US20100168245A1 - Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure - Google Patents

Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure Download PDF

Info

Publication number
US20100168245A1
US20100168245A1 US12/376,945 US37694507A US2010168245A1 US 20100168245 A1 US20100168245 A1 US 20100168245A1 US 37694507 A US37694507 A US 37694507A US 2010168245 A1 US2010168245 A1 US 2010168245A1
Authority
US
United States
Prior art keywords
fenoterol
disorder
compound
pulmonary
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/376,945
Other languages
English (en)
Inventor
Irving W. Wainer
Weizhong Zhu
Khalid Chakir
Rui Ping Xiao
Darrell R. Abernethy
Farideh M. Beigi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US12/376,945 priority Critical patent/US20100168245A1/en
Assigned to THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEIGI, FARIDEH M., ABERNETHY, DARRELL R., XIAO, RUI PING, CHAKIR, KHALID, WAINER, IRVING W., ZHU, WEIZHONG
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEIGI, FARIDEH M., ABERNETHY, DARRELL R., XIAO, RUI PING, CHAKIR, KHALID, WAINER, IRVING W., ZHU, WEIZHONG
Publication of US20100168245A1 publication Critical patent/US20100168245A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/54Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C217/56Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains not further substituted by singly-bound oxygen atoms
    • C07C217/60Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains not further substituted by singly-bound oxygen atoms linked by carbon chains having two carbon atoms between the amino groups and the six-membered aromatic ring or the condensed ring system containing that ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/46Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C215/56Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains further substituted by hydroxy groups
    • C07C215/58Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains further substituted by hydroxy groups with hydroxy groups and the six-membered aromatic ring, or the condensed ring system containing that ring, bound to the same carbon atom of the carbon chain
    • C07C215/60Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains further substituted by hydroxy groups with hydroxy groups and the six-membered aromatic ring, or the condensed ring system containing that ring, bound to the same carbon atom of the carbon chain the chain having two carbon atoms between the amino groups and the six-membered aromatic ring or the condensed ring system containing that ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present disclosure relates to the field of pharmaceutical compositions and in particular, to the preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure.
  • Fenoterol 5-[1-hydroxy-2[[2-(4-hydroxyphenyl)-1-methylethyl]-amino]ethyl] 1,2-benzenediol, is a ⁇ 2-adrenergic receptor agonist that has traditionally been used for the treatment of pulmonary disorders such as asthma.
  • This drug has two chiral (asymmetric) carbons that can each be independently arranged in an R or S configuration, so that the drug exists in distinct (R,R), (R,S), (S,R) and (S,S) forms known as stereoisomers.
  • Fenoterol is commercially available as a racemic mixture of the (R,R)- and (S,S)-compounds.
  • Fenoterol is known to act as an agonist that binds to and activates the ⁇ 2-adrenergic receptor. This activity has led to its clinical use in the treatment of asthma because this agonist's activity dilates constricted airways. Additional therapeutic uses for fenoterol remain to be thoroughly explored. Pharmacological studies of the class of drugs that includes fenoterol have shown that only one of the enantiomers is responsible for generating brochodilation. For example, studies have demonstrated that the primary bronchodilatory activity for racemic ( ⁇ )-fenoterol resides in the (R,R)-isomer of fenoterol. Further, it has recently become apparent that the inactive enantiomer may be associated with adverse effects. For instance, the diastereomer (S,S)-fenoterol has been demonstrated to cause adverse side effects or development of tolerance often associated with ⁇ 2-adrenergic receptor agonist treatment.
  • fenoterol compositions that were effective at treating disorders such as asthma, chronic obstructive pulmonary disease, or congestive heart failure, but had reduced side effects such as hypersensitivity and drug resistance (tolerance).
  • R 1 -R 3 independently are hydrogen, acyl, alkoxy carbonyl, amino carbonyl (carbamoyl) or a combination thereof;
  • R 4 is H or lower alkyl
  • R 5 is lower alkyl
  • X, Y 1 , Y 2 and Y 3 independently are hydrogen, —OR 6 and —NR 7 R 8 ;
  • R 6 is independently hydrogen, lower alkyl, acyl, alkoxy carbonyl or amino carbonyl
  • R 7 and R 8 independently are hydrogen, lower alkyl, alkoxy carbonyl, acyl or amino carbonyl.
  • compositions containing, and methods of using (R,R)-fenoterol and fenoterol analogues are also provided.
  • the disclosed (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues e.g., (R,R)-methoxyfenoterol, (R,R)-napthylfenoterol, and (R,S)-napthylfenoterol
  • FIG. 1 illustrates the chromatographic separation of (S,S)- and (R,R)-fenoterol.
  • FIG. 2A provides an ultraviolet spectra of (R,R)- and (S,S)-fenoterol.
  • FIG. 2B illustrates a circular dichroism spectra of (R,R)- and (S,S)-fenoterol.
  • FIG. 3 provides frontal chromatographic elution profiles of [ + H]-( ⁇ )-fenoterol produced by the addition of (R,R)-fenoterol to the running buffer.
  • FIG. 4A is a graph including dose-response curves generated by treating freshly isolated rat ventricular myocytes with ( ⁇ )-fenoterol, (R,R)-fenoterol or (S,S)-fenoterol.
  • FIG. 4B is a graph including dose-response curves of T 50% relaxation in freshly isolated rat ventricular myocytes generated by treatment with ( ⁇ )-fenoterol, (R,R)-fenoterol or (S,S)-fenoterol.
  • FIG. 5 illustrates the chemical structures of the stereoisomers of fenoterol and fenoterol analogs (compounds 2-7).
  • FIG. 6 illustrates the chemical structures of compounds 47-51.
  • FIG. 7 illustrates the effect of fenoterol and fenoterol analogs on cell contraction in single ventricular myocytes.
  • FIG. 8 is a graph illustrating time-dependent mean plasma concentration of (R,R)-fenoterol, (R,R)-methoxyfenoterol and (R,S)-napthylfenoterol after administration (5 mg/mL).
  • the optically active fenoterol analogues are substantially purified from a racemic mixture.
  • an optically active fenoterol analogue is purified to represent greater than 90%, often greater than 95% of the composition.
  • These analogues can be used to treat pulmonary disorders such as asthma and chronic obstructive pulmonary disease that have previously been treated with ( ⁇ )-fenoterol.
  • Use of the disclosed fenoterol analogues with an equal to higher efficacy than ( ⁇ )-fenoterol can possibly reduce adverse effects previously observed with ( ⁇ )-fenoterol.
  • fenoterol analogues use of a lower concentration of the fenoterol analogues to obtain a therapeutic effective response is expected to reduce side-effects such as hypersensitivity and drug resistance (tolerance) observed with the commercially available ( ⁇ )-fenoterol. Also, purification of the analogues removes contaminants such as the inactive enantiomer which can be responsible for these adverse effects.
  • (R,R)-fenoterol is the active component of commercially available ( ⁇ )-fenoterol. It is specifically contemplated that (R,R)-fenoterol as well as disclosed (R,R)- and (R,S)-fenoterol analogues can be used to treat cardiac disorders such as congestive heart failure. Use of substantially optically pure (R,R)-fenoterol or (R,R)- or (R,S)-fenoterol analogues to treat congestive heart failure is expected to reduce the incidence of side-effects caused by physiologically less active forms of the drug.
  • AR adrenergic receptor
  • IAM-PC immobilized artificial membrane chromatographic support
  • UV ultraviolet
  • any quantitative values are approximate whether the word “about” or “approximately” or the like are stated or not.
  • the materials, methods, and examples described herein are illustrative only and not intended to be limiting. Any molecular weight or molecular mass values are approximate and are provided only for description. Except as otherwise noted, the methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Loudon, Organic Chemistry , Fourth Edition, New York: Oxford University Press, 2002, pp.
  • Acyl A group of the formula RC(O)— wherein R is an organic group.
  • Acyloxy A group having the structure —OC(O)R, where R may be an optionally substituted alkyl or optionally substituted aryl. “Lower acyloxy” groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms.
  • Alkoxy A radical (or substituent) having the structure —O—R, where R is a substituted or unsubstituted. alkyl. Methoxy (—OCH 3 ) is an exemplary alkoxy group. In a substituted alkoxy, R is alkyl substituted with a non-interfering substituent. “Thioalkoxy” refers to —S—R, where R is substituted or unsubstituted alkyl. “Haloalkyloxy” means a radical —OR where R is a haloalkyl.
  • Alkoxy carbonyl A group of the formula —C(O)OR, where R may be an optionally substituted alkyl or optionally substituted aryl. “Lower alkoxy carbonyl” groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms.
  • Alkyl An acyclic, saturated, branched- or straight-chain hydrocarbon radical, which, unless expressly stated otherwise, contains from one to fifteen carbon atoms; for example, from one to ten, from one to six, or from one to four carbon atoms. This term includes, for example, groups such as methyl, ethyl, n-propyl, isopropyl, isobutyl, t-butyl, pentyl, heptyl, octyl, nonyl, decyl, or dodecyl.
  • the term “lower alkyl” refers to an alkyl group containing from one to ten carbon atoms.
  • alkyl groups can either be unsubstituted or substituted.
  • An alkyl group can be substituted with one or more substituents (for example, up to two substituents for each methylene carbon in an alkyl chain).
  • substituents include, for instance, amino groups, amide, sulfonamide, halogen, cyano, carboxy, hydroxy, mercapto, trifluoromethyl, alkyl, alkoxy (such as methoxy), alkylthio, thioalkoxy, arylalkyl, heteroaryl, alkylamino, dialkylamino, alkylsulfano, keto, or other functionality.
  • Amino carbonyl (carbamoyl): A group of the formula —OCN(R)R′—, wherein R and R′ are independently of each other hydrogen or a lower alkyl group.
  • Asthma is a disease of the respiratory system in which the airways constrict, become inflamed, and are lined with excessive amounts of mucus, often in response to one or more “triggers,” such as exposure to an environmental stimulant (or allergen), cold air, exercise, or emotional stress.
