added to a suspension of iodonium salt from step a (15.7 g, 36.86 mmol) and copper powder (3.59 g, 56.44 mmol) in CH2C12 (100 mL) at 0 °C. The ice bath was removed and the flask was covered with aluminum foil. After stirring at rt for 18 h, the reaction mixture was filtered over Celite and the pad rinsed with CH2CI2. The combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (100% CH2CI2 to 10% methanol in CH2CI2) to give a brown solid (14.60 g).
Step c: A solution of 1 , 1-dichloromethyl-methyl ether (5.3 mL, 58.56 mmol) and tin tetrachloride (20.58 mL, 175.68 mmol) in CH2CI2 (20 mL) was added to a solution of diphenyl ether (14.0 g, 29.28 mmol) in CH2C12 (60 mL) at -78 °C. Upon completion of the addition, the temperature was raised to 0 °C. After stirring at 0 °C for 4 h, 2 M HC1 (80 mL) was added and the reaction mixture was partitioned between EtOAc and water. The layers were separated and the organics were washed with a saturated solution of NaHC03, brine, dried (Na2S04), filtered and concentrated under reduced pressure to give a brown foam (13.20 g, 89%).
Step d: NaH2P04 »H20 (47.75 g, 346.1 mmol) followed by sodium chlorite (80%, 57.18, 506 mmol) were added to a solution of aldehyde (13.10 g, 26.62 mmol) in t-butanol (120 mL), 2-methyl-2-butene (60 mL) and water (120 mL) at rt. The heterogeneous mixture becomes a clear solution after 20 minutes. After stirring at rt for 4 h, the reaction mixture was partitioned between CH2CI2 and water, and the layers separated. The aqueous phase was extracted with CH2CI2 and the combined organic extracts were washed with 0.3 N HC1, water (2X), dried (Na2S04), filtered and concentrated under reduced pressure (15.5g).
Step e: Neat BBr (1 1.44 mL, 118.76 mmol) was added to a solution of anisole (15.5 g, 29.69 mmol) in CH2CI2 (150 mL) at -50 °C. The amber reaction mixture was warmed to 0
°C and stirred for 4 h, then warmed to rt and stirred 4h. Water and ice were carefully added and most of the CH2CI2 was removed under reduced pressure. The residual mixture was partitioned between EtOAc and water and the layers separated. The organic layer was washed with water (3X), dried (Na2S04), filtered and concentrated under reduced pressure to give the desired carboxylic acid intermediate (15.10 g).
Intermediate 3:
Step a: A 50 L, 4-neck flask was equipped with an overhead stirrer, temperature probe, and cooling bath. The flask was charged with 3,5-dimethylphenol (2497 g, 20.5 mol), water (14 L), and 50% (wt/wt) aqueous sodium hydroxide (1636 g, 20.5 mol). The mixture was stirred 1.5 h to complete dissolution. The mixture was cooled to 4 °C using an ice/water bath (note 2). Formaldehyde (37% aqueous solution, 1496 g, 18.5 mol) was added in one portion. The mixture was stirred cold throughout the daytime and allowed to warm slowly overnight. The reaction mixture was diluted with dichloromethane (5 L) and ethyl acetate (5 L). Added HC1 (1.5 L of 12 M, 18.0 mol) over 30 minutes to pH 5. After stirring for 6 h, the solid was collected by filtration. The filter cake was washed with water (1.2 L) and dichloromethane (2 L). The solid was dried in a vacuum oven (50 °C, -30 in Hg) to a constant weight, giving 1222 g (39%> yield) of the benzyl alcohol. Step b: A 12 L flask was equipped with an overhead stirrer, heating mantle, temperature probe, condenser with a nitrogen bubbler on the outlet, and an addition funnel. The flask was charged with paraformaldehyde (488 g, 16.3 mol), potassium carbonate (94 g, 0.7 mol), and 2-propanol (4.5 L). The mixture was heated to 50 °C then the heating mantle was turned off. Diisopropylphosphite (2260 g, 13.6 mol) was added from the addition funnel at a rate that would maintain the temperature at 50-60 °C. The mixture was cooled to 35 °C over 2 h then filtered through Celite. The pad was washed with 2-propanol (2 x 200 mL). The mixture was concentrated under reduced pressure. The colorless residual oil (2950 g) was dissolved in dichloromethane (9 L). The solution was washed with 1 N HC1 (1.35 L) and saturated aqueous NaHCCb (2.25 L), and dried over MgS04 (1 kg). The mixture was filtered through Celite and the pad washed with dichloromethane (2 x 500
mL). The filtrate was concentrated under reduced pressure, giving diisopropyl hydroxymethylphosphonate as a colorless oil weighing 2836 g.
Step c: A 22 L, four-neck flask was equipped with an overhead stirrer, cooling bath, temperature probe, 2 L addition funnel, and a nitrogen bubbler. The flask was charged with diisopropyl hydroxymethylphosphonate (1408g, 6.67 mol), triethylamine (1350 g, 13.34 mol), and dichloromethane (4 L). The resulting solution was cooled to 5 °C using an ice/water bath. A solution of /?-toluenesulfonyl chloride (1335 g, 7.0 mol) in dichloromethane (10 L) was added from the addition funnel at a rate that would keep the temperature below 10 °C. The mixture was stirred in an ice bath for 1.75 h then at ambient temperature (~20 °C) for 15 h. The reaction mixture was washed with 1 M HC1 (6 L) followed by saturated aqueous NaHC03 (6 L). The organic layer was dried over MgS04 (400 g) and filtered. The filtrate was concentrated under reduced pressure to give the tosylate as a yellow oil weighing 2111 g (90% yield).
Step d: A 22 L, four-neck flask was equipped with an overhead stirrer, temperature probe, heating mantle, and condenser with a nitrogen bubbler on the outlet. The flask was charged with the tosylate (2054 g, 5.45 mol), DMSO (2 L), the benzyl alcohol from step a (928 g, 5.62 mol), cesium carbonate (2841 g, 8.72 mol), and DMSO (2.5 L). The mixture was heated to 55 °C over 2 h and maintained at 50-60 °C for 6 h. The reaction mixture was cooled to 20 °C overnight then cooled to 5 °C (ice/water bath). Ethyl acetate (3.6 L) was added followed by slow addition of 1% (wt/vol) aqueous NaCl solution (7.2 L). The phases were separated and the aqueous layer was extracted with ethyl acetate (2.7 L). The combined organic layers were washed with brine (2 x 2.7 L), dried over MgS04 (500 g), and filtered, rinsing with ethyl acetate (500 mL). The filtrate was concentrated under reduced pressure, obtaining intermediate 3 as a thick, dark amber syrup weighing 1935 g.
Step a TFA (58.6 mL, 0.789 mol) was added to an heterogeneous mixture of benzyl alcohol from intermediate 3/step a (20 g, 0.132 mol) and anisole (42.8 mL, 0.395 mol) in CH2CI2 (200 mL) at -20 °C. The ice bath was removed and the reaction mixture was allowed to warm to rt over 1 h. The clear solution was poured into a mixture of ice (200
g) and a concentrated solution of NH4OH (60 mL). After stirring for 5 minutes, the mixture was extracted with CH2CI2 (2X). The combined organic extracts were washed with brine, dried (Na2S04), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (10% to 30% EtOAc in hexanes) to give a yellow oil (16.48 g, 52%).
Step b: Trifluoromethanesulfonic anhydride was added to a solution of phenol (16.48 g, 68.1 mmol) and pyridine (10.95 mL), 136.2 mmol) in CH2C12 (680 mL) at 0 °C. After 15 minutes at 0 °C, the ice bath was removed and the reaction allowed to warm to rt. After stirring at rt for 1 h, the orange solution was washed with 10%> hydrochloric acid, water, dried (Na2S04), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (5% EtOAc in hexanes) to give the aryl triflate as a clear oil (19.76 g, 78%).
Step c: A mixture of aryl triflate (19.76 g, 52.8 mmol), Pd(OAc)2 (1.18 g, 5.28 mmol), diphenylphosphinopropane (2.18 g, 5.28 mmol), triethylamine (14.7 mL, 105.7 mmol) in methanol (50 mL) and DMF (100 mL) was charged in a bomb. The bomb was sealed, evacuated and charged with 60 psi of CO. After heating the bomb at 90 °C for 16 h, the cooled bomb was opened. The black reaction mixture was filtered over Celite and rinsed with EtOAc. The combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (5% EtOAc in hexanes) to give the methyl benzoate as a yellow solid (6.65 g, 44%).
Step d: L1AIH4 (1.87 g, 49.2 mmol) was added in small portion to a solution of methyl benzoate (6.65 g, 23.4 mmol) in THF (230 mL) at 0 °C. After stirring at 0 °C for 90 minutes, the grey slurry was quenched carefully with 1 N NaoH. The solids were filtered off over Celite and the pad was rinsed with EtOAc. The combined filtrates were extracted with EtOAc (2X). The combined organic extracts were washed with brine, dried (Na2S04), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (20 to 40% EtOAc in hexanes) to give the benzylic alcohol as a white solid (4.7 g, 78%).
Step e: A mixture of CBr4 (9.13 g, 27.5 mmol) and PPh3 (7.22 g, 27.5 mmol) in THF (160 mL) was stirred at rt for 5 minutes. A solution of the benzylic alcohol (4.7 g, 18.4 mmol) in THF (24 mL) was added to the stirring solution of CBr4 and PPh3. The off white heterogeneous mixture was stirred at rt for 90 minutes. The solids were filtered off over Celite and rinsed with ether. The combined organic extracts were concentrated under
reduced pressure and the residue was purified by column chromatography to give the benzyl bromide as a white solid (4.68 g, 83%).
Intermediate 4 was synthesized according to the procedures described for the synthesis of intermediate 1/step b, then intermediate 2/step c-e.
Example El
Ethyl 3- [5-(2,6-dimethyl-4-phosphonomethoxy-benzyl)-2-hydroxy-phenyl] - propanoate
Step a: Methanesulfonic acid (0.46 mL, 7.2 mmol) was added to a solution of intermediate 3 (990 mg, 3 mmol) and methyl 3-(2-hydroxyphenyl)-propanoate (810 mg, 4.5 mmol) in CH2CI2 (30 mL) at -40 °C. The bath was removed and the reaction mixture was allowed to warm to 0 °C. Upon reaching 0 °C, water was added and the layers were separated. The aqueous phase was extracted with CH2CI2 and the combined organics were washed with water, dried (Na2S04), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (10% to 25 %> EtOAc in CH2CI2) to give the lactone (500 mg, 34%>) and the methyl carboxylate (350 mg, 24%>).
Step b: Bromotrimethylsilane (1 mL, 3.6 mmol) was added to a solution of methyl carboxylate (350 mg, 0.71 mmol) in CH2CI2 (20 mL) at rt. After stirring at rt for 16 h, the reaction mixture was concentrated under reduced pressure. The residue was taken up in EtOAc and washed with water, brine, dried (Na2S04), filtered, and concentrated under reduced pressure to give the lactone phosphonic acid.
Step c: Asolution of HC1 in dioxane (4 N, 0.25 mL, 1 mmol) was added to a solution of lactone phosphonic acid (100 mg, 0.26 mmol) in ethanol (4 mL) at rt. After stirring at rt for 1 h, the reaction mixture was concentrated under reduced pressure and azeotrpoed with ethanol. The residue was taken up in ether and sonicated. The solid was collected by filtration, rinsed with ether and dried to give the ethyl carboxylate (90 mg, 82%); 1H
NMR (500 MHz, CD3OD) δ: 6.72 (s, 2H), 6.68 (s, 1H), 6.65-6.58 (m, 2H), 4.19 (d, J = 10.0 Hz, 2H), 4.02 (q, J= 7.0 Hz, 2H), 3.87 (s, 2H), 2.79 (t, J= 8.0 Hz, 2H), 2.55 (t, J = 8.0 Hz, 2H), 2.20 (s, 6H), 1.18 (t, J= 7.0 Hz, 3H). 31P NMR (202.3 MHz, CD3OD) δ:
19.2 ppm. LCMS m/z: 423.4 [C21H27O7P +1] .Anal for C21H27O7P; Calcd: C:59.71, H:6.44; Found: C:59.73, H:6.18.
Example Al
3- [5-(2,6-Dimethyl-4-phosphonomethoxy-benzyl)-2-hydroxy-phenyl] -propanoic acid
1 N NaOH (0.79 mL, 0.79 mmol) was added to a solution of lactone phosphonic acid (Example El/step b, 100 mg, 0.26 mmol) in methanol at rt. After 1 h at rt, additional IN NaOH was added (0.8 mL). After stirring at rt for 2.5 days, most of the methanol was removed under reduced pressure. The residual solution was diluted with water and washed with ether, and acidified to pH 1 with 2 N hydrochloric acid at 0 °C. After 30 minutes, the precipitate was collected by filtration, rinsed with water and dried to give the carboxylic acid (80 mg, 78%). 1H NMR (500 MHz, CD3OD) δ: 6.74 (s, 1H), 6.71 (s, 2H), 6.62-6.59 (m, 2H), 4.17 (d, J= 10.0 Hz, 2H), 3.87 (s, 2H), 2.79 (t, J= 8.0 Hz, 2H), 2.55
(t, J= 8.0 Hz, 2H), 2.20 (s, 6H). 31P NMR (202.3 MHz, CD3OD) δ: 18.69 ppm. LCMS m/z: 395.4 [C19H23O7P +1]+.
Example E2
Ethyl [5-(2,6-dimethyl-4-phosphonomethoxy-benzyl)-2-hydroxy-phenyl]-acetate
The title compound was prepared according to the procedure described for the synthesis of example El . Step b gave a 50/50 mixture of lactone and the title compound; 1H NMR (500 MHz, CD3OD) δ: 6.75-6.66 (m, 4H), 6.65 (d, J= 6.5 Hz, 1H), 4.18 (d, J= 10.0 Hz, 2H), 4.10 (q, J = 7.3 Hz, 2H), 3.90 (s, 2H), 3.51 (s, 2H), 2.21 (s, 6H), 1.21 (t, J = 7.3 Hz, 3H). 31P NMR (202.3 MHz, CD3OD) δ: 19.13 ppm. LCMS m/z: 409.6 [C2oH2507P +1]+. Anal for C2oH2507P; Calcd: C:56.34, H:6.38; Found: C:56.76, H:6.14.
Example A2
[5-(2,6-Dimethyl-4-phosphonomethoxy-benzyl)-2-hydroxy-phenyl]-acetic acid
The title compound was prepared according to the procedure described for the synthesis of example Al; 1H NMR (500 MHz, CD3OD) δ: 6.78 (s, 1H), 6.71 (s, 2H), 6.65-6.60 (m, 2H), 4.06 (d, J= 10.0 Hz, 2H), 3.89 (s, 2H), 3.51 (s, 2H), 2.20 (s, 6H). 31P NMR (202.3 MHz, CD3OD) δ: 15.94 ppm. LCMS m/z: 381.6 [Ci8H2i07P +1]+. Anal for C2oH2507P + 0.4 NaCl; Calcd: C:53.55, H:5.24; Found: C:53.17, H:4.78.
Example A3
[2-Hydroxy-5-[4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl]- phenyl] -acetic acid
Step a: A solution of NaCN (288 mg, 5.88 mmol) in water (10 mL) was added to a solution of benzaldehyde intermediate (intermediate in the synthesis of intermediate 4, 1.10 g, 2.94 mmol), ethyl chloroformate (0.3 mL, 3.09 mmol) and tetrabutyl ammonium bromide (47 mg, 0.15 mmol) in CH2C12 (10 mL) at rt. After stirring at rt for 16 h, the biphasic reaction mixture was partitioned between CH2C12 and water, and the layers separated. The organics were washed with a saturated solution of NaHC03, brine, dried (Na2S04), filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (5% methanol in CH2C12) to give the (ethoxycarbonyloxy)- benzyl-cyanide (1.10 g, 79%).
Step b: Pd/C (400 mg) was added to a solution of (ethoxycarbonyloxy)-benzyl-cyanide (1.10 g, 2.32 mmol) in ethanol (35 mL) in a pressure vessel. The vessel was sealed, degassed and pressurize with 50 psi of hydrogen. The sealed vessel was heated at 70 °C for 20 h. The cooled bomb was opened, and the black reaction mixture was filtered over Celite and rinsed with EtOAc. The combined filtrates were concentrated under reduced
pressure and the residue was purified by column chromatography (5% EtOAc in hexanes) to give the benzyl cyanide (1.10 g, 100%).
Step c: A solution of potassium hydroxide (567 mg, 10.1 mmol) in water (4 mL) was added to a solution of benzyl cyanide (391 mg, 1.01 mmol) in ethanol (8 mL) and the resulting solution was heated at reflux for 16 h. The cooled reaction mixture was concentrated under reduced pressure. The residue was taken up in water and washed with ether. The aqueous phase was acidified to pH 1 with cone hydrochloric acid. The white precipitate was collected by filtration, rinsed with water and dried to give the diacid (331 mg, 87%).
Step d: Neat boron tribromide (0.51 mL, 5.27 mmol) was added over 2 min to a suspension of diacid (331 mg, 0.88 mmol) in CH2CI2 at rt. After stirring at rt for 18 h, the reaction mixture was poured into ice/water (50 mL) and the pH adjusted to 14 with solid NaOH. The layers were separated and the organic phase was extracted with 1 N NaOH. The combined aqueous extracts were acidified to pH 1 with cone HC1. The precipitate was collected by filtration, rinsed with water and dried to give the title compound (168 mg, 53%); 1H NMR (500 MHz, DMSO-d6) δ: 9.21 (s, 1H), 6.91 (s, 2H), 6.78 (d, J= 1.5 Hz, 1H), 6.64 (d, J= 8.0 Hz, 1H), 6.60 (dd, J= 8.0, 1.5 Hz, 1H), 3.82 (s, 2H), 3.37 (s, 2H), 2.92 (d, J= 17.5 Hz, 2H), 2.16 (s, 6H), 1.21 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-dg) δ: 44.15 ppm. LCMS m/z: 363.6 [Ci9H2305P +1]+. Anal for Ci9H2305P + H20; Calcd: C:60.00, H:6.62; Found: C:60.00, H:6.60.
Example E3-1
Ethyl [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - phenyl] -acetate
A solution of HC1 in dioxane (4 N, 0.034 mL, 0.138 mmol) was added to a solution of diacid from example A3 (50 mg, 0.138 mmol) in ethanol at rt. After stirring at rt for 22 h, the reaction mixture was concentrated under reduced pressure and the residue was partitioned between EtOAc and water. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic extracts were washed with brine, dried (Na2S04), filtered and concentrated under reduced pressure to give the title compound as
a yellow solid (52 mg, 97%); 1H NMR (500 MHz, DMSO-d6) δ: 9.26 (s, 1H), 6.91 (s, 2H), 6.75 (s, 1H), 6.65-6.60 (m, 2H), 4.09 (q, J= 7.0 Hz, 2H), 3.83 (s, 2H), 3.44 (s, 2H), 2.92 (d, J= 17.5 Hz, 2H), 2.16 (s, 6H), 1.21 (d, J= 14.0 Hz, 3H), 1.13 (t, J= 7.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 44.16 ppm. LCMS m/z: 391.6 [C21H27O5P +1]+. Anal for C2iH2705P; Calcd: C:62.29, H:7.27; Found: C:63.44, H:7.40.
Example E3-2
Isopropyl [2-Hydroxy-5-[4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -phenyl] -acetate
The title compound was prepared according to the procedure described for the synthesis of compound E3-1 using 2-propanol. The compound was purified by reverse-phase column chromatography (0 to 30% acetonitrile in water); 1H NMR (500 MHz, DMSO-d6) δ: 9.24 (s, 1H), 6.91 (s, 2H), 6.74 (s, 1H), 6.66 (s, 2H), 4.83 (hept, J = 6.4 Hz, 1H), 3.82 (s, 2H), 3.40 (s, 2H), 2.92 (d, J = 17.5 Hz, 2H), 2.16 (s, 6H), 1.21 (d, J = 14.0 Hz, 3H), 1.13 (d, J = 6.4 Hz, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ: 44.08 ppm. LCMS m/z: 405.6 [C22H2905P +1]+. Anal for C22H2905P + 0.5 EtOAc + 0.5 NaCl; Calcd: C:60.34, H:6.96; Found: C:60.03, H:60.65.
Example E3-3
Propyl [2-Hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - phenyl] -acetate
The title compound was prepared according to the procedure described for the synthesis of compound E3-1 using propanol. The compound was purified by reverse-phase column chromatography (0 to 30% acetonitrile in water); 1H NMR (500 MHz, DMSO-d6) δ: 9.25 (s, 1H), 6.91 (s, 2H), 6.76 (s, 1H), 6.65-6.60 (m, 2H), 3.92 (t, J = 6.5 Hz, 2H), 3.83 (s, 2H), 3.45 (s, 2H), 2.92 (d, J = 17.5 Hz, 2H), 2.16 (s, 6H), 1.51 (tq, J = 6.5, 7.3 Hz, 2H), 1.21 (d, J = 14.0 Hz, 3H), 0.82 (t, J = 7.3 Hz, 3H). 31P NMR (121.46 MHz, DMSC ¾) δ:
44.11 ppm. LCMS m/z: 405.6 [C22H2905P +1]+. Anal for C22H2905P + 0.6 H20; Calcd: C:63.63, H:7.33; Found: C:63.53, H:7.14.
Example A4
3-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]
-2-hydroxy-benzyl] -benzoic acid
Step a: A solution of 2-methoxy-phenyl-magnesium bromide (1 M in THF, 32 mL, 32mmol) was added to a solution of methyl 3-carboxaldehyde-benzoate (5.0 g, 30.5 mmol) in CH2C12 (30 mL) at rt over 90 minutes. Upon completion of the addition, the reaction mixture was quenched with saturated aqueous solution of NH4C1 and diluted with EtOAc. The layers were separated and the organics were washed with water then brine, dried (Na2S04), filtered, concentrated to dryness and purified by column
chromatography (20% EtOAc in hexanes) to give an oil (9.52 g); Rf =0.7 (20% EtOAc in hexanes).
Step b: TFA (4.53 mL, 61 mmol) was added to a solution of carbinol from step a (9.52 g, 30.5 mmol) and triethylsilane (9.85 mL, 61 mmol) in CH2C12 (300 mL) at rt. After stirring at rt for 4 h, water (100 mL) was added and the reaction mixture was stirred vigourously for 5 minutes. The layers were separated and the organics were dried
(Na2S04), filtered, concentrated to dryness and purified by column chromatography
(100% hexanes to 10% EtOAc in hexanes) to give an oil (7.08 g, 91% for 2 steps); Rf = 0.9 (10% EtOAc in hexanes).
Step c: Fuming nitric acid (1.17 mL, 27.6 mmol) was added to neat acetic anhydride (2.97 mL) at -20 °C. Iodine (1.15 g, 4.59 mmol) was then added followed by TFA (2.13 mL). After stirring at -20 °C for 15 minutes, the cold bath was removed and the reaction mixture was stirred at rt. After 2 h at rt, all the iodine was consumed and the orange vapors were blown away under a stream of nitrogen. The black reaction mixture was concentrated under reduced pressure and the residue was taken up in acetic anhydride (8.95 mL) and the black solution cooled to -20 °C. A solution of methyl 3-(2- methoxybenzyl)benzoate (7.08 g, 27.6 mmol) in acetic anhydride (2.97 mL) and TFA
(2.34 mL) was added to the black solution at -20 °C and the resulting mixture was placed
in a fridge at 5 °C overnight. The resulting blue solution was then stirred at rt for 4 h and concentrated under reduced pressure. The residue was taken up in methanol (8.95 mL) and a solution of NaHS03 (1.49 g in 14,9 mL) followed by a solution of NaBF4 (16.38 g in 74.7 mL) were added. After stirring at rt for 1 h, the solvent was decanted and the brown sludge was washed with water. The solids were taken up in hexanes and sonicated for 1 h. The solvents were decanted and the residue was washed with hexanes. The brown oil was purified by column chromatography (100% CH2CI2 to 10% methanol in CH2CI2) to give a brown solid (2.61 g, 87% based on iodine); Rf = 0.6 (10% methanol in CH2CI2). Step d: A solution of ethyl 3,5-dibromo-4-hydroxy-benzyl-methylphosphinate (372 mg, 1 mmol) and triethylamine (0.15 mL, 1.1 mmol) in CH2CI2 (4 mL) was added to a suspension of iodonium salt from step c (850 mg, 1.3 mmol) and copper powder (127 mg, 2 mmol) in CH2CI2 (8 mL) at 0 °C. The ice bath was removed and the flask was covered with aluminum foil. After stirring at rt for 18 h, the reaction mixture was filtered over Celite and the pad rinsed with CH2CI2. The combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (100% CH2CI2 to 10% methanol in CH2C12) to give a brown solid (436 mg, 70%); Rf = 0.5 (10% methanol in CH2CI2).