  • This airway narrowing causes symptoms such as wheezing, shortness of breath, chest tightness, and coughing.
  • the disorder is a chronic or recurring inflammatory condition in which the airways develop increased responsiveness to the various stimuli, characterized by bronchial hyper-responsiveness, inflammation, increased mucus production, and intermittent airway obstruction.
  • Carbamate A group of the formula —OC(O)N(R), wherein R is H, or an aliphatic group, such as a lower alkyl group or an aralkyl group
  • Cardiac Disorder or Disease In general, a cardiac disorder/disease is a class of disorders/diseases that involve the heart and/or blood vessels (arteries and veins). In a particular example, cardiac disorder/disease includes congestive heart failure.
  • Chronic Obstructive Pulmonary Disease A group of respiratory tract diseases including chronic bronchitis, emphysema and bronchiectasis that are characterized by airflow obstruction or limitation that is not fully reversible.
  • the airflow limitation is usually progressive and associated with an abnormal inflammatory response of the lungs to noxious particles or gases.
  • Congestive Heart Failure Heart failure in which the heart is unable to maintain an adequate circulation of blood in the bodily tissues or to pump out the venous blood returned to it by the veins.
  • a derivative such as a fenoterol analogue
  • a biological activity such as ⁇ 2-adrenergic receptor stimulation
  • a molecule from which it was derived such as a fenoterol
  • Isomers Compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed “isomers”. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that are not mirror images of one another are termed “diastereomers” and those that are non-superimposable mirror images of each other are termed “enantiomers.” When a compound has an asymmetric center, for example, if a carbon atom is bonded to four different groups, a pair of enantiomers is possible.
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or ( ⁇ ) isomers, respectively).
  • a chiral compound can exist as either an individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a “racemic mixture.”
  • the compounds described herein may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)-stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof.
  • the methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see, e.g., March, Advanced Organic Chemistry, 4th edition, New York: John Wiley and Sons, 1992, Chapter 4).
  • parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like
  • solid compositions for example, powder, pill, tablet, or capsule forms
  • conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • non-toxic auxiliary substances such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • Phenyl groups may be unsubstituted or substituted with one, two or three substituents, with substituent(s) independently selected from alkyl, heteroalkyl, aliphatic, heteroaliphatic, thioalkoxy, halo, haloalkyl (such as —CF 3 ), nitro, cyano, —OR (where R is hydrogen or alkyl), —N(R)R′ (where R and R′ are independently of each other hydrogen or alkyl), —COOR (where R is hydrogen or alkyl) or —C(O)N(R′)R′′ (where R′ and R′′ are independently selected from hydrogen or alkyl).
  • pulmonary disorder/disease includes any disorder/disease pertaining to the lungs.
  • pulmonary disorder/disease includes asthma and chronic obstructive pulmonary disease.
  • purified does not require absolute purity; rather, it is intended as a relative term.
  • a purified preparation is one in which a desired component such as an (R,R)-enantiomer of fenoterol is more enriched than it was in a preceding environment such as in a ( ⁇ )-fenoterol mixture.
  • a desired component such as (R,R)-enantiomer of fenoterol is considered to be purified, for example, when at least about 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% of a sample by weight is composed of the desired component.
  • Purity of a compound may be determined, for example, by high performance liquid chromatography (HPLC) or other conventional methods.
  • HPLC high performance liquid chromatography
  • the specific fenoterol analogue enantiomers are purified to represent greater than 90%, often greater than 95% of the other enantiomers present in a purified preparation.
  • the purified preparation may be essentially homogeneous, wherein other stereoisomers are less than 1%.
  • a compound described herein may be obtained in a purified form or purified by any of the means known in the art, including silica gel and/or alumina chromatography. See, e.g., Introduction to Modern Liquid Chromatography, 2nd Edition, ed. by Snyder and Kirkland, New York: John Wiley and Sons, 1979; and Thin Layer Chromatography , ed. by Stahl, New York: Springer Verlag, 1969.
  • a compound includes purified fenoterol or fenoterol analogue with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% of a sample by weight relative to other contaminants.
  • a compound includes at least two purified stereoisomers each with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% of a sample by weight relative to other contaminants.
  • a compound can include a substantially purified (R,R)-fenoterol analogue and a substantially purified (R,S)-fenoterol analogue.
  • subject includes both human and veterinary subjects, for example, humans, non-human primates, dogs, cats, horses, rats, mice, and cows.
  • mammal includes both human and non-human mammals.
  • Treating or treatment With respect to disease, either term includes (1) preventing the disease, e.g., causing the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease, (2) inhibiting the disease, e.g., arresting the development of the disease or its clinical symptoms, or (3) relieving the disease, e.g., causing regression of the disease or its clinical symptoms.
  • Therapeutically Effective Amount A quantity of a specified agent sufficient to achieve a desired effect in a subject being treated with that agent. For example, this may be the amount of (R,R)-fenoterol or (R,R)- or (R,S)-fenoterol analogue useful in preventing, reducing, and/or inhibiting, and/or treating a cardiac disorder such as congestive heart failure.
  • a therapeutically effective amount of an agent is an amount sufficient to prevent, reduce, and/or inhibit, and/or treat the disorder in a subject without causing a substantial cytotoxic effect in the subject.
  • the effective amount of an agent useful for preventing, reducing, and/or inhibiting, and/or treating a disorder in a subject will be dependent on the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition.
  • R 1 -R 3 independently are hydrogen, acyl, alkoxy carbonyl, amino carbonyl or a combination thereof;
  • R 4 is H or lower alkyl
  • R 5 is lower alkyl
  • X and Y independently are selected from hydrogen, lower —OR 6 and —NR 7 R 8 ;
  • R 6 is lower alkyl or acyl
  • R 7 and R 8 independently are hydrogen, lower alkyl, alkoxy carbonyl, acyl or amino carbonyl.
  • Y may be —OH
  • R 5 is a 1- or 2-napthyl derivative optionally having 1, 2 or 3 substituents. Examples of such R 5 groups are represented by the formula
  • Y 1 , Y 2 and Y 3 independently are hydrogen, lower —OR 6 and —NR 7 R 8 ;
  • R 6 is independently for each occurrence selected from lower alkyl and acyl
  • R 7 and R 8 independently are hydrogen, lower alkyl, alkoxy carbonyl, acyl or amino carbonyl (carbamoyl).
  • R 7 and R 8 independently are hydrogen, lower alkyl, alkoxy carbonyl, acyl or amino carbonyl (carbamoyl).
  • compounds at least one of Y 1 , Y 2 and Y 3 is —OCH 3 .
  • Particular R 5 groups include those represented by the formulas
  • R 6 is lower alkyl, such as methyl, ethyl, propyl or isopropyl or acyl, such as acetyl.
  • R 5 groups include
  • R 4 is lower alkyl and R 5 is
  • X and Y independently are selected from H, lower alkyl —OR 6 and —NR 7 R 8 ;
  • R 6 is lower alkyl
  • R 7 and R 8 independently are hydrogen or lower alkyl.
  • R 4 is selected from ethyl, n-propyl, and isopropyl and R 5 has the formula
  • X is H, —OR 6 or —NR 7 R 8 .
  • R 6 may be methyl or R 7 and R 8 are hydrogen.
  • R 5 has the formula
  • R 4 is selected from methyl, ethyl, n-propyl and isopropyl and R 5 represents
  • R 1 -R 3 examples include, without limitation, acyl, acyloxy and alkoxy carbonyl groups.
  • Compounds having such cleavable groups are referred to as “prodrugs.”
  • the term “prodrug,” as used herein, means a compound which includes a substituent that is convertible in vivo (e.g., by hydrolysis) to a hydroxyl group.
  • Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs , Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology , Vol.
  • An exemplary (R,R)-compound has the chemical structure of:
  • solvates such as hydrates
  • pharmaceutically acceptable salts and/or different physical forms of (R,R)-fenoterol or any of the fenoterol analogues herein described.
  • Solvate means a physical association of a compound with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including by way of example covalent adducts and hydrogen bonded solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolable solvates. Representative solvates include ethanol associated compound, methanol associated compounds, and the like. “Hydrate” is a solvate wherein the solvent molecule(s) is/are H 2 O.
  • the disclosed compounds also encompass salts including, if several salt-forming groups are present, mixed salts and/or internal salts.
  • the salts are generally pharmaceutically-acceptable salts that are non-toxic. Salts may be of any type (both organic and inorganic), such as fumarates, hydrobromides, hydrochlorides, sulfates and phosphates. In an example, salts include non-metals (e.g., halogens) that form group VII in the periodic table of elements. For example, compounds may be provided as a hydrobromide salt.
  • salt-forming groups include, but are not limited to, a carboxyl group, a phosphonic acid group or a boronic acid group, that can form salts with suitable bases.
  • These salts can include, for example, nontoxic metal cations which are derived from metals of groups IA, IB, IIA and IIB of the periodic table of the elements.
  • alkali metal cations such as lithium, sodium or potassium ions, or alkaline earth metal cations such as magnesium or calcium ions can be used.
  • the salt can also be a zinc or an ammonium cation.