Step e: Neat BBr3 (0.67 mL, 6.96 mmol) was added to a solution of compound from step d (436 mg, 0.696 mmol) in CH2CI2 (15 mL) at 0 °C. The ice bath was removed and the amber reaction mixture was stirred at rt for 22 h. Water and ice were carefully added and the layers were separated. The aqueous layer was extracted with EtOAc. The combined organic extracts were dried (Na2S04), filtered and concentrated under reduced pressure. The brown solid was taken up in EtOH (15 mL) and a solution of KOH (195 mg, 3.48 mmol) in water (15 mL) was added. After stirring at rt for 18 hours, additional KOH (233 mg) was added and the mixture was heated at 60 °C until clear solution. The cooled solution was concentrated under reduced pressure and the brown solid was purified by CI 8 column chromatography (100% water to 10% Methanol in water). The fractions containing the product were pooled and partially concentrated under reduced pressure. The pH was brought down to 1 with cone HC1 and a precipitate formed. After stirring at rt for 1 h the solids were collected by filtration and rinsed with water to give the title compound as an off white solid (205 mg); 1H NMR (500 MHz, DMSO-d6) δ: 9.23 (s, 1H), 7.80 (s, 1H), 7.73 (d, J= 8.0 Hz, 1H), 7.62 (s, 2H), 7.45 (d, J= 8.0 Hz, 1H), 7.38 (t,
J= 8.0 Hz, 1H), 6.71 (d, J= 9.0 Hz, 1H), 6.65 (d, J= 3.0 Hz, 1H), 6.37 (dd, J= 9.0, 3.0 Hz, 1H), 3.90 (s, 2H), 3.10 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14.0 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.85 ppm. LCMS m/z: 571.6 ^HigB^OeP +1] Anal for C22H19Br206P + 0.4 H20 + 0.1 NaCl; Calcd: C:45.31, H:3.42; Found: C:44.96, H:3.03.
Example E4-1
Methyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzylj-benzoate
A solution of HCl in dioxane (4 M, 0.022 mL, 0.088 mmol) was added to a solution of carboxylic acid (50 mg, 0.088 mmol) in methanol (1.75 mL) at rt, and the resulting solution was heated at 40 °C. After 4 days, the cooled reaction mixture was concentrated under reduced pressure and the residue partitioned between EtOAc and water. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic extracts were washed with brine, dried (Na2S04), filtered, concentrated under reduced pressure to give a brown solid (46 mg, 89%); 1H NMR (500 MHz, DMSO-d6) δ: 9.23 (s, 1H), 7.81 (s, 1H), 7.75 (d, J= 8.0 Hz, 1H), 7.62 (s, 2H), 7.49 (d, J= 8.0 Hz, 1H), 7.41 (t, J= 8.0 Hz, 1H), 6.71 (d, J= 9.0 Hz, 1H), 6.64 (d, J= 3.0 Hz, 1H), 6.36 (dd, J= 9.0, 3.0 Hz, 1H), 3.89 (s, 2H), 3.82 (s, 3H), 3.11 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.94 ppm. LCMS m/z: 585.4 ^Hi^OeP +1]+. Anal for C23H2iBr206P + 0.1 EtOAc; Calcd: C:47.40, H:3.71; Found: C:47.42, H:4.00.
The following compounds were synthesized according to the procedure described above using the appropriate starting materials.
Example E4-2
Ethyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzyl] -benzoate
1H NMR (500 MHz, DMSO-d6) δ: 9.23 (s, 1H), 7.80 (s, 1H), 7.75 (d, J= 8.0 Hz, 1H), 7.62 (s, 2H), 7.47 (d, J= 8.0 Hz, 1H), 7.40 (t, J= 8.0 Hz, 1H), 6.71 (d, J= 9.0 Hz, 1H), 6.63 (d, J= 3.0 Hz, 1H), 6.37 (dd, J= 9.0, 3.0 Hz, 1H), 4.29 (q, J= 7.0 Hz, 2H), 3.89 (s, 2H), 3.82 (s, 3H), 3.10 (d, J= 17.0 Hz, 2H), 1.29 (t, J= 7.0 Hz, 3H), 1.28 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.75 ppm. LCMS m/z: 599.4 [C^H^BrjOgP +1]+. Anal for C24H23Br206P + 0.5 H20 + 0.1 EtOAc; Calcd: C:47.57, H:4.06; Found: C:47.53, H:3.87.
Example E4-3
Isopropyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-ph
hydroxy-benzyl] -benzoate
1H NMR (500 MHz, DMSO-d6) δ: 9.22 (s, 1H), 7.78 (s, 1H), 7.74 (d, J= 7.5 Hz, 1H),
7.62 (s, 2H), 7.46 (d, J= 8.0 Hz, 1H), 7.39 (t, J= 7.5 Hz, 1H), 6.71 (d, J= 9.0 Hz, 1H),
6.63 (d, J= 3.0 Hz, 1H), 6.37 (dd, J= 9.0, 3.0 Hz, 1H), 5.10 (hept, J= 6.3 Hz, 2H), 3.89 (s, 2H), 3.82 (s, 3H), 3.10 (d, J= 17.0 Hz, 2H), 1.30 (d, J= 6.3 Hz, 3H), 1.28 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 43.01 ppm. LCMS m/z: 613.4
[C25H25Br206P +1]+. Anal for C25H25Br206P + 0.7 TFA + 0.2 EtOAc; Calcd: C:46.03, H:3.88; Found: C:46.41, H:3.47.
Example E5
Methyl 4- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzyl] -benzoate
1H NMR (500 MHz, DMSO-d6) δ: 9.24 (s, 1H), 7.86 (d, J= 8.5 Hz, 2H), 7.62 (s, 2H), 7.33 (d, J= 8.5 Hz, 2H), 6.72 (d, J= 8.5 Hz, 1H), 6.61 (d, J= 3.0 Hz, 1H), 6.37 (dd, J
8.5, 3.0 Hz, 1H), 3.90 (s, 2H), 3.82 (s, 3H), 3.10 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14. Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.79 ppm. LCMS m/z: 585.4
[C23H2iBr206P +1]+. Anal for C23H2iBr206P + 0.1 EtOAc; Calcd: C:47.40, H:3.71; Found: C:47.21, H:3.44.
Example A5
4-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]
-2-hydroxy-benzyl] -benzoic acid
1H NMR (300 MHz, DMSC /6) δ: 9.21 (s, 1H), 7.81 (d, J= 8.5 Hz, 2H), 7.61 (s, 2H), 7.29 (d, J= 8.5 Hz, 2H), 6.71 (d, J= 9.0 Hz, 1H), 6.59 (d, J= 3.0 Hz, 1H), 6.35 (dd, J 9.0, 3.0 Hz, 1H), 3.88 (s, 2H), 3.08 (d, J= 17.0 Hz, 2H), 1.26 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.72 ppm. LCMS m/z: 571.6 [C22Hi9Br206P +1]+. Anal for ¾2Η19ΒΓ206Ρ + 0.8 H20; Calcd: C:45.20, H:3.55; Found: C:44.85, H:3.16.
Example E6
Methyl 5- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzyl]-2-fluoro-benzoate
1H NMR (500 MHz, DMSO-d6) δ: 9.25 (s, 1H), 7.70 (dd, J= 7.0, 2.5 Hz, 1H), 7.62 (s, 2H), 7.50-7.46 (m, 1H), 7.23 (t, J= 8.0 Hz, 1H), 6.71 (d, J= 8.5 Hz, 1H), 6.64 (d, J= 3.0 Hz, 1H), 6.37 (dd, J= 8.5, 3.0 Hz, 1H), 3.85 (s, 2H), 3.82 (s, 3H), 3.10 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14.0 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.92 ppm.
LCMS m/z 603.4 [C23H20Br2FO6P +1]+.
Example A6
5-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydroxy- benzyl]-2-fluoro-benzoic acid
1H NMR (500 MHz, DMSO-d6) δ: 9.24 (s, 1H), 7.69 (dd, J= 7.0, 2.5 Hz, 1H), 7.62 (s, 2H), 7.50-7.46 (m, 1H), 7.18 (t, J= 8.0 Hz, 1H), 6.71 (d, J= 8.5 Hz, 1H), 6.65 (d, J= 3.0 Hz, 1H), 6.36 (dd, J= 8.5, 3.0 Hz, 1H), 3.84 (s, 2H), 3.10 (d, J= 17.0 Hz, 2H), 1.28 (d, J = 14.0 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.96 ppm.
LCMS m/z 589.1 [C22H18Br2F06P +1]+.
Anal for C22H18Br2F06P + 1.2 H20; Calcd: C:43.33, H:3.37; Found: C:43.38, H:3.41.
Example E7
Methyl 4- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzyl]-2-fluoro-benzoate
1H NMR (500 MHz, DMSO-d6) δ: 9.29 (s, 1H), 7.78 (t, J= 7.8 Hz, 1H), 7.62 (s, 2H), 7.14 (t, J= 9.8 Hz, 2H), 6.73 (d, J= 8.5 Hz, 1H), 6.65 (d, J= 3.0 Hz, 1H), 6.38 (dd, J = 8.5, 3.0 Hz, 1H), 3.90 (s, 2H), 3.82 (s, 3H), 3.08 (d, J= 17.0 Hz, 2H), 1.25 (d, J= 14.0 Hz, 3H).
31P NMR (121.46 MHz, DMSC /6) δ: 42.61 ppm.
LCMS m/z: 603.4 ^HzoB^FOeP +1]+.
Example A7
4-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydroxy- benzyl]-2-fluoro-benzoic acid
1H NMR (300 MHz, DMSO- 6) δ: 9.27 (s, 1H), 7.74 (t, J= 8.1 Hz, 1H), 7.62 (s, 2H), 7.09 (t, J= 10.8 Hz, 2H), 6.72 (d, J= 8.5 Hz, 1H), 6.63 (d, J= 3.0 Hz, 1H), 6.38 (dd, J = 8.5, 3.0 Hz, 1H), 3.87 (s, 2H), 3.09 (d, J= 17.0 Hz, 2H), 1.27 (d, J= 14.0 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.86 ppm.
LCMS m/z: 589.1 [C22H18Br2F06P +1]+.
Anal for C22Hi8Br2F06P + 0.7 H20; Calcd: C:43.98, H:3.25; Found: C:43.99, H:2.94.
Example A8
5 '- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] - 2'-hydroxy-biphenyl-3-carboxylic acid
Step a: A mixture of palladium acetate and X-phos in THF (3 mL) was degassed and stirred at rt. After 30 minutes, this solution was added to an heterogeneous mixture of methyl 3-bromo-benzoate (6.85g, 31.85 mmol), 2-methoxyphenyl boronic acid (7.26 g, 47.78 mmol) and Κ3Ρ04·Η20 (21.98 g, 95.56 mmol) in THF (32 mL). After stirring at rt for 18 h, the insolubles were removed by filtration through Celite and rinsed with EtOAc. The combined filtrates were concentrated under reduced pressure and the residue was purified by column chromatography (5% EtOAc in hexanes) to give a white solid (6.53 g, 85%); Rf = 0.5 (10% EtOAc in hexanes).
The title compound was prepared according to the procedure described for the synthesis of example A4; 1H NMR (500 MHz, DMSO-d6) δ: 9.47 (s, 1H), 8.08 (s, 1H), 7.86 (d, J = 8.0 Hz, 1H), 7.71 (d, J= 8.0 Hz, 1H), 7.66 (s, 2H), 7.52 (t, J= 8.0 Hz, 1H), 6.91 (d, J = 8.5 Hz, 1H), 6.73 (d, J= 3.0 Hz, 1H), 6.59 (dd, J= 8.5, 3.0 Hz, 1H), 3.12 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.89 ppm. LCMS
m/z: 557.4 [C2iHi7Br206P +1] . Anal for C2iHi7Br206P + 0.2 EtOAc; Calcd: C:45.64, H:3.27; Found: C:45.62, H:3.36.
Example E8
Methyl 5 '- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] - 2'-hydroxy-biphenyl-3-carboxylate
The title compound was prepared according to the procedure described for the synthesis of example E4; 1H NMR (500 MHz, DMSO-d6) δ: 9.50 (s, 1H), 8.11 (d, J = 2.0 Hz, 1H), 7.89 (dd, J= 8.0, 2.0 Hz, 1H), 7.74 (d, J= 8.0 Hz, 1H), 7.66 (s, 2H), 7.55 (t, J= 8.0 Hz, 1H), 6.90 (d, J= 8.5 Hz, 1H), 6.76 (d, J= 3.0 Hz, 1H), 6.59 (dd, J= 8.5, 3.0 Hz, 1H), 3.86 (s, 3H), 3.12 (d, J= 17.0 Hz, 2H), 1.28 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-dg) δ: 42.89 ppm. LCMS m/z: 571.6 [¾2Η19ΒΓ206Ρ +1]+. Anal for ¾2Η19ΒΓ206Ρ + 0.3 EtOAc; Calcd: C:46.71, H:3.62; Found: C:46.87, H:3.30.
Example E9-1
Methyl 5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino] -acetate
Step a: EDCI (90 mg, 0.47 mmol) was added to an heterogeneous mixture of methyl glycine hydrochloride (99 mg, 0.79 mmol), carboxylic acid intermediate 2 (200 mg, 0.39 mmol), HOBT (72 mg, 0.47 mmol) and diisopropylethylamine (0.26 mL, 1.50 mmol) in CH2C12 (6 mL) at rt. After stirring at rt for 16 h, the clear reaction mixture was partitioned between CH2C12 and 0.5 N HCl. The layers were separated and the organics were washed with 0.5 N HCl, a saturated solution of NaHC03 (2X), dried (Na2S04), filtered, concentrated under reduced pressure and the residue was purified by column
chromatography (7% methanol in CH2C12) to give the amide as a foam (60 mg, 26%).
I l l
Step b: Bromotrimethylsilane (0.27 mL, 2.07 mmol) was added to a solution of phosphinate (200 mg, 0.35 mmol) in CH2CI2 at 0 °C. The ice bath was removed and the reaction mixture was stirred at rt. After 16 h, the solution was concentrated under reduced pressure. The residue was taken up in 5/1 acetonitrile/water, stirred at rt for 20 minutes and concentrated under reduced pressure. The residue was azeotroped with methanol
(3x), dried under high vacuum, taken up in ether, sonicated and the product was collected by filtration to give a white powder (130 mg, 68%); 1H NMR (500 MHz, DMSO-d6) δ: 11.70 (s, 1H), 9.13 (s, 1H), 7.67 (s, 2H), 7.30 (d, J= 3.0 Hz, 1H), 6.95-6.90 (m, 2H), 4.05 (s, 2H), 3.65 (s, 3H), 3.14 (d, J= 17.0 Hz, 2H), 1.31 (d, J= 14.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.97 ppm. LCMS m/z: 552.4 [Ci8H18Br2N07P +1]+. Anal for Ci8H18Br2N07P; Calcd: C:38.21, H:3.69, N:2.42; Found: C:37.90, H:3.31, N:2.21.
Example E9-2
Ethyl 5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydrox y-benzoylamino] -acetate
Step a: Thionyl chloride (0.05 mL, 0.71 mmol) was added to a solution of carboxylic acid intermediate 2 (120 mg, 0.236 mmol) in chcloroform (4 mL). The reaction mixture was refluxed for 2 h, cooled to rt, concentrated under reduced pressure, azeotroped with CH2C12 (2X) and dried under high vacuum. The residue was taken up in CH2C12 (4 mL) and a solution of ethyl glycine hydrochloride (40 mg, 0.28 mmol) and triethylamine (0.13 mL, 0.94 mmol) in CH2C12 (4 mL) was added at rt. After stirring at rt for 16 h, the reaction mixture was quenched with water and the CH2C12 removed under reduced pressure. The residue was partitioned between EtOAc and 1 N HC1 and the layers were separated. The organics were washed with water, dried (Na2S04), filtered, concentrated under reduced pressure and purified by column chromatography to give the desired amide (50 mg, 36%).
Step b: The title compound was prepared according to the procedure described for the synthesis of example E9-1, step b (38 mg, 80%); 1H NMR (300 MHz, DMSO-d6) δ: 11.68 (s, 1H), 9.18 (m, 1H), 7.74 (s, 2H), 7.37 (d, J= 3.0 Hz, 1H), 7.00-6.97 (m, 2H), 4,19 (q, J = 7.1 Hz, 2H), 4.10 (d, J = 5.7 Hz, 2H), 3.20 (d, J= 17.0 Hz, 2H), 1.37 (d, J =
14.0 Hz, 3H), 1.27 (t, J= 7.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.97 ppm. LCMS m/z: 566.4 [Ci9H20Br2NO7P +1]+. Anal for Ci9H20Br2NO7P; Calcd: C:40.38, H:3.57, N:2.48; Found: C:40.23, H:3.60, N:2.32.
Example E9-3
Isopropyl 5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -acetate
The title compound was prepared according to the procedure described for the synthesis of example E9-1; 1H NMR (300 MHz, DMSO-d6) δ: 11.60 (s, 1H), 9.09 (m, 1H), 7.66 (s, 2H), 7.28 (d, J= 3.0 Hz, 1H), 7.00-6.90 (m, 2H), 4,97 (hept, J= 6.3 Hz, 1H), 3.98 (d, J = 5.4 Hz, 2H), 3.12 (d, J= 17.0 Hz, 2H), 1.29 (d, J= 14.0 Hz, 3H), 1.19 (d, J= 6.3 Hz, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.96 ppm. LCMS m/z: 580.6
[C20H22Br2NO7P +1]+. Anal for C20H22Br2NO7P; Calcd: C:41.48, H:3.83, N:2.42; Found: C:41.24, H:3.86, N:2.39.
Example E9-4
Propyl 5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydroxy- benzoylamino] -acetate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, CD3OD) δ: 7.65 (s, 2H), 7.26 (d, J= 2.4 Hz, 1H), 7.00-6.90 (m, 2H), 4,15-4.07 (m, 4H), 3.21 (d, J= 16.5 Hz, 2H), 1.67 (dt, J= 7.5, 7.5, 2H), 1.48 (d, J= 14.1 Hz, 3H), 0.94 (d, J= 7.5 Hz, 3H). 31P NMR (121.46 MHz, DMSO- d6) δ: 42.96 ppm. LCMS m/z: 580.6 [C20H22Br2NO7P +1]+. Anal for C20H22Br2NO7P; Calcd: C:41.48, H:3.83, N:2.42; Found: C:41.24, H:3.85, N:2.28.
Example E9-5
Benzyl 5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydroxy- benzoylamino] -acetate
The title compound was prepared according to the procedure described for the synthesis of example E9-1 1H NMR (300 MHz, CD3OD) δ: 7.66 (s, 2H), 7.40-7.26 (m, 5H),7.26 (d, J= 2.7 Hz, 1H), 6.95-6.85 (m, 2H), 5.19 (s, 2H), 4,14 (s, 2H), 3.22 (d, J= 16.8 Hz, 2H), 1.67 (dt, J= 7.5, 7.5, 2H), 1.48 (d, J= 14.4 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 43.01 ppm. LCMS m/z: 628.6 [C24H22Br2N07P +1]+. Anal for C24H22Br2N07P + 2.7 H20 + 0.1 CH3OH; Calcd: C:42.63, H:4.13, N:2.06; Found: C:42.26, H:3.72, N:2.03.
Example A9
5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydroxy- benzoylamino] -acetic acid
An aqueous solution of NaOH (2 N, 0.45 mL, 0.91 mmol) was added to a solution of compound from example 9-1 (100 mg, 0.18 mmol) in methanol at 0 °C. The ice bath was removed and the reaction mixture was stirred at rt. After 5 h at rt, additional NaOH was added (2 N, 0.27 mL). After stirring at rt for 16 h, most of the methanol was removed under reduced pressure and the aqueous solution partitioned between aqueous NaOH (2 N, 1 mL), water and ether. The layer were partitioned and the aqueous layer was washed with ether (2X), acidified with cone HC1 to pH 1 and extracted with EtOAc (2X). The combined EtOAc extracts were washed with water (2X) and concentrated under reduced pressure to give the title compound (60 mg, 62%); 1H NMR (300 MHz, DMSO-d6) δ:
11.67 (s, 1H), 9.06 (m, 1H), 7.67 (s, 2H), 7.31 (d, J= 2,7 Hz, 1H), 6.95-6.85 (m, 2H), 3.94 (d, J= 5.7 Hz, 2H), 3.12 (d, J= 17.1 Hz, 2H), 1.29 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.94 ppm. LCMS m/z: 538.1 [Ci7Hi6Br2N07P +1]+. Anal for Ci7H16Br2N07P; Calcd: C:38.02, H:3.00, N:2.61; Found: C:38.16, H:3.31, N:2.36.
Example El 0-1
(5) Ethyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -propanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.61 (s, 1H), 9.03 (d, J= 7.2 Ηζ,ΙΗ), 7.66 (s, 2H), 7.42 (d, J= 3.3 Hz, 1H), 6.90 (d, J= 8.7 Hz, 1H), 6.79 (dd, J= 8.7, 3.3 Hz, 1H), 4.43 (dq, J = 7.2, 7.2 Hz, 1H), 4,11 (q, J= 7.2 Hz, 2H), 3.12 (d, J= 16.8 Hz, 2H), 1.39 (d, J= 7.2 Hz, 3H), 1.29 (d, J= 14.1 Hz, 3H), 1.18 (t, J= 7.2 Hz, 3H). 31P
NMR (121.46 MHz, DMSO-d6) δ: 42.98 ppm. LCMS m/z: 580.6 [C2oH22Br2N07P +1]+. Anal for C2oH22Br2N07P + 0.3 H20, + 0.2 CH3OH; Calcd: C:41.05, H:3.99, N:2.37;
Found: C:40.98, H:4.00, N: 1.98.
Example El 0-2
(5) Isopropyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino]-propanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (500 MHz, DMSO-d6) δ: 11.62 (s, 1H), 9.03 (d, J= 7.2
Ηζ,ΙΗ), 7.67 (s, 2H), 7.42 (d, J= 3.0 Hz, 1H), 6.91 (d, J= 9.0 Hz, 1H), 6.80 (dd, J= 9.0, 3.0 Hz, 1H), 4.92 (hept, J= 7.5 Hz, 1H, 4.41 (dq, J= 7.5, 7.5 Hz, 1H), 3.14 (d, J= 17.0 Hz, 2H), 1.39 (d, J= 7.5 Hz, 3H), 1.30 (d, J= 14.0 Hz, 3H), 1.20(d, J= 7.5 Hz, 6H). LCMS m/z: 594.4 [C2iH24Br2N07P +1]+.
Example A10
(5) 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydroxy- benzoylamino] -propanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.65 (s, 1H), 9.01 (d, J= 7.2 Ηζ,ΙΗ), 7.66 (s, 2H), 7.42 (d, J= 3.3 Hz, 1H), 6.89 (d, J= 9.0 Hz, 1H), 6.79 (dd, J= 9.0, 3.3 Hz, 1H), 4.40 (dq, J= 7.2, 7.2 Hz, 1H), 4,11 (q, J= 7.2 Hz, 2H), 3.12 (d, J= 17.1 Hz, 2H), 1.38 (d, J= 7.2 Hz, 3H), 1.29 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.90 ppm. LCMS m/z: 552.4 [Ci8Hi8Br2N07P +1]+. Anal for Ci8Hi8Br2N07P; Calcd: C:39.23, H:3.29, N:2.54; Found: C:38.92, H:3.43, N:2.41.
Example Ell
Ethyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydr oxy-benzoylamino]-2-methyl-propanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.50 (s, 1H), 8.84 (s,lH), 7.66 (s, 2H), 7.43 (d, J= 3.0 Hz, 1H), 6.88 (d, J= 9.0 Hz, 1H), 6.79 (dd, J= 9.0, 3.3 Hz, 1H), 4,06 (q, J= 7.0 Hz, 2H), 3.13 (d, J= 17.1 Hz, 2H), 1.46 (s, 6H), 1.29 (d, J= 14.1 Hz, 3H), 1.12 (t, J = 7.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.97 ppm. LCMS m/z: 594.4 [C2iH24Br2N07P +1]+. Anal for C2iH24Br2N07P + 0.3 H20 + 0.3 CH3OH; Calcd: C:42.06, H:4.28, N:2.30; Found: C:41.75, H:3.84, N: 1.88.