  • the salt can also be formed with suitable organic amines, such as unsubstituted or hydroxyl-substituted mono-, di- or tri-alkylamines, in particular mono-, di- or tri-alkylamines, or with quaternary ammonium compounds, for example with N-methyl-N-ethylamine, diethylamine, triethylamine, mono-, bis- or tris-(2-hydroxy-lower alkyl)amines, such as mono-, bis- or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine or tris(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxy-lower alkyl)amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine, or N-methyl-D-glucamine, or quaternary ammonium compounds such as tetrabutylam
  • Exemplary compounds disclosed herein possess at least one basic group that can form acid-base salts with inorganic acids.
  • basic groups include, but are not limited to, an amino group or imino group.
  • inorganic acids that can form salts with such basic groups include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid.
  • Basic groups also can form salts with organic carboxylic acids, sulfonic acids, sulfo acids or phospho acids or N-substituted sulfamic acid, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2-phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid, and, in addition, with amino acids, for example with ⁇ -amino acids, and also with methanesulfonic acid, ethanesulfonic acid, 2-hydroxymethanesulfonic acid, ethane-1,2-disulfonic acid
  • fenoterol is provided as a hydrobromide salt and exemplary fenoterol analogues are provided as their fumarate salts.
  • employing a pharmaceutically acceptable salt may also serve to adjust the osmotic pressure of a composition.
  • the compounds used in the method are provided are polymorphous.
  • the compounds can be provided in two or more physical forms, such as different crystal forms, crystalline, liquid crystalline or non-crystalline (amorphous) forms.
  • any of the above described compounds e.g., (R,R)-fenoterol or fenoterol analogues or a hydrate or pharmaceutically acceptable salt thereof
  • a medicament for ⁇ 2-adrenergic receptor stimulation in a subject or for the treatment of pulmonary and cardiac disorders e.g., asthma and congestive heart failure.
  • Formulations suitable for such medicaments, subjects who may benefit from same and other related features are described elsewhere herein.
  • the disclosed fenoterol analogues can be synthesized by any method known in the art. Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure , Fifth Edition, Wiley-Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis , Fourth Edition, New York: Longman, 1978).
  • Compounds as described herein may be purified by any of the means known in the art, including chromatographic means, such as HPLC, preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. Most typically the disclosed compounds are purified via open column chromatography or prep chromatography.
  • Scheme III describes an exemplary synthesis for the chiral building blocks used in Scheme II.
  • the (R)- and (S)-3′,5′-dibenzyloxyphenyl-bromohydrin enantiomers were obtained by the enantiospecific reduction of 3,5-dibenzyloxy- ⁇ -bromoacetophenone using boron-methyl sulfide complex (BH 3 SCH 3 ) and either (1R,2S)- or (1S,2R)-cis-1-amino-2-indanol.
  • BH 3 SCH 3 boron-methyl sulfide complex
  • the disclosed (R,R)-fenoterol and fenoterol analogues can be useful, at least, for the treatment of pulmonary disorders such as asthma and chronic obstructive pulmonary disease (COPD) and cardiac disorders such as congestive heart failure. Accordingly, pharmaceutical compositions comprising at least one disclosed fenoterol compound or analogue are also described herein.
  • Formulations for pharmaceutical compositions are well known in the art. For example, Remington's Pharmaceutical Sciences , by E. W. Martin, Mack Publishing Co., Easton, Pa., 19th Edition, 1995, describes exemplary formulations (and components thereof) suitable for pharmaceutical delivery of (R,R)-fenoterol and disclosed fenoterol analogues. Pharmaceutical compositions comprising at least one of these compounds can be formulated for use in human or veterinary medicine. Particular formulations of a disclosed pharmaceutical composition may depend, for example, on the mode of administration (e.g., oral or parenteral) and/or on the disorder to be treated (e.g., pulmonary disorder or cardiac disorder such as congestive heart failure). In some embodiments, formulations include a pharmaceutically acceptable carrier in addition to at least one active ingredient, such as a fenoterol compound.
  • a pharmaceutically acceptable carrier in addition to at least one active ingredient, such as a fenoterol compound.
  • compositions useful for the disclosed methods and compositions are conventional in the art.
  • the nature of a pharmaceutical carrier will depend on the particular mode of administration being employed.
  • parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • solid compositions such as powder, pill, tablet, or capsule forms conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • compositions to be administered can optionally contain minor amounts of non-toxic auxiliary substances or excipients, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like; for example, sodium acetate or sorbitan monolaurate.
  • auxiliary substances or excipients such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like; for example, sodium acetate or sorbitan monolaurate.
  • excipients include, nonionic solubilizers, such as cremophor, or proteins, such as human serum albumin or plasma preparations.
  • compositions may be formulated as a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts are non-toxic salts of a free base form of a compound that possesses the desired pharmacological activity of the free base. These salts may be derived from inorganic or organic acids. Non-limiting examples of suitable inorganic acids are hydrochloric acid, nitric acid, hydrobromic acid, sulfuric acid, hydriodic acid, and phosphoric acid.
  • Non-limiting examples of suitable organic acids are acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, methyl sulfonic acid, salicylic acid, formic acid, trichloroacetic acid, trifluoroacetic acid, gluconic acid, asparagic acid, aspartic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenesulfonic acid, and the like. Lists of other suitable pharmaceutically acceptable salts are found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Company, Easton, Pa., 1995. A pharmaceutically acceptable salt may also serve to adjust the
  • the dosage form of a disclosed pharmaceutical composition will be determined by the mode of administration chosen.
  • oral dosage forms may be employed.
  • Oral formulations may be liquid such as syrups, solutions or suspensions or solid such as powders, pills, tablets, or capsules. Methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art.
  • compositions comprising a disclosed compound may be formulated in unit dosage form suitable for individual administration of precise dosages.
  • the amount of active ingredient such as (R,R)-fenoterol administered will depend on the subject being treated, the severity of the disorder, and the manner of administration, and is known to those skilled in the art.
  • the formulation to be administered will contain a quantity of the extracts or compounds disclosed herein in an amount effective to achieve the desired effect in the subject being treated.
  • the compositions are provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.0 mg, about 2.5 mg, 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg (such as about 2 mg to about 10 mg) active ingredient is administered two to four times a day, such as two times, three times or four times.
  • the present disclosure includes methods of treating disorders including pulmonary and cardiac disorders.
  • the pulmonary disorder is asthma or chronic obstructive pulmonary disease.
  • the cardiac disorder is congestive heart failure.
  • Disclosed methods includes administering (R,R)-fenoterol or a disclosed fenoterol analogue (and, optionally, one or more other pharmaceutical agents) to a subject in a pharmaceutically acceptable carrier and in an amount effective to treat the pulmonary and/or cardiac disorder.
  • Routes of administration useful in the disclosed methods include but are not limited to oral and parenteral routes, such as intravenous (iv), intraperitoneal (ip), rectal, topical, ophthalmic, nasal, and transdermal. Formulations for these dosage forms are described above.
  • an effective amount of (R,R)-fenoterol or a disclosed fenoterol analogue will depend, at least, on the particular method of use, the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition.
  • a “therapeutically effective amount” of a composition is a quantity of a specified compound sufficient to achieve a desired effect in a subject being treated. For example, this may be the amount of a (R,R)-fenoterol necessary to prevent, inhibit, reduce or relieve the pulmonary and/or cardiac disorder and/or one or more symptoms of disorder in a subject.
  • a therapeutically effective amount of (R,R)-fenoterol or a disclosed fenoterol analogue is an amount sufficient to prevent, inhibit, reduce or relieve the pulmonary and/or cardiac disorder and/or one or more symptoms of the disorder without causing a substantial cytotoxic effect on host cells.
  • Therapeutically effective doses of a disclosed fenoterol compound or pharmaceutical composition can be determined by one of skill in the art, with a goal of achieving concentrations that are at least as high as the EC 50 of the applicable compound disclosed in the examples herein.
  • An example of a dosage range is from about 0.001 to about 10 mg/kg body weight orally in single or divided doses.
  • a dosage range is from about 0.005 to about 5 mg/kg body weight orally in single or divided doses (assuming an average body weight of approximately 70 kg; values adjusted accordingly for persons weighing more or less than average).
  • compositions are, for example, provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.5 mg, about 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg active ingredient is administered two to four times a day, such as two times, three times or four times.
  • the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound, the metabolic stability and length of action of that compound, the age, body weight, general health, sex and diet of the subject, mode and time of administration, rate of excretion, drug combination, and severity of the condition of the subject undergoing therapy.
  • Phenylmethylsulfonyl fluoride (PMSF), benzamidine, leupeptin, pepstatin A, MgCl 2 , EDTA, Trizma-Hydrochloride (Tris-HCl), ( ⁇ )-propranolol and minimal essential medium (MEM) were obtained from Sigma Aldrich (St. Louis, Mo.).