Example All
2-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydr oxy-benzoylamino] -2-methyl-propanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.52 (s, 1H), 8.89 (s,lH), 7.66 (s, 2H), 7.40 (d, J= 2.7 Hz, 1H), 6.88 (d, J= 9.0 Hz, 1H), 6.79 (dd, J= 9.0, 2.7 Hz, 1H),
3.12 (d, J= 16.5 Hz, 2H), 1.46 (s, 6H), 1.29 (d, J= 14.4 Hz, 3H). 31P NMR (121.46 MHz, DMSO-dg) δ: 42.96 ppm. LCMS m/z: 566.1 [Ci9H2oBr2N07P +1]+. Anal for
Ci9H2oBr2N07P; Calcd: C:40.38, H:3.57, N:2.48; Found: C:40.09, H:3.45, N:2.30.
Example E12-1
(5) Ethyl 2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino]-3-methyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.41 (s, 1H), 8.90 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.29 (d, J= 3.0 Hz, 1H), 7.00-6.85 (m, 2H), 4.35 (dd, J= 5.7, 7.5 Hz, 1H), 4.20-4.03 (m, 2H), 3.13 (d, J= 16.8 Hz, 2H), 2.23-2.09 (m, 1H), 1.28 (d, J= 14.1 Hz, 3H), 1.18 (t, J= 7.2 Hz, 3H), 0.92 (d, J= 7.2 Hz, 6H). 31P NMR (121.46 MHz, DMSO-dg) δ: 42.80 ppm. LCMS m/z: 608.9 [C22H26Br2N07P +1]+. Anal for
C22H26Br2N07P; Calcd: C:43.52, H:4.32, N:2.31; Found: C:43.41, H:4.13, N:2.15.
Example E12-2
(5) Propyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino]-3-methyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.50 (s, 1H), 8.99 (d, J= 7.5 Hz, 1H), 7.74 (s, 2H), 7.38 (d, J= 3.3 Hz, 1H), 7.05-6.95 (m, 2H), 4.45 (dd, J= 5.7, 7.5 Hz,
1H), 4.20-4.03 (m, 2H), 3.21 (d, J= 17.1 Hz, 2H), 2.30-2.08 (m, 1H), 1.72-1.60 (m, 2H), 1.37 (d, J= 14.1 Hz, 3H), 1.00 (d, J= 7.2 Hz, 6H), 0.95 (t, J= 7.5 Hz, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.76 ppm. LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for C23H28Br2N07P; Calcd: C:44.47, H:4.54, N:2.25; Found: C:44.21, H:4.25, N:2.14.
Example E12-3
(5) Isopropyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino]-3-methyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.40 (s, 1H), 8.99 (d, J= 7.8 Hz, 1H), 7.66 (s, 2H), 7.28 (d, J= 2.7 Hz, 1H), 7.00-6.93 (m, 2H), 4.92 (hept, J= 6.9 Hz, 1H), 4.32 (dd, J= 5.7, 7.5 Hz, 1H), 3.12 (d, J= 17.1 Hz, 2H), 2.20-2.10 (m, 1H), 1.28 (d, J= 14.1 Hz, 3H), 1.00 (d, J= 7.0 Hz, 6H), 0.91 (d, J= 6.9 Hz, 3H), 31P NMR (121.46 MHz, DMSO-de) δ: 42.78 ppm. LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for C23H28Br2N07P; Calcd: C:44.47, H:4.54, N:2.25; Found: C:44.12, H:4.38, N:2.17.
Example A12
(5) 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydroxy- benzoylamino] -3-methyl-butanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.38 (s, 1H), 8.83 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.31 (d, J= 3.0 Hz, 1H), 7.00-6.83 (m, 2H), 4.35 (dd, J= 5.7, 7.5 Hz, 1H), 3.12 (d, J= 16.5 Hz, 2H), 2.23-2.09 (m, 1H), 1.29 (d, J= 14.4 Hz, 3H), 0.91 (d, J = 6.9 Hz, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.96 ppm. LCMS m/z: 580.6
[C2oH22Br2N07P +1]+. Anal for C2oH22Br2N07P; Calcd: C:41.48, H:3.83, N:2.42; Found: C:41.23, H:3.58, N:2.26.
Example E13
(5) Ethyl 2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino] -3-phenyl-propanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.41 (s, 1H), 8.99 (d, J= 7.2 Hz, 1H), 7.66 (s, 2H), 7.32-7.18 (m, 6H), 6.98-6.80 (m, 2H), 4.72-4.62 (m, 1H), 4.08 (q, J = 7.2 Hz, 2H), 3.12 (d, J= 17.7 Hz, 2H), 1.28 (d, J= 13.8 Hz, 3H), 1.13 (t, J= 7.2 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.76 ppm. LCMS m/z: 656.6 [C26H26Br2N07P +1]+. Anal for C26H26Br2N07P; Calcd: C:47.01, H:4.10, N:2.11; Found: C:46.80, H:3.61, N:2.03.
Example A13
(5) 2- [5- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -3-phenyl-propanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.43 (s, 1H), 8.95 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.35-7.18 (m, 6H), 6.90-6.80 (m, 2H), 4.7-4.58 (m, 1H), 3.12 (d, J = 16.8 Hz, 2H), 1.29 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSC /6) δ: 42.96 ppm.
LCMS m/z: 628.6 [C24H22Br2N07P +1]+. Anal for C24H22Br2N07P + H20; Calcd:
Example E14
(5) Ethyl 2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino]-4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.60 (s, 1H), 8.95 (d, J= 7.2 Hz, 1H), 7.66 (s, 2H), 7.43 (d, J= 3.3 Hz, 1H), 6.90 (d, J= 8.7 Hz, 1H), 6.79 (dd, J= 8.7, 3.3 Hz, 1H), 4.50-4.42 (m, 1H), 4.11 (q, J= 7.2 Hz, 2H), 3.12 (d, J= 17.1 Hz, 2H), 1.80-1.58 (m, 3H), 1.29 (d, J= 14.1 Hz, 3H), 1.17 (t, J= 7.2 Hz, 3H), 0.91 (d, J= 5.7 Hz, 3H), 0.87 (d, J= 6.0 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.99 ppm. LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for C23H28Br2N07P + CF3COOH; Calcd: C:40.84, H:3.98, N: 1.90; Found: C:40.83, H:4.01, N: 1.99.
Example A14
(5) 2- [5- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -4-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.67 (s, 1H), 8.91 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.47 (d, J= 3.0 Hz, 1H), 6.89 (d, J= 9.0 Hz, 1H), 6.77 (dd, J= 9.0, 3.0 Hz, 1H), 4.50-4.48 (m, 1H), 3.12 (d, J= 16.8 Hz, 2H), 1.80-1.58 (m, 3H), 1.29 (d, J = 14.4 Hz, 3H), 0.90 (d, J= 6.0 Hz, 3H), 0.7 (d, J= 6.3 Hz, 3H). 31P NMR (121.46 MHz, DMSO-dg) δ: 43.03 ppm. LCMS m/z: 594.6 [C2iH24Br2N07P +1]+. Anal for
Example E15
(5) Ethyl 2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino]-4-methylsulfanyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.57 (s, 1H), 8.95 (d, J= 6.9 Hz, 1H), 7.66 (s, 2H), 7.42 (d, J= 3.3 Hz, 1H), 6.91 (d, J= 9.0 Hz, 1H), 6.79 (dd, J= 9.0, 3.3 Hz, 1H), 4.62-4.52 (m, 1H), 4.12 (q, J= 7.2 Hz, 2H), 3.13 (d, J= 16.5 Hz, 2H), 2.12-1.98 (m, 4H), 1.29 (d, J= 14.1 Hz, 3H), 1.18 (t, J= 7.2 Hz, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.99 ppm. LCMS m/z: 640.4 [C22H26Br2N07PS +1]+. Anal for
C22H26Br2N07PS + CF3COOH; Calcd: C:38.27, H:3.61, N: 1.86; Found: C:38.58, H:3.68, N: 1.82.
Example A15
(5) 2- [5- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -4-methylsulfanyl-butanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.64 (s, 1H), 8.98 (d, J= 7.2 Hz, 1H), 7.66 (s, 2H), 7.45 (d, J= 3.3 Hz, 1H), 6.89 (d, J= 9.0 Hz, 1H), 6.77 (dd, J= 9.0, 3.3 Hz, 1H), 4.58-4.45 (m, 1H), 3.13 (d, J= 16.8 Hz, 2H), 2.12-1.98 (m, 4H), 1.29 (d, J = 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.98 ppm. LCMS m/z: 612.4
[C2oH22Br2N07PS +1]+. Anal for C2oH22Br2N07PS + CF3COOH; Calcd: C:36.43, H:3.20, N: 1.93; Found: C:36.62, H:3.40, N: 1.75.
Example E16
(5) Ethyl l-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoyl]-pyrrolidine-2-carboxylate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6, 80 °C) δ: 9.50-9.40 (m, 1H), 7.66 (s, 2H), 6.85 (d, J= 5.4 Hz, 1H), 6.75 (d, J= 5.4 Hz, 1H), 6.50 (s, 1H), 4.45-4.38 (m, 1H), 4.12-3.98 (m, 2H), 3.60-3.40 (m, 2H), 3.12 (d, J= 10.2 Hz, 2H), 2.90-2.70 (m, 1H), 2.40- 2.25 (m, 1H), 1.90-1.80 (m, 2H), 1.31 (d, J= 8.4 Hz, 3H), 1.20-1.05 (m, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.95 ppm.
LCMS m/z: 606.6 [C22H24Br2N07P +1]+.
Anal for C22H24Br2N07P + 0.7 CF3COOH; Calcd: C:41.03, H:3.63, N:2.04; Found: C:41.05, H:3.71, N: 1.94.
Example A16
(5) l-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoyl] -pyrr olidine-2-carboxylic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6, 80 °C) δ: 9.50-9.40 (m, 1H), 7.66 (s, 2H), 6.85 (d, J= 5.4 Hz, 1H), 6.75 (d, J= 5.4 Hz, 1H), 6.56 (s, 1H), 4.40-4.30 (m, 1H), 3.45-3.35 (m, 2H), 3.12 (d, J= 10.2 Hz, 2H), 2.90-2.70 (m, 1H), 2.30-2.15 (m, 1H), 2.00- 1.80 (m, 2H), 1.33 (d, J= 8.4 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 43.01 ppm. LCMS m/z: 578.6 [C2oH2oBr2N07P +1]+. Anal for C2oH2oBr2N07P + 0.2 CF3COOH; Calcd: C:42.02, H:3.63, N:2.38; Found: C:42.31, H:3.61, N:2.14.
Example E17
Ethyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino]-propanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.88 (s, 1H), 8.94 (m, 1H), 7.74 (s, 2H), 7.44 (d, J= 3.3 Hz, 1H), 6.95 (d, J= 9.0 Hz, 1H), 6.85 (dd, J= 9.0, 3.3 Hz, 1H), 4.14 (q, J= 7.2 Hz, 2H), 3.60-3.50 (m, 2H), 3.20 (d, J= 17.4 Hz, 2H), 2.65 (t, J= 6.8 Hz, 2H), 1.37 (d, J= 14.4 Hz, 3H), 1.24 (t, J= 7.2 Hz, 3H). 31P NMR (121.46 MHz, DMSO- d6) δ: 42.82 ppm. LCMS m/z: 580.6 [C2oH22Br2N07P +1]+. Anal for C2oH22Br2N07P; Calcd: C:41.48, H:3.83, N:2.42; Found: C:41.25, H:3.70, N:2.27.
Example A17
3-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydroxy- benzoylamino] -propanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.90 (s, 1H), 8.94 (m, 1H), 7.74 (s, 2H), 7.45 (d, J= 3.3 Hz, 1H), 6.95 (d, J= 9.0 Hz, 1H), 6.85 (dd, J= 9.0, 3.3 Hz, 1H), 3.60-3.50 (m, 2H), 3.20 (d, J= 16.8 Hz, 2H), 2.60-2.50 (m, 2H), 1.37 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.70 ppm. LCMS m/z: 552.4
[Ci8Hi8Br2N07P +1]+. Anal for Ci8Hi8Br2N07P; Calcd: C:39.23, H:3.29, N:2.54; Found: C:38.95, H:3.19, N:2.41.
Ethyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydr oxy-benzoylamino]-4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.78 (s, 1H), 8.80 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.47 (d, J= 3.3 Hz, 1H), 6.85 (d, J= 9.0 Hz, 1H), 6.67 (dd, J= 9.0, 3.3 Hz, 1H), 4.25-4.10 (m, 1H), 4.00 (q, J= 7.1 Hz, 2H), 3.12 (d, J= 16.8 Hz, 2H), 2.60-2.45 (m, 2H), 1.90-1.75 (m, 1H), 1.28 (d, J= 13.8 Hz, 3H), 1.09 (t, J= 7.1 Hz, 3H), 0.88 (d, J = 7.0 Hz, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.80 ppm. LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for C23H28Br2N07P; Calcd: C:44.47, H:4.54, N:2.25; Found: C:44.25, H:4.46, N:2.19.
Example A18
3-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydr oxy-benzoylamino] -4-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.82 (s, 1H), 8.80 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.49 (d, J= 3.3 Hz, 1H), 6.85 (d, J= 8.7 Hz, 1H), 6.68 (dd, J= 8.7, 3.3 Hz, 1H), 4.25-4.10 (m, 1H), 3.12 (d, J= 17.1 Hz, 2H), 2.50-2.40 (m, 2H), 1.90-1.75 (m, 1H), 1.29 (d, J= 14.1 Hz, 3H), 0.87 (d, J= 7.2 Hz, 6H). 31P NMR (121.46 MHz, DMSO- d6) δ: 42.88 ppm. LCMS m/z: 594.4 [C2iH24Br2N07P +1]+. Anal for C2iH24Br2N07P; Calcd: C:42.52, H:4.08, N:2.36; Found: C:42.43, H:4.18, N:2.25.
Example E19
Ethyl 3- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydr oxy-benzoylamino] -butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.85 (s, 1H), 8.75 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.48 (d, J= 3.3 Hz, 1H), 6.85 (d, J= 9.3 Hz, 1H), 6.66 (dd, J= 9.3, 3.3 Hz, 1H), 4.42-4.30 (m, 1H), 4.04 (q, J= 7.2 Hz, 2H), 3.12 (d, J= 16.8 Hz, 2H), 3.70-3.50 (m, 2H), 1.28 (d, J= 13.8 Hz, 3H), 1.09 (t, J= 7.1 Hz, 3H), 1.20-1.10 (m, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.67 ppm. LCMS m/z: 594.4 [C2iH24Br2N07P +1]+. Anal for C2iH24Br2N07P; Calcd: C:42.52, H:4.08, N:2.36; Found: C:42.47, H:4.19, N:2.21.
Example A19
3-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydr oxy-benzoylamino]-butanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSC /6) δ: 11.85 (s, 1H), 8.75 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.49 (d, J= 3.0 Hz, 1H), 6.84 (d, J= 9.0 Hz, 1H), 6.66 (dd, J= 9.0, 3.3 Hz, 1H), 4.40-4.30 (m, 1H), 3.12 (d, J= 18.3 Hz, 2H), 3.65-3.50 (m, 2H), 1.29 (d, J = 14.1 Hz, 3H), 1.09 (t, J= 7.1 Hz, 3H), 1.20-1.10 (d, J= 6.6 Hz, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.79 ppm. LCMS m/z: 566.1 [Ci9H2oBr2N07P +1]+. Anal for Ci9H2oBr2N07P + 0.6 H20; Calcd: C:39.62, H:3.71, N:2.43; Found: C:39.60, H:3.73, N:2.13.
Example E20-1
(5) Methyl 2- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -3-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.42 (s, 1H), 8.94 (d, J= 7.5 Hz,
1H), 7.66 (s, 2H), 7.30 (d, J= 3.0 Hz, 1H), 7.00-6.85 (m, 2H), 4.43 (dd, J= 7.5, 5.7 Hz, 1H), 3.65 (s, 3H), 3.13 (d, J= 16.8 Hz, 2H), 2.00-1.85 (m, 1H), 1.50-1.35 (m, 1H), 1.29 (d, J= 14.1 Hz, 3H), 1.30-1.10 (m, 1H), 0.95-0.80 (m, 6H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.96 ppm. LCMS m/z: 608.9 [C22H26Br2N07P +1]+. Anal for
C22H26Br2N07P; Calcd: C:39.11, H:3.64, N: 1.85; Found: C:38.90, H:3.92, N: 1.86.
Example E20-2
(5) Ethyl 2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2- hydroxy-benzoylamino] -3-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.40 (s, 1H), 8.93 (d, J= 7.8 Hz, 1H), 7.66 (s, 2H), 7.30 (d, J= 3.0 Hz, 1H), 7.00-6.85 (m, 2H), 4.41 (dd, J= 7.8, 5.7 Hz, 1H), 4.20-4.05 (m, 2H), 3.13 (d, J= 17.1 Hz, 2H), 2.00-1.85 (m, 1H), 1.50-1.35 (m, 1H), 1.29 (d, J= 14.4 Hz, 3H), 1.30-1.15 (m, 1H), 0.95-0.83 (m, 6H). 31P NMR (121.46 MHz, DMSO-dg) δ: 42.95 ppm. LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for
C23H28Br2N07P; Calcd: C:44.47, H:4.54, N:2.25; Found: C:44.65, H:4.29, N: 1.99.
Example A20
(5) 2- [5- [2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2- hydroxy-benzoylamino] -3-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6) δ: 11.37 (s, 1H), 8.89 (d, J= 7.5 Hz, 1H), 7.66 (s, 2H), 7.30 (d, J= 3.0 Hz, 1H), 7.00-6.85 (m, 2H), 4.39 (dd, J= 7.5, 4.8 Hz, 1H), 3.12 (d, J= 16.8 Hz, 2H), 2.00-1.85 (m, 1H), 1.55-1.40 (m, 1H), 1.29 (d, J= 14.1 Hz, 3H), 1.30-1.10 (m, 1H), 0.95-0.80 (m, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ:
42.96 ppm. LCMS m/z: 594.4 [C2iH24Br2N07P +1] . Anal for C2iH24Br2N07P; Calcd: C:41.88, H:4.18, N:2.33; Found: C:41.57, H:3.96, N:2.19.
Example E21
(5) Ethyl 2-([5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]- 2-hydroxy-benzoyl]-methyl-amino)-3-methyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6, 80 °C) δ: 7.66 (s, 2H), 6.86 (d, J= 5.1 Hz, 1H), 6.76 (d, J= 3.9 Hz, 1H), 4.18-4.02 (m, 2H), 4.20-4.03 (m, 3H), 3.12 (d, J= 10.2 Hz, 2H), 2.30-2.10 (m 1H), 2.08 (s, 3H), 1.30 (d, J= 8.4 Hz, 3H), 1.18 (t, J= 8.7 Hz, 3H), 1.10-0.90 (m, 6H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.80 ppm.
LCMS m/z: 622.6 [C23H28Br2N07P +1]+. Anal for C23H28Br2N07P + 0.8 CF3COOH; Calcd: C:41.47, H:4.07, N: 1.97; Found: C:41.34, H:4.16, N: 1.90.
Example A21
(5) 2-([5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]- 2-hydroxy-benzoyl] -methyl-amino)-3-methyl-butanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A9; 1H NMR (300 MHz, DMSO-d6, 80 °C) δ: 9.40-9.30 (m, 1H), 7.64 (s, 2H), 7.07-6.65 (m, 2H), 6.38 (s, 1H), 3.40-2.80 (m, 1H), 3.13 (d, J= 9.9 Hz, 2H), 2.80 (s, 3H), 2.25-2.05 (m 1H), 1.20-1.05 (m, 3H), 1.10-0.70 (m, 6H).
31P NMR (121.46 MHz, DMSC /6) δ: 43.05 ppm.
LCMS m/z: 594.6[C2iH24Br2N07P +1]+.
Anal for C2iH24Br2N07P + 0.3 CF3COOH; Calcd: C:42.85, H:4.14, N:3.01; Found: C:42.92, H:4.28, N:2.86.
Example E22
(5) Ethyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -propanoate
Step a: Diisopropylethylamine (0.35 mL, 2.02 mmol) was added dropwise to a suspension of ethyl alanine hydrochloride (163 mg, 1.06 mmol) in CH2CI2 (5.3 mL) at 0 °C. After stirring at 0 °C for 10 minutes, the carboxylic acid intermediate 4 (200 mg, 0.53 mmol), HO AT (89 mg, 0.64 mmol) and EDCI (122 mg, 0.64 mmol) was added to the clear solution and the ice bath was removed. After stirring at rt for 16 h, the clear reaction mixture was partitioned between CH2CI2 and 10% hydrochloric acid. The layers were separated and the organics were washed with 10%> hydrochloric acid, a saturated solution of NaHCC"3 (2X), dried (Na2S04), filtered, concentrated under reduced pressure and the residue was purified by column chromatography (3%> methanol in CH2CI2) to give the amide as a foam (78 mg, 31 >).
Step b: Bromotrimethylsilane (0.28 mL, 2.1 mmol) was added to a solution of
phosphinate (101 mg, 0.21 mmol) in CH2CI2 at 0 °C. The ice bath was removed and the reaction mixture was stirred at rt. After 16 h, the solution was concentrated under reduced pressure. The residue was taken up in 5/1 acetonitrile/water (6 mL), stirred at rt for 20 minutes and concentrated under reduced pressure. The residue was partitioned between EtOAc and water. The layers were separated and the aqueous phase was extracted with EtOAc. The combined organic extracts were washed with brine, dried (Na2S04), filtered, and concentrated under reduced pressure to give a white foam (64 mg, 67%>); 1H NMR
(500 MHz, DMSO-dg) δ: 7.71 (s, 1H), 6.88 (s, 2H), 6.80 (m, 1H), 6.69 (m, 1H), 4.46 (m, 1H), 4,10 (m, 2H), 3.81 (s, 2H), 2.65 (d, J= 19.5 Hz, 2H), 2.06 (s, 6H), 1.39 (d, J= 7.0 Hz, 3H), 1.17 (m, 3H), 0.94 (d, J= 12.0 Hz, 3H). LCMS m/z: 448.9 [C23H30NO6P +1]+. Anal for C23H30NO6P + 2.7 H20, + 0.2 EtOAc; Calcd: C:55.64, H:7.26, N:2.73; Found: C:55.56, H:6.54, N:2.53.
(5) 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -propanoic acid
An aqueous solution of NaOH (2 N, 0.25 mL, 0.5 mmol) was added to a solution of compound from example E22 (42 mg, 0.094 mmol) in methanol at 0 °C. The ice bath was removed and the reaction mixture was stirred at rt. After stirring at rt for 16 h, most of the methanol was removed under reduced pressure and the aqueous solution partitioned between aqueous 1 N NaOH , and ether. The layer were partitioned and the aqueous layer was washed with ether (2X), acidified with 1 N HCl to pH 1 and extracted with EtOAc (2X). The combined EtOAc extracts were washed with brine, dried (Na2S04), filtered, and concentrated under reduced pressure to give the title compound as a white foam (27 mg, 68%); 1H NMR (500 MHz, DMSO- 6) δ: 8.93 (s, 1H), 7.72 (s, 1H), 6.98 (s, 2H), 6.90 (d, J= 9.0 Hz, 1H), 6.84 (d, J= 9.0 Hz, 1H), 4.45 (m, 1H), 3.95 (s, 2H), 2.97 (d, J = 18.0 Hz, 2H), 2.21 (s, 6H), 1.44 (d, J= 7.5 Hz, 3H), 1.30-1.20 (m, 3H). LCMS m/z: 420.9 [C2iH26N06P +1]+. Anal for C2iH26N06P + 1.3 H20, + 0.5 EtOAc; Calcd: C:56.74, H:6.75, N:2.88; Found: C:56.63, H:6.39, N:2.59.
Example E23
(5) Ethyl 2-[2-hydroxy-5-[4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -3-phenyl-propanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (500 MHz, DMSO-d6) δ: 11.33 (s, 1H), 8.84 (d, J= 7.5 Hz, 1H), 7.51 (s, 1H), 7.20-7.10 (m, 5H), 6.84 (s, 2H), 6.82 (d, J= 8.0 Hz, 1H), 6.70 (d, J = 8.0 Hz, 1H), 4.60 (q, J= 6.5 Hz, 1H), 4,00 (q, J= 7.0 Hz, 2H), 3.79 (s, 2H), 2.83 (d, J = 17.5 Hz, 2H), 2.07 (s, 6H), 1.12 (d, J= 14.0 Hz, 3H), 1.04 (d, J= 7.0 Hz, 3H). LCMS m/z: 524.9 [C29H34N06P +1]+. Anal for C29H34N06P + 0.8 H20; Calcd: C:64.75, H:6.67, N:2.60; Found: C:65.07, H:6.70, N:2.83.
Example A23
(5) 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -3-phenyl-propanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 11.43 (s, 1H), 8.86 (d, J= 7.5 Hz, 1H), 7.59 (s, 1H), 7.30-7.15 (m, 5H), 6.92 (s, 2H), 6.89 (d, J= 8.4 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.64 (m, 1H), 3.87 (s, 2H), 2.91 (d, J= 17.4 Hz, 2H), 2.15 (s, 6H), 1.20 (d, J = 13.5 Hz, 3H). LCMS m/z: 496.4 [C27H30NO6P +1]+. Anal for C27H30NO6P + 0.8 H20 + 0.1 EtOAc; Calcd: C:63.44, H:6.30, N:2.70; Found: C:63.42, H:6.14, N:2.74.
Example E24-1
(5) Methyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (500 MHz, DMSO-d6) δ: 11.70 (s, 1H), 8.94 (d, J= 7.5 Hz, 1H), 7.76 (s, 1H), 6.99 (s, 2H), 6.90-6.80 (m, 2H), 4.57 (m, 1H), 3.96 (s, 2H), 3.70 (s, 3H), 2.98 (d, J= 18.0 Hz, 2H), 2.21 (s, 6H), 1.83-1.63 (m, 3H), 1.27 (d, J= 14.0 Hz, 6H), 0.97 (d, J= 6.5 Hz, 3H), 0.93 (d, J= 6.5 Hz, 3H). LCMS m/z: 476.6 [C25H34NO6P +1]+. Anal for C25H34NO6P + 0.3 H20 + 0.3 EtOAc; Calcd: C:62.02, H:7.35, N:2.76; Found: C:61.90, H:7.28, N:2.86.
Example E24-2
(5) Ethyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (300 MHz, DMSO-d6) δ: 11.64 (s, 1H), 8.86 (m, 1H), 7.69 (s, 1H), 6.92 (m, 1H), 6.90-6.75 (m, 1H), 4.47 (m, 1H), 4.20-4.00 (m, 2H), 3.88 (s, 2H), 2.91 (d, J= 17.4 Hz, 2H), 2.14 (s, 6H), 1.80-1.50 (m, 3H), 1.30-1.10 (m, 6H), 1.00-0.80 (m, 6H). LCMS m/z: 490.6 [C26H36N06P +1]+. Anal for C26H36N06P + 0.6 H20; Calcd:
C:62.41, H:7.49, N:2.80; Found: C:62.31, H:7.31, N:2.89.
Example E24-3
(5) Propyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (500 MHz, DMSO-d6) δ: 11.68 (s, 1H), 8.93 (d, J= 7.0 Hz, 1H), 7.45 (s, 1H), 6.98 (s, 2H), 6.88 (d, J= 8.5 Hz, 1H), 6.84 (d, J= 8.5 Hz, 1H), 4.53 (m, 1H), 4.10-4.00 (m, 2H), 3.94 (s, 2H), 2.97 (d, J= 17.5 Hz, 2H), 2.20 (s, 6H), 1.83-1.57 (m, 5H), 1.26 (d, J= 14.0 Hz, 6H), 1.00-0.85 (m, 9H). LCMS m/z: 504.6 [C27H38N06P +1]+. Anal for C27H38N06P + 0.5 H20; Calcd: C:63.27, H:7.67, N:2.73; Found: C:63.36, H:7.69, N:2.79.
Example A24
(5) 2- [2-Hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -4-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (500 MHz, DMSO-d6) δ: 11.75 (s, 1H), 8.88 (d, J= 7.5 Ηζ,ΙΗ), 7.77 (s, 1H), 6.98 (s, 2H), 6.90-6.80 (m, 2H), 4.50 (m, 1H), 3.95 (s, 2H), 2.98 (d, J= 17.5 Hz, 2H), 2.21 (s, 6H), 1.80-1.60 (m, 3H), 1.26 (d, J= 14.0 Hz, 6H), 0.97 (d, J = 6.0 Hz, 6H), 0.93 (d, J= 6.0 Hz, 6H). LCMS m/z: 462.6 [C24H32NO6P +1]+. Anal for C24H32NO6P + 1.3 H20 + 0.3 EtOAc; Calcd: C:59.19, H:7.29, N:2.74; Found: C:59.20, H:7.41, N:2.72.
Example E25-1
(5) Ethyl 2-[2-hydroxy-5-[4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -3-methyl-butanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (500 MHz, DMSO-d6) δ: 11.44 (s, 1H), 8.86 (s, 1H), 7.68 (s, 1H), 6.98 (m, 3H), 6.89 (m, 2H), 4.41 (m, 1H), 4.25-4,10 (m, 2H), 3.95 (s, 2H), 2.98 (d, J = 16.0 Hz, 2H), 2.20 (s, 6H), 1.35-1.15 (m, 6H), 1.05-0.90 (m, 6H). LCMS m/z: 476.6 [C25H34N06P +1]+. Anal for C25H34NO6P + 0.8 H20; Calcd: C:61.29, H:7.32, N:2.86; Found: C:61.27, H:7.33, N:2.56.
Example E25-2
(5) Pivaloyloxy methyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6- dimethyl-benzyl] -benzoylamino] -3-methyl-butanoate
Step a: A mixture N-Boc-Valine (1.55 g, 7.13 mmol), iodomethyl pivalate (1.73 g, 7.13 mmol) and Cs2C03 (2.32 g, 7.13 mmol) in DMF (50 mL) was heated at 60 °C. After 3 h at 60 °C, the yellow heterogeneous mixture was partitioned between EtOAc and water. The layers were separated and the organics were washed with, water (2X), A saturated solution of NaHC03, 10% hydrochloric acid, brine, dried (Na2S04), filtered and concentrated under reduced pressure to give a clear oil (1.685 g, 71%).
Step b: TFA ( 7.5 mL) was added to a solution of pivaloyloxymethyl N-Boc-valinate (500 mg) in CH2C12 (7.5 mL) at 0 °C. After stirring at 0 °C for 2 h, the reaction mixture was concentrated under reduced pressure and azeotroped with toluene to give the free amine.
The title compound was prepared according to the procedure described for the synthesis of example E22 using pivaloyloxymethyl valinate; 1H NMR (500 MHz, DMSO- 6) §:
11.46 (s, 1H), 8.88 (d, J= 7.0 Hz, 1H), 7.67 (s, 1H), 6.98 (m, 3H), 6.88 (d, J= 8.0 Hz, 1H), 5.86 (d, J= 6.0 Hz, 1H), 5.74 (d, J= 6.0 Hz, 1H), 4.40 (m, 1H), 3.94 (s, 2H), 2.97 (d, J= 17.5.0 Hz, 2H), 2.20 (s, 6H), 1.26 (d, J= 14.0 Hz, 3H), 1.16 (s, 9H), 0.98 (d, J = 5.0 Hz, 1H). LCMS m/z: 562.6 [C29H40NO8P +1]+. Anal for C29H40NO8P + 1.2 H20; Calcd: C:59.72, H:7.33, N:2.40; Found: C:59.97, H:7.82, N:2.87.
Example A25
(5) 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -3-methyl-butanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (500 MHz, DMSO-d6) δ: 11.41 (s, 1H), 8.80 (d, J= 8.0 Hz, 1H), 7.69 (s, 1H), 6.98 (m, 3H), 6.88 (d, J= 8.0 Hz, 1H), 4.40 (m, 1H), 3.95 (s, 2H), 2.97 (d, J= 18.0 Hz, 2H), 2.20 (s, 6H), 1.26 (d, J= 13.5 Hz, 3H), 0.97 (d, J= 6.5 Hz, 6H). LCMS m/z: 448.9 [C23H30NO6P +1]+. Anal for C23H30NO6P + 0.9 H20 + 0.2 EtOAc; Calcd: C:59.39, H:6.99, N:2.91; Found: C:59.16, H:6.66, N:2.80.
Example E26
(5) Ethyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -3-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (300 MHz, DMSO-d6) δ: 11.42 (s, 1H), 8.88 (d, J= 7.5 Hz, 1H), 7.67 (s, 1H), 6.98 (m, 3H), 6.88 d, J= 8.0 Hz, 1H), 4.47 (m, 1H), 4.25-4.10 (m, 2H), 2.98 (d, J= 17.5 Hz, 2H), 2.20 (s, 6H), 1.95 (m, 1H), 1.50 (m, 1H), 1.30-1.20 (m, 7H), 1.00-0.90 (m, 6H). LCMS m/z: 490.6 [C26H36N06P +1]+. Anal for C26H36N06P + 0.5 H20 + 0.1 EtOAc; Calcd: C:62.50, H:7.51, N:2.76; Found: C:62.32, H:7.57, N:3.06.
Example A26
(5) 2- [2-Hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -3-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 11.40 (s, 1H), 8.83 (d, J= 8.0 Hz, 1H), 7.68 (s, 1H), 6.98 (m, 3H), 6.87 d, J= 8.5 Hz, 1H), 4.45 (m, 1H), 2.97 (d, J= 18.0 Hz, 2H), 2.20 (s, 6H), 1.95 (m, 1H), 1.50 (m, 1H), 1.26 (d, J= 13.5 Hz, 3H), 1.30-1.20 (m, 1H), 1.00-0.90 (m, 6H). LCMS m/z: 462.6 [C24H32N06P +1]+. Anal for C24H32N06P + 1.4 H20 + 0.2 EtOAc; Calcd: C:59.06, H:7.27, N:2.78; Found: C:59.07, H:7.42, N:2.71.
Example E27
(R) Ethyl 2- [2-hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl- benzyl] -benzoylamino] -4-methyl-pentanoate
The title compound was prepared according to the procedure described for the synthesis of example E22; 1H NMR (500 MHz, DMSO-d6) δ: 11.67 (s, 1H), 8.92 (d, J= 7.5 Hz, 1H), 7.75 (s, 1H), 6.98 (s, 2H), 6.89 (d, J= 8.5 Hz, 1H), 6.84 (d, J= 8.5 Hz, 1H), 4.52 (m, 1H), 4.20-4.10 (m, 2H), 3.95 (s, 2H), 2.97 (d, J= 17.5 Hz, 2H), 2.20 (s, 6H), 1.80-1.60 (m, 3H), 1.26 (d, J= 13.5 Hz, 3H), 1.22 (t, J= 7.0 Hz, 3H), 0.97 (d, J= 6.5 Hz, 3H), 0.93 (d, J= 6.5 Hz, 3H). LCMS m/z: 490.9 [C26H36N06P +1]+. Anal for C26H36N06P + 0.8 H20 + 0.1 EtOAc; Calcd: C:61.84, H:7.55, N:2.73; Found: C:61.93, H:7.49, N:2.68.
Example A27
(R) 2- [2-Hydroxy-5- [4-(hydroxy-methyl-phosphinoylmethyl)-2,6-dimethyl-benzyl] - benzoylamino] -4-methyl-pentanoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (500 MHz, DMSO-d6) δ: 11.77 (s, 1H), 8.89 (d, J= 8.0 Hz, 1H), 7.78 (s, 1H), 6.99 (s, 2H), 6.90-6.80 (m, 2H), 4.51 (m, 1H), 3.96 (s, 2H), 2.98 (d, J = 17.5 Hz, 2H), 2.22 (s, 6H), 1.80-1.60 (m, 3H), 1.27 (d, J= 14.0 Hz, 3H), 0.98 (d, J= 6.0 Hz, 3H), 0.94 (d, J= 6.0 Hz, 3H). LCMS m/z: 462.6 [C24H32N06P +1]+. Anal for
C24H32N06P + 1 H20 + 0.1 EtOAc + 0.2 Et20; Calcd: C:60.16, H:7.37, N:2.78; Found: C:60.19, H:7.20, N:2.72.
Example E28
Methyl 4-( [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phi
hydroxy-benzoylamino]-methyl)-benzoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.80 (s, 1H), 9.35 (m, 1H), 7.92 (d, J= 8.4 Hz, 2H), 7.66 (s, 2H), 7.45-7.38 (m, 3H), 6.90 (d, J= 9.0 Hz, 1H), 6.82 (dd, J = 9.0, 2.7 Hz, 1H), 4.54 (d, J= 6.0 Hz, 2H), 3.82 (s, 3H), 3.12 (d, J= 17.1 Hz, 2H), 1.29 (d, J = 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.98 ppm. LCMS m/z: 628.6 [C24H22Br2N07P +l]+.
Anal for C24H22Br2N07P; Calcd: C:45.96, H:3.54, N:2.23; Found: C:45.94, H:3.44, N:2.17.
Example A28
4-([5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydroxy- benzoylamino] -methyl)-benzoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 11.82 (s, 1H), 9.33 (m, 1H), 7.89 (d, J= 8.4 Hz, 2H), 7.66 (s, 2H), 7.45-7.38 (m, 3H), 6.90 (d, J= 9.0 Hz, 1H), 6.79 (dd, J = 9.0, 3.0 Hz, lH), 4.56 (d, J= 6.0 Hz, 2H), 3.12 (d, J= 17.1 Hz, 2H), 1.29 (d, J= 13.8 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.92 ppm. LCMS m/z: 614.4
[C23H2oBr2N07P +1]+. Anal for C23H20Br2NO7P + 0.5 H20; Calcd: C:44.40, H:3.40, N:2.25; Found: C:44.06, H:3.36, N:2.25.
Example E29
Methyl 3-( [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hyd roxy-benzoylamino]-methyl)-benzoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.89 (s, 1H), 9.42 (m, 1H), 7.99 (s, 1H), 7.92 (d, J= 7.8 Hz, 1H), 7.74 (s, 2H), 7.66 (m, 1H), 7.55 (m, 1H), 7.49 (d, J= 3.3 Hz, 1H), 6.98 (d, J= 9.0 Hz, 1H), 6.87 (dd, J= 9.0, 3.3 Hz, 1H), 4.62 (d, J= 5.7 Hz, 2H), 3.91 (s, 3H), 3.20 (d, J= 17.1 Hz, 2H), 1.37 (d, J= 14.4 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 43.00 ppm.
LCMS m/z: 628.6 [C24H22Br2N07P +1]+.
Anal for C24H22Br2N07P; Calcd: C:45.96, H:3.54, N:2.23; Found: C:45.75, H:3.52, N:2.12.
Example A29
3-([5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hyd roxy-benzoylamino] -methyl)-benzoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 11.83 (s, 1H), 9.33 (m, 1H), 7.89 (s, 1H), 7.82 (d, J= 7.8 Hz, 1H), 7.65 (s, 2H), 7.55 (m, 1H), 7.46 (d, J= 7.8 Hz, 1H), 7.42 (d, J= 3.3 Hz, 1H), 6.89 (d, J= 8.7 Hz, 1H), 6.79 (dd, J= 8.7, 3.3 Hz, 1H), 4.53 (d, J = 5.7 Hz, 2H), 3.11 (d, J= 17.1 Hz, 2H), 1.28 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-de) δ: 42.93 ppm. LCMS m/z: 614.4 [C23H2oBr2N07P +1]+. Anal for
Example E30
Methyl 4-(2-[5-[2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-h ydroxy-benzoylamino]-ethyl)-benzoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.83 (s, 1H), 8.90 (m, 1H), 7.88 (d, J= 8.7 Hz, 2H), 7.66 (s, 2H), 7.38 (d, J= 8.7 Hz, 2H), 7.33 (d, J= 3.3 Hz, 2H), 6.86 (d, J = 9.0 Hz, 1H), 6.77 (dd, J= 9.0, 3.3 Hz, 1H), 3.82 (s, 3H), 3.60-3.50 (m, 2H), 3.12 (d, J = 17.1 Hz, 2H), 2.91 (m, 2H), 1.29 (d, J= 14.4 Hz, 3H). 31P NMR (121.46 MHz, DMSO- d6) δ: 42.92 ppm. LCMS m/z: 642.1 [C25H24Br2N07P +1]+. Anal for C25H24Br2N07P; Calcd: C:46.83, H:3.77, N:2.18; Found: C:47.00, H:3.82, N:2.08.
Example A30
4-(2-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-h ydroxy-benzoylamino] -ethyl)-benzoic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 11.84 (s, 1H), 8.90 (m, 1H), 7.85 (d, J= 8.1 Hz, 2H), 7.65 (s, 2H), 7.40-7.30 (m, 3H), 6.85 (d, J= 9.0 Hz, 1H), 6.76 (dd, J = 9.0, 3.3 Hz, 1H), 3.60-3.45 (m, 2H), 3.11 (d, J= 17.1 Hz, 2H), 2.95 (m, 2H), 1.28 (d, J = 13.8 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.97 ppm. LCMS m/z: 628.6
[C24H22Br2N07P +1]+. Anal for C24H22Br2N07P; Calcd: C:45.96, H:3.54, N:2.23; Found: C:46.20, H:3.75, N:2.03.
Example E31
Ethyl 1- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydr oxy-benzoyl]-piperidine-4-carboxylate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 9.63 (s, 1H), 7.72 (s, 2H), 6.89 (d, J = 9.3 Hz, 1H), 6.77 (dd, J= 9.3, 3.0 Hz, 1H), 6.49 (d, J= 3.0 Hz, 1H), ), 4.13 (q, J= 7.2 Hz, 2H), 3.50-3.25 (m, 2H), 3.19 (d, J= 17.1 Hz, 2H), 3.20-2.85 (m, 2H), 2.00-1.75 (m, 2H), 1.60-1.40 (m, 2H), 1.36 (d, J= 14.1 Hz, 3H), 1.24 (t, J= 7.1 Hz, 3H).
31P NMR (121.46 MHz, DMSO-d6) δ: 42.86 ppm. LCMS m/z: 620.4 ^sHjeB^NOvP +1]+. Anal for CzsHjeB^NC^P; Calcd: C:44.61, H:4.23, N:2.26; Found: C:44.96, H:3.93, N: 1.99.
Example A31
l-[5-[2,6-Dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy]-2-hydr oxy-benzoyl] -piperidine-4-carboxylic acid
The title compound was prepared according to the procedure described for the synthesis of example A22; 1H NMR (300 MHz, DMSO-d6) δ: 9.53 (s, 1H), 7.64 (s, 2H), 6.81 (d, J = 8.7 Hz, 1H), 6.69 (dd, J= 8.7, 3.0 Hz, 1H), 6.39 (d, J= 3.0 Hz, 1H), ), 3.50-3.25 (m, 2H), 3.10 (d, J= 17.4 Hz, 2H), 3.10-2.80 (m, 2H), 1.90-1.70 (m, 2H), 1.55-1.35 (m, 2H), 1.28 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSC /6) δ: 42.93 ppm.
LCMS m/z: 592.4 [CZ^B^NOTP +1]+. Anal for Cz^B^NC^P; Calcd: C:42.66, H:3.75, N:2.37; Found: C:42.39, H:3.84, N:2.16.
Example E32
Methyl 4- [5- [2,6-dibromo-4-(hydroxy-methyl-phosphinoylmethyl)-phenoxy] -2-hydr oxy-benzoylamino] -benzoate
The title compound was prepared according to the procedure described for the synthesis of example E9-2; 1H NMR (300 MHz, DMSO-d6) δ: 11.25 (s, 1H), 10.65 (s, 1H), 7.94 (d, J= 8.7 Hz, 2H), 7.83 (d, J= 8.7 Hz, 2H), 7.67 (s, 2H), 7.28 (d, J= 3.0 Hz, 1H), 6.98 (d, J = 9.0 Hz, 1H), 6.90 (dd, J= 9.0, 3.0 Hz, 1H), 4.54 (d, J= 6.0 Hz, 2H), 3.82 (s, 3H), 3.13 (d, J= 17.1 Hz, 2H), 1.30 (d, J= 14.1 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.95 ppm. LCMS m/z: 614.4 [CzsHjoB^NC^P +1]+. Anal for CzsHjoB^NC^P; Calcd: C:45.05, H:3.29, N:2.28; Found: C:44.97, H:3.16, N:2.21.
Example A33
3,5-dimethyl-4-(3-isopropyl-4-hydroxybenzyl)benzyl alcohol
Methyl 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)benzoate was prepared from 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)phenol (Chiellini et al., Bioorg. Med. Chem. Lett. 10:2607 (2000)) according to the procedure described for the synthesis of intermediate 4/step b-c.
A mixture of methyl 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)benzoate
(1.52 g, 4.26 mmol) in methanol (8.0 mL) and 4 N HCl-dioxane (3.2 mL, 12.8 mmol) was heated at 100 °C for 5 min in a microwave oven. The solvent was removed under reduced pressure and the residue was dissolved in THF (25 mL). The solution was cooled to 0 °C and to it was slowly added DIBAL (14.7 mL, 14.7 mmol). The reaction mixture was stirred at 0 °C for 2 h, quenched with saturated sodium potassium tartrate and diluted with hexanes (20 mL). The reaction mixture was stirred at room temperature for 2 h and the organic layer was separated. The organic solution was dried over MgS04, filtered and concentrated under reduced pressure to give the title compound (1.01, 83%) as white
solid: 1H NMR (300 MHz, CD3OD): δ 7.05 (s, 2H), 6.84 (d, J= 2.1 Hz, 1H), 6.58 (m, 2H), 4.55 (s, 2H), 3.96 (s, 2H), 3.22 (m, 1H), 2.25 (s, 6H), 1.14 (d, J= 7.0 Hz, 6H); TLC conditions: Uniplate silica gel, 250 microns; Mobile phase = ethyl acetate-hexanes (1 :3); Rf = 0.4.
Example E33-1
3,5-Dimethyl-4-(3-isopropyl-4-hydroxybenzyl)benzyl Phosphate Monoammonium Salt
Step a: To a mixture of 3,5-dimethyl-4-(3-isopropyl-4-hydroxybenzyl)benzyl alcohol (0.80 g, 2.82 mmol) and di-tert-butyl diethylphosphoramidate (0.98 g, 3.94 mmol) in DMF (20.0 mL) at rt was added 5-methylthiotetrazole (0.46 g, 3.84 mmol). The reaction mixture was stirred at rt for 40 min and cooled to 0 °C. To it was added t-butyl hydrogen peroxide (1.16 mL, 8.46 mmol). The reaction mixture was stirred at rt for 2 h, quenched with aqueous NH4C1 and extracted with ethyl acetate. The organic solution was dried over MgS04, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel, eluting with 30% ethyl acetate in hexanes in to afford di-tert-butyl 3,5-dimethyl-4-(4-hydroxy-3-isopropylbenzyl)benzyl phosphate (0.74 g, 55%) as a white solid: 1H NMR (300 MHz, CD3OD): δ 7.11 (s, 2H), 6.85 (s, 1H), 6.58 (m, 2H), 4.98 (d, J= 8.4 Hz, 2H), 3.99 (s, 2H), 3.22 (m, 1H), 2.27 (s, 6H), 1.51 (s, 18H), 1.15 (d, J = 7.0 Hz, 6H); TLC conditions: Uniplate silica gel, 250 microns; Mobile phase = ethyl acetate-hexanes (1 :3); Rf = 0.3.
Step b: A mixture of di-tert-butyl 3,5-dimethyl-4-(4-hydroxy-3-isopropylbenzyl)benzyl phosphate (0.44 g, 0.92 mmol) and 1 N HCl-dioxane (8.0 mL, 8.0 mmol) was stirred at room temperature for 3 h. The solvent was removed under reduced pressure and the residue was dissolved in ether (10 mL). The organic solution was washed with water (10 mL), dried in MgS04 and filtered. The solvent was removed under reduced pressure and the residue was dissolved in THF (6 mL). The organic solution was cooled to 0 °C and to it was added ammonium hydroxide (28%, 0.64 mL, 4.6 mmol). The solvent was removed under reduced pressure and the residue was treated with hexanes (10 mL). The solvent
was decanted and the residue was dissolved in acetone (3 mL). The solution was filtered through a mini-filter and the solvent was removed under reduced pressure to give the title compound (0.27 g, 77%) as a white solid: 1H NMR (300 MHz, CD3OD): δ 7.12 (s, 2H), 6.85 (d, J= 2.0 Hz, 1H), 6.58 (m, 2H), 4.87 (d, J= 8.4 Hz, 2H), 3.95 (s, 2H), 3.22 (m, 1H), 2.23 (s, 6H), 1.15 (d, J= 7.0 Hz, 6H); LC-MS m/z = 363 [Ci9H2805P - H]+.