  • Egg phosphatidylcholine lipids (PC) were obtained from Avanti Polar Lipids (Alabaster, Ala.).
  • ( ⁇ )-fenoterol was purchased from Sigma Aldrich and [ 3 H]-( ⁇ )-fenoterol was acquired from Amersham Biosciences (Boston, Mass.).
  • the organic solvents n-hexane, 2-propanol and triethylamine were obtained as ultra pure HPLC grade solvents from Carlo Erba (Milan, Italy).
  • Fetal bovine serum and penicillin-streptomycin were purchased from Life Technologies (Gaithersburg, Md.), and [ 125 I]-( ⁇ )-iodocyanopindolol (ICYP) was purchased from NEN Life Science Products, Inc. (Boston, Mass.).
  • a Rheodyne model 7125 injector with 20 ⁇ l sample loop was used to inject 0.2-0.3 mg ( ⁇ )-fenoterol onto the chromatographic system.
  • the mobile phase was n-hexane/2-propanol (88/12 v/v) with 0.1% triethylamine, the flow rate was 1 mL/minute and the temperature of the system was maintained at 25° C.
  • the stereochemical configurations of the resolved (R,R)-fenoterol and (S,S)-fenoterol were established using circular dichroism (CD) measurements obtained with a JASCO® J-800 spectropolarimeter.
  • CD circular dichroism
  • the (R,R)-fenoterol and (S,S)-fenoterol were dissolved in 2-propanol and the measurements were obtained using 1 cm path length at room temperature.
  • the liquid chromatography column containing the immobilized ⁇ 2-AR was prepared using a previously described technique (Beigi et al., Anal. Chem., 76: 7187-7193, 2004). In brief, cellular membranes were obtained from a HEK 293 cell line that had been transfected with cDNA encoding human ⁇ 2-AR. An aliquot of a cell pellet suspension corresponding to 5-7 mg total protein, as determined by the micro BCA method, was used to create the column.
  • the membranes were prepared in 10 mL buffer composed of Tris-HCl [50 mM, pH 7.4] containing MgCl 2 (2 mM), benzamidine (1 mM), leupeptin (0.03 mM) pepstatin A (0.005 mM) and EDTA (1 mM).
  • IAM-PC immobilized artificial membrane chromatographic support
  • 80 ⁇ M PC 80 ⁇ M PC were added to the membrane preparation and the resulting mixture was stirred at room temperature for 3 hours, transferred into (5 cm length) nitrocellulose dialysis membrane (MW cutoff 10,000 Da, Pierce Chemical, Rockford, Ill.) and placed in 1 L of dialysis buffer composed of Tris-HCl [50 mM, pH 7.4] containing EDTA (1 mM), MgCl 2 (2 mM), NaCl (300 mM) and PMSF (0.2 mM) at 4° C. for 24 hours. The dialysis step was repeated twice using fresh buffer.
  • the mixture was centrifuged at 120 ⁇ g for 3 minutes, the supernatant was discarded and the pellet of IAM support containing the immobilized receptor-bearing membranes was collected.
  • the pellet was resuspended in 2 mL chromatographic running buffer, composed of Tris-HCl [10 mM, pH 7.4] containing EDTA (1 mM) and MgCl 2 (2 mM) and the suspension was pumped through a HR 5/2 chromatographic glass column (Amersham Pharmacia Biotech, Uppsala, Sweden) at a flow rate of 0.3 mL/minutes using a peristaltic pump.
  • the end adaptors were assembled producing a total gel-bed volume of 0.4 mL.
  • the column was stored at 4° C. when not in use.
  • the column containing the immobilized ⁇ 2-AR stationary phase was placed in a chromatographic system composed of a HPLC pump (10-AD, Shimadzu Inc., Columbia, Md.), a manually controlled FPLC injector (Amersham Biotechnology, Uppsala, Sweden) with a 50 ⁇ L sample loop, the packed immobilized receptor column and an on-line radioactive flow detector (IN/US, Tampa, Fla.), all connected sequentially.
  • HPLC pump 10-AD, Shimadzu Inc., Columbia, Md.
  • FPLC injector Analogenor
  • sample loop 50 ⁇ L sample loop
  • the packed immobilized receptor column packed immobilized receptor column
  • an on-line radioactive flow detector I/US, Tampa, Fla.
  • the running buffer was composed of Tris-HCl [10 mM, pH 7.4] containing EDTA (1 mM) and MgCl 2 (2 mM) and 0.05 nM [ 3 H]-( ⁇ )-fenoterol, the marker ligand.
  • (R,R)-fenoterol or (S,S)-fenoterol was added to the running buffer in sequential concentrations of 0.1, 80.0, 240, and 700 nM, and applied to the column.
  • the immobilized receptor column was equilibrated with about 80 mL of running buffer, without the added (R,R)-fenoterol or (S,S)-fenoterol respectively, in between each sample injection. All chromatographic studies were carried out at room temperature at a flow rate of 0.2 mL/minutes.
  • V i is the solute elution volume
  • V min is the elution volume at the saturation point
  • P is the number of available binding sites
  • M is the concentration of the marker ligand
  • K d is the dissociation constant of the ligand.
  • HEK293 cells were harvested in lysis buffer, Tris-HCl [5 mM, pH 7.4] containing EGTA [5 mM], and homogenized with 15 strokes on ice. Samples were centrifuged at 30,000 ⁇ g for 15 minutes to pellet membranes. Membranes were resuspended in binding buffer, Tris-HCl [20 mM, pH 7.4] containing NaCl (120 mM), KCl (5.4 mM), CaCl 2 (1.8 mM), MgCl 2 (0.8 mM), and glucose (5 mM) and stored in aliquots at ⁇ 80° C.
  • Binding assays were performed on 5-10 ⁇ g of membrane protein using saturating amounts (1-300 pM) of the ⁇ -AR-specific ligand [ 125 I]cyanopindolol (ICYP).
  • ICYP 125 I]cyanopindolol
  • the 5-10 ⁇ g of membrane protein were pretreated with 50 ⁇ M of GTP ⁇ s (non-hydrolyzable guanosine triphosphate) and then incubated with 125 ICYP (50 pM) and different concentrations of fenoterol or its isomers in a total volume of 250 ⁇ L.
  • Nonspecific binding was determined in the presence of 20 ⁇ M propranolol. Reactions were conducted in 250 ⁇ L of binding buffer at 37° C. for 1 hour.
  • the binding reaction was terminated by addition of ice-cold Tris-HCl [10 mM, pH 7.4] to the membrane suspension, followed by rapid vacuum filtration through glass-fiber filters (Whatman GF/C). Each filter was washed three times with an additional 7 mL of ice-cold Tris-HCl [10 mM, pH 7.4].
  • the radioactivity of the wet filters was determined in a gamma counter. All assays were performed in duplicate, and receptor density was normalized to milligrams of membrane protein. K d and the maximal number of binding sites (B max ) for ICYP were determined by Scatchard analysis of saturation binding isotherms. Data from competition studies were analyzed using 1- or 2-site competition binding curves with GRAPHPAD PRISM® Software (GRAPHPAD PRISM is a registered trademark of GraphPad Software, Inc., San Diego, Calif.).
  • the assignment of the absolute configuration of the isolated fractions was accomplished using their chiroptical properties.
  • the ultraviolet (UV) spectra of both fractions contained identical maxima at about 280 and 230, indicating the same UV chromophores for the two enantiomers.
  • the circular dichroism (CD) spectrum shows, for the less retained enantiomeric fraction, negative CD bands at about 280 and 215 nm, while the spectrum is positive at 230 and 200 nm. The sign of the CD bands is reversed for the most retained fenoterol fraction, this confirming the enantiomeric nature of the two fractions.
  • the lowest energy UV and CD spectra of the two enantiomeric fractions are presented in FIGS. 2A and 2B , respectively.
  • the sign of the lowest energy CD band can be used to assign the absolute configuration to the separated fenoterol enantiomers, by applying the Brewster-Buta/Smith-Fontana sector rule for chiral benzylic derivatives (Brewster and Buta, J. Am. Chem. Soc., 88: 2233-2240, 1996).
  • This sector rule is used to predict the sign of the CD band related to the 1 L b electronic transition of the benzylic compounds, with either hydroxyl or amine moieties and has been primarily applied to conformationally mobile aromatic compounds containing a single stereogenic center. In the case of fenoterol, there are two stereogenic centers.
  • the immobilized ⁇ 2-AR column was equilibrated with the running buffer containing the [ 3 H]-fenoterol, the marker ligand, before the initiation of the displacement studies. It was assumed that the binding data calculated using frontal displacement chromatography reflects the binding of (R,R)-fenoterol and (S,S)-fenoterol to the active state of the receptor. In frontal chromatography, the initial flat portion of the chromatographic trace represents the binding of a marker ligand that is specific for the immobilized target, in this study the ⁇ 2-AR, as well as non-specific binding to other sites on the immobilized membrane fragments. Saturation of specific binding sites produces a breakthrough front followed by a plateau representing the establishment of a new equilibrium.
  • the isolated cells were resuspended in HEPES buffer solution [20 mM, pH 7.4] containing, NaCl (137 mM), KCl (5.4 mM), MgCl 2 (1.2 mM), NaH 2 PO 4 (1.0 mM), CaCl 2 (1.0 mM), and glucose (20 mM). All studies were performed within 8 hours of cell isolation.
  • the cells were placed on the stage of an inverted microscope (Zeiss model IM-35, Zeiss, Thornwood, N.Y.), perfused with the HEPES-buffered solution at a flow rate of 1.8 mL/minutes, and electrically stimulated at 0.5 Hz at 23° C.
  • Cell length was monitored by an optical edge-tracking method using a photodiode array (Model 1024 SAQ, Reticon, Boston, Mass.) with a 3 ms time resolution. Cell contraction was measured by the percent shortening of cell length following electrical stimulation.
  • NMR multiplicities were reported by using the following abbreviations: s, singlet; d, doublet; t, triplet; q, quartet; p, pentet; m, multiplet; apt., apparent; and br, broad.
  • Low resolution mass spectra were obtained on a Finnigan LCQ Duo LC MS/MS atmospheric pressure chemical ionization (API) quadrupole ion trap MS system equipped with both electrospray (ESI) and atmospheric pressure chemical ionization (APCI) probes.
  • Analytical HPLC data was obtained using a Waters 2690 Separations Module with PDA detection.
  • 3′,5′-Dibenzyloxy- ⁇ -bromoacetophenone 46.
  • a solution of 2.4 mL (46 mmol) of Br 2 in 45 mL of CHCl 3 was added dropwise over 1 h to a chilled, stirring solution of 9.66 g (29 mmol) of 3′,5′-dibenzyloxyacetophenone (45) in 40 mL of CHCl 3 .
  • the resulting solution was allowed to warm to room temperature over 1 hour with good stirring, then poured into 100 mL of cold H 2 O and transferred to a separatory funnel where the CHCl 3 fraction was isolated, washed with brine solution, dried (Na 2 SO 4 ), filtered, and concentrated to 10.8 g.
  • Solvents were removed in vacuo and the resulting residue taken up in 30 mL of CHCl 3 and washed with 25 mL of 0.2 M sulfuric acid followed by 20 mL of brine, then dried (Na 2 SO 4 ), filtered, and evaporated.