Example E33-2
Cis-(S)-4-[4-[4-(3-chlorophenyl)-2-oxo-2-lambda*5*-[l,3,2]dioxaphosphinan-2- yloxymethyl] -2,6-dimethyl-benzyl] -2-isopropyl-phenol
A solution of t-BuMgCl (2 M in THF, 0.47 mL, 0.93 mmol) was added to a solution of example A33 (218 mg, 066 mmol) in THF at 0 °C. After stirring at 0 °C for 15 minutes, tra/?5-4-(3-chlorophenyl)-2-(4-nitrophenoxy)-2-oxido-l ,3,2-dioxaphosphorinane (294 mg, 0.80 mmol, Boyer et al., J. Med. Chem. 49:7711 (2006)). The ice bath was removed and the reaction mixture was stirred at rt for 16 h. A saturated solution of NH4C1 was added and the mixture was partitioned between water and ether. The layers were separated and the organics were dried (Na2S04), filtered, concentrated under reduced pressure and purified by preparative TLC (35%> acetone in hexanes) to give the title compound (70 mg, 18%); 1H NMR (300 MHz, DMSO-d6) δ: 9.02 (s, 1H), 7.49 (s, 1H), 7.48-7.35 (m, 3H),
7.08 (s, 2H), 6.87 (d, J= 2.1 Hz, 2H), 6.62 (d, J= 8.1 Hz, 2H), 6.45 (dd, J= 8.1, 2.1 Hz, 1H), 5.73 (m,lH), 5.07 (dd, J= 8.7, 2.1 Hz, 2H), 4.60-4.40 (m, 2H), 3.89 (s, 2H), 3.11 (hept, J= 6.6 Hz, 1H), 2.30-2.10 (m, 2H), 2,21 (s, 6H), 1.10 (d, J= 6.6 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: 42.95 ppm. LCMS m/z: 515.2 [C28H32C105P +1]+. Anal for C28H32C105P; Calcd: C:65.30, H:6.26; Found: C:65.41, H:6.49.
Example A34
3,5-dimethyl-4-(3-isopropyl-4-hydroxybenzyl)phenol
To a solution of 3,5-dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)phenol (0.30 g, 0.95 mmol) in methanol (6.0 mL) was added 2 N HC1 (1.4 mL, 2.8 mmol). The reaction mixture was stirred at room temperature for 72 h, diluted with water (15 mL) and extracted with ethyl acetate (10 mL). The organic solution was dried over MgS04, filtered and concentrated under reduced pressure to afford 3,5-dimethyl-4-(3-isopropyl-4- hydroxybenzyl)phenol (0.23 g, 89%) as a colorless oil: 1H NMR (300 MHz, CD3OD): δ 6.84 (d, J= 2.1 Hz, 1H), 6.58 (m, 2H), 6.53 (s, 2H), 3.87 (s, 2H), 3.23 (m, 1H), 2.17 (s, 6H), 1.15 (d, J = 7.0 Hz, 6H); TLC conditions: Uniplate silica gel, 250 microns; Mobile phase = acetone-hexanes (1 :3); Rf = 0.5.
Example E34-1
3,5-Dimethyl-4-(4-hydroxy-3-isopropylbenzyl)phenyl Phosphate Monoammonium Salt
The title compound was prepared from 3,5-dimethyl-4-(3-isopropyl-4- hydroxybenzyl)phenol (Example A34) according to the procedure described for the synthesis of Example E33-1. 1H NMR (300 MHz, CD3OD): δ 6.98 (s, 2H), 6.88 (d, J = 2.1 Hz, 1H), 6.54 (m, 2H), 3.92 (s, 2H), 3.23 (m, 1H), 2.22 (s, 6H), 1.17 (d, J= 7.0 Hz, 6H); LC-MS m/z = 351 [Ci8H2505P + H]+. Anal. Calcd for (Ci8H2505P + 0.5 H20): C, 57.44; H, 7.23; N, 3.72. Found: C, 57.28; H, 7.58; N, 3.78.
Example E34-2
Di(pivaloyloxymethyl) [3,5-Dimethyl-4-(4-hydroxy-3-isopropylbenzyl)]- phenoxylphosphate
Diisopropylethylamine (0.6 mL, 3.42 mmol) followed by iodomethyl pivalate (0.84 g, 3.42 mmol) were added to a solution of phosphate E34-1 (300 mg, 0.86 mmol) in acetonitrile at 0 °C. After stirring at rt for 24 h, the volatiles were removed under reduced pressure and the residue was purified by column chromatography (30% etAc in hexanes) to give the title compound as an oil (100 mg, 20%); 1H NMR (500 MHz, CDC13) δ: 7.26 (s, 1H), 6.91 (s, 2H), 6.59 (d, J= 8.5 Hz, 2H), 6.50 (d, J= 8.5 Hz, 2H), 5.77-6.90 (m,4H), 4.80 (s, 1H), 3.90 (s, 2H), 3.15 (hept, J= 7.0 Hz, 1H), 2,22 (s, 6H), 1.23-1.19 (m, 24H). LCMS m/z: 579.9 [C30H43O9P +1]+. Anal for C30H43O9P + 0.2 Et20; Calcd: C:62.34, H:7.64; Found: C:62.30, H:7.72.
Example E34-3
Cis-(S)-2-[4-(3-chlorophenyl)-2-oxo-2-lambda*5*[l,3,2]dioxaphosphinan-2-yloxy- 2,6-dimethylbenzyl]-6-isopropylphenol
The title compound was prepared according to the procedure described for the synthesis of example E33-2; 1H NMR (300 MHz, DMSO-d6) δ: 8.99 (s, 1H), 7.45-7.30 (m, 4H), 6.88 (s, 2H), 6.81 (d, J= 2.1 Hz, 2H), 6.60 (d, J= 8.1 Hz, 2H), 6.43 (dd, J= 8.1, 2.1 Hz, 1H), 5.81 (m,lH), 4.70-4.40 (m, 2H), 3.84 (s, 2H), 3.07 (hept, J = 6.9 Hz, 1H), 2.30-2.10 (m, 2H), 2,18 (s, 6H), 1.05 (d, J= 6.9 Hz, 3H). 31P NMR (121.46 MHz, DMSO-d6) δ: - 11.48 ppm. LCMS m/z: 501.9 [C27H3oC105P +1]+. Anal for C27H30ClO5P + 0.1 H20; Calcd: C:64.50, H:6.05; Found: C:64.48, H:6.28.
Example A35
4-(4-Aminomethyl-2,6-dimethyl-benzyl)-2-isopropyl-phenol Hydrochloric acid salt
3,5-Dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)benzyl alcohol was prepared according to the procedure described for the synthesis of example A33 omitting the acid treatment to remove the MOM protecting group.
Step a: 3,5-Dimethyl-4-(3-isopropyl-4-methoxymethoxybenzyl)benzyl bromide was prepared according to the procedure described for the synthesis of intermediate 4/step e. Trityl amine (530 mg, 2.04 mmol) was added to a solution of benzyl bromide (400 mg, 1.02 mmol) in acetonitrile (10 mL) at rt. After stirring at rt for 12 h, the solvent was removed under reduced pressure and the residue purified by column chromatography (4% EtOAc in hexanes) to give the protected benzyl amine (320 mg, 55%).
Step b: Hydrochloric acid (4 N, 0.56 mL, 2.25 mmol) was added to a solution of trityl amine (320 mg, 0.56 mmol) in methanol at 0 °C. After stirring at rt for 72 h, the volatiles were removed under reduced pressure. The residue was azeotroped with methanol then taken up in EtOAc, sonicated, and the precipitate was collected by filtration, rinsed with EtOAc and dried to give the title compound as a white solid (120 mg, 67%); 1H NMR (300 MHz, CD3OD): δ 7.11 (s, 2H), 6.80 (d, J= 2.1 Hz, 1H), 6.60-6.50 (m, 2H), 3.99 (s, 2H), 3.97 (s, 2H), 3.19 (hept, J= 6.9 Hz, 1H), 2.27 (s, 6H), 1.11 (d, J= 6.9 Hz, 6H) LCMS m/z: 284.3 [Ci9H25NO +1]+. Anal for Ci9H25NO + 1.2 HC1 + 0.4 EtOAc; Calcd: C:68.27, H:8.18, N:3.86; Found: C:68.31, H:7.92, N:3.69.
Example E35
Cis-(S)-4-[4-(3-chlorophenyl)-2-oxo-2-lambda*5*[l,3,2]dioxaphosphinan-2- ylamino]methyl-2,6-dimethylbenzyl]-2-isopropyl-l-phenol
Diisopropylethylamine (0.04 mL, 0.24 mmol) was added to a solution of benzylamine hydrochloride (example A35, 40 mg, 0.12 mmol) in THF (2 mL) at 0 °C. After stirring at 0 °C for 20 minutes, tra/?5-4-(3-chlorophenyl)-2-(4-nitrophenoxy)-2-oxido-l,3,2- dioxaphosphorinane (40 mg, 0.15 mmol, Boyer et ah, J. Med. Chem. 49:7711 (2006)) was added. After stirring at rt for 2 h, the reaction mixture was quenched with water and extracted with EtOAc. The organics were dried (MgS04), filtered, concentrated under reduced pressure and the residue was purified by column chromatography (80 % acetone in hexanes) to give the title compound as a white solid; 1H NMR (500 MHz, CD30D) δ: 7.33 (s, 1H), 7.30 (d, J = 7.8, 1.5 Hz, 2H), 7.26 (t, J = 7.8 Hz, 1H), 7.19 (d, J = 7.8 Hz,
1H), 7.06 (s, 2H), 6.83 (d, J = 1.5 Hz, 2H), 6.56 (d, J = 8.0 Hz, 2H), 6.50 (dd, J = 8.0, 1.5 Hz, 1H), 5.58 (dd, J = 10.5, 2.5 Hz, 1H), 4.64 (t, J = 10.5 Hz, 1H), 4.40 (m, 1H), 4.12 (d, J = 14.0Hz, 1H), 3.93 (s, 2H), 3.18 (hept, J = 7.0 Hz, 1H), 2,21 (s, 6H), 2.15-2.00 (m, 2H), 1.12 (d, J = 7.0 Hz, 3H). LCMS m/z: 514.6 [C28H33CINO4P +1]+. Anal for C28H33CINO4P; Calcd: C:65.43, H:6.47, N:2.73; Found: C:65.40, H:6.41, N:2.56.
Example A36
[4-(3'-benzyl-4'-hydroxy-benzyl)-3,5-dimethylphenoxy]-methylphosphonic acid
The compound above was prepared as described in example 38 in US 7514419
Example E36
di(pivaloyloxymethyl) [-4-(3'-Benzyl-4'-hydroxy-benzyl)-3,5-dimethyl
phenoxy] methylphosphonate
Diethylisopropylamine (19.5 mL, 118.2 mmol) was added to a solution of crude phosphonic acid (example 38 in US 7514419, 59.1 mmol) in acetonitrile at rt (300 mL). The orange turbid mixture was heated at 40 °C and iodomethyl pivalate (28.6 g, 118.2 mmol) was added. The reaction was monitored by HPLC (note 1). After 3.5 h, iodomethyl pivalate (14.3 g, 59.1 mmol) and diethylisopropylamine (9.75 mL, 59.1 mmol) were added. The reaction mixture was stirred at 40 °C overnight. Iodomethyl pivalate (14.3 g, 59.1 mmol) and diethylisopropylamine (9.75 mL, 59.1 mmol) were added. After stirring for an additional 2 hours (Note 2) the cooled reaction mixture was poured into a mixture of ethyl acetate (150 mL) and water (300 mL). The layers were separated and the organics were washed sequentially with water (150 mL), a saturated solution of sodium bicarbonate (100 mL), and brine (100 mL) then dried (MgS04), filtered and concentrated to dryness to a brown oil. The residue was taken up in 1/1
acetone /hexanes (400 mL) and filtered through a pad of silica (note 3). The pad was rinsed with 1/1 acetone/hexanes (100 mL). The orange filtrate was concentrated to dryness and the orange oil was taken up in acetone (60 mL). Hexanes (300 mL) was added and the cloudy solution was stirred at rt. After a few minutes a solid appears and the mixture turns slowly into a thick slurry. After stirring at rt overnight, the tan solid was collected by filtration, rinsed with 1/6 acetone/hexanes (100 mL), air dried then dried under high vacuum to give an off white solid 24.06 g (68.7% yield).
1H NMR (500 MHz, DMSO-d6) δ: 9.12 (s, 1H), 7.25-7.20 (m, 2H), 7.14-7.11 (m, 3H), 6,68 (dd, J= 8.5, 2.0 Hz, 1H), 6.72 (s, 2H), 6.64 (d, J= 8.0 Hz, 1H), 6.52 (dd, J= 8.5, 2.5 Hz, 1H), 5.70-5.65 (m, 4H), 4.43 (d, J= 10 Hz, 2H), 3.77-3.74 (m, 4H), 2.11 (s, 6H), 1.12 (s, 18 H).
31P NMR (121.46 MHz, DMSO-d6) δ: 21.42 ppm.
LCMS m/z: 641.4 [Css^NOgP +1]+. Anal for Css^NOgP; Calcd: C:65.61, H:7.08; Found: C:65.55, H:6.70, N:7.16.
Example A37
[3,5-dimethyl-4-(4'-hydroxy-3'-iso-propylbenzyl)phenoxy]-methylphosphonic acid
The compound above was prepared as described in example 7 in US 7514419.
Example E37
Cis (S)-2-[(3,5-dimethyl-4-(4'-hydroxy-3'-iso-propylbenzyl)- phenoxy)methyl]-4-(3-chlorophenyl)-2-oxo-2λ5-[l,3,2]-dioxaphosphonane
Example A38
2- [3,5-dimethyl-4- [3 '-benzyl-4 '-hydroxy-benzyl] henyl] -ethylphosphonic acid
The compound above was prepared as described in example 42 in US 7514419.
Example A39
[3,5-dimethyl-4-[3'-(4-fluoro-benzyl)-4'-hydroxy-benzyl]- phenoxy] methylphosphonic acid
The compound above was prepared as described in example 40 in US 7514419.
Example 40
[3,5-dichloro-4-(4'-hydroxy-3'-/so-propylbenzyl)phenoxy] methylphosphonic acid The compound above was prepared as described in example 7-5 in US 7514419.
Example 41
[3,5-dibromo-4-(3'-iS0-propyl-4'-hydroxyphenoxy)-phenoxy] methylphosphonic acid
The compound above was prepared as described in example 8-1 in US 7514419.
Example 42
[4-(4'-hydroxy-3'-iso-propylbenzyl)-2,3,5-trimethylphenoxy]methylphosphonic Acid
The compound above was prepared as described in example 61-1 in US 7514419.
B Biological assays:
1. Receptor Binding
[00271] The purpose of these studies is to determine the affinity of T3 and various thyromimetics for human thyroid hormone receptors TRa and TRP and to assess oral bioavailability.
[00272] Methods: Baculoviruses expressing TRal, TRpi and RXRa are generated using cDNA and other reagents from Invitrogen (Carlsbad, CA). To produce TR/RXR heterodimer proteins, the sf9 insect cells are first grown to a density of 1 5x105 cells/ mL. TRal or TRpi and RXRa baculovirus stocks are added to the cell culture with a ratio of 1 to 1 (multiplicity of infection =10). The cells are harvested three days after the infection. The cells are lysed in assay buffer (50 mM NaCl, 10% Glycerol, 20 mM tris, pH 7.6 2 mM EDTA, 5 mM β mercaptoethanol and 1.25% CHAPS) and the lysates are assayed for
125
T3 binding as follows: I-T3 is incubated with the lysates of TR and RXR recombinant
125 baculoviruses coinfected cells (50 μΐ) in assay buffer for one h and then the I-T3
125
TR/RXR complex is separated from free I-T3 by a mini gel filtration (Sephadex G50) column. The bound 125I-T3 is counted with a scintillation counter.
[00273] Binding of compounds to either the TRal or TRpi are also performed by means of scintillation proximity assays (SPA). The SPA assay, a common method used for the quantitation of receptor-ligand equilibria, makes use of special beads coated with a scintillant and a capture molecule, copper, which binds to the histidine-tagged a or β receptor. When labeled T3 is mixed with receptor and the SPA beads, radioactive counts are observed only when the complex of protein and radiolabeled ligand is captured on the surface of the bead. Displacement curves are also generated with labeled T3 and increasing concentrations of unlabeled thyromimetics of interest.
2. Stability in rat whole blood
[00274] Objective: To assess the metabolic stability of various compounds of the invention in whole blood isolated from the rat.
[00275] Methods: Fresh blood was collected from an anesthetized Sprague Dawley rat (male; -250 g; Harlan Laboratories, Livermore, CA) by cardiac puncture and transferred into tubes containing the anticoagulant lithium EDTA. Test compounds were dissolved in DMSO at a concentration of 10 mM and diluted into whole blood in Eppendorf tubes to yield a final concentration of 10 μΜ. The mixtures were incubated at 37 °C in a Thermomixer (Eppendorf) and 50 μΐ aliquots were removed at appropriate time intervals and quenched by addition to 100 μΐ methanol or 150 μΐ acetonitrile. The quenched samples were clarified by centrifugation (Eppendorf Microfuge, 4000 rpm, room temperature). Aliquots of the supematants were filtered through a 0.45-micron filter and transferred to vials for quantification of test compound concentration by reverse phase HPLC.
[00276] HPLC analysis was conducted on an Agilent 1100 system with use of a Phenomenex C8 column (4.6 x 150 mm, 5 μΜ) at 40°C. The column was eluted at a flow rate of 1.5 mL/min with a linear gradient from 20 mM potassium phosphate buffer pH 6.2 to 80% acetonitrile over 15 minutes. The column effluent was monitored at 265 nm. Concentrations of test compounds in the samples were determined by comparison with
authentic standards prepared in the appropriate matrix. The temporal profile of whole blood test compound concentrations was plotted and half-life calculated using the equation: ty2 = ln2/k, where k is the pseudo first-order rate constant of the disappearance of test compound as a function of incubation time.
[00277] Results: Half lives for a selection of test compounds evaluated are shown in the table below:
[00278] Significance: Several of the compounds tested showed a high degree of metabolic instability with half- lives in whole rat blood of <30 minutes. Metabolic instability reduces the exposure time to active test compound in vivo and may confer a favorable therapeutic index.
3. Pharmacokinetics after intravenous administration to the rat or dog
[00279] Objective: To determine the pharmacokinetics of select compounds of the invention following intravenous administration to the rat and dog.
[00280] Methods:
[00281] (a) Rats. Male Sprague Dawley rats (380-450 g; Harlan Laboratories, Livermore, CA) were fasted overnight prior to test compound administration. Food was returned 3 hours after compound administration. Test compounds were dissolved in saline and administered via the penile vein while the animals (n = 4/test compound) were under halothane anesthesia. The dosing volume was 1 mL/kg body weight. Blood was collected from a distal tail vein into lithium heparin containing tubes at 0.083, 0.25, 0.5, 1, 2, 4, and 8 hours after compound administration. Plasma was prepared from the blood samples by centrifugation (14,000 rpm, Eppendorf Microfuge) and stored at -70 °C until analysis by LC-MS/MS.
[00282] (b) Dogs. Two male and two female Beagle dogs (10-14 kg; Marshall Farms; North Rose, NY) were fasted overnight prior to test compound administration. Food was returned 3 hours after compound administration. Test compounds were dissolved in phosphate-buffered saline and administered intravenously. The dosing volume was 0.5
mL/kg body weight. Blood was collected via cephalic venipuncture into K3EDTA vacutainers at 0.0833, 0.25, 1, 2, 4, 6, 8, 12, and 24 hours after compound administration. Plasma was prepared from the blood samples by centrifugation (4,000 rpm, Eppendorf Microfuge) and stored at -70 °C until analysis by LC-MS/MS.
[00283] (c) LC-MS/MS analysis. On the day of analysis, the plasma samples were thawed at 4 °C for 4.5 hr. Plasma proteins from plasma samples (50 μί) were precipitated by addition of 100 of acetonitrile containing 200 ng/mL of a proprietary internal standard. After 20 min of centrifugation (Eppendorf microfuge, 4,000 rpm, 4 °C) the resulting supernatant was collected and analyzed by LC-MS/MS. A 10 aliquot of the supernatant was injected onto a Gemini C18 column (5 μιη, 2 x 50 mm, Phenomenex) fitted with a Gemini C18 guard column (5 μιη, 4.0 x 2.0 mm, Phenomenex, Torrance, CA) and eluted with a gradient consisting of mobile phase A (5 mM ammonium acetate in 5% acetonitrile) and B (1% ammonium hydroxide in 100% acetonitrile) at a flow rate of 0.5 mL/min (0 min, 0% B; 0-0.1 min, 0-50 % B; 0.1-1.7 min, 50-60 % B; 1.7-1.8 min, 60-90 % B; 1.8-3.5 min, 90 % B; 3.5-3.6 min, 90-0 % B; 3.6-5.5 min, 0 % B). The injector temperature was set at 4 °C. Test compounds were detected using the negative MS/MS mode and quantified by comparison of peak areas to standard curves obtained by spiking known concentrations of the analytes to heparinized blank rat plasma. Calibration curves ranged from 5 to 6000 ng/mL. The limit of quantification (LOQ) was -10 ng/mL.
[00284] (d) Pharmacokinetic analysis. The temporal profile of plasma drug concentrations was analyzed by non-compartmental methods using WinNonLin version 1.1 (Scientific Consulting, Inc., Cary, NC). Extrapolation of the plasma concentration- time plot was performed to estimate the zero time intercept by fitting a natural log-linear line to the first two data points. Area under the curve (AUC) values were determined by trapezoidal summation of the plasma concentration-time profile to the last measurable time point. The t½ values were calculated by the first order rate constant associated with the terminal log-linear portion of the curve as defined by at least three data points of the elimination phase.
[00285] Results: The pharmacokinetic parameters determined from the temporal profile of plasma concentrations of select test compounds in the rat and dog are shown in the table below:
[00286] Significance: The compounds evaluated showed relatively low exposure, a short half-life, and a moderate to high clearance rate in the rat. For Compound A34-1, this same general profile was also observed in a higher species, the dog. Compounds with this profile are expected to show an improved therapeutic index relative to compounds with high exposure, low clearance and a long circulating half-life.
4. Pharmacokinetics of A36 and E36 following single intravenous and oral doses in rats
[00287] Objective: The purpose of this study was to evaluate the oral bioavailability (OBAV) and other pharmacokinetic parameters of A36 in male CD IGS rats following oral and intravenous administrations of E36 and A36, respectively.
[00288] Methods: For the IV formulation, A36 was prepared as a solution in PBS at drug concentrations of 1, 3 and 10 mg/mL and dosing volume at 1 mL/kg (1, 3 and 10 mg/kg). Oral (PO) solutions of E36 were prepared by dissolving the prodrug in PEG-400 at drug concentrations of 0.5, 1.5, 5, 15 and 50 mg/mL and dosing volume of 2 mL/kg (1,
3, 10, 30 and 100 mg/kg). Oral solutions of A36 were prepared by dissolving of the compound in PEG-400 at drug concentrations of 3 and 30 mg/mL and dosing volume of 1 mL/kg (3 and 30 mg/kg).
[00289] Male CD IGS rats were purchased from Charles Rivers Laboratories (San Diego, CA) at approximately 200-450 g. Animals were housed 2 per cage under a 12- hour lighting cycle (7 am-7 pm light) and controlled temperature (~22°C). Rats were fed Teklad 7001 rat chow (Harlan Teklad, Madison, WI) and allowed water ad libitum unless otherwise specified in the protocol.
[00290] Three groups (n=4 per group) of male CD IGS rats (200-250 g) were dosed intravenously (IV) via the lateral tail vein with 1, 3 and 10 mg/kg of A36 as a solution in phosphate buffer saline. The animals were lightly anesthetized with isoflurane during the IV dosing procedure. In a separate study, four groups (n=4 per group) of CD IGS rats (400-450 g) were administered an oral gavage of 1, 3, 10, 30 and 100 mg/kg of E36 as a solution in PEG-400. Two additional groups (n=4 per group) of rats were given an oral gavage of 3 and 30 mg/kg of A36 as a PEG-400 solution. Animals had free access to food prior to and during the IV and oral evaluation. Blood (-200 μί) samples (lithium heparin) were taken from the lateral tail vein or retro sinus orbital at 5 min and 0.5, 1, 2,
4, 8, 12 and 24 hr following IV administration of A36 or at 0.5, 1, 2, 3, 4, 8, 12 and 24 hr
post PO administration of E36 and A36. The blood collection tubes were preloaded with the esterase inhibitor paraoxon (10 mM final concentration). Blood samples were centrifuged (Eppendorf Micro fuge, 14,000 rpm, room temperature, 1 min) to obtain plasma. The plasma samples were stored at -80°C prior to LC -MS/MS analysis.