  • 4-Benzyloxyphenylacetone (34). To 10.0 g (41.3 mmol) of 4-benzyloxyphenylacetic acid (31) was added, 20 mL of acetic anhydride and 20 mL of pyridine, which was heated to reflux with stirring under argon atmosphere for 6 hours. Solvents were evaporated and residue dissolved in CHCl 3 (50 mL) and washed with 1N NaOH (2 ⁇ 50 mL). Dried organic layer (MgSO 4 ), filtered, and evaporated to 11.8 g of an amber oil. Vacuum distillation at 0.1 mm Hg in an oil-bath set to 170° C.
  • Phenylacetone 35.
  • a solution of 20.4 g (0.15 mol) of phenylacetic acid, acetic anhydride (70 mL) and pyridine (70 mL) was heated to reflux with stirring under argon atmosphere for 6 hours.
  • Solvents were evaporated and residue dissolved in CHCl 3 (100 mL), washed with 1N NaOH (2 ⁇ 100 mL) and dried the organic layer (MgSO 4 ), filtered, and evaporated to give 20.4 g.
  • Vacuum distillation at 0.1 mm Hg in an oil bath set to 160° C., followed by silica gel chromatography eluting with 1/1 hexanes/CH 2 Cl 2 gave 5.5 g (27%).
  • 1 H (CDCl 3 ) 2.15 (s, 3H), 3.70 (s, 2H), 7.20-7.36 (m, 5H).
  • reaction mixture was diluted with 30 mL of CH 2 Cl 2 , cooled in an ice bath and 80 mL of 5% NaOH (in water) was added. Fractions were separated, organics (Na 2 SO 4 ) dried and evaporated to 638 mg (71%) of (R)-15.
  • the mixture was filtered through Celite, the filter cake rinsed with isopropanol, and the filtrate concentrated.
  • the reaction was cooled and the solvent removed.
  • the crude material was purified by open column chromatograph or preparative chromatograph.
  • HPLC 1 Column: Varian Sunfire C18 100 ⁇ 4.6; 70/30/0.1 water/acetonitrile/TFA; 1.0 mL/min; Det: 278 nm; 2.76 min (fumarate, 6.99%), 3.57 min (90.11%); Purity: 97.1%.
  • HPLC 1 Column: Varian Sunfire C18 100 ⁇ 4.6; 70/30/0.1 water/acetonitrile/TFA; 1.0 mL/min; Det: 278 nm; 2.79 min (fumarate, 3.34%), 3.56 min (96.11%); Purity: 99.5%
  • a step in the synthesis of the 4 stereoisomers of 1-6 was the coupling of the epoxide formed from either (R)- or (S)-3′,5′-dibenzyloxyphenylbromohydrin with the (R)- or (S)-enantiomer of the appropriate benzyl-protected 2-amino-3-benzylpropane (1-5) or the (R)- or (S)-enantiomer of N-benzyl-2-aminoheptane (6), Scheme I.
  • fenoterol analogues have an equivalent if not greater binding affinity for ⁇ 2-adrenergic receptors than fenoterol.
  • ⁇ 1 -adrenergic receptor binding was done on rat cortical membrane following a previously described procedure (Beer et al., Biochem. Pharmacol. 37: 1145-1151, 1988).
  • male Sprague-Dawley rats weighing 250-350 g were decapitated and their brains quickly removed.
  • the cerebral cortices were dissected on ice, weighed and promptly transferred to a 50 ml test tube containing approximately 30 ml of 50 mM Tris-HCl, pH 7.8 (at room temperature).
  • the tissues were homogenized with a polytron and centrifuged at 20,000 ⁇ g for 12 min at 4° C.
  • the pellet was washed again in the same manner and resuspended at a concentration of 20 mg (original wet wt) per 1 ml in the assay buffer (20 mM Tris-HCl, 10 mM MgCl 2 , 1 mM EDTA, 0.1 mM ascorbic acid at pH 7.8).
  • 30 nM ICI 118-551 was also added to the assay buffer.
  • HEK 293 cells stability transfected with cDNA encoding human ⁇ 2 -AR (provided by Dr. Brian Kobilka, Stanford Medical Center, Palo Alto, Calif.) were grown in Dulbecco's Modified Eagle Medium (DMEM) containing 10% fetal bovine serum (FBS), 0.05% penicillin-streptomycin, and 400 ⁇ g/ml G418 as previously described (Pauwels et al., Biochem. Pharmacol. 42: 1683-1689, 1991).
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • penicillin-streptomycin 0.05% penicillin-streptomycin
  • 400 ⁇ g/ml G418 400 ⁇ g/ml G418 as previously described (Pauwels et al., Biochem. Pharmacol. 42: 1683-1689, 1991).
  • the cells were scraped from the 150 ⁇ 25 mm plates and centrifuged at 500 ⁇ g for 5 minutes.
  • the pellet was homogenized in 50 mM Tris-HCl, pH 7.7, with a Polytron, centrifuged at 27,000 ⁇ g, and resuspended in the same buffer. The latter process was repeated, and the pellet was resuspended in 25 mM Tris-HCl containing 120 mM NaCl, 5.4 mM KCl, 1.8 mM CaCl 2 , 0.8 mM MgCl 2 , and 5 mM glucose, pH 7.4.
  • the binding assays contained 0.3 nM [ 3 H]CGP-12177 in a volume of 1.0 ml. Nonspecific binding was determined by 1 ⁇ M propranolol.
  • binding affinities expressed as K i values, were determined using membranes obtained from a HEK 293 cell line stably transfected with cDNA encoding human ⁇ 2 -AR (Pauwels et al., Biochem. Pharmacol. 42: 1683-1689, 1991) with [ 3 H]CGP-12177 as the marker ligand.
  • the resulting IC 50 values and Hill coefficients were calculated for each test compound using GraphPad Prism® software and K i values were calculated using the Cheng-Prusoff transformation ( Biochem Pharmacol 22: 3099-3108, 1973):
  • K i IC 50 /(1+ L/K d )+ Eqn. 1.
  • L is the concentration of [ 3 H]CGP-12177 and K d is the binding affinity of the [ 3 H]CGP-12177.
  • (R,R)-5 had the highest relative affinity of the tested compounds, although the difference between (R,R)-5 and (R,R)-1 did not reach statistical significance, Table 1.
  • the minimal effect of transforming the p-OH moiety into a methyl ether is consistent with previous data from Schirrmacher et al. ( Bioorg. Med. Chem.
  • Binding affinities expressed as K i values, for the ⁇ 1 -AR were determined using rat cortical membranes with [ 3 H]-CGP-12177 as the marker ligand (Beer et al., Biochem. Pharmacol. 37: 1145-1151, 1988.). The calculated K i for (R,R)-5 was 3,349 nM and the binding affinities for the all of the remaining test compounds were >10,000 nM, Table 1. Unlike the data from the ⁇ 2 -AR binding studies, there was no clear trend which could be associated with the stereochemistry of the compounds.
  • the relative selectivity of the compounds for the ⁇ 2 -AR and ⁇ 1 -AR was determined using the ratio K i ⁇ 1 /K i ⁇ 2 , Table 1. Of particular interest were the ratios for the four compounds with sub-micromolar affinity for the ⁇ 2 -AR, (R,R)-1, (R,R)-2, (R,R)-5 and (R,S)-5, which were 46, 43, 14 and 46, respectively. The results for (R,R)-1 and (R,R)-2 are consistent with previously reported K i ⁇ 1 /K i ⁇ 2 ratio of 53 for the ⁇ 2 -AR-selective agonist (R,R)-TA-2005, compound 49, FIG. 6 .
  • This example illustrates the pharmacological activities of the compounds with sub-micromolar affinity for the ⁇ 2 -AR and (R,S)-1 and (S,S)-1.
  • This example illustrates the use of Comparative Molecular Field Analysis (CoMFA) to analyze the disclosed compounds.
  • the disclosed compounds were analyzed using Comparative Molecular Field Analysis, a 3D QSAR technique applicable to the analysis of the relative activities of stereoisomers and/or enantiomers at a selected target.
  • CoMFA was performed as implemented in SYBYL 7.2. (TRIPOS Inc., St. Louis, Mo.). Molecular models of all derivatives were prepared in HyperChem v. 6.03 (HyperCube Inc., Gainesville, Fla.) using ModelBuild procedure to ensure the same conformation of the scaffold. Models were extracted to SYBYL and the partial atomic charges (Gasteiger-Huckel type) were calculated. Ligand models were aligned using as a common substructure of the two asymmetric carbon atoms in the core of the fenoterol molecule (—C*—CH 2 —NH—C*—CH 2 —).
  • the model was used to identify the regions responsible for the discrimination between the stereoisomers.
  • the CoMFA procedure produced several distinct asymmetric regions located in close proximity of each chiral center.
  • the first chiral center (carrying the ⁇ hydroxyl group) is surrounded by an electropositive region behind the molecule.
  • An electropositive region can be associated with hydrogen bond formation and indicates favorable donor properties or unfavorable acceptor properties of the pseudoreceptor.
  • the location of the electropositive field indicates that the orientation of the ⁇ -OH moiety behind the plane of the model (the S configuration at the chiral center) would hinder H bond formation with the receptor.
  • the electropositive region is closely associated with a steric unfavorable region behind the first chiral center.
  • the CoMFA model also demonstrated the effect of the second chiral center.
  • the preferred configuration can be derived from the binding data, where for compounds 1-4 and 6 the (R,R)-isomers had the higher affinities relative to their respective (R,S)-isomers, while the K i values for (R,R)-5 and (R,S)-5 were equivalent, Table 1.
  • the more active isomers are those with the methyl moiety on the stereogenic center on the aminoalkyl portion of the molecules pointing out of the plane of the figure of the CoMFA model. This is depicted by a steric disfavoring region behind the second chiral center of the molecule, and indicates a preference for the R configuration at this site.
  • the large electropositive region encompassing the area close to the —OH or OCH 3 substituents represents H-bond donor properties of the pseudoreceptor to these moieties.
  • a large electronegative region and another electropositive region, both located parallel on two sides of the aromatic system most likely represent ⁇ - ⁇ or ⁇ -hydrogen bond interactions between the ⁇ 2 -AR and electron-rich aromatic moieties, such as the naphthyl ring. This is consistent with the increased affinity of compounds 1, 2 and 5 relative to the other compounds examined in this study. The role of this interaction is suggested by the observation that the K i values for (R,R)-5 and (R,S)-5 were equivalent to (R,R)-1 and (R,R)-2, Table 1.
  • Two steric regions are located close to the electrostatic regions and one favors and the other disfavors bulkiness in the respective areas. This indicates that the binding of the aminoalkyl portions of the molecules are also sterically restricted.
  • a key aspect in this process is the interaction of the hydroxyl moiety on the chiral carbon of the agonist with the Asn-293 residue in TM6, and for this interaction an R-configuration is preferable at the chiral carbon (Eimerl et al., Biochem. Pharmacol. 36: 3523-3527, 1987; Kikkawa et al., Mol. Pharmacol. 53: 128-134, 1998; and Swaminath et al. J. Biol. Chem. 279: 686-691, 2004). Since the “catechol” portion of the fenoterol molecule was not altered in this study, it follows that in the CoMFA model, an R-configuration at the first stereogenic center is preferred in most stable complexes.
  • the methoxy moiety of the ligand was also located in close proximity to three other polar residues, histidine 296 (H296) in TM6, tryptophan 109 (W109) in TM3 and asparagine 312 (N312) in TM7, each of which can interact with an aromatic group on the aminoalkyl portion of (R,R)-49.
  • the cloud of ⁇ -electrons of aromatic rings can act as hydrogen bond acceptors, although it has been estimated that the interaction would be about half as strong as a normal hydrogen bond (Levitt and Perutz J. Mol. Biol. 201: 751-754, 1998).