[00291] Sample Preparation and LC-MS/MS Analysis
[00292] On the day of analysis, the plasma samples were thawed at 4°C for 4.5 hr. Plasma proteins from plasma samples (50 μί) were precipitated by addition of 100 of acetonitrile containing 200 ng/mL of a proprietary internal standard. After 20 min of centrifugation (Eppendorf microfuge, 4,000 rpm, 4°C) the resulting supernatant was collected and analyzed by LC-MS/MS.
[00293] A 10 aliquot of the supernatant was injected onto a Gemini CI 8 column (5 μιη, 2 x 50 mm, Phenomenex) fitted with a Gemini CI 8 guard column (5 μιη, 4.0 x 2.0 mm, Phenomenex, Torrance, CA) and eluted with a gradient consisting of mobile phase A (5 mM ammonium acetate in 5% acetonitrile) and B (1% ammonium hydroxide in 100% acetonitrile) at a flow rate of 0.5 mL/min (0 min, 0% B; 0-0.1 min, 0-50 % B; 0.1-1.7 min, 50-60 % B; 1.7-1.8 min, 60-90 % B; 1.8-3.5 min, 90 % B; 3.5-3.6 min, 90-0 % B; 3.6-5.5 min, 0 % B). The injector temperature was set at 4°C. A36, the monoprodrug, and E36 were detected using the negative MS/MS mode (411.1/63.1, A36) and quantified by comparison of peak areas to standard curves obtained by spiking known concentrations of the analytes to heparinized blank rat plasma. Calibration curves ranged from 5 to 6000 ng/mL of A36 with a tentative limit of quantification (LOQ) of 5 ng/mL.
[00294] WinNonLin Analysis
[00295] The temporal profile of plasma drug concentrations was analyzed by non- compartmental methods [1, 2] using WinNonLin version 5.2 (Pharsight Corp, Mountain View, CA) [3]. Area under the curve (AUC) values were determined by trapezoidal summation of the plasma concentration-time profile to the last measurable time point. The t½ values were calculated by the first order rate constant associated with the terminal log-linear portion of the curve as defined by at least three data points of the elimination phase. For IV bolus analysis, extrapolation of the plasma concentration-time plot was performed to estimate the zero time intercept by fitting a natural log-linear line to the first two data points. The mean absorption time (MAT) value is the difference between mean residence time MRTPo and MRTiy. The oral bioavailability (OBAV) value of A36 was calculated by comparison of the dose normalized AUC values of the plasma profile of
A36 following PO administration of E36 or A36 with the group averaged AUC value of A36 following IV administration of A36.
[00296] Statistical Data Analysis
[00297] Results in tables and graphs are expressed as means ± standard deviations (s.d.) or means ± standard error of the mean (s.e.m.).
[00298] Results: Key pharmacokinetic parameters (m ± s.d., n=4-5) of A36 following IV administration of 1 mg/kg of A36 (MW 412.2) and after oral administration of 1 mg/kg of the prodrug E36 (MW 649.7) to freely feeding CD IGS rats are summarized in the table below. The oral bioavailability of A36 when dosed orally itself was 1-3% and was 1-4% following administration of the prodrug. The plasma AUC and Cmax values of A36 were dose proportional over the oral (1-100 mg/kg) and IV (1-10 mg/kg) dose ranges evaluated in CD IGS rats.
[00299] Conclusions: In male CD IGS rats, the plasma clearance of A36 was low (0.22 L/hr/kg versus 3.13 L/hr/kg hepatic blood flow [4]) and the volume of distribution was below total body water volume (0.37 L/kg versus 0.6 L/kg volume of total body water [4]). The plasma elimination half-life of A36 following an IV bolus administration of A36 was 1.67 hr. The plasma AUC of A36 was dose proportional over the IV dosing range.
[00300] Following oral administration of 1, 3, 10, 30 and 100 mg/kg of E36, the plasma concentrations of A36 reached plasma Cmax values of 0.06, 0.09, 0.33, 1.19 and 3.19 μg/mL, respectively. The Tmax of A36 after oral dosing of E36 was 2-4 hr, and the MAT of A36 was approximately 4 hr over the dosing range. The plasma elimination
half-life of A36 following PO administration of the prodrug was 3.68 hr. The plasma AUC and Cmax values of A36 were dose proportional over the oral dosing range of E36. E36 as a solution in PEG-400 was poorly orally bioavailable (1-4%) in CD IGS rats: the prodrug did not improve the oral bioavailability of A36.
5. Pharmacokinetics of A36 and E36 following single intravenous and oral doses in beagle dogs
[00301] Objective: The purpose of this study was to evaluate the oral bioavailability (OBAV) and other pharmacokinetic parameters of A36 in beagle dogs following oral and intravenous administrations of E36 and A36, respectively.
[00302] Methods: For the IV formulation, A36 was prepared as a solution in PBS at a drug concentration of 2 mg/mL and dosing volume at 0.5 mL/kg (1 mg/kg). Oral (PO) solutions of E36 were prepared by dissolving the prodrug in PEG-400 at a drug concentration of 1.5 mg/mL and dosing volume of 2 mL/kg (3 mg/kg). An oral formulation of A36 was prepared by dissolving the compound in PEG-400 at a drug concentration of 3 mg/mL and dosing volume of 1 mL/kg (3 mg/kg).
[00303] Male and female beagle dogs were purchased from Marshall Farms (North Rose, NY) and Harlan (Indianapolis, IN) at approximately 9-15 kg. Animals were individually housed under a 12-hour lighting cycle (7 am-7 pm light) and controlled temperature (-22° C). The dogs were fed twice daily with Teklad 8563 chow (Harlan Teklad, Madison, WI) and allowed water ad libitum unless otherwise specified in the protocol.
[00304] A group of overnight (12 hr) fasted beagle dogs (2 male, 2 female) were administered an IV bolus of 1 mg/kg of E36 via the cephalic vein. Blood (EDTA) (3 mL) was sampled by a contralateral cephalic or a saphenous venipuncture at 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 hr post dose. Separate groups of overnight fasted animals were administered an oral gavage of 3 mg/kg of either A36 or E36. Blood (EDTA) (3 mL) was collected from the contralateral cephalic or saphenous vein at 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 hr post dose. In both IV and oral legs of the study, food was returned after 4 hr post dose. Blood samples were centrifuged (Eppendorf Micro fuge, 14,000 rpm, room temperature, 1 min) to obtain plasma. The plasma samples were stored in a -80°C freezer prior to LC-MS/MS analysis.
[00305] Sample Preparation and LC-MS/MS Analysis
[00306] On the day of analysis, the plasma samples were thawed at 4°C for 4.5 hr. Plasma proteins from plasma samples (50 μί) were precipitated by addition of 100 μΐ, of acetonitrile containing 200 ng/mL of a proprietary internal standard. After 20 min of centrifugation (Eppendorf microfuge, 4,000 rpm, 4°C) the resulting supernatant was collected and analyzed by LC-MS/MS under the conditions described below.
[00307] A 10 aliquot of the supernatant was injected onto a Gemini CI 8 column (5 μιη, 2 x 50 mm, Phenomenex) fitted with a Gemini CI 8 guard column (5 μιη, 4.0 x 2.0 mm, Phenomenex, Torrance, CA) and eluted with a gradient consisting of mobile phase A (5 mM ammonium acetate in 5% acetonitrile) and B (1% ammonium hydroxide in 100% acetonitrile) at a flow rate of 0.5 mL/min (0 min, 0% B; 0-0.1 min, 0-50 % B; 0.1-1.7 min, 50-60 % B; 1.7-1.8 min, 60-90 % B; 1.8-3.5 min, 90 % B; 3.5-3.6 min, 90-0 % B; 3.6-5.5 min, 0 % B). The injector temperature was set at 4°C. A36 was detected using the negative MS/MS mode (411.1/63.1) and quantified by comparison of peak areas to standard curves obtained by spiking known concentrations of the analytes to EDTA- treated blank dog plasma. Calibration curves ranged from 5 to 6000 ng/mL of A36 with a limit of quantification (LOQ) of approximately 5 ng/mL.
[00308] WinNonLin Analysis
[00309] The temporal profile of plasma drug concentrations was analyzed by non- compartmental methods [1, 2] using WinNonLin version 5.2 (Pharsight Corp, Mountain View, CA) [3]. Area under the curve (AUC) values were determined by trapezoidal summation of the plasma concentration-time profile to the last measurable time point. The plasma t½ values were calculated by the first order rate constant associated with the terminal log-linear portion of the curve as defined by at least three data points of the elimination phase. For IV bolus analysis, extrapolation of the plasma concentration-time plot was performed to estimate the zero time intercept by fitting a natural log-linear line to the first two data points. The mean absorption time (MAT) value is the difference between mean residence time MRTPo and MRTiy. The oral bioavailability (OBAV) value of A36 was calculated by comparison of the dose normalized AUC values of the plasma profile of A36 following PO administration with the group averaged AUC value of A36 following IV administration of A36.
[00310] Statistical Data Analysis
[00311] Results in tables and graphs are expressed as means ± standard deviations (s.d.) for n=4 and means ± ½ range for n=2.
[00312] Results'. Key pharmacokinetic parameters (mean ± s.d., n=4) of A36 following IV administration of 1 mg/kg of A36 and after oral administration of 3 mg/kg of the prodrug E36 to overnight fasted beagle dogs are summarized in the table below. The absolute oral bioavailability of A36 was 6% and 19% when administered as A36 and E36, respectively. No obvious sex differences in the pharmacokinetic parameters were observed in this small sample size comparison.
[00313] Conclusions: The plasma clearance of A36 in the beagle dog was low relative to its hepatic blood flow (0.14 L/hr/kg versus 2.62 L/hr/kg hepatic blood flow), and the volume of distribution was below total body water volume (0.20 L/kg versus 0.6 L/kg volume of total body water). The plasma half-life of A36 was similar following IV administration of A36 and PO administration of the prodrug E36 (1.93 hr IV A36 versus 1.86 hr PO E36). Following oral administration of 3 mg/kg of E36, the plasma concentrations of A36 reached a Cmax of 1.14 μg/mL. The Tmax of A36 after oral dosing of E36 was 1.50 hr. The MAT of A36 was approximately 1.79 hr. The bioavailability of A36 when dosed orally itself was 6%> but improved to 19%> when administered as the prodrug E36. The pharmacokinetic parameters in male (n=2) and female (n=2) dogs were reported together as n=4 as no obvious sex differences in the disposition of A36 were observed in this small sample size evaluation.
6. Pharmacokinetics of A36 and E36 following single intravenous and oral doses in cynomolgus monkeys
[00314] Objective: A36 is a human thyroid hormone receptor agonist and the active metabolite of E36, a prodrug currently under evaluation for the treatment of hyperlipidemia. The purpose of this study was to evaluate the oral bioavailability (OBAV) and other pharmacokinetic parameters of A36 in male and female cynomolgus monkeys following oral and intravenous administrations of E36 and A36, respectively.
[00315] Methods: The intravenous dose formulation of A36 was prepared by dissolving an appropriate amount of A36 in a solution of 10% Captisol in Dulbecco's phosphate-buffered saline (DPBS, IX, pH 7.4) to obtain a drug concentration of 1 mg/mL at a dosing volume of 1 mL/kg (1 mg/kg). The oral dose formulations of E36 were prepared by first dissolving E36 in appropriate amounts of Transcutol (30%> of total vehicle), then slowly adding required Labrasol (70%> of total vehicle), and continuously mixing until a clear solution was obtained. The oral dosing concentrations of E36 prepared were 0.02, 0.06, 0.2, 0.6 and 2 mg/mL at a dosing volume of 5 mL/kg (0.1, 0.3, 1, 3 and 10 mg/kg).
[00316] The in-life portion of this study was conducted at Covance Research Products (Alice, TX). The male and female cynomolgus monkeys (2-4 kg) used in this evaluation were from the Covance stock colony. Animals in the Covance stock colonies were maintained and monitored for good health in accordance with Covance SOPs and at the discretion of the laboratory animal veterinarian. Prior to assignment to the study, the health of each animal is assured in accordance with Covance SOPs. During the two-day acclimation and the test period, animals were housed in individual cages. Animals were not commingled for at least 24 hours after dose administration to allow monitoring of any test article-related effects.
[00317] Non-certified primate diet and tap water were provided ad libitum unless otherwise specified for dose administration. Fruits and other appropriate treats may have also been provided. Environmental controls for the animal room were set to maintain a temperature of 18 to 26°C and a 12-hour light/12-hour dark cycle. Humidity was not monitored.
[00318] Protocol: The protocol for the two phase study is summarized in the table below.
[00319] In the first phase, three fasted male and three female cynomolgus monkeys were administered an IV bolus via the saphenous vein of 1 mg/kg of A36 as a solution in phosphate buffer saline. Blood samples were collected from the femoral vein predose and at 0.083, 0.25, 0.5, 1, 2, 4, 8, 12, 24, 36, 48 and 72 hr post dose and transferred into tubes containing potassium EDTA anticoagulant and the esterase inhibitor, paraoxon. After dose administration, but before the needle was removed from the animal, the dose apparatus will be flushed with approximately 2 mL of saline.
[00320] In the second phase of the study, the same set of three fasted male and female animals from the first phase was administered an oral gavage of 3 mg/kg of E36 as a solution in 70% Labrasol/30% Transcutol. The oral doses were administered via nasogastric intubation. Prior to withdrawing the gavage tube, the tube was flushed with approximately 5 mL of water. Blood samples were collected predose and at 0.25, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 24, 36, 48 and 72 hr post dose and processed as previously described. Additional groups of monkeys consisting of one male and one female were orally administered 0.1, 0.3, 1 and 10 mg/kg of E36 (70% Labrasol/30%> Transcutol) with blood sampled as before.
[00321] Blood samples were centrifuged to obtain plasma. The plasma samples were stored at -70°C prior to LC-MS/MS analysis.
[00322] Sample Preparation and LC-MS/MS Analysis
[00323] On the day of analysis, the plasma samples were thawed at 4°C for 4.5 hr. Plasma proteins from plasma samples (50 μί) were precipitated by addition of 100 μΙ_, of
acetonitrile containing 200 ng/mL of a proprietary internal standard. After 20 min of centrifugation (Eppendorf microfuge, 4,000 rpm, 4°C) the resulting supernatant was collected and analyzed by LC-MS/MS.
[00324] A 10 aliquot of the supernatant was injected onto a Gemini CI 8 column (5 μιη, 2 x 50 mm, Phenomenex) fitted with a Gemini CI 8 guard column (5 μιη, 4.0 x 2.0 mm, Phenomenex, Torrance, CA) and eluted with a gradient consisting of mobile phase A (5 mM ammonium acetate in 5% acetonitrile) and B (1% ammonium hydroxide in 100% acetonitrile) at a flow rate of 0.5 mL/min (0 min, 0% B; 0-0.1 min, 0-50 % B; 0.1-1.7 min, 50-60 % B; 1.7-1.8 min, 60-90 % B; 1.8-3.5 min, 90 % B; 3.5-3.6 min, 90-0 % B; 3.6-5.5 min, 0 % B). The injector temperature was set at 4°C. A36, the monoprodrug, and E36 were detected using the negative MS/MS mode (411.1/63.1, A36) and quantified by comparison of peak areas to standard curves obtained by spiking known concentrations of the analytes to heparinized blank rat plasma. Calibration curves ranged from 5 to 6000 ng/mL of A36 with a limit of quantification (LOQ) of ~ 0.5 ng/mL.
[00325] WinNonLin Analysis
[00326] The temporal profile of plasma drug concentrations was analyzed by non- compartmental methods [1, 2] using WinNonLin version 5.2 (Pharsight Corp, Mountain View, CA) [3]. Area under the curve (AUC) values were determined by trapezoidal summation of the plasma concentration-time profile to the last measurable time point. The t½ values were calculated by the first order rate constant associated with the terminal log-linear portion of the curve as defined by at least three data points of the elimination phase.
[00327] For IV bolus analysis, extrapolation of the plasma concentration-time plot was performed to estimate the zero time intercept Co by fitting a natural log-linear line to the first two data points. The mean absorption time (MAT) value is the difference between mean residence time MRTpo and MRTiy. The oral bioavailability (OBAV) of A36 was calculated in a true crossover fashion by comparison of the area under the curve (AUC) values of the plasma concentrations of A36 following oral administration of E36 with the AUC values of A36 following IV administration of A36.
[00328] Statistical Data Analysis
[00329] Results in tables and graphs are expressed as means ± standard deviations (s.d.) or means ± standard error of the mean (s.e.m.) for sample size n>2 and means ± ½ range for n=2.
[00330] Results: The key pharmacokinetic parameters (mean ± s.d., n=6) of A36 following IV administration of 1 mg/kg of A36 and after oral administration of 3 mg/kg of the prodrug E36 to fasted cynomolgus monkeys are summarized in the table below (male and female data combined). Emesis occurred in monkeys (at 1 and 2 hr post dose in one animal and at 4 hr post dose for eight animals, and again at 6 hr post dose for one of the eight animals) administered the oral formulation of E36 and was attributed to the vehicle. Dose-related systemic exposure of A36 was observed following oral administration range of 0.1 to 10 mg/kg of E36. No obvious sex differences in the IV pharmacokinetic parameters of A36 were observed in this small sample size comparison. The plasma bioavailability of A36 following oral administration of a 3 mg/kg dose of E36 to male and female monkeys was 3.86%.
[00331] Mean (±½ range or s.d.) plasma Cmax values of 0.93 ± 0.93, 2.67 ± 0.09, 22.45 ± 3.85, 86.93 ± 56.08 and 230 ± 74 ng/mL following oral administration of 0.1, 0.3, 1, 3 and 10 mg/kg of E36 were observed at a Tmax of 4 hr across the dosing range. Plasma concentrations of E36 were not detected but levels of the monoprodrug up to 97 ng/mL were observed in animals orally administered E36.
[00332] Conclusions: In cynomolgus monkeys, the plasma clearance of A36 was low (0.088 L/hr/kg versus 2.62 L/hr/kg hepatic blood flow) and the volume of distribution was below total body water volume (0.20 L/kg versus 0.6 L/kg volume of total body water). The plasma elimination half-life of A36 following an IV bolus administration of A36 was 6.06 hr. No obvious sex differences in the IV pharmacokinetic parameters of A36 were observed in this small sample size comparison.
[00333] Following oral administration of 0.1, 0.3, 1, 3 and 10 mg/kg of E36, the plasma concentrations of A36 reached plasma Cmax values of 0.93, 2.67, 22.4, 86.9 and 230 ng/mL, respectively. The Tmax of A36 after oral dosing of E36 was 4 hr independent of the dosing range. The MAT of A36 was approximately 7.81 hr after oral administration of 3 mg/kg of E36. The plasma elimination half- life of A36 following PO administration of the 3 mg/kg of the prodrug was 5.91 hr. The plasma AUC and Cmax values of A36 increased in a dose-related manner. Higher systemic exposure of the monoprodrug as measured by plasma Cmax and AUC was observed in female monkeys after oral administration of 3 and 10 mg/kg of E36. The oral bioavailability of A36 following administration of the 3 mg/kg of E36 as a solution in 70% Labrasol/30% Transcutol, a vehicle which caused emesis (at 1 and 2 hr post dose in one animal and at 4 hr post dose for three animals used in the bioavailability assessment), was 3. 6%> (8n=6).
7. Acute effects on gene expression in the liver, pituitary, and heart of the sprague dawley rat
[00334] Objective: To determine the effects of single doses of E36 on the expression of thyroid hormone-sensitive genes in the liver, pituitary, and heart of the Sprague Dawley rat and to compare with the effects of thyroid hormone, T3.
[00335] Methods: Groups (n=6) of male Sprague Dawley rats (225-250 g) ( Harlan (Livermore, CA), maintained on a 12: 12 hr light/ dark cycle (lights on at 7 am), and fed an ad-libitum diet of standard rat chow (Harlan 7001) prior to the study), were administered single oral doses of vehicle, E36 (0.1 - 10 mg/kg) or T3 (0.12 mg/kg) dissolved initially in 100% transcutol and then diluted with labrasol to yield a final solution consisting of 30% Transcutol/70% Labrasol and a total volume of 2 mL/kg. The formulations were prepared just prior to initiation of treatment and stored at 4 °C. At 3, 8, and 24 hours following drug administration, animals were anesthetized and the liver, pituitary, and heart were excised and snap frozen. mRNA levels of various thyroid hormone-sensitive genes were analyzed by RT-PCR.
[00336] Experimental protocol: Rats were fasted overnight prior to administration of E36 or T3 by gavage according to the schedule shown below:
[00337] Food was withheld for the remainder of the study. At each of the time points following vehicle or drug administration (3, 8, or 24 hours), animals were anesthetized with 2.5% isoflurane (Hospira, Lake Forest, IL), the abdominal cavity was opened, the diaphragm was cut, and the heart quickly removed and freeze-clamped in liquid nitrogen. The pituitary and liver were also removed and either freeze-clamped or snap-frozen in liquid nitrogen. All samples were stored at -80°C until processed for mRNA analysis of the following genes: malic enzyme (ME), cytochrome P450 isozyme 7A (CYP7A), and sterol regulatory element binding protein lc (SREBPlc) in liver; thyroid hormone β (TSH ) in pituitary; myosin heavy chain β (MHC ) and deiodinase 1 (ID-1) in heart.
[00338] b) mRNA analysis. Frozen tissues (10-50 mg) were transferred to Lysing Matrix D tubes containing 1.4 mm ceramic spheres (QBiogene, Irvine, CA). Following addition of 1 ml of Trizol (Invitrogen, Carlsbad, CA), the tissues were homogenized with a FastPrep tissue disruptor (QBiogene, Irvine, CA). Chloroform (200 μΐ) was added to each sample and mixed thoroughly. The samples were then centrifuged at 12,000 rpm for 15 minutes in an Eppendorf table-top centrifuge. The resulting RNA-containing supernatants were carefully transferred to new tubes and mixed with 70% ethanol (400 μΐ). Total RNA was further purified by means of RNeasy RNA purification reagents (Qiagen, Valencia, CA) used according to the instructions provided by the manufacturer. Following incubation with RNase-free DNase I (Invitrogen, Carlsbad, CA) for 15 minutes at 25 °C, the RNA samples were used as templates to synthesize first strand cDNA with reagents from Invitrogen used according to the manufacturer's instruction. The RT-PCR reactions contained 5 ng of cDNA, 200 nM of each primer, and 5 μΐ of SYBR supermix. The total volume of the reaction was 10 μΐ. The following qRT-PCR protocol was used: 1
cycle at 95 °C for 3 minutes followed by 40 cycles at 95 °C for 15 seconds and 60 °C for 45 seconds. All reactions were carried out in an iCycler IQ RT- PCR Detection System (Bio-Rad Laboratories, Inc., Hercules, CA). The RT-PCR threshold numbers were adjusted based on the actual curves and the ratios between vehicle and compound treated samples were calculated using the 2"AACt method (1).
[00339] Primers for RT-PCR reactions were as follows:
ME, 5'-GCTCTATCCTCCTTTGAATAC-3 ' (forward) and
5 '-ATAATTAGTGCTGTACATCTG-3 ' (reverse);
CYP7a, 5 ' -GTTTCGACATGCTCTCGCTAT-3 ' (forward) and
5 '-GACCAGAATAACCTCAGACTC-3 (reverse);
SREBP-lc, 5 ' -GGAGCCATGGATTGCACATT-3 ' (forward) and
5 '-AGGAAGGCTTCCAGAGAGGA-3 ' (reverse);
XSHJ_, 5 '-AGGAGAGAGTGTGCCTACTGC-3 ' (forward) and
5 '-GGTATTTCCACCGTTCTGTAG-3 ' (reverse);
MHCB. 5 ' -CAGGCCAAGCGCAACCACCTG-3 ' (forward), and
5 '-ACTCTGGAGGCTCTTCACTTG-3 ' (reverse);
ID-1 , 5 ' -GTGGACACAATGCAGAACCAG-3 ' (forward) and
5 '-ACTTCCTCAGGATTGTAGTTC-3 ' (reverse)
[00340] Results: In liver, E36 and T3 treatment resulted in similarly increased mRNA levels of malic enzyme (ME), and cytochrome P450 isozyme 7 A (CYP7A), and similarly reduced mRNA levels of sterol regulatory element binding protein 1C (SREBPlc). E36 treatment resulted in dose-dependent decreases in pituitary mRNA levels of thyroid stimulating hormone β (TSH ) with a reduction of -20% observed at the highest dose. In contrast, T3 treatment led to profound reductions in pituitary TSH (-80%) mRNA levels. In the heart, E36 treatment resulted in modest, dose-dependent reduction in mRNA levels of myosin heavy chain β (MHCP; ~30%> reduction at the highest dose), but had no obvious effect on mRNA levels of deiodinase-1 (ID-1). However, T3 treatment led to profound reductions in heart MHC (-70%) mRNA levels and a marked increase in heart ID-1 mRNA levels (-700%).