  • the higher affinity and subtype selectivity for (R,R)-5 relative to (R,R)-3 and (R,R)-4 or (R)-7 is consistent with the greater ⁇ electron distribution in the napthyl ring relative to the other aromatic rings.
  • the CoMFA model also identified a large electronegative region and another electropositive region, both located parallel to the aromatic system, which are most likely associated with ⁇ - ⁇ or ⁇ -hydrogen bond interactions between the ⁇ 2 -AR and electron-rich aromatic moieties, such as the naphthyl ring.
  • ⁇ - ⁇ or ⁇ -hydrogen bond interactions between the ⁇ 2 -AR and electron-rich aromatic moieties, such as the naphthyl ring.
  • Y308, H296, W109 and N312 were identified as possible sources of ⁇ - ⁇ and/or ⁇ -hydrogen bond interactions.
  • the estimated distances between the p-methoxy moiety on (R,R)-49 and W109 and N312 were 4.80 ⁇ and 3.45 ⁇ , respectively.
  • the interactions suggested by the CoMFA model may represent the source of the increase affinities for (R,R)-1, (R,R)-2 and (R,R)-5, relative to the other (R,R)-isomers, but not the observed ⁇ 1 / ⁇ 2 selectivity.
  • the binding of an agonist to the ⁇ 2 -AR has been described as a multistep interrelated process, in which sequential interactions between the agonist and receptor produce corresponding conformational changes (Kobilka, Mol. Pharm. 65: 1060-1062, 2004).
  • the CoMFA model reflects the final agonist/ ⁇ 2 -AR complex and, in order to discern the effect of the steric restricted site, it is necessary to consider the effect that interaction with this site has on the outcome of the binding process.
  • a detailed description of the present CoMFA model is disclosed in Jozwiak et al. ( J. Med. Chem., 50 (12): 2903-2915, 2007) which is hereby incorporated by reference in its entirety.
  • the chiral selector determines the enantioselectivity of the process. If the chirality of the chiral selector places the interaction perpendicular to the plane of the ligand, no enantioselectivity is observed. As a deviation from the perpendicular increases, so does the enantioselectivity relative to the R or S configuration.
  • the observed K i ⁇ 2 values and subtype selectivity indicate the following: 1) the K i ⁇ 2 -AR values represent the sum total of the ⁇ -hydrogen bond and ⁇ - ⁇ interactions between the 1-naphthyl moieties and Y308 and H296, as well as additional non- ⁇ 2 -AR specific interactions with other residues such as W109 and N312; 2) the steric volume swept out by (R,S)-5 increases the probability of interactions of Y308 and H296 with the ⁇ cloud of the naphtyl moiety relative to the (R,R)-5; and 3) the steric volume swept out by (R,R)-5 increases the probability of interactions with non- ⁇ 2 -AR specific sites relative to (R,S)-5.
  • a difference between compounds 3 and 5 is the steric areas swept out by the aromatic substituents.
  • the phenyl ring produces a smaller, more linear area
  • compound 5 the 1-naphthyl ring system produces a relatively larger and broader area.
  • (R,R)-2 and (R,S)-5 are chosen as possible candidates for the development of a new selective ⁇ 2 -AR agonist.
  • These compounds may have increased and extended systemic exposures relative to the commercially available rac-1 due to changes in molecular hydrophobicity, metabolic profile and transporter interactions.
  • the present example provides a pharmacophore model which may be used as a structural guide for the design of new compounds with ⁇ 2 -AR selectivity which can be tested for use in the treatment of a desired condition, including congestive heart failure.
  • This example demonstrates the plasma concentrations of (R,R)-Fenoterol, (R,R)-Methoxyfenoterol and (R,S)-Naphtylfenoterol administered as an intravenous (IV) bolus to male Sprague-Dawley rats.
  • Plasma samples were collected over six hours at the following nine timepoints: prior to administration of the desired dose; 5.00-5.30 minutes after dose; 15.00-16.30 minutes after dose; 30.00-33.00 minutes after dose; 60-65 minutes after dose; 120-125 minutes after dose; 240-245 minutes after dose; 300-305 minutes after dose; and 360-365 minutes after dose.
  • Urine was collected for 0-6 hours and 6 to 24 hours from 3 rats in each treatment group.
  • alpha micro rate constant associated with the distribution phase
  • beta micro rate constant associated with the elimination phase
  • AUC area under the curve
  • T1/2 (K10) half-life associated with the rate constant K10
  • K10 elimination rate—rate at which the drug leaves the system from the central compartment
  • K12 rate at which drug enters tissue compartment from the central compartment
  • K21 rate at which drug enters central compartment from tissue compartment
  • V1 volume of distribution of the central compartment
  • V2 volume of distribution of the tissue compartment
  • Vss volume of distribution at steady state
  • Cl chlor
  • Tables 5-7 and FIG. 8 illustrate the individual plasma concentrations of (R,R)-fenoterol, (R,R)-methoxyfenoterol and (R,S)-naphtylfenoterol after IV administration to rats (5 mg/kg).
  • the average concentration of (R,R)-fenoterol in plasma was dramatically lower (1.34 ⁇ g/ml) five minutes after IV administration to rats (5 mg/kg) compared to either the average concentration of (R,R)-methoxyfenoterol (2.12 ⁇ g/ml) or (R,S)-naphtylfenoterol (2.11 ⁇ g/ml).
  • a cardiac disorder such as congestive heart failure is treated by administering a therapeutic effective dose of (R,R)-fenoterol or one or more of the fenoterol analogues disclosed above (see Sections III and IV).
  • a subject who has been diagnosed with congestive heart failure is identified.
  • a therapeutic effective dose of (R,R)-fenoterol or the respective fenoterol analogue is administered to the subject.
  • a therapeutic effective dose of the (R,R)-fenoterol analogue including a OCH 3 group or a naphthyl derivative is administered to the subject.
  • a therapeutic effective dose of the (R,S)-fenoterol analogue including a napthyl derivative is administered to the subject.
  • the fenoterol analogue is prepared and purified as described in Section III.B and Example 5.
  • the amount of (R,R)-fenoterol or fenoterol analogue or a pharmaceutical composition thereof administered to reduce, inhibit, and/or treat congestive heart failure depends on the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition (see Section V).
  • a therapeutically effective amount of an agent is the amount sufficient to prevent, reduce, and/or inhibit, and/or treat the cardiac disorder (e.g., congestive heart failure) in a subject without causing a substantial cytotoxic effect in the subject.
  • (R,R)-fenoterol a disclosed fenoterol analogue (such as an (R,R)-fenoterol analogue including a OCH 3 group or a naphthyl derivative or an (R,S)-fenoterol analogue including a napthyl derivative) or pharmaceutical composition is provided at a dosage range from about 0.001 to about 10 mg/kg body weight orally in single or divided doses.
  • the dosage range is from about 0.005 to about 5 mg/kg body weight orally in single or divided doses (assuming an average body weight of approximately 70 kg; values adjusted accordingly for persons weighing more or less than average).
  • a disclosed fenoterol compound or pharmaceutical composition is provided by oral administration in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.0 mg to about 10 mg, more particularly about 2.5 mg, about 5 mg, or about 10 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg active ingredient is administered two to four times a day.
  • a tablet containing about 1 mg to about 10 mg active ingredient is administered two times a day.
  • a pulmonary disorder such as asthma or chronic obstructive pulmonary disease is treated by administering a therapeutic effective dose of the fenoterol analogues disclosed above (see Sections III-V).
  • a subject who has been diagnosed with or displays one of the symptoms associated with asthma or chronic obstructive pulmonary disease is identified.
  • a therapeutic effective dose of the desired fenoterol analogue is administered to the subject.
  • a therapeutic effective dose of the (R,R)-fenoterol analogue including a OCH 3 group or a naphthyl derivative is administered to the subject.
  • a therapeutic effective dose of the (R,S)-fenoterol analogue including a napthyl derivative is administered to the subject.
  • the fenoterol analogue is prepared and purified as described in Section III.B and Example 5.
  • the amount of the fenoterol analogue administered to prevent, reduce, inhibit, and/or treat the pulmonary disorder depends on the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition.
  • a therapeutically effective amount of an agent will be the amount sufficient to prevent, reduce, and/or inhibit, and/or treat the pulmonary disorder in a subject without causing a substantial cytotoxic effect in the subject.
  • (R,R)-fenoterol a disclosed fenoterol analogue (such as an (R,R)-fenoterol analogue including a OCH 3 group or a naphthyl derivative or an (R,S)-fenoterol analogue including a napthyl derivative) or pharmaceutical composition is provided at a dosage range from about 0.001 to about 10 mg/kg body weight orally in single or divided doses.
  • the dosage range is from about 0.005 to about 5 mg/kg body weight orally in single or divided doses (assuming an average body weight of approximately 70 kg; values adjusted accordingly for persons weighing more or less than average).
  • a disclosed fenoterol compound or pharmaceutical composition is provided by oral administration in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.0 mg to about 10 mg, more particularly about 2.5 mg, about 5 mg, or about 10 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg active ingredient is administered two to four times a day.
  • a tablet containing about 1 mg to about 10 mg active ingredient is administered two times a day.
US12/376,945 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure Abandoned US20100168245A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/376,945 US20100168245A1 (en) 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US83716106P 2006-08-10 2006-08-10
US92782507P 2007-05-03 2007-05-03
PCT/US2007/075731 WO2008022038A1 (en) 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)- or (r,s)-fenoterol analogues and their use in treating congestive heart failure
US12/376,945 US20100168245A1 (en) 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/075731 A-371-Of-International WO2008022038A1 (en) 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)- or (r,s)-fenoterol analogues and their use in treating congestive heart failure

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/333,866 Continuation US8703826B2 (en) 2006-08-10 2011-12-21 Preparation of (R,R)-fenoterol and (R,R)-or (R,S)-fenoterol analogues and their use in treating congestive heart failure

Publications (1)

Publication Number Publication Date
US20100168245A1 true US20100168245A1 (en) 2010-07-01

Family

ID=38917814

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/376,945 Abandoned US20100168245A1 (en) 2006-08-10 2007-08-10 Preparation of (r,r)-fenoterol and (r,r)-or (r,s)-fenoterol analogues and their use in treating congestive heart failure
US13/333,866 Active US8703826B2 (en) 2006-08-10 2011-12-21 Preparation of (R,R)-fenoterol and (R,R)-or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US14/199,208 Active 2027-11-22 US9522871B2 (en) 2006-08-10 2014-03-06 Preparation of (R,R)-fenoterol and (R,R)-or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US15/347,354 Active US9908841B2 (en) 2006-08-10 2016-11-09 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US15/874,747 Active US10308591B2 (en) 2006-08-10 2018-01-18 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US16/390,982 Active US10562843B2 (en) 2006-08-10 2019-04-22 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure

Family Applications After (5)

Application Number Title Priority Date Filing Date
US13/333,866 Active US8703826B2 (en) 2006-08-10 2011-12-21 Preparation of (R,R)-fenoterol and (R,R)-or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US14/199,208 Active 2027-11-22 US9522871B2 (en) 2006-08-10 2014-03-06 Preparation of (R,R)-fenoterol and (R,R)-or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US15/347,354 Active US9908841B2 (en) 2006-08-10 2016-11-09 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US15/874,747 Active US10308591B2 (en) 2006-08-10 2018-01-18 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US16/390,982 Active US10562843B2 (en) 2006-08-10 2019-04-22 Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure

Country Status (11)

Country Link
US (6) US20100168245A1 (pt)
EP (1) EP2064174B1 (pt)
JP (2) JP5302194B2 (pt)
KR (1) KR101378067B1 (pt)
CN (1) CN103739503A (pt)
AU (1) AU2007286051B2 (pt)
BR (1) BRPI0716495A2 (pt)
CA (1) CA2660707C (pt)
IL (1) IL196965A (pt)
MX (1) MX2009001534A (pt)
WO (1) WO2008022038A1 (pt)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110294802A1 (en) * 2007-12-17 2011-12-01 Mcinally Thomas Pharmaceutically acceptable salts of methyl (3-{ [[3-(6- amino- 2-butoxy-8-oxo-7, 8-dihydro-9h-purin-9-yl) propyl] (3- morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy
US10251868B2 (en) * 2011-09-06 2019-04-09 Novartis Ag Benzothiazolone compound

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2660707C (en) 2006-08-10 2014-07-08 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Preparation of (r,r)-fenoterol and (r,r)- or (r,s)-fenoterol analogues and their use in treating congestive heart failure
CN101531602B (zh) * 2008-03-10 2014-08-27 北京德众万全医药科技有限公司 一种hplc法分析分离酒石酸福莫特罗中间体的方法
MX2012010364A (es) * 2010-03-08 2013-02-26 Univ Tennessee Res Foundation Antagonistas de los receptores beta-adrenérgicos y usos de los mismos.
EP2544676B1 (en) 2010-03-10 2018-09-19 The United States of America, as represented by The Secretary, Department of Health and Human Services The use of fenoterol and fenoterol analogues in the treatment of glioblastomas and astrocytomas
JP6130495B2 (ja) 2012-05-25 2017-05-17 アメリカ合衆国 カンナビノイド受容体活性関連障害及び疾患を調節する方法
US9784726B2 (en) 2013-01-08 2017-10-10 Atrogi Ab Screening method, a kit, a method of treatment and a compound for use in a method of treatment
WO2017059268A1 (en) * 2015-10-01 2017-04-06 Mitchell Woods Pharmaceuticals, Inc. Methods of reducing chemoresistance and treating cancer
GB201714734D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714736D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714740D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
GB201714745D0 (en) 2017-09-13 2017-10-25 Atrogi Ab New compounds and uses
KR101987841B1 (ko) 2019-03-13 2019-06-11 김도현 완구총
CN110376313B (zh) * 2019-08-20 2022-06-07 广州健康元呼吸药物工程技术有限公司 一种检测富马酸福莫特罗或其相关制剂中杂质的方法
EP4146183A1 (en) * 2020-05-05 2023-03-15 Irving W. Wainer Method for treating symptoms of viral infections
GB202205895D0 (en) 2022-04-22 2022-06-08 Atrogi Ab New medical uses