[00341] Conclusions: Administration of single oral doses of E36 or T3 to Sprague Dawley rats resulted in the expected changes in mRNA levels of thyroid hormone- sensitive genes ME, CYP7A, and SREBPlc in the liver. In contrast, treatment with E36 had significantly reduced impact on the expression of genes in the pituitary (TSH ) and in the heart (MHC , ID-1) compared to T3 treatment.
8. Effects on plasma cholesterol levels and thyroid function indicators after 14 days of once-daily oral administration to beagle dogs
[00342] Objective: To determine the effects of once-daily oral administration of E36 for 14 days on plasma cholesterol levels and thyroid function indicators in beagle dogs.
[00343] Methods: Male and female beagle dogs were purchased from Marshall Farms (North Rose, NY) at approximately 9-15 kg in body weight. Animals were housed individually under a 12-hour lighting cycle (7 am-7 pm light) and controlled temperature (-22° C). The dogs were fed twice-daily with Teklad 8563 chow (Harlan Teklad, Madison, WI) and allowed water ad libitum. The twelve beagle dogs (9-15 kg) were randomized into 6 dosing groups (1 male and 1 female/group) and gavaged once-daily with a PEG-400 solution of E36 at doses of 0.1, 0.3, 1, 3, or 10 mg/kg or with vehicle for 14 days. E36 was dissolved in 100% PEG-400. The formulation was prepared just prior to initiation of treatment and stored at 4°C. A fresh formulation was prepared for each 7- day treatment period. At the end of the treatment cycle (Cycle 1), the dogs were washed out for 6 weeks and then entered into a second 14-day treatment cycle. Cycle 2 employed the same dosing paradigm as Cycle 1 , but animals were randomized to Cycle 2 in such a way that the combined dosing groups from the two cycles each consisted of 4 different animals (2 males, 2 females). Blood samples were collected at baseline and appropriate time intervals thereafter and analyzed for total plasma cholesterol levels, serum levels of total T4 (tT4), free T4 (fT4), total T3 (tT3), free T3 (fT3), and thyroid stimulating hormone (TSH), and for plasma drug levels.
[00344] Experimental protocol: In Cycle 1, 6 male and 6 female dogs were randomized into the following 6 treatment groups consisting of 1 male and 1 female animal each: vehicle, 0.1, 0.3, 1, 3, and 10 mg/kg/day of E36 (A36 equivalents). After a 14 day dosing period, the dogs were washed out for 6 weeks and then entered into a second 14-day treatment cycle. Cycle 2 employed the same dosing paradigm as Cycle 1, but animals were randomized into different dosing groups for Cycle 2 in such a way that the combined dosing groups from the two cycles each consisted of 4 different animals (2 males, 2 females).
Weeks 1-2 (Cycle 1)
Weeks 3-8
Washout (the length of the washout allowed full recovery of cholesterol levels and indicators of thyroid hormone function).
Weeks 8-10 (Cycle 2)
**Baseline = average of values obtained on D -37, -34, -30, -27, -23, -16 and prior to dosing on Dl.
***Cholesterol and TFT values remained relative stable over the course of the extended baseline sampling period.
[00345] In both cycles, vehicle and drugs were administered once-daily by gavage at approximately 8:30 am by means of a gastric feeding tube (#10) inserted through a bite block. The dosing volume was 2 mL/kg. Blood samples were obtained in the conscious state via cephalic venipuncture. Blood samples (1-3 mL) were collected into lithium heparin-containing vials and into serum separator tubes (Becton Dickinson). Plasma was prepared from the heparinized blood samples by centrifugation in an Eppendorf Microfuge (14,000 rpm, 2 min, room temperature). Serum was prepared according to the instructions of the serum separator tube manufacturer. Plasma and serum samples were stored at "70°C until analysis.
[00346] Analyses:
[00347] Plasma cholesterol. Total plasma cholesterol was measured using an Infinity cholesterol reagent (Thermo Electron Corporation, Waltham, MA) and with use of a standard curve prepared from a 300 mg/dL cholesterol standard. Average values ± the standard error of the mean (SEM) were calculated for all treatment groups.
[00348] Thyroid function tests (TFTs). Serum samples were shipped on dry ice to the Diagnostic Center for Population and Animal Health (Lansing, MI) and assayed for total T4 (tT4), free T4 (f 4), total T3 (tT3), free T3 (fT3), and thyroid stimulating hormone (TSH). Average values ± the standard error of the mean (SEM) were calculated for all treatment groups.
[00349] Plasma drug levels. On the day of analysis, the plasma samples were thawed at 4°C for 4.5 hr. Plasma proteins from plasma samples (50 μί) were precipitated by addition of 100 \ L of acetonitrile containing 200 ng/mL of an internal standard (A37). After 20 min of centrifugation (Eppendorf microfuge, 4,000 rpm, 4°C) the resulting supernatant was collected and analyzed by LC-MS/MS. A 10 aliquot of the supernatant was injected onto a Gemini C18 column (5 μιη, 2 x 50 mm, Phenomenex) fitted with a Gemini C18 guard column (5 μιη, 4.0 x 2.0 mm, Phenomenex, Torrance, CA) and eluted with a gradient consisting of mobile phase A (5 mM ammonium acetate in 5% acetonitrile) and B (1% ammonium hydroxide in 100% acetonitrile) at a flow rate of 0.5 mL/min (0 min, 0% B; 0-0.1 min, 0-50 % B; 0.1-1.7 min, 50-60 % B; 1.7-1.8 min, 60-90 % B; 1.8-3.5 min, 90 % B; 3.5-3.6 min, 90-0 % B; 3.6-5.5 min, 0 % B). The injector temperature was set at 4°C. E36 and A36 were detected using the negative MS/MS mode and quantified by comparison of peak areas to standard curves obtained by spiking known concentrations of the analytes to heparinized blank rat plasma. Calibration curves ranged from 5 to 6000 ng/mL. The limit of quantification (LOQ) was -10 ng/mL. Average values ± the standard error of the mean (SEM) were calculated for all treatment groups.
[00350] Results: Treatment with E36 resulted in progressive, dose-dependent reductions of total plasma cholesterol levels, with an average reduction at the end of treatment of -20 mg/dL or -15 % from baseline at the lowest dose evaluated (0.1 mg/kg/day), and of -60 mg/dL or -38% from baseline at the highest dose evaluated (10 mg/kg/day). Levels of tT4 on Day 15 were reduced by <10% from baseline in all E36 treatment groups with exception of the 1 and 10 mg/kg/day groups in which a -29% and a -44%) reduction was observed, respectively. Levels of fT4 on Day 15 were reduced by -13-32%) from baseline in the E36 treatment groups but the reductions were not dose- dependent. Moreover, a ~19%> reduction in fT4 levels was observed in the vehicle-treated group. Changes from baseline in tT3 levels in the E36-treated groups on Day 15 were no greater than that observed in the vehicle-treated group, whereas fT3 levels were reduced
in a dose-related manner by -8-45% in the E36 treatment groups with only a -4% reduction observed in the vehicle-treated group. No meaningful changes in TSH levels were observed in any of the treatment groups.
[00351] Conclusions: Once-daily oral treatment of beagle dogs for 14 days with E36 (0.1-10 mg/kg) reduced total plasma cholesterol levels in a dose dependent manner by 15- 38%. Serum TSH levels were essentially unaffected by treatment with E36. Other indicators of thyroid function in general did not undergo major changes except in the highest dose group (10 mg/kg/day) at which -45 % reductions in tT4 and fT3 were observed.
9. Effects on cardiovascular function following seven days of once-daily oral administration to male Sprague Dawley rats
[00352] Objective: To evaluate the effects of E36 administered orally once-daily for 7 days at doses of 0.03 to 10 mg/kg/day on cardiovascular function (heart rate and first derivative of left ventricular pressure) in the Sprague Dawley rat and to compare with the effects of thyroid hormone, T3.
[00353] Methods: Male Sprague-Dawley (SD) rats (200-220g body weight) were purchased from Harlan (Livermore, CA), maintained on a 12: 12 hr light/ dark cycle (lights on at 7 am), and fed an ad libitum diet of standard rat chow (Harlan 7001) for the duration of the study. E36 was dissolved in 100%) PEG-400. T3 was dissolved in water. The formulations were prepared just prior to initiation of treatment and stored at 4°C. Vehicle (100% PEG-400), E36 (0.03 -10 mg/kg) or T3 (0.2 mg/kg) were administered to Sprague Dawley rats (200-220 g) orally once-daily for 7 days. On the 8th day, animals were anesthetized and instrumented with subcutaneous electrodes and a carotid manometer for the measurement of heart rate (HR) and the first derivative of ventricular pressure (LV dP/dt), respectively.
[00354] Protocol: Fifty-four rats were randomized into 9 groups of 6 animals and orally dosed daily at -9:30 am for 7 days according to the schedule below:
[00355] On the 8 day, animals were anesthetized with isoflurane 2.5% (02 carrier) placed in a dorsal recumbent position and body temperature maintained with a circulating- water heating pad at 37 °C. Needle electrodes were placed subcutaneous ly to allow continuous recording of an electrocardiogram (ECG; Gould Instrument Systems). The ECG signal was processed with a Gould Biotach amplifier generating heart rate (HR) measurements on a beat-to-beat basis, and expressed as beats per minute (bpm). The right common carotid artery was exposed via a midline incision, ligated distally, and cannulated with a pressure-calibrated 2.5F Millar catheter tip manometer interfaced to a transducer amplifier (Gould). The pressure signal was further processed using a differential amplifier (Gould) allowing continuous recording of the first derivative of left ventricular pressure (LV dP/dt). All analog signals were digitally acquired at 400 hz using the CODAS acquisition system (Dataq Inc), and average values (HR, LV dP/dt,) determined in the playback mode.
[00356] Results: The administration of T3 to Sprague Dawley rats resulted in marked and significant increases in HR and LV dP/dt relative to vehicle-treated animals. In contrast to administration of T3, the administration of E36 did not result in meaningful changes in HR or LV dP/dt.
[00357] Conclusions: E36 administered at once-daily oral doses of up to 10 mg/kg/day for 7 days is devoid of chronotropic and inotropic effects in the Sprague Dawley rat. This is in contrast to T3 treatment, which was associated with marked effects on cardiac function.
10. Effects on plasma cholesterol levels and thyroid function indicators after 14 days of once-daily oral administration followed by 14 days of alternate day administration to beagle dogs
[00358] Objective: To determine the effects of oral administration of E37 once-daily for 14 days followed by alternate day dosing for 14 days on plasma cholesterol levels and indicators of thyroid function in beagle dogs.
[00359] Methods: Male and female Beagle dogs were purchased from Marshall Farms (North Rose, NY) at approximately 9-15 kg. Animals were housed individually under a 12-hour lighting cycle (7 am-7 pm light) and controlled temperature (-22° C). The dogs were fed twice-daily with Teklad 8563 chow (Harlan Teklad, Madison, WI) and allowed water ad libitum. The twelve beagle dogs (9-15 kg) were randomized into 6 dosing groups (1 male and 1 female/group) and gavaged once-daily with a 0.5% CMC/1% Lutrol F68 suspension of E37 at doses of 0.1, 0.3, 1, 3, or 10 mg/day or with vehicle for 14 days. E37 was administered as a suspension in 0.5%> CMC/1% Lutrol in deionized water. To prepare the vehicle, the required amount of CMC was weighed and dissolved in deionized water using a Waring blender. The required amount of Lutrol F68 was weighed and slowly added to the Waring blender while mixing. The contents of the blender were mixed until dissolved and stored refrigerated. To prepare the dosing formulations, the required amount of E37 was weighed into a beaker and the required volume of vehicle slowly added to the beaker while stirring using a magnetic stir bar and stir plate. The contents of the beaker were stirred until a fine paste was obtained. Vehicle was added to the beaker while stirring until a uniform E37 suspension was obtained. At the end of the treatment cycle (Cycle 1), the dogs were washed out for 4 weeks and then entered into a second 14-day treatment cycle. Cycle 2 employed the same dosing paradigm as Cycle 1, but animals were randomized to Cycle 2 in such a way that the combined dosing groups from the two cycles consisted of 4 different animals (2 males, 2 females) each. At the conclusion of Cycle 2, dosing was continued on alternate days for an additional 14-day period (Cycle 2 Extension). Blood samples were collected at baseline and appropriate time intervals thereafter and analyzed for total plasma cholesterol levels, serum levels of total T4 (tT4), free T4 (fT4), total T3 (tT3), free T3 (fT3), and thyroid stimulating hormone (TSH).
[00360] Experimental protocol: In Cycle 1, 6 male and 6 female dogs were randomized into the following 6 treatment groups consisting of 1 male and 1 female
animal each: vehicle, 0.1, 0.3, 1, 3, and 10 mg/kg/day of E37 (A37 equivalents). After a 14 day dosing period, the dogs were washed out for 4 weeks and then entered into a second 14-day treatment cycle. Cycle 2 employed the same dosing paradigm as Cycle 1, but animals were randomized to Cycle 2 in such a way that the combined dosing groups from the two cycles each consisted of 4 different animals (2 males, 2 females). At the conclusion of Cycle 2, dosing was continued on alternate days for an additional 14-day period.
[00361] Vehicle and drug were administered by gavage at approximately 8:30 am by means of a gastric feeding tube (#10) inserted through a bite block. The dosing volume was 2 mL/kg. Blood samples were obtained in the conscious state via cephalic venipuncture. Blood samples (1-3 mL) were collected into lithium heparin-containing vials and into serum separator tubes (Becton Dickinson). Plasma was prepared from the heparinized blood samples by centrifugation in an Eppendorf Micro fuge (14,000 rpm, 2 min, room temperature). Serum was prepared according to the instructions of the serum separator tube manufacturer. Plasma and serum samples were stored at "70°C until analysis.
[00362] A detailed protocol is shown below:
Weeks 1-2 (Cycle 1)
Weeks 3-6:
Washout (the length of the washout selected allowed full recovery of cholesterol levels and thyroid hormone function indicators).
[00363] Analyses:
[00364] Plasma cholesterol. Total plasma cholesterol was measured using an Infinity cholesterol reagent (Thermo Electron Corporation, Waltham, MA) and with use of a standard curve prepared from a 300 mg/dL cholesterol standard. Average values ± the standard error of the mean (SEM) were calculated for all treatment groups.
[00365] Thyroid function tests (TFTs). Serum samples were shipped on dry ice to the Diagnostic Center for Population and Animal Health (Lansing, MI) and assayed for total T4 (TT4), total T3 (TT3), free T4 (fT4), free T3 (fT3), and thyroid stimulating hormone (TSH). Average values ± the standard error of the mean (SEM) were calculated for all treatment groups.
[00366] Muscle Assays. In addition, Gastrocniemius muscle mass may be measured, e.g., in rats after treating with T3, KB-141 and the compound of interest.
[00367] Results: Treatment with E37 for 14 days resulted in progressive, dose- dependent reductions of total plasma cholesterol levels, with an average reduction on Day 15 of -28 mg/dL or -22% from baseline at a dose of 0.3 mg/kg/day and of -71 mg/dL or -47%) from baseline at the highest dose evaluated (10 mg/kg/day). The lowest dose of E37 evaluated, 0.1 mg/kg/day, had minimal effects on total plasma cholesterol levels. During the alternate day dosing period of Cycle 2 (Cycle 2 Extension), total plasma cholesterol levels in the E37 treatment groups remained reduced relative to vehicle- treated animals to a similar or greater extent than observed after once-daily dosing. Once-daily treatment with E37 resulted in dose-dependent reductions of serum tT4 (-20- 54%>), dose-related reductions of fT4 (~8-39%>), and non-dose-dependent reductions of fT3 (-15-32%) from baseline levels. There were no meaningful changes in tT3 levels relative to baseline levels that exceeded those observed in the vehicle-treated group in any of the E37 treatment groups. Effects of once-daily E37 treatment on serum TSH levels
were variable, with full suppression in some animals and up to 4-fold elevations in others on Day 8. On Day 15, TSH levels were reduced from baseline in a non-dose-dependent manner by -6-27% in the E37-treated groups. During the Cycle 2 Extension, levels of tT4 and fT4 that were suppressed by once-daily treatment gradually recovered to levels that approached those of vehicle-treated animals. A more variable recovery of fT3 and TSH levels was observed that did not extend to all E37 dose groups.
[00368] Conclusions: Once-daily oral treatment of beagle dogs for 14 days with E37 (0.1 - 10 mg/kg) resulted in dose-dependent reductions of average total plasma cholesterol levels (up to 47% from baseline) that were accompanied by dose-dependent reductions of serum tT4 levels, dose-related reductions in fT4 levels, and considerable fluctuations in serum TSH levels. Levels of tT3 were unaffected by treatment whereas fT3 levels were reduced in a non-dose-dependent manner. A switch from once-daily to alternate day dosing of E37 in the Cycle 2 extension did not compromise cholesterol lowering efficacy but resulted in a gradual recovery of levels of tT4 and fT4 and, in some dose groups, of levels of fT3 and TSH that were suppressed by once-daily oral E37 treatment. Alternate day dosing is thus an effective alternative to once-daily dosing that has reduced impact on the thyroid hormone axis. In addition, E37 had no detectable effects on muscle mass which suggests that unlike other T3 mimetics it did not stimulate muscle proteolysis.
Claims
WHAT IS CLAIMED:
I . A phosphorus-containing thyroid hormone agonist compound with a
pharmacokinetic half-life in a non-rodent mammalian species less than or equal to 8, preferably 6, more preferably 4, or more preferably 2 hrs.
2. A compound of claim 1, wherein the pharmacokinetic half-life is measured with reference to an active prodrug of the phosphorus-containing thyroid hormone agonist in plasma.
3. A compound of claim 1, which when administered at a lipid lowering (ED50) dose, causes a less than 40% decrease in circulating total T4 levels, bound and unbound, in a non-rodent mammalian species when measured within 24 hours after a fourteen day dose administration of said compound compared to baseline T4 levels.
4. A phosphorus-containing thyroid hormone agonist compound, which when administered at a lipid lowering (ED50) dose, causes a decrease in circulating T4 levels of less than 20% in a non-rodent mammalian species when measured within 24 hours after a fourteen day dose administration of said compound compared to baseline T4 levels.
5. A compound of any of claims 1-4, which at maximally effective dose, exhibits no significant liver adverse findings (frank necrosis (biopsy), liver function test (LFT).
6. A compound of any of claims 1-4, which shows increase in oxygen consumption of less than 15%, 10%, 5%.
7. A compound of any of claims 1-4, which does not significantly change mRNA levels of genes in muscle, pituitary, and/or heart by more than statistical significance, 1 - 5 fold, etc.
8. A compound of any of claims 1-4, which does not change T4:T3 ratio by more than 40%>; and/or does not cause T4:T3 ratio to drop below 15: 1.
9. A compound of any of claims 1-4, having a pharmacodynamic effect on a lipid component/ gene product in in vitro assay.
10. A compound of any of claims l-4,which accumulates in the liver preferentially to other tissues at a ratio of at least 2: 1, preferably with 10, 20, 50, 100 to 1 or more liver specificity as assayed by gene expression, in vivo signature genes in liver v. heart, muscle, kidney (PNAS).
I I . A compound of any of claims 1-10, designed with a metabolically unstable substituent.
12. A compound of claim 11 , wherein the metabolite does not activate the thyroid hormone receptor at doses < 1 μΜ.
13. A compound of claim any of claims 1-12, wherein said thyroid hormone agonist is a compound of Formula I:
(Ar1)-G-(Ar2)-T-E
wherein:
1 2
Ar and Ar are substituted aryl or heteroaryl groups;
1 2
G is an atom or group of atoms that links Ar and Ar through 1-2 contiguous atoms;
T is an atom or group of atoms linking Ar to E through 1 -4 contiguous atoms or is absent; and
E is a functional group with a pKa < 4.0 containing a phosphorus atom, and prodrug thereof.
14. A compound of claim 13, wherein said thyroid hormone agonist is a compound of Formula II:
wherein:
m is an integer from 0-3;
n is an integer from 0-2;
p is an integer from 0-1;
R\ R2, R6, and R7 are each independently selected from:
R1 is selected from:
1 7
or R and R are taken together along with the carbons to which they are attached to form:
E is a functional group with a pKa < 4.0 containing a phosphorus atom; and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
wherein:
p is an integer from 0-1;
Each Ra is independently selected from:
E is P(0)YRnY'Ru or P(0)YRnY";
Y" is -Ci-Ce-alkyl;
Y and Y' are each independently selected from the group consisting of -0-, and
-NRV-;
when Y is -O- and Y" is -Ci-Ce-alkyl, or when Y and Y' are both -0-, R11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy, -C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry, -alkyl-S-S-alkylhydroxy, and -alkyl- S -S - S -alkylhydroxy ;
when Y and Y' are both -NRV-, then R11 attached to -NRV- is independently selected from the group consisting of -H, -[C(Rz)2]q-COORy, -C(Rx)2COORy,
-[C(Rz)2]q-C(0)SRy, and -cycloalkylene-COORy;
when Y is -O- and Y' is NRV, then R11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl,
-C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy,
-C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry, -alkyl-S-S-alkylhydroxy, and -alkyl-S-S-S- alkylhydroxy; and R11 attached to -NRV- is independently selected from the group consisting of -H, -[C(Rz)2]q-COORy, -C(Rx)2COORy, -[C(Rz)2]q-C(0)SRy, and
-cycloalkylene-COORy;
or when Y and Y' are independently selected from -O- and -NRV-, then R11 and R11 together form a cyclic group comprising -alkyl-S-S-alkyl-, or together R11 and R11 are the group:
V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both Y groups attached to the phosphorus; or together V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or together Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or together W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; Z is selected from the group consisting of -CHRzOH, -CHRzOC(0)Ry,
-CHRzOC(S)Ry, -CHRzOC(S)ORy, -CHRzOC(0)SRy, -CHRzOC02Ry, -ORz,
-SRZ, -CHRZN3, -CH2aryl, -CH(aryl)OH, -CH(CH=CRz 2)OH,
-CH(C≡CRz)OH, -Rz, -NR -OCORy, -OC02Ry, -SCORy, -SC02Ry,
-NHCOR2, -NHC02Ry, -CH2NHaryl, -(CH2)q-ORz, and -(CH2)q-SRz; q is an integer 2 or 3;
Each Rz is selected from the group consisting of Ry and -H;
Each Ry is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
Each Rx is independently selected from the group consisting of -H, and alkyl, or together Rx and Rx form a cycloalkyl group;
Each Rv is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl; with the provisos that: a) V, Z, W, W are not all -H; and b) when Z is -Rz, then at least one of V, W, and W is not -H, alkyl, aralkyl, or heterocycloalkyl; and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
16. A compound of claim 11 , wherein the metabolic instability is mediated by the deactivating enzyme selected from the group consisting of esterase, deiodinase, carboxylesterase, aldehyde oxidase, glutathione transferase, cysteine β -lyase, and phosphatase.
17. A compound of claim 16 wherein the metabolic instability is mediated by the deactivating enzyme selected from the group consisting of the deactivating enzyme is selected from esterase, deiodinase, carboxylesterase, and aldehyde oxidase.
18. A compound of claim 17 wherein the metabolically unstable substituent is contained within the R substituent of a compound of Formula III.
A compound of claim 18, wherein R is selected from:
Each Rd is independently selected from:
23. A compound of claim 19, wherein E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11], -P(0)[-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
A compound of claim 19, wherein T is selected from:
E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy][-ORn],
-P(0)[-OCH(V)CH2CH20-], -P(0)(OH)(ORe), -P(0)(ORe)(ORe),
-P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe),
-P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected
from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl; and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
A compound of claim 24, wherein
R is OH
E is selected from:
-P03H2, -P(0)[-OCH2OC(0)-t-butyl]2, -P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2, -P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2,
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-N(H)C (CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH
(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3),
-P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3), and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
26. A compound of claim 11 , wherein the metabolic instability is mediated by the deactivating enzyme selected from the group consisting of glutathione transferase, and cysteine β -lyase.
27. A compound of claim 26, wherein the metabolically unstable substituent is contained within the T substituent of a compound of Formula III.
32. A compound of claim 27, wherein E is selected from:
-PO3H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11], -P(0)[-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), ) wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl. and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
A compound of claim 27, wherein:
R is selected from:
E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy][-ORn], -P(0)[-OCH(V)CH2CH20-], -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), ) wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl. and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
A compound of claim 33, wherein
R is OH;
E is selected from:
-PO3H2, -P(0)[-OCH2OC(0)-t-butyl]2, -P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2, -P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2,
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-N(H)C (CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH
(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3),
-P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3);
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
35. A compound of claim 11 , wherein the metabolic instability is mediated by the deactivating enzyme deiodinase.
A compound of claim 35, wherein a compound of Formula III contains at least metabolically unstable iodo substituent.
Each Re is independently selected from:
40. A compound of claim 36, wherein E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11], -P(0)[-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y"), and -P(0)[-OCRz 2OC(0)ORy](Y"), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
A compound of claim 36, wherein:
With the proviso that a least R 1 , or R 3 is iodo;
E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2,
-P(0)[-OCRz 2OC(0)ORy]2, -P(0)[-N(H)CRz 2C(0)ORy]2,
-P(0)[-N(H)CRz 2C(0)ORy] [-OR11], -P(0)[-OCH(V)CH2CH20-] , -P(0)(OH)(ORe), -P(0)(ORe)(ORe), -P(0)[-OCRz 2OC(0)Ry](ORe), -P(0)[-OCRz 2OC(0)ORy](ORe), -P(0)[-N(H)CRz 2C(0)ORy](ORe), -P(0)(OH)(NH2), -P(0)(OH)(Y"), -P(0)(ORy)(Y"), -P(0)[-OCRz 2OC(0)Ry](Y' '), and -P(0)[-OCRz 2OC(0)ORy](Y' '), wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
42. A compound of claim 41 , wherein
G is selected from:
With the proviso that a least R 1 , or R 3 is iodo;
R5 is OH
E is selected from:
-PO3H2, -P(0)[-OCH2OC(0)-t-butyl]2, -P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-N(H)CH2C(0)OCH2CH3]2, -P(0)[-N(H)CH(CH3)C(0)OCH2CH3]2,
-P(0)[-N(H)C(CH3)2C(0)OCH2CH3]2,
-P(0)[-N(H)CH(CH3)C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-N(H)C (CH3)2C(0)OCH2CH3][3,4-methylenedioxyphenyl], -P(0)[-OCH
(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-], -P(0)(OH)(OCH3), -P(0)(OH)(OCH2CH3), -P(0)[-OCH2OC(0)-t-butyl](OCH3),
-P(0)[-OCH2OC(0)0-z-propyl](OCH3), -P(0)(OH)(NH2), -P(0)(OH)(CH3),
-P(0)(OH)(CH2CH3), -P(0)[-OCH2OC(0)-t-butyl](CH3), and
-P(0)[-OCH2OC(0)0-«o-propyl](CH3).
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
43. A compound of claim 11 , wherein the metabolic instability is mediated by the deactivating enzyme phosphatase.
44. A compound of claim 43, wherein the metabolically unstable substituent is contained within the E substituent of a compound of Formula III.
A compound of claim 44, wherein T is selected from:
R11 attached to -O- is independently selected from the group consisting of -H, alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted CH2-heterocycloakyl wherein the cyclic moiety contains a carbonate or thiocarbonate, optionally substituted -alkylaryl, -C(Rz)2OC(0)NRz 2, -NRz-C(0)-Ry, -C(Rz)2-OC(0)Ry, -C(Rz)2-0-C(0)ORy, -C(Rz)2OC(0)SRy, -alkyl-S-C(0)Ry,
-alkyl-S-S-alkylhydroxy, and -alkyl-S-S-S-alkylhydroxy;
or when Y and Y' are independently selected from -O- and -NRV-, then R11 and R11 together form a cyclic group comprising -alkyl-S-S-alkyl- to form a cyclic group, or together R11 and R11 are the group:
V, W, and W are independently selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted aralkyl, heterocycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, optionally substituted 1-alkenyl, and optionally substituted 1-alkynyl; or together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, wherein 0 - 1 atoms are heteroatoms and the remaining atoms are carbon, substituted with hydrogen, hydroxy, acyloxy, alkylthiocarbonyloxy,
alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both Y groups attached to the phosphorus; or together V and Z are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon
substituted by hydrogen, and said cyclic group is fused to an aryl group at the beta and gamma position to the Y attached to the phosphorus; or together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms or carbon substituted by hydrogen and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from a Y attached to the phosphorus; or together Z and W are connected via an additional 3-5 atoms to form a cyclic group, wherein 0-1 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; or together W and W are connected via an additional 2-5 atoms to form a cyclic group, wherein 0-2 atoms are heteroatoms and the remaining atoms are carbon or carbon substituted by hydrogen, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
Z is selected from the group consisting of -CHRzOH, -CHRzOC(0)Ry, -CHRzOC(S)Ry, -CHRzOC(S)ORy, -CHRzOC(0)SRy, -CHRzOC02Ry, -ORz, -SRZ, -CHRZN3, -CH2aryl, -CH(aryl)OH, -CH(CH=CRz 2)OH, -CH(C≡CRz)OH, -Rz, -NR -OCORy,
-OC02Ry, -SCORy, -SC02Ry, -NHCOR2, -NHC02Ry, -CH2NHaryl,
-(CH2)q-ORz, and -(CH2)q-SRz; q is an integer 2 or 3;
Each Rz is selected from the group consisting of Ry and -H;
Each Ry is selected from the group consisting of alkyl, aryl, heterocycloalkyl, and aralkyl;
Each Rx is independently selected from the group consisting of -H, and alkyl, or together Rx and Rx form a cycloalkyl group;
Each Rv is selected from the group consisting of -H, lower alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl;
with the provisos that: a) V, Z, W, W are not all -H; and b) when Z is -Rz, then at least one of V, W, and W is not -H, alkyl, aralkyl, or heterocycloalkyl;
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
49. A compound of claim 44, wherein E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy]2,
-P(0)[-OCH(V)CH2CH20-], wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl.
A compound of claim 44, wherein:
E is selected from:
-P03H2, -P(0)[-OCRz 2OC(0)Ry]2, -P(0)[-OCRz 2OC(0)ORy]2,
-P(0)[-OCH(V)CH2CH20-], wherein V is selected from the group consisting of optionally substituted aryl, aryl, heteroaryl, and optionally substituted heteroaryl. and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
51. A compound of claim 50, wherein
R is OH
E is selected from:
-PO3H2, -P(0)[-OCH2OC(0)-t-butyl]2, -P(0)[-OCH2OC(0)0-z-propyl]2, -P(0)[-OCH(3-chlorophenyl)CH2CH20-], -P(0)[-OCH(pyrid-4-yl)CH2CH20-],
and pharmaceutically acceptable salts and prodrugs thereof and pharmaceutically acceptable salts of said prodrugs.
A method of lowering Lp(a) using a compound from any of claims 1-51
53. A method of reducing side effects on the thyroid hormone axis using a compound from any of claims 1-51.
54. A method of preferentially administering a compound to the the liver of a subject without achieving significant impact on other tissues, the method comprising:
administering a compound of any of cliams 1-51 to a subject in need thereof, wherein the compound is preferentially delivered to the liver.
55. A compound of any of claims 1-12, wherein said thyroid hormone agonist is selected from
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US24609209P | 2009-09-25 | 2009-09-25 | |
US61/246,092 | 2009-09-25 | ||
US29081209P | 2009-12-29 | 2009-12-29 | |
US61/290,812 | 2009-12-29 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2011038207A1 true WO2011038207A1 (en) | 2011-03-31 |
Family
ID=43466956
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2010/050169 WO2011038207A1 (en) | 2009-09-25 | 2010-09-24 | Phosphorus-containing thyroid hormone receptor agonists and methods of use |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2011038207A1 (en) |
Cited By (27)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN103172480A (en) * | 2013-03-05 | 2013-06-26 | 河南科技大学 | Method for preparing iodo aromatic hydrocarbon |
WO2017185087A1 (en) * | 2016-04-22 | 2017-10-26 | Viking Therapeutics | Use of thyroid beta-agonists |
WO2017185083A1 (en) | 2016-04-22 | 2017-10-26 | Viking Therapeutics | Use of thyroid beta-agonists |
WO2017184811A1 (en) * | 2016-04-22 | 2017-10-26 | Metabasis Therapeutics, Inc. | Thyroid hormone receptor agonist and use thereof |
US9908908B2 (en) | 2012-08-30 | 2018-03-06 | Jiangsu Hansoh Pharmaceutical Co., Ltd. | Tenofovir prodrug and pharmaceutical uses thereof |
WO2018053036A1 (en) * | 2016-09-16 | 2018-03-22 | Viking Therapeutics, Inc. | Method of reducing thyroid-associated side effects |
CN110996966A (en) * | 2017-06-05 | 2020-04-10 | 维京治疗公司 | Composition for treating fibrosis |
WO2020123827A1 (en) | 2018-12-13 | 2020-06-18 | Terns, Inc. | Thrβ receptor agonist compound and preparation method and use thereof |
US10849911B2 (en) | 2015-06-10 | 2020-12-01 | Galmed Research And Development Ltd. | Low dose compositions of Aramachol salts |
US10925885B2 (en) | 2005-05-26 | 2021-02-23 | Metabasis Therapeutics, Inc. | Thyromimetics for the treatment of fatty liver diseases |
WO2021032218A1 (en) | 2019-08-19 | 2021-02-25 | 苏州闻天医药科技有限公司 | HETEROCYCLIC THR-β RECEPTOR AGONIST COMPOUND AND PREPARATION METHOD AND USE THEREFOR |
CN112457346A (en) * | 2019-08-19 | 2021-03-09 | 苏州闻天医药科技有限公司 | Fused-ring THR beta receptor agonist compound and preparation method and application thereof |
WO2021089692A1 (en) * | 2019-11-05 | 2021-05-14 | The University Of Bristol | Inhibitor of binding between podocin and keratin 8 for use in the treatment of nephrotic syndrome |
US11202789B2 (en) | 2016-11-21 | 2021-12-21 | Viking Therapeutics, Inc. | Method of treating glycogen storage disease |
US11203587B2 (en) | 2018-10-12 | 2021-12-21 | Terns, Inc. | Thyroid hormone receptor beta agonist compounds |
EP3890748A4 (en) * | 2018-12-05 | 2022-12-21 | Viking Therapeutics, Inc. | Compositions and methods for the treatment of liver disorders |
US11571431B2 (en) | 2013-12-04 | 2023-02-07 | Galmed Research And Development Ltd | Aramchol salts |
US11697666B2 (en) | 2021-04-16 | 2023-07-11 | Gilead Sciences, Inc. | Methods of preparing carbanucleosides using amides |
US11752161B2 (en) | 2020-03-27 | 2023-09-12 | Gannex Pharma Co., Ltd. | Pharmaceutical compositions, method of making and method of using thereof |
US11767337B2 (en) | 2020-02-18 | 2023-09-26 | Gilead Sciences, Inc. | Antiviral compounds |
AU2020333099B2 (en) * | 2019-08-21 | 2023-12-21 | Changchun Changcheng Pharmaceutical Technology Co., LTD | A prodrug platform useful to deliver amines, amides and phenols |
US12030903B2 (en) | 2020-02-18 | 2024-07-09 | Gilead Sciences, Inc. | Antiviral compounds |
US12054507B2 (en) | 2020-02-18 | 2024-08-06 | Gilead Sciences, Inc. | Antiviral compounds |
US12102646B2 (en) | 2018-12-05 | 2024-10-01 | Viking Therapeutics, Inc. | Compositions for the treatment of fibrosis and inflammation |
US12116380B2 (en) | 2021-08-18 | 2024-10-15 | Gilead Sciences, Inc. | Phospholipid compounds and methods of making and using the same |
US12227533B2 (en) | 2018-03-22 | 2025-02-18 | Viking Therapeutics, Inc. | Crystalline forms and methods of producing crystalline forms of a compound |
JP7670687B2 (en) | 2020-02-24 | 2025-04-30 | へパジーン セラピューティクス (エイチケイ) リミテッド | Heterocyclic thr-beta receptor agonist compounds and methods for their preparation and use - Patents.com |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1999000353A1 (en) * | 1997-06-27 | 1999-01-07 | Karo Bio Ab | Novel thyroid receptor ligands and method |
WO2005051298A2 (en) * | 2003-11-19 | 2005-06-09 | Metabasis Therapeutics, Inc. | Novel phosphorus-containing thyromimetics |
WO2006128058A2 (en) * | 2005-05-26 | 2006-11-30 | Metabasis Therapeutics, Inc. | Thyromimetics for the treatment of fatty liver diseases |
WO2006128056A2 (en) * | 2005-05-26 | 2006-11-30 | Metabasis Therapeutics, Inc. | Novel phosphinic acid-containing thyromimetics |
-
2010
- 2010-09-24 WO PCT/US2010/050169 patent/WO2011038207A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1999000353A1 (en) * | 1997-06-27 | 1999-01-07 | Karo Bio Ab | Novel thyroid receptor ligands and method |
WO2005051298A2 (en) * | 2003-11-19 | 2005-06-09 | Metabasis Therapeutics, Inc. | Novel phosphorus-containing thyromimetics |
WO2006128058A2 (en) * | 2005-05-26 | 2006-11-30 | Metabasis Therapeutics, Inc. | Thyromimetics for the treatment of fatty liver diseases |
WO2006128056A2 (en) * | 2005-05-26 | 2006-11-30 | Metabasis Therapeutics, Inc. | Novel phosphinic acid-containing thyromimetics |
Non-Patent Citations (1)
Title |
---|
FUJITAKI JAMES M ET AL: "Preclinical pharmacokinetics of a HepDirect prodrug of a novel phosphonate-containing thyroid hormone receptor agonist.", November 2008, DRUG METABOLISM AND DISPOSITION: THE BIOLOGICAL FATE OF CHEMICALS NOV 2008 LNKD- PUBMED:18703645, VOL. 36, NR. 11, PAGE(S) 2393 - 2403, ISSN: 1521-009X, XP002618644 * |
Cited By (59)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10925885B2 (en) | 2005-05-26 | 2021-02-23 | Metabasis Therapeutics, Inc. | Thyromimetics for the treatment of fatty liver diseases |
US9908908B2 (en) | 2012-08-30 | 2018-03-06 | Jiangsu Hansoh Pharmaceutical Co., Ltd. | Tenofovir prodrug and pharmaceutical uses thereof |
CN103172480A (en) * | 2013-03-05 | 2013-06-26 | 河南科技大学 | Method for preparing iodo aromatic hydrocarbon |
US11571431B2 (en) | 2013-12-04 | 2023-02-07 | Galmed Research And Development Ltd | Aramchol salts |
US10849911B2 (en) | 2015-06-10 | 2020-12-01 | Galmed Research And Development Ltd. | Low dose compositions of Aramachol salts |
US20190321379A1 (en) * | 2016-04-22 | 2019-10-24 | Hongjian Jiang | Use of thyroid beta-agonists |
CN110200978A (en) * | 2016-04-22 | 2019-09-06 | 维京治疗股份有限公司 | The application of thyroid gland beta-2-agonists |
CN109641003A (en) * | 2016-04-22 | 2019-04-16 | 维京治疗股份有限公司 | The application of thyroid gland beta-2-agonists |
US20190134069A1 (en) * | 2016-04-22 | 2019-05-09 | Viking Therapeutics, Inc. | Use of thyroid beta-agonists |
JP2019515033A (en) * | 2016-04-22 | 2019-06-06 | バイキング セラピューティクス,インコーポレーテッド | Use of thyroid beta agonists |
JP2019515915A (en) * | 2016-04-22 | 2019-06-13 | バイキング セラピューティクス,インコーポレーテッド | Use of thyroid beta agonists |
JP2019516783A (en) * | 2016-04-22 | 2019-06-20 | メタベイシス・セラピューティクス・インコーポレイテッド | Thyroid hormone receptor agonist and use thereof |
AU2017252126B2 (en) * | 2016-04-22 | 2023-02-09 | Metabasis Therapeutics, Inc. | Use of thyroid beta-agonists |
CN109071577A (en) * | 2016-04-22 | 2018-12-21 | 症变治疗公司 | Thyroid hormone receptor agonists and application thereof |
US11951114B2 (en) | 2016-04-22 | 2024-04-09 | Viking Therapeutics, Inc. | Use of thyroid beta-agonists |
US10420781B2 (en) | 2016-04-22 | 2019-09-24 | Metabasis Therapeutics Inc. | Thyroid hormone receptor agonist and use thereof |
AU2017252122B2 (en) * | 2016-04-22 | 2023-02-09 | Viking Therapeutics, Inc. | Use of Thyroid beta-Agonists |
US20220257618A1 (en) * | 2016-04-22 | 2022-08-18 | Viking Therapeutics, Inc. | Use of thyroid beta-agonists |
US11351183B2 (en) | 2016-04-22 | 2022-06-07 | Viking Therapeutics, Inc. | Use of thyroid beta-agonists |
WO2017185087A1 (en) * | 2016-04-22 | 2017-10-26 | Viking Therapeutics | Use of thyroid beta-agonists |
AU2023202321B2 (en) * | 2016-04-22 | 2025-04-24 | Viking Therapeutics, Inc. | Use of thyroid beta-agonists |
WO2017184811A1 (en) * | 2016-04-22 | 2017-10-26 | Metabasis Therapeutics, Inc. | Thyroid hormone receptor agonist and use thereof |
WO2017185083A1 (en) | 2016-04-22 | 2017-10-26 | Viking Therapeutics | Use of thyroid beta-agonists |
CN110200978B (en) * | 2016-04-22 | 2023-10-31 | 维京治疗股份有限公司 | Use of thyroid beta-agonists |
US20190255080A1 (en) * | 2016-09-16 | 2019-08-22 | Viking Therapeutics, Inc. | Method of reducing thyroid-associated side effects |
JP7656574B2 (en) | 2016-09-16 | 2025-04-03 | バイキング・セラピューティクス・インコーポレイテッド | How to Reduce Thyroid-Related Side Effects |
EP3512523A4 (en) * | 2016-09-16 | 2020-05-06 | Viking Therapeutics, Inc. | METHOD FOR REDUCING SIDE EFFECTS ASSOCIATED WITH THE THYROID |
AU2017327383B2 (en) * | 2016-09-16 | 2023-06-29 | Metabasis Therapeutics, Inc. | Method of reducing thyroid-associated side effects |
JP2019531346A (en) * | 2016-09-16 | 2019-10-31 | バイキング・セラピューティクス・インコーポレイテッド | How to reduce thyroid-related side effects |
WO2018053036A1 (en) * | 2016-09-16 | 2018-03-22 | Viking Therapeutics, Inc. | Method of reducing thyroid-associated side effects |
CN109922812A (en) * | 2016-09-16 | 2019-06-21 | 维京治疗公司 | The method for reducing side effect relevant to thyroid gland |
US11202789B2 (en) | 2016-11-21 | 2021-12-21 | Viking Therapeutics, Inc. | Method of treating glycogen storage disease |
US11707472B2 (en) | 2017-06-05 | 2023-07-25 | Viking Therapeutics, Inc. | Compositions for the treatment of fibrosis |
CN110996966A (en) * | 2017-06-05 | 2020-04-10 | 维京治疗公司 | Composition for treating fibrosis |
US12227533B2 (en) | 2018-03-22 | 2025-02-18 | Viking Therapeutics, Inc. | Crystalline forms and methods of producing crystalline forms of a compound |
US11203587B2 (en) | 2018-10-12 | 2021-12-21 | Terns, Inc. | Thyroid hormone receptor beta agonist compounds |
EP3890748A4 (en) * | 2018-12-05 | 2022-12-21 | Viking Therapeutics, Inc. | Compositions and methods for the treatment of liver disorders |
US12102646B2 (en) | 2018-12-05 | 2024-10-01 | Viking Therapeutics, Inc. | Compositions for the treatment of fibrosis and inflammation |
US11034676B2 (en) | 2018-12-13 | 2021-06-15 | Terns, Inc. | THRB receptor agonist compound and preparation method and use thereof |
EP4574821A1 (en) | 2018-12-13 | 2025-06-25 | Terns, Inc. | Thrb receptor agonist compound and preparation method and use thereof |
US12338232B2 (en) | 2018-12-13 | 2025-06-24 | Terns, Inc. | THRβ receptor agonist compound and preparation method and use thereof |
WO2020123827A1 (en) | 2018-12-13 | 2020-06-18 | Terns, Inc. | Thrβ receptor agonist compound and preparation method and use thereof |
US11084802B2 (en) | 2018-12-13 | 2021-08-10 | Terns, Inc. | THRβ receptor agonist compound and preparation method and use thereof |
EP4529954A2 (en) | 2018-12-13 | 2025-04-02 | Terns, Inc. | Thrb receptor agonist compound and preparation method and use thereof |
CN112457346A (en) * | 2019-08-19 | 2021-03-09 | 苏州闻天医药科技有限公司 | Fused-ring THR beta receptor agonist compound and preparation method and application thereof |
GB2603330B (en) * | 2019-08-19 | 2023-08-09 | Hepagene Therapeutics Hk Ltd | Heterocyclic THR-B receptor agonist compound and preparation method and use therefor |
CN112457346B (en) * | 2019-08-19 | 2022-09-06 | 和博医药有限公司 | Fused-ring THR beta receptor agonist compound and preparation method and application thereof |
GB2603330A (en) * | 2019-08-19 | 2022-08-03 | Hepagene Therapeutics Hk Ltd | Heterocyclic THR-# receptor agonist compound and preparation method and use therefor |
WO2021032218A1 (en) | 2019-08-19 | 2021-02-25 | 苏州闻天医药科技有限公司 | HETEROCYCLIC THR-β RECEPTOR AGONIST COMPOUND AND PREPARATION METHOD AND USE THEREFOR |
AU2020333099B2 (en) * | 2019-08-21 | 2023-12-21 | Changchun Changcheng Pharmaceutical Technology Co., LTD | A prodrug platform useful to deliver amines, amides and phenols |
WO2021089692A1 (en) * | 2019-11-05 | 2021-05-14 | The University Of Bristol | Inhibitor of binding between podocin and keratin 8 for use in the treatment of nephrotic syndrome |
US12054507B2 (en) | 2020-02-18 | 2024-08-06 | Gilead Sciences, Inc. | Antiviral compounds |
US12264173B2 (en) | 2020-02-18 | 2025-04-01 | Gilead Sciences, Inc. | Antiviral compounds |
US11767337B2 (en) | 2020-02-18 | 2023-09-26 | Gilead Sciences, Inc. | Antiviral compounds |
US12030903B2 (en) | 2020-02-18 | 2024-07-09 | Gilead Sciences, Inc. | Antiviral compounds |
JP7670687B2 (en) | 2020-02-24 | 2025-04-30 | へパジーン セラピューティクス (エイチケイ) リミテッド | Heterocyclic thr-beta receptor agonist compounds and methods for their preparation and use - Patents.com |
US11752161B2 (en) | 2020-03-27 | 2023-09-12 | Gannex Pharma Co., Ltd. | Pharmaceutical compositions, method of making and method of using thereof |
US11697666B2 (en) | 2021-04-16 | 2023-07-11 | Gilead Sciences, Inc. | Methods of preparing carbanucleosides using amides |
US12116380B2 (en) | 2021-08-18 | 2024-10-15 | Gilead Sciences, Inc. | Phospholipid compounds and methods of making and using the same |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2011038207A1 (en) | Phosphorus-containing thyroid hormone receptor agonists and methods of use | |
US10925885B2 (en) | Thyromimetics for the treatment of fatty liver diseases | |
CA2606498C (en) | Novel phosphinic acid-containing thyromimetics | |
US7514419B2 (en) | Phosphorus-containing thyromimetics | |
AU2006249347A1 (en) | Novel phosphorus-containing thyromimetics | |
EP3445769A1 (en) | Thyroid hormone receptor agonist and use thereof | |
US20120046364A1 (en) | Novel Sulfonic Acid-Containing Thyromimetics, and Methods for Their Use | |
MXPA06004880A (en) | Novel phosphorus-containing thyromimetics | |
HK1119709A (en) | Novel phosphorus-containing thyromimetics | |
HK1119409A (en) | Novel phosphinic acid-containing thyromimetics |
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: 10761103 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: 10761103 Country of ref document: EP Kind code of ref document: A1 |