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3966814A (en) * 1969-12-12 1976-06-29 Boehringer Ingelheim Gmbh 1-Phenyl-2-(Naphthylalkyl-amino)-ethanols and salts thereof
US5795567A (en) * 1990-07-26 1998-08-18 Monsanto Company Polyamines and method for preparation thereof
US6015837A (en) * 1996-08-29 2000-01-18 New York Medical College Method for treating scoliosis with β2-adrenoceptor agonists
US6589508B1 (en) * 1991-04-05 2003-07-08 Sepracor Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R) formoterol
US6664424B2 (en) * 1998-07-01 2003-12-16 Warner-Lambert Company Stereoisomers with high affinity for adrenergic receptors
US6747043B2 (en) * 2002-05-28 2004-06-08 Theravance, Inc. Alkoxy aryl β2 adrenergic receptor agonists
US20040192783A1 (en) * 1991-04-05 2004-09-30 John Morley Use of B-2 bronchodilator drugs
US6866839B2 (en) * 1991-04-05 2005-03-15 Sepracor Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R)-formoterol
US20050107417A1 (en) * 2003-07-28 2005-05-19 Boehringer Ingelheim International Gmbh Medicaments for inhalation comprising a novel anticholinergic and a betamimetic
US7045658B2 (en) * 2001-03-22 2006-05-16 Glaxo Group Limited Formailide derivatives as beta2-adrenoreceptor agonists

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL296701A (pt) *
AT235270B (de) 1962-08-18 1964-08-25 Chemie Linz Ag Verfahren zur Herstellung von N-substituierten Phenyläthanolaminderivaten und deren Salzen
NL125650C (pt) 1962-11-30 1900-01-01
SE354279B (pt) * 1969-06-13 1973-03-05 Draco Ab
CH564509A5 (pt) * 1969-12-12 1975-07-31 Boehringer Sohn Ingelheim
DE2010883A1 (de) * 1970-03-07 1971-09-23 Troponwerke Dinklage & Co, 5000 Köln-Mülheim Pharmakologisch wirksame Aminoalkohole und Verfahren zu ihrer Darstellung
AT308139B (de) 1970-07-27 1973-06-25 Degussa Verfahren zur Herstellung von neuen basisch substituierten Alkyltheophyllinen, ihren Isomeren und Säureadditionssalzen
IT1044214B (it) * 1971-04-22 1980-03-20 Zambeletti Spa L 2 idrossi 2 feniletilamine so stituite dotate di proprieta vasoattive e processo per la loro preparazione
JPS532194A (en) 1976-06-25 1978-01-10 Sekisui Jushi Kk Packing method by plastic band
US5795564A (en) 1991-04-05 1998-08-18 Sepracor, Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R)-formoterol
WO1998001099A2 (en) 1996-07-05 1998-01-15 E.C. Pesterfield Assoc., Inc. Method of treating retinal ischemia and glaucoma
WO1999016430A1 (en) 1997-09-30 1999-04-08 E.C. Pesterfield Associates, Inc. Galenic forms of r-or rr-isomers of adrenergic beta-2 agonists
AU6167599A (en) 1998-09-30 2000-04-17 United States Of America, Represented By The Secretary, Department Of Health And Human Services, The Use of a beta2 adrenergic receptor agonist in the treatment of cardiovascular disease
AU2004285961A1 (en) 2003-11-03 2005-05-12 Myogen, Inc. 1,4-dihydropyridine compounds, pharmaceutical compositions, and methods for the treatment of cardiovascular disease
CA2560538A1 (en) * 2004-03-22 2005-10-06 Myogen, Inc. (r)-enoximone sulfoxide and its use in the treatment of pde-iii mediated diseases
EP1781331A1 (en) 2004-08-09 2007-05-09 Université Catholique de Louvain Use of agonists and antagonists of beta-adrenoceptors for treating arterial diseases
MX2007008459A (es) * 2005-01-11 2007-07-25 Glaxo Group Ltd Sales de cinamato de un agonista beta-2 adrenergico.
CA2660707C (en) * 2006-08-10 2014-07-08 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Preparation of (r,r)-fenoterol and (r,r)- or (r,s)-fenoterol analogues and their use in treating congestive heart failure

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3966814A (en) * 1969-12-12 1976-06-29 Boehringer Ingelheim Gmbh 1-Phenyl-2-(Naphthylalkyl-amino)-ethanols and salts thereof
US5795567A (en) * 1990-07-26 1998-08-18 Monsanto Company Polyamines and method for preparation thereof
US6589508B1 (en) * 1991-04-05 2003-07-08 Sepracor Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R) formoterol
US20040192783A1 (en) * 1991-04-05 2004-09-30 John Morley Use of B-2 bronchodilator drugs
US6866839B2 (en) * 1991-04-05 2005-03-15 Sepracor Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R)-formoterol
US20050131072A1 (en) * 1991-04-05 2005-06-16 Sepracor Inc. Methods and compositions for treating pulmonary disorders using optically pure (R,R)-formoterol
US6015837A (en) * 1996-08-29 2000-01-18 New York Medical College Method for treating scoliosis with β2-adrenoceptor agonists
US6664424B2 (en) * 1998-07-01 2003-12-16 Warner-Lambert Company Stereoisomers with high affinity for adrenergic receptors
US7045658B2 (en) * 2001-03-22 2006-05-16 Glaxo Group Limited Formailide derivatives as beta2-adrenoreceptor agonists
US6747043B2 (en) * 2002-05-28 2004-06-08 Theravance, Inc. Alkoxy aryl β2 adrenergic receptor agonists
US20050107417A1 (en) * 2003-07-28 2005-05-19 Boehringer Ingelheim International Gmbh Medicaments for inhalation comprising a novel anticholinergic and a betamimetic

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110294802A1 (en) * 2007-12-17 2011-12-01 Mcinally Thomas Pharmaceutically acceptable salts of methyl (3-{ [[3-(6- amino- 2-butoxy-8-oxo-7, 8-dihydro-9h-purin-9-yl) propyl] (3- morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy
US8673907B2 (en) * 2007-12-17 2014-03-18 Astrazeneca Ab Pharmaceutically acceptable salts of methyl (3-{ [[3-(6-amino- 2-butoxy-8-oxo-7,8-dihydro-9H-purin-9-yl) propyl] (3-morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy
US10251868B2 (en) * 2011-09-06 2019-04-09 Novartis Ag Benzothiazolone compound

Also Published As

Publication number Publication date
US9522871B2 (en) 2016-12-20
IL196965A (en) 2015-10-29
JP5302194B2 (ja) 2013-10-02
US20170081276A1 (en) 2017-03-23
JP2010500381A (ja) 2010-01-07
EP2064174B1 (en) 2016-10-26
KR101378067B1 (ko) 2014-04-10
AU2007286051B2 (en) 2013-01-10
WO2008022038A1 (en) 2008-02-21
US10308591B2 (en) 2019-06-04
US9908841B2 (en) 2018-03-06
MX2009001534A (es) 2009-04-08
US8703826B2 (en) 2014-04-22
JP2013224316A (ja) 2013-10-31
KR20090051171A (ko) 2009-05-21
IL196965A0 (en) 2009-11-18
US20140187645A1 (en) 2014-07-03
US20160009636A9 (en) 2016-01-14
US20190248732A1 (en) 2019-08-15
US20120157543A1 (en) 2012-06-21
US10562843B2 (en) 2020-02-18
CN103739503A (zh) 2014-04-23
EP2064174A1 (en) 2009-06-03
US20180141894A1 (en) 2018-05-24
CA2660707C (en) 2014-07-08
CA2660707A1 (en) 2008-02-21
BRPI0716495A2 (pt) 2014-03-04
AU2007286051A1 (en) 2008-02-21

Similar Documents

Publication Publication Date Title
US10562843B2 (en) Preparation of (R,R)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
US10925840B2 (en) Use of fenoterol and fenoterol analogues in the treatment of glioblastomas and astrocytomas
AU2013202127B2 (en) Preparation of (R,R-)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
AT414238B (de) Neues hydroxyliertes 3-phenoxy-3-phenyl-1- aminopropan und dieses aminopropan enthaltende pharmazeutische formulierung
AU2014224073A1 (en) Preparation of (R,R-)-fenoterol and (R,R)- or (R,S)-fenoterol analogues and their use in treating congestive heart failure
CN101528666B (zh) (r,r)-非诺特罗和(r,r)-或(r,s)-非诺特罗类似物的制备及其在治疗充血性心力衰竭中的应用
JP2004517881A (ja) 置換フェルバメート誘導化合物

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WAINER, IRVING W.;ZHU, WEIZHONG;CHAKIR, KHALID;AND OTHERS;SIGNING DATES FROM 20070905 TO 20070928;REEL/FRAME:019917/0345

AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WAINER, IRVING W.;ZHU, WEIZHONG;CHAKIR, KHALID;AND OTHERS;SIGNING DATES FROM 20070905 TO 20070928;REEL/FRAME:022252/0512

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION