US20120226067A1 - Processes for intermediates for macrocyclic compounds - Google Patents

Processes for intermediates for macrocyclic compounds Download PDF

Info

Publication number
US20120226067A1
US20120226067A1 US13/412,148 US201213412148A US2012226067A1 US 20120226067 A1 US20120226067 A1 US 20120226067A1 US 201213412148 A US201213412148 A US 201213412148A US 2012226067 A1 US2012226067 A1 US 2012226067A1
Authority
US
United States
Prior art keywords
boc
bts
ddz
none
val
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
US13/412,148
Inventor
Eric Marsault
Luc Ouellet
Hamid R. Hoveyda
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US13/412,148 priority Critical patent/US20120226067A1/en
Publication of US20120226067A1 publication Critical patent/US20120226067A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C213/00Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C213/08Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton by reactions not involving the formation of amino groups, hydroxy groups or etherified or esterified hydroxy groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2214Motilins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C41/00Preparation of ethers; Preparation of compounds having groups, groups or groups
    • C07C41/01Preparation of ethers
    • C07C41/18Preparation of ethers by reactions not forming ether-oxygen bonds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/30Preparation of carboxylic acid esters by modifying the acid moiety of the ester, such modification not being an introduction of an ester group
    • C07C67/333Preparation of carboxylic acid esters by modifying the acid moiety of the ester, such modification not being an introduction of an ester group by isomerisation; by change of size of the carbon skeleton
    • C07C67/343Preparation of carboxylic acid esters by modifying the acid moiety of the ester, such modification not being an introduction of an ester group by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D273/00Heterocyclic compounds containing rings having nitrogen and oxygen atoms as the only ring hetero atoms, not provided for by groups C07D261/00 - C07D271/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0821Tripeptides with the first amino acid being heterocyclic, e.g. His, Pro, Trp
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0827Tripeptides containing heteroatoms different from O, S, or N

Definitions

  • the present invention relates to novel conformationally-defined macrocyclic compounds, pharmaceutical compositions comprising same and intermediates used in their manufacture. More particularly, the invention relates to macrocyclic compounds that have been demonstrated to selectively antagonize the activity of the motilin receptor. The invention further relates to macrocyclic compounds useful as therapeutics for a range of gastrointestinal disorders, in particular those in which malfunction of gastric motility or increased motilin secretion is observed, such as hypermotilinemia, irritable bowel syndrome and dyspepsia.
  • GI gastrointestinal
  • absorption, secretion, blood flow and motility Mulvihill, et al. in Basic and Clinical Endocrinology, 4 th edition, Greenspan, F. S.; Baxter, J. D., eds., Appleton & Lange: Norwalk, Conn., 1994, pp 551-570. Since interactions between the brain and GI system are critical to the proper modulation of these functions, these peptides can be produced locally in the GI tract or distally in the CNS.
  • motilin a linear 22-amino acid peptide
  • motilin plays a critical regulatory role in the GI physiological system though governing of fasting gastrointestinal motor activity.
  • the peptide is periodically released from the duodenal mucosa during fasting in mammals, including humans. More precisely, motilin exerts a powerful effect on gastric motility through the contraction of gastrointestinal smooth muscle to stimulate gastric emptying, decrease intestinal transit time and initiate phase III of the migrating motor complex in the small bowel (Itoh, Z., Ed., Motilin, Academic Press: San Diego, Calif., 1990, ASIN: 0123757304; Nelson, D. K. Dig. Dis. Sci. 1996, 41, 2006-2015; Peeters, T. L.; Vantrappen, G.; Janssens, J. Gastroenterology 1980, 79, 716-719).
  • Motilin exerts these effects through receptors located predominantly on the human antrum and proximal duodenum, although its receptors are found in other regions of the GI tract as well (Peeters, T. L.; Bormans, V.; Vantrappen, G. Regul. Pept. 1988, 23, 171-182). Therefore, motilin hormone is involved in motility of both the upper and lower parts of the GI system (Williams et al. Am. J. Physiol. 1992, 262, G50-G55).
  • motilin and its receptors have been found in the CNS and periphery, suggesting a physiological role in the nervous system that has not yet been definitively elucidated (Depoortere, I.; Peeters, T. L. Am. J. Physiol. 1997, 272, G994-999 and O'Donohue, T. L. et al. Peptides 1981, 2, 467-477).
  • motilin receptors in the brain have been suggested to play a regulatory role in a number of CNS functions, including feeding and drinking behavior, micturition reflex, central and brain stem neuronal modulation and pituitary hormone secretion (Itoh, Z. Motilin and Clinical Applications.
  • motilin Due to the critical and direct involvement of motilin in control of gastric motility, agents that either diminish (hypomotility) or enhance (hypermotility) the activity at the motilin receptor, are a particularly attractive area for further investigation in the search for new effective pharmaceuticals towards these indications.
  • Antagonists of the motilin receptor are potentially extremely useful as therapeutic treatments for diseases associated with hypermotility and hypermotilinemia, including irritable bowel syndrome, dyspepsia, gastroesophogeal reflux disorders, Crohn's disease, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, diabetes mellitus, obesity, malabsorption syndrome, carcinoid syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome, gastric stasis and eating disorders leading to obesity.
  • diseases associated with hypermotility and hypermotilinemia including irritable bowel syndrome, dyspepsia, gastroesophogeal reflux disorders, Crohn's disease, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, diabetes mellitus, obesity, malabsorption syndrome, carcinoid syndrome, diarrhea, atrophic colitis or gastritis,
  • peptidic compounds have been described as antagonists of the motilin receptor (Depoortere, I.; Macielag, M. J.; Galdes, A.; Peeters, T. L. Eur. J. Pharmacol. 1995, 286, 241-247; U.S. Pat. Nos. 5,470,830; 6,255,285; 6,586,630; 6,720,433; U.S. 2003/0176643; WO 02/64623).
  • These peptidic antagonists suffer from the known limitations of peptides as drug molecules, in particular poor oral bioavailability and degradative metabolism.
  • Cyclization of peptidic derivatives is a method employed to improve the properties of a linear peptide both with respect to metabolic stability and conformational freedom. Cyclic molecules tend to be more resistant to metabolic enzymes. Such cyclic tetrapeptide motilin antagonists have been reported (Haramura, M. et al J. Med. Chem. 2002, 45, 670-675, U.S. 2003/0191053; WO 02/16404).
  • the macrocyclic motilin antagonists of the present invention comprise elements of both peptidic and non-peptidic structures in a combination which has not been pursued for this application previously.
  • antagonists of the present invention are different.
  • the known motilin antagonists which are cyclic peptides it was found that such derivatives containing D-amino acids were devoid of activity.
  • the tripeptidomimetic compounds of the present invention the D-stereochemistry is required for two of the three building elements.
  • the motilin antagonists of the present invention are also distinct from the prior art in that they comprise a tether element to fulfill the dual role of controlling conformations and providing additional sites for interaction either through hydrophobic interactions, hydrogen bonding or dipole-dipole interactions.
  • the present invention is directed to compounds of formula (I):
  • Z 1 , Z 2 and Z 3 are independently selected from the group consisting of O, N and NR 10 , wherein R 10 is selected from the group consisting of hydrogen, lower alkyl, and substituted lower alkyl; R 1 is independently selected from the group consisting of lower alkyl substituted with aryl, lower alkyl substituted with substituted aryl, lower alkyl substituted with heteroaryl and lower alkyl substituted with substituted heteroaryl; R 2 is hydrogen;
  • R 3 is independently selected from the group consisting of alkyl and cycloalkyl with the proviso that when Z 1 is N, R 3 can form a four, five, six or seven-membered heterocyclic ring together with Z 1;
  • R 4 is hydrogen
  • R 5 and R 6 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl and substituted heteroaryl, with the proviso that at least one of R 5 and R 6 is hydrogen
  • X is selected from the group consisting of O, NR 8 , and N(R 9 ) 2 + ;
  • G 1 and G 2 each independently represent a covalent bond or a bivalent radical selected from the group consisting of —O—, —S—, —SO—, —SO 2 —, —C( ⁇ O)—, —C( ⁇ O)—O—, —O—C( ⁇ O)—, —C( ⁇ O)NH—, —NH—C( ⁇ O)—, —SO 2 —NH—, —NH—SO 2 —, —CR 42 R 43 —, —CH ⁇ CH— with a configuration Z or E, and —C ⁇ C—;with the proviso that G 1 is bonded closer to U than G 2 ; K 1
  • the invention also proposes compounds of formula (1) which are antagonists of the motilin receptor .
  • the invention proposes a method of treating a disorder associated with the motilin receptor or motility dysfunction in humans and other mammals, comprising administering a therapeutically effective amount of a compound of formula (1).
  • R 1 is selected from the group consisting of —(CH 2 ) q R 11 , and —CHR 12 R 13
  • R 11 , R 12 and R 13 are selected from the group consisting of:
  • R a and R b are chosen from the group consisting of Cl, F, CF 3 , OCH 3 , OH, and C(CH 3 ) 3 and CH 3 .
  • R 3 in formula (I) is selected from the group consisting of:
  • the tether portion (T) of formula (I) is preferably selected from the group consisting of:
  • L 1 is O, NH or NMe
  • L 2 is CH or N
  • L 3 is CH or N
  • L 4 is 0 or CH 2
  • L 5 is CH or N is CR 52 R 53 or 0
  • R 46 is H or CH 3
  • R 52 , R 53 , R 54 , R 55 , R 56 and R 57 are independently selected from hydrogen, lower alkyl, substituted lower alkyl, hydroxy, alkoxy, aryloxy, amino, and oxo; or R 52 together with R 53 or R 54 together with R 55 or R 56 together with R 57 can independently form a three to seven-membered cyclic ring comprising carbon, oxygen, sulfur and/or nitrogen atoms; (X) is the site of a covalent bond to X in formula (I); and (Z 3 ) is the site of a covalent bond to Z 3 in formula (I).
  • heterocyclic ring may contain a second nitrogen atom, or an oxygen, or sulfur atom; n 2 is selected from 0, 1, 2 or 3
  • R 7 is optionally present and is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido.
  • amino, guanidine, ureido and amidino encompass substituted derivatives thereof as well.
  • the invention provides a method of treating a disorder associated with hypermotility or hypermotilinemia in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (1).
  • FIG. 1 depicts Scheme 1 presenting a general synthetic strategy to conformationally-defined macrocycles of the present invention.
  • FIG. 2 depicts the standard procedure for the synthesis of tether T8 of Example 16.
  • FIG. 3 depicts the standard procedure for the synthesis of tether T9 of Example 17.
  • FIG. 4 depicts the standard procedure for the synthesis of Ddz-propargylamine of Example 18.
  • FIG. 5A depicts the standard procedure for the synthesis of tether T10 of Example 19.
  • FIG. 5B depicts the second synthetic route to tether T10 of Example 19.
  • FIG. 6 depicts the standard procedure for the synthesis of Tether T11 of Example 20.
  • FIG. 7 depicts the standard procedure for the synthesis of tether T12 of Example 26.
  • FIG. 8 depicts the procedure for synthesis of PPh 3 -DIAD adduct of Example 29-C.
  • FIG. 9 depicts the standard procedure for attachment of tethers via reductive amination of Example 30.
  • FIG. 10 depicts the standard procedure for the synthesis of tether T28 of Example 32.
  • FIG. 11 the standard procedure for the synthesis of tether T32 of Example 36.
  • FIGS. 12A , 12 B depict the standard procedure for the synthesis of tether T33a and T33b of Example 37.
  • FIG. 13 depicts the standard procedure for the synthesis of tether T34 of Example 38.
  • FIG. 14 depicts the standard procedure for the synthesis of tether T35 of Example 39.
  • FIG. 15 depicts the standard procedure for the synthesis of tether T36 of Example 40.
  • FIG. 16 depicts the standard procedure for the synthesis of tether T37 of Example 41.
  • FIG. 17 depicts the standard procedure for the synthesis of tether T38 of Example 42.
  • Chiral T38 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • FIG. 18 depicts the standard procedure for the synthesis of tether T39 of Example 43.
  • Chiral T39 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • FIG. 19 depicts the standard procedure for the synthesis of tether T40 of Example 44.
  • Chiral T40 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • FIG. 20 depicts the standard procedure for the synthesis of tether T41 of Example 45.
  • FIG. 21 depicts the standard procedure for the synthesis of tether T42 of Example 46.
  • FIG. 22 depicts Scheme 2 of the thioester strategy for macrocyclic compounds of the present invention.
  • FIG. 23 depicts the competitive binding curve for compound 8.
  • FIG. 24 depicts the competitive binding curve for compound 11
  • Specifically preferred compounds of the present invention include, but are not limited to:
  • the present invention is directed to a method of treating irritable bowel syndrome, dyspepsia, Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, diabetes mellitus, obesity, postgastroenterectomy syndrome, atrophic colitis or gastritis, gastric stasis, gastrointestinal dumping syndrome, celiac disease and eating disorders leading to obesity in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (I).
  • DMF, DCM and THF used are of DriSolv® (EM Science, now EMD Chemicals, Inc., part of Merck KgaA, Darmstadt, Germany) or synthesis grade quality except for (i) deprotection, (ii) resin capping reactions and (iii) washing.
  • NMP used for the amino acid (AA) coupling reactions is of analytical grade.
  • DMF was adequately degassed by placing under vacuum for a minimum of 30 min prior to use.
  • Tyr(3tBu) was synthesized following the method reported in JP2000 44595.
  • Cpa was made using literature methods (Tetrahedron: Asymmetry 2003, 14, 3575 ⁇ 3580) or obtained commercially.
  • Boc- and Fmoc-protected amino acids and side chain protected derivatives, including those of N-methyl and unnatural amino acids, were obtained from commercial suppliers or synthesized through standard methodologies known to those in the art.
  • Ddz-amino acids were either synthesized by standard procedures or obtained commercially from Orpegen (Heidelberg, Germany) or Advanced ChemTech (Louisville, Ky., USA).
  • Bts-amino acids were synthesized as described in Example 6. Hydroxy acids were obtained from commercial suppliers or synthesized from the corresponding amino acids by literature methods.
  • Analytical TLC was performed on pre-coated plates of silica gel 60F254 (0.25 mm thickness) containing a fluorescent indicator.
  • concentration/evaporated under reduced pressure indicates evaporation utilizing a rotary evaporator under either water aspirator pressure or the stronger vacuum provided by a mechanical oil vacuum pump as appropriate for the solvent being removed.
  • “Dry pack” indicates chromatography on silica gel that has not been pre-treated with solvent, generally applied on larger scales for purifications where a large difference in R f exists between the desired product and any impurities.
  • “dried in the standard manner” is that the resin is dried first in air (1 h), and subsequently under vacuum (oil pump usually) until full dryness is attained ( ⁇ 30 min to O/N).
  • reaction mixture was washed with Et 2 O. Washing is continued until the absence of non-polar impurities in the aqueous layer is confirmed by TLC (typically 3 ⁇ 100 mL).
  • the aqueous solution was then cooled to 0° C., acidified to pH 2.0 with 1 N HCl until no additional cloudiness forms, and extracted with EtOAc (3 ⁇ 100 mL).
  • EtOAc 3 ⁇ 100 mL
  • a mixture of DCM and EtOAc may be used as the extraction solvent, depending on the solubility of the product obtained from different amino acids or derivatives. Note that DCM cannot be used solely as solvent because of the emulsion formed during extraction.
  • Gly, Ala, D-Ala, ⁇ -Ala and GABA Use 1.5 eq of amino acid per eq of Bts-Cl, in order to prevent dibetsylation.
  • Met Carry out the reaction under N 2 to prevent oxidation.
  • Gln and Asn Due to the solubility of Bts-Gln and Bts-Asn, the work-up required is modified from the standard procedure: Upon completion of the reaction, the reaction mixture was washed with diethyl ether. Washing is continued until the absence of non-polar impurities in the aqueous layer is confirmed by TLC (typically 3 ⁇ 100 mL). The aqueous phase was then cooled to 0° C.
  • the compounds of Formula I can be synthesized using traditional solution synthesis techniques or solid phase chemistry methods. In either, the construction involves four phases: first, synthesis of the building blocks, including one to four moieties, comprising recognition elements for the biological target receptor, plus one tether moiety, primarily for control and definition of conformation. These building blocks are assembled together, typically in a sequential fashion, in a second phase employing standard chemical transformations. The precursors from the assembly are then cyclized in the third stage to provide the macrocyclic structures. Finally, a post-cyclization processing stage involving removal of protecting groups and optional purification then provides the desired final compounds (see FIG. 1 ). This method has been previously disclosed in WO 01/25257 and U.S. patent application Ser. No. 09/679,331. A general synthetic strategy is shown in FIG. 1 .
  • tether component required for compounds of the invention are synthesized as described in WO01/25257, U.S. Provisional Pat. Appl. Ser. No, 60/491,248 or herein.
  • a standard procedure for the synthesis of tether B is shown in FIG. 2 .
  • Step T8-1 Chlorotrimethylsilane (116 mL, 0.91 mol, 1.5 eq) was added to a suspension of 2-hydroxycinnamic acid (100 g, 0.61 mol, 1.0 eq) in MeOH (500 mL, HPLC grade) over 30 min at 0° C. The resulting mixture was stirred at rt O/N. The reaction was monitored by TLC (EtOAc/MeOH: 98/2). Heating the reaction mixture in a hot water can accelerate the process if necessary. After the reaction was completed, the reaction mixture was evaporated under reduced pressure to afford methyl 2-hydroxycinnamate as a white solid (108.5 g) in quantitative yield. The identity of this intermediate compound is confirmed by NMR.
  • Step T8-2 3,4-Dihydro-2H-pyran (DHP, 140 mL, 1.54 mol, 2.52 eq) was added dropwise to 2-bromoethanol (108 mL, 1.51 mol, 2.5 eq) in a 2 L three-neck flask with mechanical stirring at 0° C. over 2 h. The resulting mixture was stirred for additional 1 h at rt.
  • DHP 3,4-Dihydro-2H-pyran
  • 2-bromoethanol 108 mL, 1.51 mol, 2.5 eq
  • Methyl 2-hydroxycinnamate from Step T8-1 (108 g, 0.61 mol, 1.0 eq), potassium carbonate (92.2 g, 0.67 mol, 1.1 eq), potassium iodide (20 g, 0.12 mol, 0.2 eq) and DMF (300 mL, spectrometric grade) were added to the above flask.
  • the reaction mixture was stirred at 70° C. (external temperature) for 24 h.
  • the reaction was monitored by TLC (DCM/Et 2 O: 95/5).
  • the reaction was allowed to coolto rt and Et 2 O (450 mL) was added.
  • the inorganic salts were removed by filtration and washed with Et 2 O (3 ⁇ 50 mL).
  • Step T8 ⁇ 3 DIBAL (1.525 L, 1.525 mol, 2.5 eq, 1.0 M in DCM) was added slowly to a solution of the above crude ester from Step T8-2 (0.61 mol based on the theoretical yield) in anhydrous DCM (610 mL) at ⁇ 35° C. with mechanical stirring over 1.5 h.
  • Step T8-4 To a mixture of the allylic alcohol from Step T8-3 (28 g, 0.100 mol, 1.0 eq) and collidine (0.110 mol, 1.1 eq) in 200 mL of anhydrous DMF under N 2 was added anhydrous LiCl (4.26 g, 0.100 mol, 1.0 eq.) dissolved in 100 mL of anhydrous DMF. The mixture was then cooled to 0° C., and MsCI (12.67 g, 0.110 mol, 1.1 eq., freshly distilled over P 2 O 5 ), was added dropwise. The reaction was allowed to warm to rt and monitored by TLC (3:7 EtOAc/hex).
  • Step T8-5 PPh 3 (25.9 g, 0.099 mol, 1.5 eq) was added at 0° C. to a solution of the allylic azide from Step T8-4 (20.0 g, 0.066 mol, 1.0 eq.) in 100 mL of THF. The solution was stirred for 30 min at 0° C. and 20 h at rt. Water (12 mL) was then added and the resulting solution was heated at 60° C. for 4 h. The solution was cooled to rt, 2N HCl (15 mL) added and the mixture stirred for 90 min at 50° C. The separated organic phase was extracted with 0.05 N HCl (2 ⁇ 100 mL).
  • the combined aqueous phase was washed with Et 2 O (5 ⁇ 150 mL) and toluene (4 ⁇ 150 mL) (more extraction could be necessary, follow by TLC), which were combined and back-extracted with 0.05 N HCl (1 ⁇ 100 mL).
  • This acidic aqueous phase from back-extraction was combined with the main aqueous phase and washed with ether (5 ⁇ 150 mL) again.
  • the pH of the aqueous phase was then adjusted to 8-9 by the addition of sodium hydroxide (5 N). Care must be exercised to not adjust the pH above 9 due to the reaction conditions required by the next step.
  • the aqueous phase was concentrated under reduced pressure (aspirator, then oil pump) or lyophilized to dryness.
  • Step T8-6 A mixture of the crude amino alcohol from Step T8-5 (0.5 mol based on the theoretical yield), Ddz-OPh (174 g, 0.55 mol, 1.1 eq) and Et 3 N (70 mL, 0.5 mol, 1.0 eq) in DMF (180 mL) was stirred for 24 h at 50° C. Additional DMF is added if required to solubilize all materials. The reaction was monitored by TLC (hex/EtOAc: 50/50, ninhydrin detection).
  • reaction mixture was diluted with Et 2 O (1.5 L) and water (300 mL).
  • the separated aqueous phase was extracted with Et 2 O (2 ⁇ 150 mL).
  • the combined organic phase was washed with water (3 ⁇ 500 mL) and brine (1 ⁇ 500 mL), dried over MgSO 4 , filtered and the filtrate concentrated under reduced pressure.
  • the layers were monitored by TLC to ensure no product was lost into the aqueous layer. If so indicated, perform one or more additional extractions with Et 2 O of the aqueous phase to recover this material.
  • FIG. 3 A standard procedure for the synthesis of tether T9 is shown in FIG. 3 .
  • Tether T9 can also be synthesized from T8 by reduction as in step T9-3 or with other appropriate hydrogenation catalysts known to those in the art.
  • FIG. 4 A standard procedure for the synthesis of Ddz propargylamine is shown in FIG. 4 .
  • a solution of propargylamine (53.7 g, 0.975 mol, 1.5 eq) in degassed DMF (Drisolv, 388 mL) was treated with Ddz-N 3 (170.9 g, 0.65 mol, 1.0 eq), tetramethylguanidine (TMG, 81.4 mL, 0.65 mol, 1.0 eq) and DIPEA (113.1 mL, 0.65 mol, 1.0 eq) and stirred at 50° C. O/N.
  • TLC conditions:25/75 EtOAc/hex.
  • FIG. 5A A standard procedure for the synthesis of tether T10 is shown in FIG. 5A .
  • Two alternative routes to this tether have been developed.
  • the first synthetic approach proceeded starting from the commercially available monobenzoate of resorcinol (T10-0). Mitsunobu reaction under standard conditions with the protected amino alcohol from Example 9, followed by saponification of the benzoate provided T 10 - 1 in good yield after recrystallization. Alkylation of the phenol with 2-bromoethanol using the optimized conditions shown permitted the desired product Ddz-T10 to be obtained after dry pack purification in 42% yield.
  • a second synthetic route to T10 is shown in FIG. 5B .
  • FIG. 6 A standard procedure for the synthesis of tether T11 is shown in FIG. 6 .
  • FIG. 7 A standard procedure for the synthesis of tether T12 is shown in FIG. 7 .
  • the resin was filtered and washed 2 ⁇ DCM, 1 ⁇ toluene, 1 ⁇ EtOH, 1 ⁇ toluene, 1 ⁇ (DCM/MeOH), 1 ⁇ (THF/MeOH), 1 ⁇ (DCM/MeOH), 1 ⁇ (THF/MeOH), 2 ⁇ DCM, then dried in the standard manner.
  • the resin was filtered and washed DCM (2 ⁇ ), toluene (1 ⁇ ), EtOH (1 ⁇ ), toluene (1 ⁇ ), DCM/MeOH (1 ⁇ ), 1 ⁇ THF/MeOH (1 ⁇ ), DCM/MeOH (1 ⁇ ), THF/MeOH (1 ⁇ ), 2 ⁇ DCM, then dried in the standard manner.
  • FIG. 8 A procedure for the synthesis of PPh 3 -DIAD adduct is shown in FIG. 8 .
  • Example 29 In certain instances, the Mitsunobu process of Example 29 cannot be applied or is not efficient for incorporation of the tether. Hence, reductive amination has been developed as an alternative that can be employed for tether incorporation as illustrated hereinbelow for one of the preferred tethers. Similar chemistry can be used to incorporate other tethers of the present invention.
  • the Tether (30-2) with the amine protected as its Ddz derivative was efficiently oxidized to the corresponding aldehyde 30-2 using SO 3 ⁇ pyr in DMSO-Et 3 N-DCM.
  • This aldehyde (0.14 mmol, 56 mg, 1.5 eq based upon loading of resin support) was dissolved in a 1:3 mixture of TMOF-MeOH (DriSolv, 4 mL) at rt.
  • FIG. 10 A standard procedure for the synthesis of tether T28 is shown in FIG. 10 .
  • FIG. 11 A standard procedure for the synthesis of tether T32 is shown in FIG. 11 .
  • FIGS. 12A and 12B A standard procedure for the synthesis of tether T 33 a and T 33 b is shown in FIGS. 12A and 12B .
  • tether T34 ( FIG. 13 ), tether T35 ( FIG. 14 ), tether T36 ( FIG. 15 ), tether T37 ( FIG. 16 ), tether T38 ( FIG. 17 ), tether T39 ( FIG. 18 ), tether T40 ( FIG. 19 ), tether T41 ( FIG. 20 ) and tether T42 ( FIG. 21 ).
  • FIG. 22 presents a scheme depicting a thioester strategy for macrocyclic compounds of the present invention.
  • amino acids indicated can be replaced by corresponding hydroxy acids and coupled to the next building block utilizing methods known to those in the art.
  • the resin containing the cyclization precursor is combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, Foster City, Calif., commercially supplied MP-carbonate resin was treated with 3 ⁇ THF (1 L per 400 g) and dried O/N at 30° C. in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin).
  • a 0.2 M DIPEA solution in THF was then added to the combined resins (1 mL/60 mg MP-carbonate resin) and the suspension agitated O/N at rt. Subsequently, the resin was filtered and rinsed 2 ⁇ THF.
  • the combined filtrates are collected together in an appropriate vessel, then the volatile contents evaporated under reduced pressure [in addition to the standard methods, solvent can also be removed in vacuo using centrifugal evaporation (ThermoSavant Discovery®, SpeedVac® or comparable) (Thermo Electron Corporation, Waltham, Mass.)] to provide the crude macrocycles.
  • solvent can also be removed in vacuo using centrifugal evaporation (ThermoSavant Discovery®, SpeedVac® or comparable) (Thermo Electron Corporation, Waltham, Mass.)] to provide the crude macrocycles.
  • Example 47 Except for the cyclization itself and subsequent work-up, this procedure is identical to that of Example 47.
  • the resin containing the cyclization precursor was combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, commercially supplied MP-carbonate resin was treated with THF (3 ⁇ , 1 L per 400 g) and dried O/N at 30° C. in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin). To this was added THF (1 mL per 100 mg resin) and silver trifluoroacetate (1 eq relative to the initial loading of the resin). Finally, an amount of DIPEA sufficient to obtain a 0.2 M solution was added. The reaction mixture was agitated at rt O/N.
  • a ring-closing metathesis (RCM) strategy as developed by Grubbs et al. can also be used to access some of the macrocyclic compounds of the invention (see for example US 5,811,515; Grubbs, R. H. et al. J. Org. Chem. 2001, 66, 5291-5300; Princestner, A. Angew. Chem. Int. Ed. 2000, 39, 3012 ⁇ 3043).
  • the reaction was best performed after formation of the macrocyclic ring.
  • further reaction of amino moieties on side chains examples was typically efficiently done by reaction of the partially protected macrocycle.
  • acylation, sulfonylation, alkylation (via reductive amination), guanidine and urea formation were performed via standard methods.
  • Table 1 hereinbelow, shows a representative, but by no means exclusive, summary of the chemical synthesis of several representative compounds of the invention.
  • HPLC analyses are performed on a Waters Alliance® system 2695 running at 1 mL/min using an Xterra MS C18 column 4.6 ⁇ 50 mm (3.5 ⁇ m).
  • a Waters 996 PDA provided UV data for purity assessment (Waters Corporation, Milford, Mass.).
  • the second part (40%) went to an evaporative light scattering detector (ELSD, Polymer Laboratories, now part of Varian, Inc., Palo Alto, Calif., PL-ELS-1000TM) for purity assessment and the last portion (10%) to a chemiluminescence nitrogen detector (CLND, Antek® Model 8060, Antek Instruments, Houston, Tex., part of Roper Industries, Inc., Duluth, Ga.) for quantitation and purity assessment. Data was captured and processed utilizing the most recent version of the Waters Millenium® software package (Milford, Mass.).
  • ELSD evaporative light scattering detector
  • CLND chemiluminescence nitrogen detector
  • An example LC method suitable for compounds of the present invention uses MeOH as solvent A, H 2 O as solvent B and 1% TFA/H 2 O as solvent D.
  • Initial mobile-phase composition is 5% A, 85% B and 10% D. Details of the standard gradient method are shown below:
  • the compounds of the present invention were evaluated for their ability to interact at the human motilin receptor utilizing a competitive radioligand binding assay as described in Method B1. Further characterization of the interaction can be performed utilizing the functional assays described in Methods B2, B3 and B4. Some of these methods can be conducted, if so desired, in a high throughput manner to permit the simultaneous evaluation of many compounds.
  • Other assays have also been described that are suitable for HTS, such as that based upon the stable expression of a synthetic gene for the human motilin receptor.
  • the compounds are preferably tested in the Aequorin assay as described in Method B2, although the procedure of Method B3 is also applicable.
  • the representative compounds examined act as antagonists at the motilin receptor and are devoid of agonist activity at the concentrations studied.
  • Method B2 measures the actual signaling event, which makes it more relevant to the effect that is desired, whereas the assay of Method B3 simply measures GTP turnover.
  • a common and scientifically-accepted ex vivo assay for the measurement of agonist or antagonist activity at the motilin receptor is the contraction of rabbit duodenum or other gastrointestinal smooth muscle tissue.
  • A2-A4 Agonists are defined as compounds that induce>50% contraction relative to the motilin peptide, whereas antagonists are defined as compounds that cause>50% inhibition of the response to motilin.
  • Gastric motility is generally measured in the clinical setting as the time required for gastric emptying and subsequent transit time through the GI tract.
  • Gastric emptying scans are well known to those skilled in the art an, briefly, comprise use of an oral contrast agent, such as barium, or a radiolabeled meal. Solid and liquids can be measured independently.
  • a test food or liquid is radiolabeled with an isotope ( 99m Tc) and after ingestion or administration, transit time through the GI tract and gastric emptying are measured by visualization using gamma cameras.
  • Example Method B1 Competitive Radioligand Binding Assay (Motilin Receptor)
  • the reaction is initiated by addition of 10 ⁇ l of [ 125 I]-motilin (final conc. 0.04-0.06 nM) to each well. Plates are sealed with TopSeal-A, vortexed gently and incubated at room temperature for 2 hours.
  • the reaction is arrested by filtering samples through pre-soaked (0.3% polyethyleneimine, 2 h) Multiscreen Harvest plates using a Tomtec® Harvester (Tomtec, Hamden, Conn.)), washed 9 times with 500 ⁇ L of cold 50 mM Tris-HCl (pH 7.4), and than plates are air-dried in a fumehood for 30 minutes. A bottom seal is applied to the plates prior to the addition of 25 ⁇ L of MicroScint-0TM to each well. Plates are then sealed with TopSeal-A® and counted for 30 sec per well on a TopCount® Microplate Scintillation and Luminescence Counter (PerkinElmer, Wellesley, Mass.) where results are expressed as counts per minute (cpm).
  • K i values were calculated using a K d value of 0.16 nM for [ 125 I]-motilin (previously determined during membrane characterization).
  • Example Method B2 Aequorin Functional Assay (Motilin Receptor)
  • Compounds were provided as dry films at a quantity of approximately 1.2 ⁇ mol in pre-formatted 96-well plates. Compounds were dissolved in 100% DMSO at a concentration of 10 mM and stored at ⁇ 20° C. until further use. Daughter plates were prepared at a concentration of 500 ⁇ M in 30% DMSO with 0.1% BSA and stored at ⁇ 20° C. until testing. On the test day, compounds were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO concentration in the assay was 0.6%.
  • Cells are collected from culture plates with Ca 2+ and Mg 2+ -free phosphate buffered saline (PBS) supplemented with 5 mM EDTA, pelleted for 2 minutes at 1000 ⁇ g, resuspended in assay buffer (see above) at a density of 5 ⁇ 10 6 cells/mL and incubated overnight in the presence of 5 ⁇ M coelenterazine. After loading, cells were diluted with assay buffer to a concentration of 5 ⁇ 10 5 cells/mL.
  • PBS Ca 2+ and Mg 2+ -free phosphate buffered saline
  • assay buffer see above
  • agonist testing 50 ⁇ l of the cell suspension was mixed with 50 ⁇ l of the appropriate concentration of test compound or motilin (reference agonist) in 96-well plates (duplicate samples). The emission of light resulting from receptor activation was recorded using the Functional Drug Screening System 6000 ‘FDSS 6000’ (Hamamatsu Photonics K.K., Japan).
  • an approximate EC80 concentration of motilin i.e. 0.5 nM; 100 ⁇ L was injected onto the cell suspension containing the test compounds (duplicate samples) 15 ⁇ 30 minutes after the end of agonist testing and the consequent emission of light resulting from receptor activation was measured as described in the paragraph above.
  • E the measured RLU value at a given agonist concentration (C)
  • E max the maximal response
  • EC 50 the concentration producing 50% stimulation
  • n is the slope index.
  • results for agonist testing results for each concentration of test compound were expressed as percent activation relative to the signal induced by motilin at a concentration equal to the EC 80 (i.e. 0.5 nM). For antagonist testing, results for each concentration of test compound were expressed as percent inhibition relative to the signal induced by motilin at a concentration equal to the EC 80
  • CHO membranes were immobilized into 96-well FlashPlate® microplates.
  • Test compound, GTP ⁇ S, motilin and [ 35 S]-GTP ⁇ S were combined in each well according to the Assay Volumes described above.
  • the reaction is initiated by addition of 100 mL of [ 36 S]-GTP ⁇ S to each well. Each plate is sealed (TopSeal-ATM) and incubated in the dark at room temperature for 150 min. Then, plates are counted for 30 seconds per well on the TopCount® NXT.
  • Top and Bottom correspond to the top and bottom values of the dose-response curve calculated by GraphPadTM Prism®).
  • Duodenal segments were vertically suspended in organ chambers of 10 mL filled with Krebs buffer and connected to an isotonic force transducer, with a preload of 1 g. After a stabilization period, the muscle strips were challenged with 10 ⁇ 4 M acetylcholine and washed. This was repeated until a stable maximal contraction was obtained (2-3 times), with an interval of at least 20 minutes.
  • test compounds were added to the bath. After 15 min incubation, a dose response to motilin was recorded by adding logarithmically increasing concentrations of motilin to the bath (final concentration 10 ⁇ 9 to 10 .6 M). A blank experiment (no test compound present) was also performed. At the end of the dose response curve, a supramaximal dose of acetylcholine (10 ⁇ 4 M) was given and this response was used as a reference (100% contraction).

Abstract

The present invention is directed to novel macrocyclic compounds of formula (I) and their pharmaceutically acceptable salts, hydrates or solvates:
Figure US20120226067A1-20120906-C00001
wherein R1, R2, R3, R4, R5, R6 , n1, m, p Z1, Z2, and Z3 are as describe in the specification. The invention also relates to compounds of of formula (I) which are antagonists of the motilin receptor and are useful in the treatment of disorders associated with this receptor and with or with motility dysfunction.

Description

    RELATED APPLICATION INFORMATION
  • This application is a divisional application of U.S. patent application Ser. No. 12/273,648, filed Nov. 19, 2008, now U.S. Pat. No. 8,129,561, which is a continuation application of U.S. patent application Ser. No. 12/273,638, filed Nov. 19, 2008, which is a continuation application of U.S. patent application Ser. No. 10/872,142, filed Jun. 18, 2004, now U.S. Pat. No. 7,521,420, which claims the benefit of U.S. patent application Ser. No. 60/479,223, filed Jun. 18, 2003. The disclosure of each application is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to novel conformationally-defined macrocyclic compounds, pharmaceutical compositions comprising same and intermediates used in their manufacture. More particularly, the invention relates to macrocyclic compounds that have been demonstrated to selectively antagonize the activity of the motilin receptor. The invention further relates to macrocyclic compounds useful as therapeutics for a range of gastrointestinal disorders, in particular those in which malfunction of gastric motility or increased motilin secretion is observed, such as hypermotilinemia, irritable bowel syndrome and dyspepsia.
  • BACKGROUND OF THE INVENTION
  • A number of peptide hormones are involved in the control of the different functions in the gastrointestinal (GI) tract, including absorption, secretion, blood flow and motility (Mulvihill, et al. in Basic and Clinical Endocrinology, 4th edition, Greenspan, F. S.; Baxter, J. D., eds., Appleton & Lange: Norwalk, Conn., 1994, pp 551-570). Since interactions between the brain and GI system are critical to the proper modulation of these functions, these peptides can be produced locally in the GI tract or distally in the CNS.
  • One of these peptide hormones, motilin, a linear 22-amino acid peptide, plays a critical regulatory role in the GI physiological system though governing of fasting gastrointestinal motor activity. As such, the peptide is periodically released from the duodenal mucosa during fasting in mammals, including humans. More precisely, motilin exerts a powerful effect on gastric motility through the contraction of gastrointestinal smooth muscle to stimulate gastric emptying, decrease intestinal transit time and initiate phase III of the migrating motor complex in the small bowel (Itoh, Z., Ed., Motilin, Academic Press: San Diego, Calif., 1990, ASIN: 0123757304; Nelson, D. K. Dig. Dis. Sci. 1996, 41, 2006-2015; Peeters, T. L.; Vantrappen, G.; Janssens, J. Gastroenterology 1980, 79, 716-719).
  • Motilin exerts these effects through receptors located predominantly on the human antrum and proximal duodenum, although its receptors are found in other regions of the GI tract as well (Peeters, T. L.; Bormans, V.; Vantrappen, G. Regul. Pept. 1988, 23, 171-182). Therefore, motilin hormone is involved in motility of both the upper and lower parts of the GI system (Williams et al. Am. J. Physiol. 1992, 262, G50-G55). In addition, motilin and its receptors have been found in the CNS and periphery, suggesting a physiological role in the nervous system that has not yet been definitively elucidated (Depoortere, I.; Peeters, T. L. Am. J. Physiol. 1997, 272, G994-999 and O'Donohue, T. L. et al. Peptides 1981, 2, 467-477). For example, motilin receptors in the brain have been suggested to play a regulatory role in a number of CNS functions, including feeding and drinking behavior, micturition reflex, central and brain stem neuronal modulation and pituitary hormone secretion (Itoh, Z. Motilin and Clinical Applications. Peptides 1997, 18, 593-608; Asakawa, A.; Inui, A.; Momose, K.; et al., M. Peptides 1998, 19, 987-990 and Rosenfeld, D. J.; Garthwaite, T. L. Physiol. Behay. 1987, 39, 753-756). Physiological studies have provided confirmatory evidence that motilin can indeed have an effect on feeding behavior (Rosenfeld, D. J.; Garthwaite, T. L. Phys. Behay. 1987, 39, 735-736).
  • The recent identification and cloning of the human motilin receptor (WO 99/64436) has simplified and accelerated the search for agents which can modulate its activity for specific therapeutic purposes.
  • Due to the critical and direct involvement of motilin in control of gastric motility, agents that either diminish (hypomotility) or enhance (hypermotility) the activity at the motilin receptor, are a particularly attractive area for further investigation in the search for new effective pharmaceuticals towards these indications.
  • Peptidic agonists of the motilin receptor, which have clinical application for the treatment of hypomotility disorders, have been reported (U.S. Pat. No. 5,695,952; 5,721,353; 6,018,037; 6,380,158; 6,420,521, U.S. Appl. 2001/0041791, WO 98/42840; WO 01/00830 and WO 02/059141). Derivatives of erythromycin, commonly referred to as motilides, have also been reported as agonists of the motilin receptor (U.S. Pat. Nos. 4,920,102; 5,008,249; 5,175,150; 5,418,224; 5,470,961; 5,523,401, 5,554,605; 5,658,888; 5,854,407; 5,912,235; 6,100,239; 6,165,985; 6,403,775).
  • Antagonists of the motilin receptor are potentially extremely useful as therapeutic treatments for diseases associated with hypermotility and hypermotilinemia, including irritable bowel syndrome, dyspepsia, gastroesophogeal reflux disorders, Crohn's disease, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, diabetes mellitus, obesity, malabsorption syndrome, carcinoid syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome, gastric stasis and eating disorders leading to obesity.
  • A variety of peptidic compounds have been described as antagonists of the motilin receptor (Depoortere, I.; Macielag, M. J.; Galdes, A.; Peeters, T. L. Eur. J. Pharmacol. 1995, 286, 241-247; U.S. Pat. Nos. 5,470,830; 6,255,285; 6,586,630; 6,720,433; U.S. 2003/0176643; WO 02/64623). These peptidic antagonists suffer from the known limitations of peptides as drug molecules, in particular poor oral bioavailability and degradative metabolism.
  • Cyclization of peptidic derivatives is a method employed to improve the properties of a linear peptide both with respect to metabolic stability and conformational freedom. Cyclic molecules tend to be more resistant to metabolic enzymes. Such cyclic tetrapeptide motilin antagonists have been reported (Haramura, M. et al J. Med. Chem. 2002, 45, 670-675, U.S. 2003/0191053; WO 02/16404).
  • Other motilin antagonists, which are non-peptidic and non-cyclic in nature have also been reported (U.S. Pat. Nos. 5,972,939; 6,384,031; 6,392,040; 6,423,714; 6,511,980; 6,624,165; 6,667,309; U.S. 2002/0111484; 2001/041701; 2002/0103238; 2001/0056106, 2002/0013352; 2003/0203906 and 2002/0002192)
  • The macrocyclic motilin antagonists of the present invention comprise elements of both peptidic and non-peptidic structures in a combination which has not been pursued for this application previously.
  • Indeed, the structural features of antagonists of the present invention are different. In particular, within the known motilin antagonists which are cyclic peptides, it was found that such derivatives containing D-amino acids were devoid of activity. In contrast, for the tripeptidomimetic compounds of the present invention, the D-stereochemistry is required for two of the three building elements.
  • The motilin antagonists of the present invention are also distinct from the prior art in that they comprise a tether element to fulfill the dual role of controlling conformations and providing additional sites for interaction either through hydrophobic interactions, hydrogen bonding or dipole-dipole interactions.
  • SUMMARY OF THE INVENTION
  • In a first aspect, the present invention is directed to compounds of formula (I):
  • Figure US20120226067A1-20120906-C00002
  • and pharmaceutically acceptable salts, hydrates or solvates thereof wherein:
    Z1, Z2 and Z3 are independently selected from the group consisting of O, N and NR10, wherein R10 is selected from the group consisting of hydrogen, lower alkyl, and substituted lower alkyl;
    R1 is independently selected from the group consisting of lower alkyl substituted with aryl, lower alkyl substituted with substituted aryl, lower alkyl substituted with heteroaryl and lower alkyl substituted with substituted heteroaryl;
    R2 is hydrogen;
  • R3 is independently selected from the group consisting of alkyl and cycloalkyl with the proviso that when Z1 is N, R3 can form a four, five, six or seven-membered heterocyclic ring together with Z1;
  • R4 is hydrogen;
    R5 and R6 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl and substituted heteroaryl, with the proviso that at least one of R5 and R6 is hydrogen;
    X is selected from the group consisting of O, NR8, and N(R9)2 +;
      • wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and amidino; and
      • R9 is selected from the group consisting of hydrogen, lower alkyl, and substituted lower alkyl;
        m, n1 and p are independently selected from 0, 1 or 2; and
        T is a bivalent radical of formula II:

  • —U—(CH2)d—W—Y—Z—(CH2)e—  (II)
      • wherein d and e are independently selected from 0, 1, 2, 3, 4 or 5;
      • wherein U is bonded to X of formula (I) and is —CH2— or —C(═O)—;
      • wherein Y and Z are each optionally present;
      • W, Y and Z are independently selected from the group consisting of: —O—, —NR28—, —S—, —SO—, —SO2—, —C(═O)—, —C(═O)—O—, —O—C(═O)—, —C(═O)—NH—, —NH—C(═O)—, —SO2—NH—, —NH—SO2—, —CR29R30—, —CH═CH— with a configuration Z or E, and -GEC-, or from a ring structure independently selected from the group:
  • Figure US20120226067A1-20120906-C00003
  • wherein any carbon atom contained within said ring structure, can be replaced by a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
    G1 and G2 each independently represent a covalent bond or a bivalent radical selected from the group consisting of —O—, —S—, —SO—, —SO2—, —C(═O)—, —C(═O)—O—, —O—C(═O)—, —C(═O)NH—, —NH—C(═O)—, —SO2—NH—, —NH—SO2—, —CR42R43—, —CH═CH— with a configuration Z or E, and —C═C—;with the proviso that G1 is bonded closer to U than G2;
    K1, K2, K3, K4, K6, K15 and K16 are independently selected from the group consisting of O, NR44 and S;
    f is selected from 1, 2, 3, 4, 5 or 6;
    R31, R32, R38, R39, R48 and R49 are independently selected from hydrogen, halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido; and
    R33, R34, R35, R36, R37, R47, R50 and R51 are independently selected from hydrogen, halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido.
  • In a second aspect, the invention also proposes compounds of formula (1) which are antagonists of the motilin receptor .
  • In a third aspect, the invention proposes a method of treating a disorder associated with the motilin receptor or motility dysfunction in humans and other mammals, comprising administering a therapeutically effective amount of a compound of formula (1).
  • While the invention will be described in conjunction with example embodiments, it will be understood that it is not intended to limit the scope of the invention to such embodiment. On the contrary, it is intended to cover all alternatives, modifications and equivalents as may be included as defined by the appended claims.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Preferably in formula (I), as depicted hereinabove, R1 is selected from the group consisting of —(CH2)qR11, and —CHR12R13
      • wherein q is 0, 1, 2 or 3; and
      • R11 and R12 are independently selected from a ring structure from the following group:
  • Figure US20120226067A1-20120906-C00004
      • wherein any carbon atom in said ring structure can be replaced a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
      • A1, A2, A3, A4 and A5 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido;
      • B1, B2, B3, and B4 are independently selected from NR14, S or O, wherein R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido;
      • R13 is as defined for as R11 and R12 or is selected from the group comprising lower alkyl, substituted lower alkyl, hydroxy, alkoxy, aryloxy, amino, carboxy, carboxyalkyl, carboxyaryl, and amido.
        wherein A1, A2, A3, A4 and A5 are most preferably selected from halogen, trifluroromethyl, C1-6 alkyl or C1-6 alkoxy.
  • Preferably, R11, R12 and R13 are selected from the group consisting of:
  • Figure US20120226067A1-20120906-C00005
  • wherein Ra and Rb are chosen from the group consisting of Cl, F, CF3, OCH3, OH, and C(CH3)3 and CH3.
  • Also preferably, R3 in formula (I), is selected from the group consisting of:
      • (CH2),CH3, —CH(CH3)(CH2)tCH3, —CH(OR15)CH3, —CH2SCH3 —CH2CH2SCH3, —CH2S(═O)CH3, —CH2CH2S(═O)CH3, —CH2S(═O)2CH3, —CH2CH2S(═O)2CH3, —(CH2)uCH(CH3)2, —C(CH3)3, and —(CH2)y—R21, wherein:
        • s and u are independently selected from 0, 1, 2, 3, 4 or 5;
        • t is independently selected from 1, 2, 3 or 4;
        • y is selected from 0, 1, 2, 3 or 4;
        • R15 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl and acyl;
        • R21 is selected from a ring structure selected from the following group:
  • Figure US20120226067A1-20120906-C00006
        • wherein any carbon atom in said ring structure can be replaced by a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
        • z is selected from 1, 2, 3, 4 or 5;
        • E1, E2 and E3 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido; and
        • J is optionally present and is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido.
  • The tether portion (T) of formula (I) is preferably selected from the group consisting of:
  • Figure US20120226067A1-20120906-C00007
  • wherein L1 is O, NH or NMe; L2 is CH or N; L3 is CH or N; L4 is 0 or CH2; L5 is CH or N is CR52R53 or 0; R46 is H or CH3;
    R52, R53, R54, R55, R56 and R57 are independently selected from hydrogen, lower alkyl, substituted lower alkyl, hydroxy, alkoxy, aryloxy, amino, and oxo; or R52 together with R53 or R54 together with R55 or R56 together with R57 can independently form a three to seven-membered cyclic ring comprising carbon, oxygen, sulfur and/or nitrogen atoms;
    (X) is the site of a covalent bond to X in formula (I); and
    (Z3) is the site of a covalent bond to Z3 in formula (I).
  • In a particularly preferred embodiment of the invention, there are provided compounds of formula (I) wherein m, n and p are O, X, Z1, Z2 and Z3 are NH and R2, R4 and R5 are hydrogen, represented by formula (III):
  • Figure US20120226067A1-20120906-C00008
  • According to another aspect of the invention, there are provided compounds of formula (I) wherein when Z1 is a nitrogen atom, R3 forms a four, five, six or seven-membered heterocyclic ring together with Z1 ,represented by formula (IV):
  • Figure US20120226067A1-20120906-C00009
  • wherein said heterocyclic ring may contain a second nitrogen atom, or an oxygen, or sulfur atom;
    n2 is selected from 0, 1, 2 or 3
  • R7 is optionally present and is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido.
  • It is to be understood, that in the context of the present invention, the terms amino, guanidine, ureido and amidino encompass substituted derivatives thereof as well.
  • Preferably, the invention provides a method of treating a disorder associated with hypermotility or hypermotilinemia in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (1).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts Scheme 1 presenting a general synthetic strategy to conformationally-defined macrocycles of the present invention.
  • FIG. 2 depicts the standard procedure for the synthesis of tether T8 of Example 16.
  • FIG. 3 depicts the standard procedure for the synthesis of tether T9 of Example 17.
  • FIG. 4 depicts the standard procedure for the synthesis of Ddz-propargylamine of Example 18.
  • FIG. 5A depicts the standard procedure for the synthesis of tether T10 of Example 19.
  • FIG. 5B depicts the second synthetic route to tether T10 of Example 19.
  • FIG. 6 depicts the standard procedure for the synthesis of Tether T11 of Example 20.
  • FIG. 7 depicts the standard procedure for the synthesis of tether T12 of Example 26.
  • FIG. 8 depicts the procedure for synthesis of PPh3-DIAD adduct of Example 29-C.
  • FIG. 9 depicts the standard procedure for attachment of tethers via reductive amination of Example 30.
  • FIG. 10 depicts the standard procedure for the synthesis of tether T28 of Example 32.
  • FIG. 11 the standard procedure for the synthesis of tether T32 of Example 36.
  • FIGS. 12A, 12B depict the standard procedure for the synthesis of tether T33a and T33b of Example 37.
  • FIG. 13 depicts the standard procedure for the synthesis of tether T34 of Example 38.
  • FIG. 14 depicts the standard procedure for the synthesis of tether T35 of Example 39.
  • FIG. 15 depicts the standard procedure for the synthesis of tether T36 of Example 40.
  • FIG. 16 depicts the standard procedure for the synthesis of tether T37 of Example 41.
  • FIG. 17 depicts the standard procedure for the synthesis of tether T38 of Example 42. Chiral T38 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • HPLC (standard gradient) tR=8.46 min
    Chiral material can be accessed by starting with the chiral epoxide. For example, the (S)-isomer of T38 was constructed in 89% overall yield from (S)-propylene oxide.
  • FIG. 18 depicts the standard procedure for the synthesis of tether T39 of Example 43. Chiral T39 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • FIG. 19 depicts the standard procedure for the synthesis of tether T40 of Example 44. Chiral T40 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
  • FIG. 20 depicts the standard procedure for the synthesis of tether T41 of Example 45.
  • FIG. 21 depicts the standard procedure for the synthesis of tether T42 of Example 46.
  • FIG. 22 depicts Scheme 2 of the thioester strategy for macrocyclic compounds of the present invention.
  • FIG. 23 depicts the competitive binding curve for compound 8.
  • FIG. 24 depicts the competitive binding curve for compound 11
  • DESCRIPTION OF PREFERRED EMBODIMENTS
  • Although preferred embodiments of the present invention have been described in detail herein and illustrated in the accompanying structures, schemes and tables, it is to be understood that the invention is not limited to these precise embodiments and that various changes and modifications may be effected therein without departing from the scope or spirit of the present invention.
  • Specifically preferred compounds of the present invention, include, but are not limited to:
  • Figure US20120226067A1-20120906-C00010
    Figure US20120226067A1-20120906-C00011
    Figure US20120226067A1-20120906-C00012
    Figure US20120226067A1-20120906-C00013
    Figure US20120226067A1-20120906-C00014
    Figure US20120226067A1-20120906-C00015
    Figure US20120226067A1-20120906-C00016
    Figure US20120226067A1-20120906-C00017
    Figure US20120226067A1-20120906-C00018
    Figure US20120226067A1-20120906-C00019
    Figure US20120226067A1-20120906-C00020
    Figure US20120226067A1-20120906-C00021
    Figure US20120226067A1-20120906-C00022
    Figure US20120226067A1-20120906-C00023
  • In addition to the preferred tethers (T) illustrated previously, other specific tethers employed for compounds of the invention are shown hereinbelow:
  • Figure US20120226067A1-20120906-C00024
    Figure US20120226067A1-20120906-C00025
  • In a preferred embodiment, the present invention is directed to a method of treating irritable bowel syndrome, dyspepsia, Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, diabetes mellitus, obesity, postgastroenterectomy syndrome, atrophic colitis or gastritis, gastric stasis, gastrointestinal dumping syndrome, celiac disease and eating disorders leading to obesity in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (I).
  • Synthetic Methods A. General Information
  • Reagents and solvents were of reagent quality or better and were used as obtained from various commercial suppliers unless otherwise noted, DMF, DCM and THF used are of DriSolv® (EM Science, now EMD Chemicals, Inc., part of Merck KgaA, Darmstadt, Germany) or synthesis grade quality except for (i) deprotection, (ii) resin capping reactions and (iii) washing. NMP used for the amino acid (AA) coupling reactions is of analytical grade. DMF was adequately degassed by placing under vacuum for a minimum of 30 min prior to use. Tyr(3tBu) was synthesized following the method reported in JP2000 44595. Cpa was made using literature methods (Tetrahedron: Asymmetry 2003, 14, 3575−3580) or obtained commercially. Boc- and Fmoc-protected amino acids and side chain protected derivatives, including those of N-methyl and unnatural amino acids, were obtained from commercial suppliers or synthesized through standard methodologies known to those in the art. Ddz-amino acids were either synthesized by standard procedures or obtained commercially from Orpegen (Heidelberg, Germany) or Advanced ChemTech (Louisville, Ky., USA). Bts-amino acids were synthesized as described in Example 6. Hydroxy acids were obtained from commercial suppliers or synthesized from the corresponding amino acids by literature methods. Analytical TLC was performed on pre-coated plates of silica gel 60F254 (0.25 mm thickness) containing a fluorescent indicator. The term “concentrated/evaporated under reduced pressure” indicates evaporation utilizing a rotary evaporator under either water aspirator pressure or the stronger vacuum provided by a mechanical oil vacuum pump as appropriate for the solvent being removed. “Dry pack” indicates chromatography on silica gel that has not been pre-treated with solvent, generally applied on larger scales for purifications where a large difference in Rf exists between the desired product and any impurities. For solid phase chemistry processes, “dried in the standard manner” is that the resin is dried first in air (1 h), and subsequently under vacuum (oil pump usually) until full dryness is attained (˜30 min to O/N).
  • B. Synthetic Methods for Building Blocks of the Invention Example 6 Standard Procedure for the Synthesis of Bts-Amino Acids
  • Figure US20120226067A1-20120906-C00026
  • To a solution of the amino acid or amino acid derivative (0.1 mol, 1.0 eq) in 0.25 N sodium hydroxide (0.08 mol, 0.8 eq) with an initial pH of approximately 9.5 (pH meter) at rt, solid Bts-Cl (0.11 mol, 1.1 eq) was added in one portion. The resulting suspension was stirred vigorously for 2-3 d. The pH of the reaction should be adjusted with 5.0 N sodium hydroxide as required to remain within the range 9.5-10.0 during this time. Typically, the pH has to be adjusted every 20−30 min during the first 5 h. Once the pH stops dropping, it is an indication that the reaction is almost complete. This can be confirmed by TLC (EtOAc:MeOH, 95:5). Upon completion, the reaction mixture was washed with Et2O. Washing is continued until the absence of non-polar impurities in the aqueous layer is confirmed by TLC (typically 3×100 mL). The aqueous solution was then cooled to 0° C., acidified to pH 2.0 with 1 N HCl until no additional cloudiness forms, and extracted with EtOAc (3×100 mL). Alternatively, a mixture of DCM and EtOAc may be used as the extraction solvent, depending on the solubility of the product obtained from different amino acids or derivatives. Note that DCM cannot be used solely as solvent because of the emulsion formed during extraction. The combined organic phases were washed with brine (2×150 mL), dried over MgSO4, filtered and evaporated under reduced pressure. DCM (1×) and hexanes (2×) were evaporated from the residue in order to ensure complete removal of the EtOAc and give the desired compound as a solid in 55-98% yield.
  • The following are modifications that have proven useful for certain amino acids:
  • Gly, Ala, D-Ala, β-Ala and GABA: Use 1.5 eq of amino acid per eq of Bts-Cl, in order to prevent dibetsylation.
    Met: Carry out the reaction under N2 to prevent oxidation.
    Gln and Asn: Due to the solubility of Bts-Gln and Bts-Asn, the work-up required is modified from the standard procedure: Upon completion of the reaction, the reaction mixture was washed with diethyl ether. Washing is continued until the absence of non-polar impurities in the aqueous layer is confirmed by TLC (typically 3×100 mL). The aqueous phase was then cooled to 0° C. and acidified to pH 2.0 with 6 N HCl. 6 N HCl was employed to minimize the volume of the solution due to the water solubility of Bts-Gln and Bts-Asn. (They are, in contrast, difficult to dissolve in DCM, EtOAc or chloroform.) The solution was maintained at 0° C. for 10 min and the product was collected by filtration as a white precipitate. The solid was washed with cold water (1×), cold brine (2×) and water (1×, 25° C.). The pH of this wash was taken, if it is not approximately 4, the solid was washed again with water. Finally, the solid was washed with cold EtOAc, then with cold Et2O (2×), and finally dried under vacuum (oil pump) (83-85% yield).
  • C. General Synthetic Strategy to Conformationally-Defined Macrocycles of the Present Invention
  • The compounds of Formula I can be synthesized using traditional solution synthesis techniques or solid phase chemistry methods. In either, the construction involves four phases: first, synthesis of the building blocks, including one to four moieties, comprising recognition elements for the biological target receptor, plus one tether moiety, primarily for control and definition of conformation. These building blocks are assembled together, typically in a sequential fashion, in a second phase employing standard chemical transformations. The precursors from the assembly are then cyclized in the third stage to provide the macrocyclic structures. Finally, a post-cyclization processing stage involving removal of protecting groups and optional purification then provides the desired final compounds (see FIG. 1). This method has been previously disclosed in WO 01/25257 and U.S. patent application Ser. No. 09/679,331. A general synthetic strategy is shown in FIG. 1.
  • D. Procedures for the Synthesis of Representative Tethers of the Present Invention
  • The important tether component required for compounds of the invention are synthesized as described in WO01/25257, U.S. Provisional Pat. Appl. Ser. No, 60/491,248 or herein. A standard procedure for the synthesis of tether B is shown in FIG. 2.
  • Step T8-1: Chlorotrimethylsilane (116 mL, 0.91 mol, 1.5 eq) was added to a suspension of 2-hydroxycinnamic acid (100 g, 0.61 mol, 1.0 eq) in MeOH (500 mL, HPLC grade) over 30 min at 0° C. The resulting mixture was stirred at rt O/N. The reaction was monitored by TLC (EtOAc/MeOH: 98/2). Heating the reaction mixture in a hot water can accelerate the process if necessary. After the reaction was completed, the reaction mixture was evaporated under reduced pressure to afford methyl 2-hydroxycinnamate as a white solid (108.5 g) in quantitative yield. The identity of this intermediate compound is confirmed by NMR. This reaction can be carried out on larger (kg) scale with similar results
    Step T8-2: 3,4-Dihydro-2H-pyran (DHP, 140 mL, 1.54 mol, 2.52 eq) was added dropwise to 2-bromoethanol (108 mL, 1.51 mol, 2.5 eq) in a 2 L three-neck flask with mechanical stirring at 0° C. over 2 h. The resulting mixture was stirred for additional 1 h at rt. Methyl 2-hydroxycinnamate from Step T8-1 (108 g, 0.61 mol, 1.0 eq), potassium carbonate (92.2 g, 0.67 mol, 1.1 eq), potassium iodide (20 g, 0.12 mol, 0.2 eq) and DMF (300 mL, spectrometric grade) were added to the above flask. The reaction mixture was stirred at 70° C. (external temperature) for 24 h. The reaction was monitored by TLC (DCM/Et2O: 95/5). The reaction was allowed to coolto rt and Et2O (450 mL) was added. The inorganic salts were removed by filtration and washed with Et2O (3×50 mL). The filtrate was diluted with hexanes (400 mL) and washed with water (3×500 mL), dried over MgSO4, filtered and the filtrate evaporated under reduced pressure. The crude ester (desired product and excess Br—C2H4-OTHP) was used for the subsequent reduction without further purification.
    Step T8−3: DIBAL (1.525 L, 1.525 mol, 2.5 eq, 1.0 M in DCM) was added slowly to a solution of the above crude ester from Step T8-2 (0.61 mol based on the theoretical yield) in anhydrous DCM (610 mL) at −35° C. with mechanical stirring over 1.5 h. The resulting mixture was stirred for 1.5 h at −35° C., then 1.5 h at 0° C. The reaction was monitored by TLC (hex/EtOAc: 50/50). When complete, Na2SO4·10 H2O (100 g, 0.5 eq) was slowly added; hydrogen evolution was observed, when it subsided water was added (100 mL). The mixture was warmed to it and stirred for 10 min, then warmed to 40° C. with hot water and stirred under reflux for 20 min. The mixture was cooled to rt, diluted with DCM (600 mL), and the upper solution decanted into a filter. The solid that remained in the flask was washed with dichloromethane (5×500 mL) with mechanical stirring and filtered. The filtrate from each wash was checked by TLC, and additional washes performed if necessary to recover additional product. In an alternative work-up procedure, after dilution with DCM (600 mL), the mixture was filtered. The resulting solid was then continuously extracted with 0.5% TEA in dichloromethane using a Soxhlet extractor. Higher yield was typically obtained by this alternative procedure, although it does require more time. The filtrate was concentrated under reduced pressure and the residue purified by dry pack (EtOAc/hex/Et3N: 20/80/0.5) to give the product alcohol as a yellowish oil (yield: 90%). The identity and purity were confirmed by NMR.
    Step T8-4: To a mixture of the allylic alcohol from Step T8-3 (28 g, 0.100 mol, 1.0 eq) and collidine (0.110 mol, 1.1 eq) in 200 mL of anhydrous DMF under N2 was added anhydrous LiCl (4.26 g, 0.100 mol, 1.0 eq.) dissolved in 100 mL of anhydrous DMF. The mixture was then cooled to 0° C., and MsCI (12.67 g, 0.110 mol, 1.1 eq., freshly distilled over P2O5), was added dropwise. The reaction was allowed to warm to rt and monitored by TLC (3:7 EtOAc/hex). When the reaction was complete, NaN3 (32.7 g, 0.500 mol, 5.0 eq.) was added. The reaction mixture was stirred at rt O/N with progress followed by NMR. When the reaction was complete, the mixture is poured into an ice-cooled water bath, and extracted with diethyl ether (3×). The combined organic phases were then washed sequentially with citrate buffer (2×), saturated sodium bicarbonate (2×), and finally with brine(1×). The organic layer was dried with MgSO4, filtered and the filtrate concentrated under reduced pressure. The allylic azide was obtained in 90% combined yield, and was of sufficient quality to use as such for the following step.
    Step T8-5: PPh3 (25.9 g, 0.099 mol, 1.5 eq) was added at 0° C. to a solution of the allylic azide from Step T8-4 (20.0 g, 0.066 mol, 1.0 eq.) in 100 mL of THF. The solution was stirred for 30 min at 0° C. and 20 h at rt. Water (12 mL) was then added and the resulting solution was heated at 60° C. for 4 h. The solution was cooled to rt, 2N HCl (15 mL) added and the mixture stirred for 90 min at 50° C. The separated organic phase was extracted with 0.05 N HCl (2×100 mL). The combined aqueous phase was washed with Et2O (5×150 mL) and toluene (4×150 mL) (more extraction could be necessary, follow by TLC), which were combined and back-extracted with 0.05 N HCl (1×100 mL). This acidic aqueous phase from back-extraction was combined with the main aqueous phase and washed with ether (5×150 mL) again. The pH of the aqueous phase was then adjusted to 8-9 by the addition of sodium hydroxide (5 N). Care must be exercised to not adjust the pH above 9 due to the reaction conditions required by the next step. The aqueous phase was concentrated under reduced pressure (aspirator, then oil pump) or lyophilized to dryness. Toluene (2×) was added to the residue and then also evaporated under reduced pressure to remove traces of water. The crude product (desired amino alcohol along with inorgnic salt) was used for the next reaction without further purification.
    Step T8-6: A mixture of the crude amino alcohol from Step T8-5 (0.5 mol based on the theoretical yield), Ddz-OPh (174 g, 0.55 mol, 1.1 eq) and Et3N (70 mL, 0.5 mol, 1.0 eq) in DMF (180 mL) was stirred for 24 h at 50° C. Additional DMF is added if required to solubilize all materials. The reaction was monitored by TLC (hex/EtOAc: 50/50, ninhydrin detection). After the reaction was complete, the reaction mixture was diluted with Et2O (1.5 L) and water (300 mL). The separated aqueous phase was extracted with Et2O (2×150 mL). The combined organic phase was washed with water (3×500 mL) and brine (1×500 mL), dried over MgSO4, filtered and the filtrate concentrated under reduced pressure. The layers were monitored by TLC to ensure no product was lost into the aqueous layer. If so indicated, perform one or more additional extractions with Et2O of the aqueous phase to recover this material. The crude product was purified by dry pack (recommended column conditions: EtOAc/hex/Et3N: 35/65/0.5 to 65/35/0.5) to give the tether Ddz-T8 as a pale yellow syrup (yield: ˜40%). The identity and purity of the product was confirmed by NMR.
  • 1H NMR (DMSO-d6): 1.6 ppm (s, 6H, 2×CH3), 3.6-3.8 ppm (wide s, 10H, 2×OCH3, 2×OCH2), 3.95 ppm (triplet, 2H, CH2N), 6-6.2 ppm (m, 2H, 2×CH), 6.2-6.5 ppm (m, 3H, 3×CH, aromatic), 6.6-7.6 ppm (m, 5H, aromatic).
  • A standard procedure for the synthesis of tether T9 is shown in FIG. 3.
  • Tether T9 can also be synthesized from T8 by reduction as in step T9-3 or with other appropriate hydrogenation catalysts known to those in the art.
  • A standard procedure for the synthesis of Ddz propargylamine is shown in FIG. 4. In a dried three-neck flask, a solution of propargylamine (53.7 g, 0.975 mol, 1.5 eq) in degassed DMF (Drisolv, 388 mL) was treated with Ddz-N3 (170.9 g, 0.65 mol, 1.0 eq), tetramethylguanidine (TMG, 81.4 mL, 0.65 mol, 1.0 eq) and DIPEA (113.1 mL, 0.65 mol, 1.0 eq) and stirred at 50° C. O/N. The reaction was monitored by TLC (conditions:25/75 EtOAc/hex. Rf: 0.25; detection: UV, ninhydrin). Upon completion, DMF was evaporated under reduced pressure until dryness and the residue dissolved in Et2O (1 L). The organic solution was washed sequentially with citrate buffer (pH 4.5, 3×), saturated aqueous sodium bicarbonate (2×), and brine (2×), then dried with MgSO4, filtered and the filtrate evaporated under reduced pressure. A pale orange solid was obtained. This solid was triturated with 1% EtOAc in hex, then collected by filtration and dried under vacuum (oil pump) to provide the desired product (153.4 g, 85.2%).
  • A standard procedure for the synthesis of tether T10 is shown in FIG. 5A. Two alternative routes to this tether have been developed. The first synthetic approach proceeded starting from the commercially available monobenzoate of resorcinol (T10-0). Mitsunobu reaction under standard conditions with the protected amino alcohol from Example 9, followed by saponification of the benzoate provided T10-1 in good yield after recrystallization. Alkylation of the phenol with 2-bromoethanol using the optimized conditions shown permitted the desired product Ddz-T10 to be obtained after dry pack purification in 42% yield.
  • A second synthetic route to T10 is shown in FIG. 5B.
  • From resorcinol, two successive Mitsunobu reactions are conducted with the appropriate two carbon synthons illustrated, themselves derived from 2-aminoethanol and ethylene glycol, respectively, through known protection methodologies. Lastly, deprotection of the silyl ether, also under standard conditions provided Boc-T10.
  • Although the yields in the two methods are comparable, the first required less mechanical manipulation and is preferred for larger scales.
  • A standard procedure for the synthesis of tether T11 is shown in FIG. 6.
  • A standard procedure for the synthesis of tether T12 is shown in FIG. 7.
  • In a 3-L flame-dried three-neck flask, a solution of (aminomethyl)phenylthiobenzyl alcohol (12-0, 96 g, 0.39 mol) in degassed DMF (1 L, 0.4 M) was prepared. To this was added DdzN3 (0.95 eq), followed by TMG (0.39 mol, 49 mL). The reaction was stirred for 10 min, then DIPEA (68 mL, 0.39 mol) added. The mixture was heated at 50° C. under N2 until TLC indicated no DdzN3 remained (48 h typically). (TLC eluent: EtOAc:Hex 50:50; detection: ninhydrin). Upon completion, to the reaction mixture was added 3 L citrate buffer and the separated aqueous layer extracted with Et2O (3×1500 mL). The combined organic phase was washed sequentially with citrate buffer (2×200 mL), water (2×200 mL) and brine (2×200 mL). The organic layer was dried over MgSO4, filtered and the filtrate evaporated under reduced pressure. A dark orange oil was obtained, which was purified by dry-pack. For this procedure, the oil was first dissolved in EtOAc:Hex:DCM:TEA (20:80:1:0.5, v/v/v/v). At this point, a little extra DCM was sometimes required to ensure complete dissolution. The solution was loaded onto the column, then the column eluted with EtOAc:Hex:DCM:Et3N (20:80:1:0.5) until all the impurities were separated out as indicated by TLC, paying particular attention to that closest to the desired product. The elution was then continued with EtOAc:Hex:Et3N 30:70:0.5 (v/v/v) and finally with EtOAc:hexanes:Et3N (50:50:0.5) to elute the desired product. After removal of the solvent from the fractions containing the product under reduced pressure, the residue was dissolved in the minimum amount of DCM, a three-fold larger volume of hexanes added, then the solvents again evaporated under reduced pressure. This treatment was repeated until an off-white foam was obtained. The latter solidified while drying under vacuum (oil pump). Alternatively, the material yielded a solid after sequential concentration with DCM (1×) and hexanes (2×). Tether Ddz-T12 was obtained as an off-white solid (85-90% yield).
  • Example 29 Standard Procedure for Attachment of Tethers Utilizing the Mitsunobu Reaction Example 29-A Using PPh3-DIAD Isolated Adduct
  • To a 0.2 M solution of the appropriate tether (1.5 eq) in THF or THF-toluene (1:1) was added the PPh3-DIAD (pre-formed by mixing equivalent amounts of the reagents and isolated by evaporation of solvent, see Example 29-C) adduct (1.0 eq.). The resultant mixture was manually agitated for 10 sec (the solution remained turbid), then added to the resin. Alternatively, the resin was added to the solution. The reaction suspension was agitated O/N (after min the mixture becomes limpid). The resin was filtered and washed 2×DCM, 1× toluene, 1×EtOH, 1× toluene, 1×(DCM/MeOH), 1×(THF/MeOH), 1× (DCM/MeOH), 1× (THF/MeOH), 2×DCM, then dried in the standard manner.
  • Example 29-B Using “PPh3-DIAD In Situ Procedure”
  • To a 0.2 M solution of the appropriate tether (4 eq) in THF or THF-toluene (1:1) was added triphenylphosphine (4 eq). The resultant mixture was manually shaken until a homogenous solution was obtained, then added to the resin. Alternatively, the resin (or IRORI™ MiniKans® (NEXUS Biosystems, Poway, Calif.), miniaturized microreactors, containing resin) was added to the solution. To this suspension was then added DIAD (3.9 eq) and the reaction agitated O/N. Note: Since the reaction is exothermic, for larger scales, the reaction should be cooled in an ice bath. In addition, an appropriate vent must be supplied to allow any pressure build-up to be released. The resin was filtered and washed DCM (2×), toluene (1×), EtOH (1×), toluene (1×), DCM/MeOH (1×), 1×THF/MeOH (1×), DCM/MeOH (1×), THF/MeOH (1×), 2×DCM, then dried in the standard manner.
  • A procedure for the synthesis of PPh3-DIAD adduct is shown in FIG. 8.
  • DIAD (1 eq) was added dropwise to a well-stirred solution of triphenylphosphine (1 eq) in THF (0.4 M) at 0° C. under nitrogen. The mixture was then maintained at 0° C. with stirring for 30 min. The white solid obtained was collected by filtration (use medium sized fritted filters), washed with cold anhydrous THF until the washes were colorless, and lastly washed once with anhydrous Et2O. The white solid product was then vacuum-dried (oil pump) and stored under nitrogen. (Note: The PPh3-DIAD adduct can be made in larger than immediately required quantity and stored under nitrogen; it is very important to store this reagent under anhydrous conditions.)
  • Example 30 Standard Procedure for Attachment of Tethers via Reductive Amination as shown in FIG. 9
  • In certain instances, the Mitsunobu process of Example 29 cannot be applied or is not efficient for incorporation of the tether. Hence, reductive amination has been developed as an alternative that can be employed for tether incorporation as illustrated hereinbelow for one of the preferred tethers. Similar chemistry can be used to incorporate other tethers of the present invention.
  • The Tether (30-2) with the amine protected as its Ddz derivative was efficiently oxidized to the corresponding aldehyde 30-2 using SO3·pyr in DMSO-Et3N-DCM. This aldehyde (0.14 mmol, 56 mg, 1.5 eq based upon loading of resin support) was dissolved in a 1:3 mixture of TMOF-MeOH (DriSolv, 4 mL) at rt. To this was added the resin containing the tripeptide (30-1, as its trifluoroacetic acid salt from the deprotection of the terminal amine), the mixture was agitated briefly to wet the resin, and then borane-pyridine complex (as the commercially available 8 M solution, 23 μL , 2 eq) was introduced to the suspension. The reaction was agitated O/N, then the resin filtered, washed with DCM (2×), THF (1×), DCM/MeOH [3:1] (1×), THF/MeOH [3:1](1×), DCM (2×) and dried in the standard manner. Care must be taken to ensure that the desired resin bound product 30-3 is not contaminated with the dialkylated material. However, even if the reaction does not proceed to completion or if a small amount of the dialkylation side product is present, the material is of sufficient purity for the macrocyclizatton reaction.
  • A standard procedure for the synthesis of tether T28 is shown in FIG. 10.
  • Henry reaction of 2-hydroxybenzaldehyde 28-0 provided 28-1 in 79% yield. This was followed by reduction first with sodium borohydride, then with catalytic hydrogenation, to give the amine, which was then protected as its Boc derivative, 28-2. Yields of these first two steps were lower on larger scales. Alkylation of 28-2 with the TBDMS ether of 2-bromoethanol, itself synthesized by standard methods, gave 28-3 in 74% yield. Deprotection of the silyl ether under standard conditions yielded the desired protected tether, Boc-T28. Alternative use of ethylene carbonate for the phenol alkylation to avoid the protection/deprotection steps, gave 73% yield.
  • A standard procedure for the synthesis of tether T32 is shown in FIG. 11.
  • A standard procedure for the synthesis of tether T33 a and T33 b is shown in FIGS. 12A and 12B.
  • The construction to the (R)-isomer of this tether (T33a) was accomplished from 2-iodophenol (33-0) and (S)-methyl lactate (33-A). Mitsunobu reaction of 33-0 and 33-A proceeded with inversion of configuration in excellent yield to give 33-1. Reduction of the ester to the corresponding alcohol (33-2) also occurred in high yield and was followed by Sonagashira reaction with Ddz-propargylamine. The alkyne in the resulting coupling product, 33-3, was reduced with catalytic hydrogenation. Workup with scavenger resin provided the desired product, Ddz-T33a.
  • The synthesis of the (S)-enantiomer (Ddz-T33b) was carried out in an identical manner in comparable yield starting from (R)-methyl lactate (33-B). See FIG. 12B.
  • Standard procedures for the synthesis of various tethers are shown in the figures: tether T34 (FIG. 13), tether T35 (FIG. 14), tether T36 (FIG. 15), tether T37 (FIG. 16), tether T38 (FIG. 17), tether T39 (FIG. 18), tether T40 (FIG. 19), tether T41 (FIG. 20) and tether T42 (FIG. 21).
  • E. Examples of Synthetic Strategies for the Macrocyclic Compounds of the Invention
  • FIG. 22 presents a scheme depicting a thioester strategy for macrocyclic compounds of the present invention.
  • It should be noted that one or more of the amino acids indicated can be replaced by corresponding hydroxy acids and coupled to the next building block utilizing methods known to those in the art.
  • Example 47 Standard Procedure for Macrocyclization with Thioester Linker
  • The resin containing the cyclization precursor is combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, Foster City, Calif., commercially supplied MP-carbonate resin was treated with 3×THF (1 L per 400 g) and dried O/N at 30° C. in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin). A 0.2 M DIPEA solution in THF was then added to the combined resins (1 mL/60 mg MP-carbonate resin) and the suspension agitated O/N at rt. Subsequently, the resin was filtered and rinsed 2×THF. The combined filtrates are collected together in an appropriate vessel, then the volatile contents evaporated under reduced pressure [in addition to the standard methods, solvent can also be removed in vacuo using centrifugal evaporation (ThermoSavant Discovery®, SpeedVac® or comparable) (Thermo Electron Corporation, Waltham, Mass.)] to provide the crude macrocycles.
  • Example 48 Standard Procedure for Silver-Assisted Macrocyclization with Thioester Linker
  • Except for the cyclization itself and subsequent work-up, this procedure is identical to that of Example 47. The resin containing the cyclization precursor was combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, commercially supplied MP-carbonate resin was treated with THF (3×, 1 L per 400 g) and dried O/N at 30° C. in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin). To this was added THF (1 mL per 100 mg resin) and silver trifluoroacetate (1 eq relative to the initial loading of the resin). Finally, an amount of DIPEA sufficient to obtain a 0.2 M solution was added. The reaction mixture was agitated at rt O/N. The solution was then filtered and the resins washed 2×THF. The filtrates are collected together in an appropriate vessel, then evaporated under reduced pressure [(the volatile contents could also be removed in vacuo using centrifugal evaporation (ThermoSavant Discovery®, SpeedVac® or comparable)] to provide the crude macrocycles. For this procedure, silver trifluoroacetate should be stored in a dessicator between uses. In addition, it is recommended to use a new bottle of THF (or a bottle that has been recently opened under N2 or Ar) to minimize formation of silver oxide.
  • Additionally, a ring-closing metathesis (RCM) strategy, as developed by Grubbs et al. can also be used to access some of the macrocyclic compounds of the invention (see for example US 5,811,515; Grubbs, R. H. et al. J. Org. Chem. 2001, 66, 5291-5300; Fürstner, A. Angew. Chem. Int. Ed. 2000, 39, 3012−3043).
  • To access certain derivatives of compounds of the present invention, additional reactions from those in the general scheme were required. For some, it was advantageous to react the functionality to be derivatized prior to the formation of the macrocyclic ring. The cyclic structure can restrict access of reagents to that functionality. For example, in the synthesis of N-methyl and N-acyl derivatives of macrocycles, where the secondary nitrogen atom of the ring is the site of derivatization, the reaction is preferred to be performed prior to the application of the appropriate cyclization protocol.
  • In other cases, for example the derivatization of side chain functionality, the reaction was best performed after formation of the macrocyclic ring. For example, further reaction of amino moieties on side chains examples was typically efficiently done by reaction of the partially protected macrocycle. In this manner, acylation, sulfonylation, alkylation (via reductive amination), guanidine and urea formation were performed via standard methods.
  • Table 1, hereinbelow, shows a representative, but by no means exclusive, summary of the chemical synthesis of several representative compounds of the invention.
  • TABLE 1
    Synthesis of Representative Compounds of the Present Invention
    Tether Additional
    AA1 AA2 AA3 Tether Attachment Steps
    1 Bts-D- Boc-D-Val Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    2 Bts-D-Phe Boc-D-Val Boc-Nva Boc-T8 Example 29 none
    3 Bts-D-Phe Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    4 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    5 Bts-D- Boc-D-Ala Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    6 Bts-D- Boc-D-Val Boc-Met Ddz-T8 Example 29 none
    Tyr(tBu)
    7 Bts-D- Boc-D-Val Boc-Nle Ddz-T8 Example 29 none
    Tyr(tBu)
    8 Bts-D- Boc-D-Val Boc-Phe Ddz-T8 Example 29 none
    Tyr(tBu)
    9 Bts-D- Boc-D-Val Boc-Val Ddz-T8 Example 29 none
    Tyr(tBu)
    10 Bts-D- Boc-D-Val Boc-Leu Ddz-T9 Example29 none
    Tyr(tBu)
    11 Bts-D-2-Nal Boc-D-Val Boc-Nva Boc-T8 Example 29 none
    12 Bts-D- Boc-D-Val Boc-Abu Ddz-T8 Example 29 none
    Tyr(tBu)
    13 Bts-D-Phe Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    14 Bts-D-2-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    15 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3Cl)
    16 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(4Cl)
    17 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none
    Trp(Boc)
    18 Bts-D- Boc-D-2-Abu Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    19 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(4F)
    20 Bts-D-Phe Boc-D-Val Boc-Leu Boc-T8 Example 29 none
    21 Bts-D-2-Nal Boc-D-Val Boc-Leu Boc-T8 Example 29 none
    22 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Tyr(OMe)
    23 Bts-D-1-Nal Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    24 Bts-D-2-Thi Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    25 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(2Cl)
    26 Bts-D- Boc-D-Val Boc-Cpa Ddz-T9 Example 29 none
    Tyr(tBu)
    27 Bts-D-4-Thz Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    28 Bts-D-3-Pal Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    29 Bts-D- Boc-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    Tyr(tBu)
    30 Bts-D- Hnva(THP) Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    34 Bts-D- Ddz-D-Tyr(tBu) Boc-Nva Ddz-T8 Example 29 None
    Tyr(tBu)
    38 Bts-D- Boc-D-Val Boc-Ala Ddz-T8 Example 29 none
    Tyr(tBu)
    39 Bts-D- Boc-D-Val Boc-□-Ala Ddz-T8 Example 29 none
    Tyr(tBu)
    40 Bts-D- Boc-D-Val Boc-Gly Ddz-T8 Example 29 none
    Tyr(tBu)
    41 Bts-D- Boc-DPhe Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    52 Bts-D- Boc-D-Val Boc-Phg Ddz-T8 Example 29 none
    Tyr(tBu)
    55 Bts-D- Ddz-D-Val Ddz-Lys(Boc) Ddz-T8 Example 29 none
    Tyr(tBu)
    56 Bts-D- Ddz-D-Val Ddz-Orn(Boc) Ddz-T8 Example 29 none
    Tyr(tBu)
    57 Bts-D- Ddz-D-Val Ddz-Ser(tBu) Ddz-T8 Example 29 none
    Tyr(tBu)
    58 Bts-D- Ddz-D-Val Ddz-Tyr(tBu) Ddz-T8 Example 29 none
    Tyr(tBu)
    59 Bts-D- Ddz--D-Val Ddz-Trp(Boc) Ddz-T8 Example 29 none
    Tyr(tBu)
    60 Bts-D- Boc-D-Val Boc-Tyr(OMe) Ddz-T8 Example 29 none
    Tyr(tBu)
    65 Bts-D- Boc-D-Val Boc-Nva Ddz-T2 Example 29 none
    Tyr(tBu)
    71 Bts-D- Boc-D-Val Boc-Nva Ddz-T10 Example 29 none
    Tyr(tBu)
    72 Bts-D- Boc-D-Val Boc-2-Nal Ddz-T8 Example 29 none
    Tyr(tBu)
    76 Bts-D- Boc-D-2-Nal Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    77 Bts-D- Boc-D-Nle Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    80 Bts-D- Boc-D-Val Boc-Ile Ddz-T8 Example 29 none
    Tyr(tBu)
    85 Bts-D- Boc-D-Val Boc-D-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    87 Bts-D-Bip Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    88 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    89 Bts-D-Hfe Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    90 Bts-D-Dip Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    91 Bts-D- Boc-D-Nva Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    92 Bts-D- Boc-D-Tle Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    96 Bts-D- Boc-β-Ala Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    97 Bts-D- Boc-D-Chg Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    98 Bts-D- Boc-D-Val Boc-Nva Ddz-T18 Example 29 none
    Tyr(tBu)
    99 Bts-D- Boc-D-Val Boc-Nva Ddz-T15 Example 29 none
    Tyr(tBu)
    109 Bts-D- Boc-D-Val Ddz-Dab(Boc) Ddz-T9 Example 29 none
    Tyr(tBu)
    110 Bts-D- Boc-D-Val Boc-Nva Ddz-T11 Example 29 none
    Tyr(tBu)
    111 Bts-D- Boc-D-Val Hval(THP) Ddz-T9 Example 29 none
    Tyr(tBu)
    112 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none
    Tyr(tBu)
    120 Bts-D- Boc-D-Pro Boc-Nva Ddz-T8 Example 29 none
    Tyr(tBu)
    121 Bts-D- Boc-D-Val Boc-Nva Ac-T8-NH2 Example 29 none
    Tyr(tBu)
    122 Boc-D-3-Pal Boc-D-Val Boc-Nva Boc-T9 Example 30 none
    123 Boc-D-2-Pal Boc-D-Val Boc-Nva Boc-T9 Example 30 none
    124 Boc-D-4-Pal Boc-D-Val Boc-Nva Boc-T9 Example 30 none
    125 Bts-D- Boc-D-Cpg Boc-Nva Boc-T9 Example 29 none
    Tyr(tBu)
    126 Bts-D- Boc-D-Val Boc-NMeLeu Boc-T9 Example 29 none
    Tyr(tBu)
    127 Boc-D- Boc-D-Val Boc-Nva Boc-T12 Example 30 none
    His(Mts)
    128 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    Tyr(OMe)
    129 Bts-D-1-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    130 Bts-D-2-Thi Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    131 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    Phe(3Cl)
    132 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    Phe(4Cl)
    133 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none
    Phe(4F)
    134 Bts-D- Boc-D-Val Boc-Leu Boc-T2 Example 29 none
    Phe(3Cl)
    135 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    Tyr(OMe)
    136 Bts-D-1Nal Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    137 Bts-D-2-Thi Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    138 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    Phe(3Cl)
    139 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    Phe(4Cl)
    140 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none
    Phe(4F)
    141 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Tyr(OMe)
    142 Bts-D-1-Nal Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    143 Bts-D-2-Thi Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    144 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(3Cl)
    145 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(4Cl)
    146 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(4F)
    147 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none
    Tyr(OMe)
    148 Bts-D-1-Nal Boc-D-Val Boc-Cpa Boc-T11 Example 29 none
    149 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none
    Phe(3Cl)
    150 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none
    Phe(4Cl)
    151 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none
    Phe(4F)
    152 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    Tyr(OMe)
    153 Bts-D-1-Nal Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    154 Bts-D-2-Thi Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    155 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    Phe(3Cl)
    156 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    Phe(4Cl)
    157 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 none
    Phe(4F)
    158 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T11 Example 29 none
    Phe(3Cl)
    159 Bts-D- Boc-D-Ile Boc-Nva Boc-T9 Example 29 none
    Tyr(But)
    160 Bts-D- Boc-D-allolle Boc-Nva Boc-T9 Example 29 none
    Tyr(But)
    161 Boc-D- Boc-D-Val Boc-Nva Boc-T9 Example 30 none
    Phe(4CH2NHFmoc)
    162 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(2Me)
    163 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3Me)
    164 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(4Me)
    165 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3OMe)
    166 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(2OMe)
    167 Bts-D-3- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    benzothienyl
    168 Bts-D-3-Thi Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    169 Bts-D-□- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    HomoPhe(3Cl)
    170 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3,4diCl)
    171 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3,4diF)
    172 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3,4diOMe)
    173 Bts-D-1Nal Hnva(THP) Boc-Nva Boc-T9 Example 29 none
    174 Bts-D- Hnva(THP) Boc-Nva Boc-T9 Example 29 none
    Tyr(OMe)
    175 Bts-D- Boc-D-Val Boc-Nva Boc-T33b Example 29 none
    Tyr(tBu)
    176 Bts-D- Boc-D-Val Boc-Nva Boc-T33a Example 29 none
    Tyr(tBu)
    177 Bts-D- Boc-D-Val Boc-Nva Boc-T28 Example 29 none
    Tyr(tBu)
    178 Bts-D- Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    Tyr(OMe)
    179 Bts-D-1-Nal Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    180 Bts-D-2-Thi Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    181 Bts-D- Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    Phe(3Cl)
    182 Bts-D- Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    Phe(4Cl)
    183 Bts-D- Ddz-D-Val Ddz-Ser(tBu) Ddz-T9 Example 29 none
    Phe(4F)
    184 Bts-D-1-Nal Ddz-D-Val Ddz-Dap(Boc) Ddz-T11 Example 29 none
    185 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T11 Example 29 none
    Phe(4Cl)
    186 Ddz-D- Ddz-D-Val Ddz-His(Mts) Ddz-T9 Example 30 none
    Tyr(tBu)
    187 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3CF3)
    188 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(3F)
    189 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Phe(4NO2)
    190 Bts-D-3- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    benzothienyl
    191 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(3OMe)
    192 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(3,4diCl)
    193 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none
    Phe(3,4diF)
    194 Bts-D- Boc-D-Val Boc-Nva Boc-T34 Example 29 none
    Tyr(OMe)
    195 Bts-D- Boc-D-Val Boc-Nva Boc-T38 Example 29 none
    Tyr(OMe)
    196 Bts-D- Boc-D-Val Boc-Cpa Ddz-T32(Boc) Example 29 none
    Phe(3Cl)
    197 Bts-D- Boc-D-Val Boc-Cpa Boc-T34 Example 29 none
    Phe(3Cl)
    198 Bts-D- Boc-D-Val Boc-Cpa Boc-T38 Example 29 none
    Phe(3Cl)
    199 Bts-D- Boc-D-Val Boc-Cpa Boc-T41 Example 29 none
    Phe(3Cl)
    200 Bts-D- Boc-D-Val Boc-Cpa Boc-T8 Example 29 none
    Phe(3Cl)
    201 Bts-D-1-Nal Boc-D-Val Boc-Nva Boc-T8 Example 29 none
    202 Bts-D- Boc-D-Val Boc-Nva Boc-T8 Example 29 none
    Phe(3OMe)
    203 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 acetylation
    Phe(4Cl)
    204 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 guanidinylation
    Phe(4Cl)
    205 Bts-D- Boc-D-Val Boc-NMeLeu Boc-T9 Example 29 none
    Phe(3Cl)
    206 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 mesylation
    Phe(4Cl)
    207 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 TMS-
    Phe(4Cl) isocyanate
    followed by
    dilute acid
    208 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 guanidinylation
    Tyr(tBu)
    209 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 acetylation
    Tyr(tBu)
    210 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 reductive
    Tyr(tBu) amination with
    acetone
    211 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 reductive
    Phe(4Cl) amination with
    excess
    formaldehyde
    212 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 reductive
    Phe(4Cl) amination with
    acetone
    213 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Tyr(3,5dil)
    214 Bts-D- Boc-D-Val Boc-Hse(Bzl) Boc-T9 Example 29 hydrogenolysis
    Tyr(OMe) for protecting
    group removal
    215 Bts-D- Ddz-D-Val Ddz-Dap(Boc) Ddz-T9 Example 29 reductive
    Tyr(tBu) amination with
    excess
    formaldehyde
    216 Bts-D- Boc-D-Val Boc-Cpa Boc-T40 Example 29 none
    Phe(3Cl)
    217 Bts-D- Boc-D-Val Boc-Cpa Boc-T36 Example 29 none
    Phe(3Cl)
    218 Bts-D- Boc-D-Val Boc-Nva Boc-T39 Example 29 none
    Phe(3Cl)
    219 Bts-D- Boc-D-Val Boc-Nva Boc-T37 Example 29 none
    Phe(3Cl)
    220 Bts-D- Boc-D-Val Boc-Nva Boc-T39 Example 29 none
    Phe(3Cl)
    221 Bts-D- Boc-D-Val Boc-Nva Boc-T35 Example 29 none
    Phe(3Cl)
    222 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none
    Tyr(3tBu)
    223 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 acetylation
    Tyr(But)
    224 Bts-D-1-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive
    amination with
    formaldehyde
    225 Bts-D-1-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 acetylation
    226 Bts-D-1-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive
    amination with
    aldehyde
    227 Bts-D-1-Nal Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive
    amination with
    benzaldehyde
    Notes
    Any amino acid or tether designated as the Boc derivative could be substituted with the corresponding Ddz derivative.
  • D. Analytical Data for Selected Compounds of the Invention
  • 1H and 13C NMR spectra were recorded on a Varian Mercury 300 MHz spectrometer (Varian, Inc., Palo Alto, Calif.) and are referenced internally with respect to the residual proton signals of the solvent. Information about the conformation of the molecules in solution can be determined utilizing appropriate two-dimensional NMR techniques known to those skilled in the art. HPLC purifications were run on a Waters Xterra® MS C18 column, using the Waters FractionLynx® system (Waters Corporation, Milford, Mass.). Automated medium pressure chromatographic purifications were performed on an Isco CombiFlash® 16× system with disposable silica or C18 cartridges that permitted up to sixteen (1.6) samples to be run simultaneously (Teledyne Isco, Inc., Lincoln, Nebr.). MS spectra were recorded on a Waters Micromass® Platform II or ZQ™ system. HRMS spectra were recorded with a VG Micromass ZAB-ZF spectrometer. Chemical and biological information were stored and analyzed utilizing the ActivityBase® database software (ID Business Solutions Ltd., Guildford, Surrey, UK).
  • General Methods for Analytical HPLC Analyses
  • HPLC analyses are performed on a Waters Alliance® system 2695 running at 1 mL/min using an Xterra MS C18 column 4.6×50 mm (3.5 μm). A Waters 996 PDA provided UV data for purity assessment (Waters Corporation, Milford, Mass.). An LCPackings (Dionex Corporation, Sunnyvale, Calif.) splitter (50:40:10) allowed the flow to be separated in three parts. The first part (50%) went to a Micromass® Platform II MS equipped with an APCI probe for identity confirmation. The second part (40%) went to an evaporative light scattering detector (ELSD, Polymer Laboratories, now part of Varian, Inc., Palo Alto, Calif., PL-ELS-1000™) for purity assessment and the last portion (10%) to a chemiluminescence nitrogen detector (CLND, Antek® Model 8060, Antek Instruments, Houston, Tex., part of Roper Industries, Inc., Duluth, Ga.) for quantitation and purity assessment. Data was captured and processed utilizing the most recent version of the Waters Millenium® software package (Milford, Mass.).
  • An example LC method suitable for compounds of the present invention uses MeOH as solvent A, H2O as solvent B and 1% TFA/H2O as solvent D. Initial mobile-phase composition is 5% A, 85% B and 10% D. Details of the standard gradient method are shown below:
  • Time A % B % D % Curve
    0.00 5 85 10 6
    1.00 5 85 10 6
    6.00 50 40 10 6
    9.00 50 40 10 6
    14.00 90 0 10 6
    17.00 90 0 10 6
    17.50 5 85 10 6
    20.00 5 85 10 6
  • Compounds 2-6, 8-10, 56, 65 and 144 are as defined in Table (3), hereinbelow.
  • Compound 2
  • Yield: 12 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 8.83 (m,1H); 8.53 (m, 1H); 7.63 (m, 1H); 7.4-7.08 (m, 7H); 7.00-6.84 (m, 2H); 6.60 (d, 15 Hz, 1H); 6.41 (dt, 15 Hz, 5.4 Hz, 1H); 4,35 (m, 1H); 4.25-4.05 (m, 3H); 3.94 (dt, 1H, 6 Hz, 15 Hz); 3.79 (dd, 1H, 3.6 Hz, 8.4 Hz); 3.60 (m, 1H); 3.52−3.40 (bd, 1H); 3.22-3.06 (m, 4H); 1.88 (m, 2H); 1.54-1.28 (m, 2H); 1.25 (d, 3H, 4.8 Hz); 1.22 (d, 3H, 2.7 Hz); 0.92-0.80 (m, 6 H).
  • HRMS calc. for C30 H40N4O4: 520.3049; found 520.3057±0.0016
  • HPLC [standard gradient method (refers to that presented in General Methods for Analytical HPLC Analyses)] tR=9.55 min.
  • Compound 4
  • Yield: 12 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.35 (b, 1H); 8.98 (b, 1H); 5.52 (d, 1H, 8.4 Hz); 8.38 (b, 1H); 7.25 (b, 1H); 7.13-7.07 (m, 4H); 6.86 (t, 2H, 7.5 Hz); 6.57 (d, 2H, 8.7 Hz); 4.33 (b, 1H); 4.21-4.02 (m, 3H); 3.78 (dd, 1H, 3.3 Hz; 8.1 Hz); 3.65−3.54 (m, 1H); 3.31−3.23 (m, 1H); 3.13−3.02 (m, 4H); 2.78-2.2.28-2.18 (m, 1H); 2.0-1.80 (m, 2H); 1.50-1.30 (m, 3H); 1.25 (d, 3H, 4.5 Hz); 1.22 (d, 3H, 4.5 Hz); 1.01 (d, 3H, 6.6 Hz); 0.90 (d, 3H, 6.6 Hz); (t, 3H, 7.5 Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 172.22; 171.37; 157.77; 157.44; 156.04; 131.76; 130.80; 130.70; 127.88; 121.82; 115.83; 111.71; 62.13; 60.62; 54.21; 52.81; 47.13; 42.47; 33.31; 29.69; 29.30; 28.61; 20.36; 19.44; 18.72; 17.60; 13.97.
  • HRMS calc. for C30 H42N4O5:538.3155; found: 538.3145±0.0016
  • HPLC (standard gradient) tR=8.12 min.
  • Compound 5
  • Yield: 17 mg pure macrocycle was obtained (CLND quantification). 1H NMR (300 MHz, DMSO-d6) δ 9.02 (b, 1H); 8.47 (d, 1H, 8.4 Hz); 7.7 (b, 1H); 7.58 (d, 1H, 5.4 Hz); 7.28 (dd, 1H, 7.8 Hz, 0.8 Hz); 7.20 (t, 1H, 9.0 Hz, 0.8 Hz); 7.14 (d, 2H, 8.4 Hz); 6.98-6.91 (m, 3H); 6.66 (d, 8.7 Hz); 6.63 (d, 1H, 15.0 Hz); 6.43 (dt, 1H, 6.0 Hz, 15.0 Hz); 4.28-3.86 (m, 6H); 3.60-3.40 (m, 2H); 3.22-3.12 (m, 1 H0; 3.05 (d, 2H, 5.4 Hz); 1.92-1.80 (m, 1H); 1.56-1.40 (m, 1H); 1.36-1.20 (m, 2H); 1.25 (d, 3H, 6.6 Hz); 0.84 (t, 3H, 7.2Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 172.54; 171.86; 158.97; 158.56; 127.39; 155.84; 131.62; 129.73; 129.20; 129.02; 128.43; 126.30; 124.51; 122.01; 115.85; 112.88; 61.23; 52.90; 51.23; 47.08; 42.66; 36.13; 33.30; 21.14; 19.57; 17.07; 14.14; 11.49.
  • HRMS calc. for C28 H36N4O5:508.2685; found: 508.2681±0.0015
  • HPLC (standard gradient) tR=7.67 min.
  • Compound 6
  • Yield: 16 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.37 (b, 1H); 8.87 (b, 1H); 8.61 (d, 1H, 8.7 Hz); 7.62 (b, 1H); 7.27 (d, 1H, 7.8 Hz); 7.21 (t, 1H, 8.4 Hz); 7.14 (d, 2H, 8.4 Hz); 6.98-6.87 (m, 3H); 6.64 (d, 2H, 8.1 Hz); 6.70 (d, 1H, 15.6 Hz); 6.39 (dt, 1H, 6.3 Hz, 15.6 Hz); 4.44-4.36 (m, 1H); 4.34-4.08 (m, 2Hz); 4.45-3.92 (dt, 1H, 6.9 Hz, 15.6 Hz); 3.74 (dd, 1H, 3.6 Hz, 8.4 Hz); 3.54-3.26 (m, 3H); 3.22-3.02 (m, 3H); 2.60-2.36 (m, 4H); 2.24-2.14 (m, 1H); 2.02 (s, 3H); 1.96-1.89 (m, 1H); 1.80-1.66 (m, 1H); 1.01 (d, 3H, 6.3 Hz); 0.90 (d, 3H, 6.6 Hz).
  • 130 NMR (75.5 MHz, DMSO-d6) δ 171.51; 171.26; 158.90; 158.49; 157.38; 155.86; 131.63; 129.82; 129.21; 128.86; 128.63; 126.21; 121.98; 115.83; 112.83; 62.11; 61.06; 51.97; 47.10; 42.78; 30.91; 30.67; 29.34; 20.37; 19.39; 15.06.
  • HRMS calc. for C30 H40N4O5S: 568.2719; found: 568.2711±0.0017
  • HPLC Rt (general method) 7.92 min.
  • Compound 8
  • Yield: 27 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.05 (b, 1H); 8.43 (b, 1H); 8.34 (d, 1H, 9.3 Hz); 7.40 (b, 1H); 6.97 (d, 1H, 7.5 Hz); 6.92-6.74 (m, 9H); 6.67-6.54 (m, 2H); 6.33-6.25 (m, 3H); 6.10 (dt, 1H, 5.7 Hz, 16.2Hz); 4.22 (dt, 1H, 0.9 Hz, 12Hz); 3.94-6.66 (m, 4H); 3.30 (dd, 1H, 3.6 Hz, 7.8 Hz); 3.24 (m, 1H); 3.18 (m, 1H); 2.85-2.68 (m, 3H); 2.44-2.23 (m, 2H); 1.32 (o, 1H, 7.5 Hz); 0.97-0.89 (m, 1H); 0.42 (d, 3H, 6.6 Hz); 0.01 (d, 3H, 6.6 Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 171.20; 157.35; 155.88; 139.12; 131.61; 130.87; 129.74; 129.21; 128.77; 128.88; 126.85; 126.19; 121.97; 115.82; 112.84; 62.04; 61.10; 55.07; 50.01; 47.09; 42.85; 37.42; 29.11.
  • HRMS calc. For C34 H42N4O5: 586.3155; found: 586.3145±0.0017
  • HPLC Rt (general method) 9.34 min.
  • Compound 9
  • Yield: 17 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.39 (b, 1H); 8.83 (b, 1H); 8.29 (d, 1H, 9.3 Hz); 7.62 (b, 1H); 7.28 (d, 1H, 6.6 Hz); 7.20 (t, 1H, 6.9 Hz); 7.12 (d, 2H, 7.8 Hz); 6.98-6.91 (m, 2H); 6.63 (d, 2H, 8.4 Hz); 6.58 (d, 1H, 16.2Hz); 6.40 (dt, 1H, 5.7 Hz, 16.2Hz); 4.29-4.13 (m, 3H); 4.03−3.92 (m, 2H); 3.52 (m, 1H); 3.15−3.05 (m, 3H); 2.45-2.37 (m, 1H); 1.96-1.88 (m, 1H); 1.25 (dd, 2H, 4.5 Hz; 6 Hz); 1.01 (d, 3H, 6.3 Hz); 0.91 (d, 3H, 6.6 Hz); 0.86 (d, 3H, 7.2Hz); 0.81 (d, 3H, 6.6 Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 171.85; 171.17; 157.37; 155.87; 131.59; 129.88; 129.18; 128.97; 128.78; 128.51; 126.16; 121.97; 115.83; 112.85; 61.55; 61.18; 58.15; 54.22; 47.08; 42.89; 36.32; 29.35; 29.00; 20.34; 19.56; 18.73; 17.44.
  • HRMS calc. for C30 H40N4O5: 536.2998; found: 536.2990±0.0017.
  • HPLC (standard gradient) tR=8.15 min.
  • Compound 10
  • Yield: 24 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.33 (b, 1H); 8.82 (b, 1H); 8.56 (d, 1H, 8.3 Hz); 7.60 (b, 1H); 7.27 (d, 2H, 7.8 Hz); 7.20 (t, 1H, 7.8 Hz); 7.13 (d, 2H, 8.4 Hz); 6.95 (t, 2H, 7.8 Hz); 6.64 (d, 2H, 8.4 Hz); 6.57 (d, 1H, 15.4 Hz); 6.38 (dt, 1H, 15.4 Hz, 5.8 Hz); 4.26-4.10 (m, 3H); 3.96 (dt, 1H, 5.4 Hz, 8.4 Hz); 3.77 (dd, 1H, 3.7 Hz, 7.8 Hz); 3.51−3.24 (m, 3H); 3.18-3.02 (m, 3H); 1.90 (h, 1H, 6.4 Hz); 1.73-1.54 (m, 2H); 1.45 (dt, 1H, 6.7 Hz, 0.9 Hz); 0.99 (d, 3H, 6.6 Hz); 0.89 (d, 3H, 6.3 Hz); 0.87 (d, 3H, 6.0 Hz); 0.80 (d, 3H, 6.3 Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 172.23; 171.17; 157.37; 155.88; 131.62; 129.82; 129.19; 128.95; 128.59; 126.24; 121.99; 115.84; 112.88; 64.23; 61.98; 61.14; 51.43; 61.14; 51.43; 47.07; 42.81; 29.38; 24.85; 24.11; 21.00; 20.32; 19.30.
  • HRMS calc. for C31 H42N4O5 550.3155; found: 550.3150±0.0016.
  • HPLC (standard gradient) tR=8.91 min.
  • Compound 56
  • Yield: 16 mg pure macrocycle was obtained (CLND quantification).
  • 1H NMR (300 MHz, DMSO-d6) δ 9.39 (b, 1H); 8.90 (b, 1H); 8.67 (d, 1H, 8.4 Hz); 7.74 (b, 4H); 7.29-7.08 (m, 4H); 6.99-6.87 (m, 2H); 6.64 (d, 2H, 8.1 Hz); 6.61 (d, 1H, 16.5 Hz); 6.40 (dt, 1H, 5.7 Hz, 16.5 Hz); 4.40-4.06 (m, 4H); 4.02-3.95 (m, 1H); 3.79 (dd, 1H, 3.6 Hz, 7.8 Hz); 3.55-3.30 (m, 2H); 3.16−3.05 (m, 3H); 2.82-2.69 (m, 2H); 2.02-1.85 (m, 2H); 1.64-1.43 (m, 3H); 1.29-1.23 (m, 1H); 1.01 (d, 3H, 6.3 Hz); 0.91 (d, 3H, 6.3 Hz); 0.86-0.84 (m, 2H).
  • HPLC (standard gradient) tR=5.71 min.
  • Compound 65
  • Yield: 17 mg pure macrocycle was obtained (CLND quantification). 1H NMR (300 MHz, DMSO-d6) δ 9.60 (b, 1H); 9.39 (b, 1H); 8.88 (b, 1H); 8.70 (d, 1H, 7.5 Hz); 8.57 (d, 1H, 4.2Hz); 7.27 (t, 6 Hz); 6,96 (d, 2H, 8.4 Hz); 6.66 (d, 2H, 8.4 Hz); 5.78-5.68 (m, 1H); 5.42-5.33 (m, 1H); 3.96−3.89 (m, 1H); 3.80−3.57 (m, 5H); 3.41−3.34 (m, 1H); 3.10-2.90 (m, 1H); 2.78-2.66 (m, 1H); 2.21-2.10 (m, 1H); 2.06-1.93 (m, 1H); 1.70-1.60 (m, 1H); 1.52-1.41 (m, 1H); 1.39-1.26 (m, 1H); 1.25 (d, 3H, 4.8 Hz); 1.23 (d, 3H, 4.5 Hz); 0.83 (dd, 3H, 3 Hz, 4.5 Hz).
  • 13C NMR (75.5 MHz, DMSO-d6) δ 172.68; 172.63; 159.15; 158.73; 157.38; 157.25; 130.89; 124.99; 116.03; 62.51; 62.12; 54.29; 49.27; 42.47; 32.77; 30.43; 28.85; 20.46; 19.59; 18.72; 17.39; 13.90; 13.09.
  • HRMS calc. for C24H36N4O4: 444.2736; found: 444.2726±0.0013
  • HPLC (standard gradient) tR=6.80 min.
  • Compound 144
  • 1H NMR (300 MHz, CD3OD) δ 7.4 (m, 1H); 7.27 (dt, 1H, 1.5 Hz, 6.6 Hz); 7.22-7.14 (m, 2H); 7.08-6.98 (m, 2H); 6.78 9t, 2H, 6.6 Hz); 4.45-4.39 (m, 2H); 4.15 (d, 2H, 8.1 Hz); 7.74 (d, 1H, 9.3 Hz); 3.54 (d, 1H, 10.8 Hz); 3.35−3.22 (m, 2H); 3.20 (q, 1H, 1.5 Hz); 2.82-2.71 (m, 1H); 2.61-2.55 (m, 1H); 2.21-2.11 (m, 1 h); 2.02-1.94 (m, 1H); 1.74-1.40 (m, 5H); 1.04 (d, 3H, 6.6 Hz); 0.93 (d, 3H, 6.6 Hz); 0.74-0.64 9m, 1H); 0.45-0.28 (m, 2H); 0.15-0.08 (m, 1H); 0.06-0.02 (m, 1 H).
  • 13C NMR (75.5 MHz, CD3OD) δ 173.29; 172.14; 167.51; 155.47; 134.86; 134.81; 130.38; 130.31; 128.81; 128.25; 127.44; 121.63; 110.39; 107.71; 105.02; 67.10; 66.66; 62.81; 62.06; 60.10; 53.99; 41.44; 36.07; 31.91; 30.01; 29.18; 28.94; 27.79; 23.68; 23.15; 19.08; 18.25; 8.17; 4.98; 3.16.
  • HRMS: calc. for C31 H41N4O4Cl 568.2816; found 568.2802±0.0017
  • F. Mass Spectral Data for Selected Compounds of the Invention
  • TABLE 2
    Analysis of selected compounds of the invention
    Molecular Mono-
    Weight isotopic M + H
    Molecular Formula (calculated) Mass Found
    1 C30H40N4O5 536.7 536 537
    2 C30H40N4O4 520.7 520 521
    3 C30H42N4O4 522.7 522 523
    4 C30H42N4O5 538.7 538 539
    5 C28H36N4O5 508.6 508 509
    6 C30H40N4O5S 568.7 568 569
    7 C31H42N4O5 550.7 550 551
    8 C34H42N4O5 586.7 586 587
    9 C30H40N4O5 536.7 536 537
    10 C31H42N4O5 550.7 550 551
    11 C34H44N4O4 572.7 572 573
    12 C29H38N4O5 522.6 522 523
    13 C31H44N4O4 536.7 536 537
    14 C35H46N4O4 586.8 586 587
    15 C30H41N4O4Cl 557.1 556 557
    16 C30H41N4O4Cl 557.1 556 557
    17 C32H43N5O4 561.7 561 562
    18 C29H40N4O5 524.7 524 525
    19 C30H41N4O4F 540.7 540 541
    20 C31H42N4O4 534.7 534 535
    21 C35H44N4O4 584.7 584 585
    22 C31H44N4O5 552.7 552 553
    23 C34H44N4O4 572.7 572 573
    24 C28H40N4O4S 528.7 528 529
    25 C30H41N4O4Cl 557.1 556 557
    26 C31H42N4O5 550.7 550 551
    27 C27H39N5O4S 529.7 529 530
    28 C29H41N5O4 523.7 523 524
    29 C28H39N5O5 525.6 525 526
    30 C30H41N3O6 539.7 539 540
    34 C34H40N4O6 600.7 600 601
    38 C28H36N4O5 508.6 508 509
    39 C28H36N4O5 508.6 508 509
    40 C27H34N4O5 494.6 494 495
    41 C34H40N4O5 584.7 584 585
    52 C33H38N4O5 570.7 570 571
    55 C31H43N5O5 565.7 565 566
    56 C30H41N5O5 551.7 551 552
    57 C28H36N4O6 524.6 524 525
    58 C34H40N4O6 600.7 600 601
    59 C36H41N5O5 623.7 623 624
    60 C35H42N4O6 614.7 614 615
    65 C24H36N4O4 444.6 444 445
    71 C29H40N4O6 540.7 540 541
    72 C38H42N4O5 634.8 634 635
    76 C38H42N4O5 634.8 634 635
    77 C31H42N4O5 550.7 550 551
    80 C31H42N4O5 550.7 550 551
    85 C30H40N4O5 536.7 536 537
    87 C36H46N4O4 598.8 598 599
    88 C34H50N4O5 594.8 594 595
    89 C31H44N4O4 536.7 536 537
    90 C36H46N4O4 598.8 598 599
    91 C30H42N4O5 538.7 538 539
    92 C31H44N4O5 552.7 552 553
    96 C28H38N4O5 510.6 510 511
    97 C33H46N4O5 578.7 578 579
    98 C24H39N5O4 461.6 461 462
    99 C24H39N5O4 461.6 461 462
    109 C29H41N5O5 539.7 539 540
    110 C29H41N5O5 539.7 539 540
    111 C30H41N3O6 539.7 539 540
    112 C31H44N4O5 552.7 552 553
    120 C30H38N4O5 534.6 534 535
    121 C32H45N5O6 595.7 595 596
    122 C31H43N4O4Cl 571.2 570 571
    123 C29H41N5O4 523.7 523 524
    124 C29H41N5O4 523.7 523 524
    125 C30H40N4O5 536.7 536 537
    126 C32H46N4O5 566.7 566 567
    127 C30H38N6O3S 562.7 562 563
    128 C32H46N4O5 566.7 566 567
    129 C35H46N4O4 586.8 586 587
    130 C29H42N4O4S 542.7 542 543
    131 C31H43N4O4Cl 571.2 570 571
    132 C31H43N4O4Cl 571.2 570 571
    133 C31H43N4O4F 554.7 554 555
    134 C25H37N4O3Cl 477.0 476 477
    135 C31H45N5O5 567.7 567 568
    136 C34H45N5O4 587.8 587 588
    137 C28H41N5O4S 543.7 543 544
    138 C30H42N5O4Cl 572.1 571 572
    139 C30H42N5O4Cl 572.1 571 572
    140 C30H42N5O4F 555.7 555 556
    141 C32H44N4O5 564.7 564 565
    142 C35H44N4O4 584.7 584 585
    143 C29H40N4O4S 540.7 540 541
    144 C31H41N4O4Cl 569.1 568 569
    145 C31H41N4O4Cl 569.1 568 569
    146 C31H41N4O4F 552.7 552 553
    147 C31H43N5O5 565.7 565 566
    148 C34H43N5O4 585.7 585 586
    149 C30H40N5O4Cl 570.1 569 570
    150 C30H40N5O4Cl 570.1 569 570
    151 C30H40N5O4F 553.7 553 554
    152 C29H41N5O5 539.7 539 540
    153 C32H41N5O4 559.7 559 560
    154 C26H37N5O4S 515.7 515 516
    155 C28H38N5O4Cl 544.1 543 544
    156 C28H38N5O4Cl 544.1 543 544
    157 C28H38N5O4F 527.6 527 528
    158 C27H37N6O4Cl 545.1 544 545
    159 C31H44N4O5 552.7 552 553
    160 C31H44N4O5 552.7 552 553
    161 C31H45N5O4 551.7 551 552
    162 C31H44N4O4 536.7 536 537
    163 C31H44N4O4 536.7 536 537
    164 C31H44N4O4 536.7 536 537
    165 C31H44N4O5 552.7 552 553
    166 C31H44N4O5 552.7 552 553
    167 C32H42N4O4S 578.8 578 579
    168 C28H40N4O4S 528.7 528 529
    169 C31H43N4O4Cl 571.2 570 571
    170 C30H40N4O4Cl2 591.6 590 591
    171 C30H40N4O4F2 558.7 558 559
    172 C32H46N4O6 582.7 582 583
    173 C34H43N3O5 573.7 573 574
    174 C31H43N3O6 553.7 553 554
    175 C31H44N4O5 552.7 552 553
    176 C31H44N4O5 552.7 552 553
    177 C29H40N4O5 524.7 524 525
    178 C29H40N4O6 540.7 540 541
    179 C32H40N4O5 560.7 560 561
    180 C26H36N4O5S 516.7 516 517
    181 C28H37N4O5Cl 545.1 544 545
    182 C28H37N4O5Cl 545.1 544 545
    183 C28H37N4O5F 528.6 528 529
    184 C31H40N6O4 560.7 560 561
    185 C27H37N6O4Cl 545.1 544 545
    186 C31H40N6O5 576.7 576 577
    187 C31H41N4O4F3 590.7 590 591
    188 C30H41N4O4F 540.7 540 541
    189 C30H41N5O6 567.7 567 568
    190 C33H42N4O4S 590.8 590 591
    191 C32H44N4O5 564.7 564 565
    192 C31H4ON4O4Cl2 603.6 602 603
    193 C31H40N4O4F2 570.7 570 571
    194 C32H48N6O6 612.8 612 613
    195 C32H46N4O5 566.7 566 567
    196 C32H43N6O4Cl 611.2 610 611
    197 C32H45N6O5Cl 629.2 628 629
    198 C32H43N4O4Cl 583.2 582 583
    199 C27H39N4O6Cl 551.1 550 551
    200 C31H39N4O4Cl 567.1 566 567
    201 C34H42N4O4 570.7 570 571
    202 C31H42N4O5 550.7 550 551
    203 C30H40N5O5Cl 586.1 585 586
    204 C29H40N7O4Cl 586.1 585 586
    205 C32H45N4O4Cl 585.2 584 585
    206 C29H40N5O6SCl 622.2 621 622
    207 C29H39N6O5Cl 587.1 586 587
    208 C29H41N7O5 567.7 567 568
    209 C30H41N5O6 567.7 567 568
    210 C31H45N5O5 567.7 567 568
    211 C30H42N5O4Cl 572.1 571 572
    212 C31H44N5O4Cl 586.2 585 586
    213 C30H40N4O512 790.5 790 791
    214 C30H42N4O6 554.7 554 555
    215 C30H43N5O5 553.7 553 554
    216 C32H43N4O4Cl 583.2 582 583
    217 C31H40N4O4FCl 587.1 586 587
    218 C31H43N4O4Cl 571.2 570 571
    219 C30H40N4O4Cl2 591.6 590 591
    220 C31H43N4O4F 554.7 554 555
    221 C30H40N4O4FCl 575.1 574 575
    222 C34H50N4O5 594.8 594 595
    223 C32H44N4O6 580.7 580 581
    224 C36H48N4O4 600.8 600 601
    225 C37H48N4O5 628.8 628 629
    226 C39H49N5O4S 683.9 683 684
    227 C42H52N4O4 676.9 676 677
    Notes
    1. Molecular formulas and molecular weights (MW) are calculated automatically from the structure via ActivityBase ® software (IDBS, Guildford, Surrey, UK) or, for MW only, from the freeware program Molecular Weight Calculator v. 6.32
    2. M + H obtained from LC-MS analysis using the General Method as described
    3. All analyses conducted on material after preparative HPLC purification
  • Biological Methods and Results
  • The compounds of the present invention were evaluated for their ability to interact at the human motilin receptor utilizing a competitive radioligand binding assay as described in Method B1. Further characterization of the interaction can be performed utilizing the functional assays described in Methods B2, B3 and B4. Some of these methods can be conducted, if so desired, in a high throughput manner to permit the simultaneous evaluation of many compounds. Other assays have also been described that are suitable for HTS, such as that based upon the stable expression of a synthetic gene for the human motilin receptor.
  • Results for the examination of representative compounds of the present invention using Method B1 are presented in Table 3. The binding activity is listed as ranges with the following levels: A=0.001-0.10 μM; B=0.10-1.0 μM; C=1.0-10.0 μM. In addition, the assay results of two additional compounds using this Method are shown below. As can be observed, this demonstrates the activity of a representative bicyclic compound of Formula IV of the invention, which resulted from incorporation of D-proline as the second recognition building block. Significantly, the lack of binding activity obtained with compound 121, which is the linear analogue of compound 1 (Ki=level B), illustrates the critical importance of the cyclic structure to attaining the desired interaction.
  • Figure US20120226067A1-20120906-C00027
  • Competitive binding curves for two representative compounds of the invention (Compounds 8 and 11) are presented in FIG. 23 and FIG. 24, respectively.
  • For determination of functional significance of the binding, the compounds are preferably tested in the Aequorin assay as described in Method B2, although the procedure of Method B3 is also applicable. As can be seen from the data presented in Table 4, the representative compounds examined act as antagonists at the motilin receptor and are devoid of agonist activity at the concentrations studied. The functional activity is listed as ranges with the following levels: A=0.001-0.10 μM; B=0.10-1.0 μM. The higher sensitivity of the assay of Method B2, almost 100 times that of Method C, makes it the preferred one for this assessment. This is evident in the EC50 values obtained in each for the positive agonist standard, motilin. Additionally, Method B2 measures the actual signaling event, which makes it more relevant to the effect that is desired, whereas the assay of Method B3 simply measures GTP turnover.
  • TABLE 4
    Demonstration of Antagonist Activity at the Motilin Receptor
    Aequorin (Method B2)1
    Compound Binding (Ki) IC50
    142 A B
    149 A B
    167 A A
    168 A A
    212 A A
    Motilin 0.6 not applicable
    (human, porcine)2
    1Activity is listed as ranges with the following levels: A = 0.001-0.10 μM; B = 0.10-1.0 μM
    2Human and porcine motilin are the same peptide.
  • In addition, a common and scientifically-accepted ex vivo assay for the measurement of agonist or antagonist activity at the motilin receptor is the contraction of rabbit duodenum or other gastrointestinal smooth muscle tissue.A2-A4 Agonists are defined as compounds that induce>50% contraction relative to the motilin peptide, whereas antagonists are defined as compounds that cause>50% inhibition of the response to motilin. Compounds of the present invention have shown significant antagonist activity in this assay. For example, compound 144 exhibited a pA2=6.95, while compound 165 had a pA2=7.17, as calculated from the Schild plots of the response obtained at various concentrations as described in Method B4.
  • Gastric motility is generally measured in the clinical setting as the time required for gastric emptying and subsequent transit time through the GI tract. Gastric emptying scans are well known to those skilled in the art an, briefly, comprise use of an oral contrast agent, such as barium, or a radiolabeled meal. Solid and liquids can be measured independently. A test food or liquid is radiolabeled with an isotope (99mTc) and after ingestion or administration, transit time through the GI tract and gastric emptying are measured by visualization using gamma cameras. These studies are performed before and after the administration of the therapeutic agent to quantify the efficacy of the compound.
  • Example Method B1: Competitive Radioligand Binding Assay (Motilin Receptor) Materials:
      • Membranes were prepared from CHO cells stably transfected with the human motilin receptor and utilized at a quantity of 1.5 μg/assay point. [PerkinElmer™ SignalScreen® Product #6110544, PerkinElmer, Inc., Wellesley, Mass.]
      • [125I]-Motilin (PerkinElmer, #NEX-378); final concentration: 0.04-0.06 nM
      • Motilin (Bachem™, #H-4385, Bachem Bioscience Inc., King of Prussia, Pa.); final concentration: 1 μM
      • Multiscreen® Harvest plates-GF/B (Millipore™, #MAHFBI H60, Billerica, Mass.)
      • Deep-well polypropylene titer plate (Beckman Coulter™, #267006, Fullerton, Calif.)
      • TopSeal-A™ (PerkinElmer, #6005185, Wellesley, Mass.)
      • Bottom seal (Millipore™, #MATAHOPOO, Billerica, Mass.)
      • MicroScint-O™ (PerkinElmer, #6013611, Wellesley, Mass.)
      • Binding Buffer: 50 mM Tris-HCl (pH 7.4), 10 mM MgCl2, 1 mM EDTA, 0.1% BSA
    Assay Volumes:
      • 150 μL of membranes diluted in binding buffer
      • 10 μL of compound diluted in binding buffer
      • 10 μL of radioligand ([125I]-Motilin) diluted in binding buffer
  • Final Test Concentrations (N=11) for Compounds:
      • 10, 5, 2, 1, 0.5, 0.2, 0.1, 0.05, 0.02, 0.01, 0.005 μM.
    Compound Handling:
  • Compounds were provided frozen on dry ice at a stock concentration of 10 mM diluted in 100% DMSO and stored at −20° C. until the day of testing. On the test day, compounds were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO concentration in the assay was 0.5%.
  • Assay Protocol:
  • In deep-well plates, diluted cell membranes (1.5 μg/mL) are combined with 10 μL of either binding buffer (total binding, N=5), 1 μM motilin (non-specific binding, N=3) or the appropriate concentration of test compound. The reaction is initiated by addition of 10 μl of [125I]-motilin (final conc. 0.04-0.06 nM) to each well. Plates are sealed with TopSeal-A, vortexed gently and incubated at room temperature for 2 hours. The reaction is arrested by filtering samples through pre-soaked (0.3% polyethyleneimine, 2 h) Multiscreen Harvest plates using a Tomtec® Harvester (Tomtec, Hamden, Conn.)), washed 9 times with 500 μL of cold 50 mM Tris-HCl (pH 7.4), and than plates are air-dried in a fumehood for 30 minutes. A bottom seal is applied to the plates prior to the addition of 25 μL of MicroScint-0™ to each well. Plates are then sealed with TopSeal-A® and counted for 30 sec per well on a TopCount® Microplate Scintillation and Luminescence Counter (PerkinElmer, Wellesley, Mass.) where results are expressed as counts per minute (cpm).
  • Data are analyzed by GraphPad™ Prism (GraphPad Software, San Diego, Calif.) using a variable slope non-linear regression analysis. Ki values were calculated using a Kd value of 0.16 nM for [125I]-motilin (previously determined during membrane characterization).
  • D m ax = 1 - test concentration with maximal displacement - non - specific binding total binding - non - specific binding × 100
  • where total and non-specific binding represent the cpm obtained in the absence or presence of 1 μM motilin, respectively.
  • Example Method B2: Aequorin Functional Assay (Motilin Receptor) Materials:
      • Membranes were prepared using AequoScreen™ (EUROSCREEN, Belgium) cell lines expressing the human motilin receptor (cell line ES-380-A; receptor accession #AF034632). This cell line is constructed by transfection of the human motilin receptor into CHO-K1 cells co-expressing Gα16 and the mitochondrially targeted Aequorin (Ref #ES-WT-A5).
      • Motilin (Bachem™, #H-4385, Bachem Bioscience Inc., King of Prussia, Pa.)
      • Assay buffer: DMEM-F12 (Dulbeccoe's Modified Eagles Medium) with 15 mM HEPES and 0.1% BSA (pH 7.0)
      • Coelenterazine (Molecular Probes™, Leiden, The Netherlands)
  • Final Test Concentrations (N=5) for Compounds:
      • 10, 3.16, 1, 0.316, 0.1 μM.
    Compound Handling:
  • Compounds were provided as dry films at a quantity of approximately 1.2 μmol in pre-formatted 96-well plates. Compounds were dissolved in 100% DMSO at a concentration of 10 mM and stored at −20° C. until further use. Daughter plates were prepared at a concentration of 500 μM in 30% DMSO with 0.1% BSA and stored at −20° C. until testing. On the test day, compounds were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO concentration in the assay was 0.6%.
  • Cell Preparation:
  • Cells are collected from culture plates with Ca2+and Mg2+-free phosphate buffered saline (PBS) supplemented with 5 mM EDTA, pelleted for 2 minutes at 1000×g, resuspended in assay buffer (see above) at a density of 5×106 cells/mL and incubated overnight in the presence of 5 μM coelenterazine. After loading, cells were diluted with assay buffer to a concentration of 5×105 cells/mL.
  • Assay Protocol:
  • For agonist testing, 50 μl of the cell suspension was mixed with 50 μl of the appropriate concentration of test compound or motilin (reference agonist) in 96-well plates (duplicate samples). The emission of light resulting from receptor activation was recorded using the Functional Drug Screening System 6000 ‘FDSS 6000’ (Hamamatsu Photonics K.K., Japan).
  • For antagonist testing, an approximate EC80 concentration of motilin (i.e. 0.5 nM; 100 μL) was injected onto the cell suspension containing the test compounds (duplicate samples) 15−30 minutes after the end of agonist testing and the consequent emission of light resulting from receptor activation was measured as described in the paragraph above.
  • Results are expressed as Relative Light Units (RLU). Concentration response curves were analyzed using GraphPad™ Prism® (GraphPad Software, San Diego, Calif.) by non-linear regression analysis (sigmoidal dose-response) based on the equation E=Emax/(1+EC50/C)n where E is the measured RLU value at a given agonist concentration (C), Emax is the maximal response, EC50 is the concentration producing 50% stimulation and n is the slope index. For agonist testing, results for each concentration of test compound were expressed as percent activation relative to the signal induced by motilin at a concentration equal to the EC80 (i.e. 0.5 nM). For antagonist testing, results for each concentration of test compound were expressed as percent inhibition relative to the signal induced by motilin at a concentration equal to the EC80 (i.e. 0.5 nM).
  • Example Method B3: Flash Plate® Motilin [35S]-GTPγS Functional Assay Materials:
      • Membranes were prepared from CHO cells stably transfected with the human motilin receptor and utilized at a quantity of 1.5 μg/assay point. [PerkinElmer™ SignalScreen® Product #6110544, PerkinElmer, Inc. Wellesley, Mass.]
      • GTPγS Guanosine 5′-[y-thio]triphosphate tetralithium salt (Sigma, #G-8634, Sigma-Aldrich, St. Louis, Mo.)
      • [35S]-GTPγS (PerkinElmer, #NEX-030 H)
      • Motilin (Bachem™, #H-4385, Bachem Bioscience Inc., King of Prussia, Pa.)
      • 96-well FlashPlate® white polystyrene microplates (PerkinElmer, #SMP200, Wellesley, Mass.)
      • Deep-well polypropylene titer plate (Beckman Coulter™, #267006, Fullerton, Calif.)
      • TopSeal-A™ (PerkinElmer, #6005185, Wellesley, Mass.)
      • Assay Buffer: 50 mM Tris (pH 7.4), 100 mM NaCl, 10 mM MgCl2, 1 mM EDTA, 1 μM GDP, 0.1% BSA
    Assay Volumes:
      • 25 μL of compound diluted in assay buffer
      • 25 μL of assay buffer (agonist assay) or 0.6 μM motilin (0.1 μM final concentration) diluted in assay buffer (antagonist assay)
      • 100 μL of [35S]-GTPγS diluted in assay buffer
  • Final Test Concentrations (N=12) for Compounds:
  • 50, 20, 10, 5, 2, 1, 0.5, 0.2, 0.1, 0.05, 0.02, 0.01 μM.
  • Compound Handling:
  • Compounds were provided frozen on dry ice at a stock concentration of 10 mM diluted in 100% DMSO and stored at −20° C. until the day of testing. On the test day, compounds were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO concentration in the assay was 0.5%.
  • Assay Protocol:
  • CHO membranes were immobilized into 96-well FlashPlate® microplates. Test compound, GTPγS, motilin and [35S]-GTPγS were combined in each well according to the Assay Volumes described above.
  • For the assay to measure agonist activity, an additional 25 μl of buffer was added to each well in addition to 25 μL of either buffer (basal value, N=4), 1 μM (final conc.) motilin (Emax value, N=3), 25 μM (final conc.) GTPγS (non-specific value, N=4), or the appropriate concentration of test compound (N=3).
  • For the assay to measure antagonist activity, an additional 25 μL of either buffer (unstimulated control) or motilin (0.1 μM final conc.) is added to each well, in addition to either 25 μL of buffer (basal value, N=3), 1 μM (final conc.) motilin (Emax value, N=3), 25 μM (final conc.) GTPγS (non-specific value, N=4), or the appropriate concentration of test compound (N=3).
  • The reaction is initiated by addition of 100 mL of [36S]-GTPγS to each well. Each plate is sealed (TopSeal-A™) and incubated in the dark at room temperature for 150 min. Then, plates are counted for 30 seconds per well on the TopCount® NXT.
  • Data were analyzed by GraphPad™ Prism® 3.0 (GraphPad Software, San Diego, Calif.) using non-linear regression analysis (sigmoidal dose-response) for the calculation of IC50/EC50 values.
  • E ma x ( agonist ) or D ma x ( antagonist ) = Top - Bottom Bottom × 100
  • Where Top and Bottom correspond to the top and bottom values of the dose-response curve calculated by GraphPad™ Prism®).
  • Example Method B4: Rabbit Duodenum Contractility Assay
  • Duodenal segments were vertically suspended in organ chambers of 10 mL filled with Krebs buffer and connected to an isotonic force transducer, with a preload of 1 g. After a stabilization period, the muscle strips were challenged with 10−4 M acetylcholine and washed. This was repeated until a stable maximal contraction was obtained (2-3 times), with an interval of at least 20 minutes.
  • After a stable base line was reached, test compounds were added to the bath. After 15 min incubation, a dose response to motilin was recorded by adding logarithmically increasing concentrations of motilin to the bath (final concentration 10−9 to 10.6 M). A blank experiment (no test compound present) was also performed. At the end of the dose response curve, a supramaximal dose of acetylcholine (10−4 M) was given and this response was used as a reference (100% contraction).
  • The results of experiments at different concentrations of test compound were combined and analyzed to derive the pA2 value from the Schild plot.
  • It is appreciated that although specific experimental methods have been described herein for the purposes of illustration, various modifications to these experimental methods as well as alternate methods of experimentation may be used without departing from the scope of this invention.
  • TABLE 3
    Binding activity of selected compounds
    R1 R3 R6
     1
    Figure US20120226067A1-20120906-C00028
    Figure US20120226067A1-20120906-C00029
    Figure US20120226067A1-20120906-C00030
     2
    Figure US20120226067A1-20120906-C00031
    Figure US20120226067A1-20120906-C00032
    Figure US20120226067A1-20120906-C00033
     3
    Figure US20120226067A1-20120906-C00034
    Figure US20120226067A1-20120906-C00035
    Figure US20120226067A1-20120906-C00036
     4
    Figure US20120226067A1-20120906-C00037
    Figure US20120226067A1-20120906-C00038
    Figure US20120226067A1-20120906-C00039
     5
    Figure US20120226067A1-20120906-C00040
    CH3
    Figure US20120226067A1-20120906-C00041
     6
    Figure US20120226067A1-20120906-C00042
    Figure US20120226067A1-20120906-C00043
    Figure US20120226067A1-20120906-C00044
     7
    Figure US20120226067A1-20120906-C00045
    Figure US20120226067A1-20120906-C00046
    Figure US20120226067A1-20120906-C00047
     8
    Figure US20120226067A1-20120906-C00048
    Figure US20120226067A1-20120906-C00049
    Figure US20120226067A1-20120906-C00050
     9
    Figure US20120226067A1-20120906-C00051
    Figure US20120226067A1-20120906-C00052
    Figure US20120226067A1-20120906-C00053
     10
    Figure US20120226067A1-20120906-C00054
    Figure US20120226067A1-20120906-C00055
    Figure US20120226067A1-20120906-C00056
     11
    Figure US20120226067A1-20120906-C00057
    Figure US20120226067A1-20120906-C00058
    Figure US20120226067A1-20120906-C00059
     12
    Figure US20120226067A1-20120906-C00060
    Figure US20120226067A1-20120906-C00061
    Figure US20120226067A1-20120906-C00062
     13
    Figure US20120226067A1-20120906-C00063
    Figure US20120226067A1-20120906-C00064
    Figure US20120226067A1-20120906-C00065
     14
    Figure US20120226067A1-20120906-C00066
    Figure US20120226067A1-20120906-C00067
    Figure US20120226067A1-20120906-C00068
     15
    Figure US20120226067A1-20120906-C00069
    Figure US20120226067A1-20120906-C00070
    Figure US20120226067A1-20120906-C00071
     16
    Figure US20120226067A1-20120906-C00072
    Figure US20120226067A1-20120906-C00073
    Figure US20120226067A1-20120906-C00074
     17
    Figure US20120226067A1-20120906-C00075
    Figure US20120226067A1-20120906-C00076
    Figure US20120226067A1-20120906-C00077
     18
    Figure US20120226067A1-20120906-C00078
    Figure US20120226067A1-20120906-C00079
    Figure US20120226067A1-20120906-C00080
     19
    Figure US20120226067A1-20120906-C00081
    Figure US20120226067A1-20120906-C00082
    Figure US20120226067A1-20120906-C00083
     20
    Figure US20120226067A1-20120906-C00084
    Figure US20120226067A1-20120906-C00085
    Figure US20120226067A1-20120906-C00086
     21
    Figure US20120226067A1-20120906-C00087
    Figure US20120226067A1-20120906-C00088
    Figure US20120226067A1-20120906-C00089
     22
    Figure US20120226067A1-20120906-C00090
    Figure US20120226067A1-20120906-C00091
    Figure US20120226067A1-20120906-C00092
     23
    Figure US20120226067A1-20120906-C00093
    Figure US20120226067A1-20120906-C00094
    Figure US20120226067A1-20120906-C00095
     24
    Figure US20120226067A1-20120906-C00096
    Figure US20120226067A1-20120906-C00097
    Figure US20120226067A1-20120906-C00098
     25
    Figure US20120226067A1-20120906-C00099
    Figure US20120226067A1-20120906-C00100
    Figure US20120226067A1-20120906-C00101
     26
    Figure US20120226067A1-20120906-C00102
    Figure US20120226067A1-20120906-C00103
    Figure US20120226067A1-20120906-C00104
     27
    Figure US20120226067A1-20120906-C00105
    Figure US20120226067A1-20120906-C00106
    Figure US20120226067A1-20120906-C00107
     28
    Figure US20120226067A1-20120906-C00108
    Figure US20120226067A1-20120906-C00109
    Figure US20120226067A1-20120906-C00110
     29
    Figure US20120226067A1-20120906-C00111
    Figure US20120226067A1-20120906-C00112
    Figure US20120226067A1-20120906-C00113
     30
    Figure US20120226067A1-20120906-C00114
    Figure US20120226067A1-20120906-C00115
    Figure US20120226067A1-20120906-C00116
     34
    Figure US20120226067A1-20120906-C00117
    Figure US20120226067A1-20120906-C00118
    Figure US20120226067A1-20120906-C00119
     38
    Figure US20120226067A1-20120906-C00120
    Figure US20120226067A1-20120906-C00121
    CH3
     39
    Figure US20120226067A1-20120906-C00122
    Figure US20120226067A1-20120906-C00123
    H
     40
    Figure US20120226067A1-20120906-C00124
    Figure US20120226067A1-20120906-C00125
    H
     41
    Figure US20120226067A1-20120906-C00126
    Figure US20120226067A1-20120906-C00127
    Figure US20120226067A1-20120906-C00128
     52
    Figure US20120226067A1-20120906-C00129
    Figure US20120226067A1-20120906-C00130
    Figure US20120226067A1-20120906-C00131
     55
    Figure US20120226067A1-20120906-C00132
    Figure US20120226067A1-20120906-C00133
    Figure US20120226067A1-20120906-C00134
     56
    Figure US20120226067A1-20120906-C00135
    Figure US20120226067A1-20120906-C00136
    Figure US20120226067A1-20120906-C00137
     57
    Figure US20120226067A1-20120906-C00138
    Figure US20120226067A1-20120906-C00139
    Figure US20120226067A1-20120906-C00140
     58
    Figure US20120226067A1-20120906-C00141
    Figure US20120226067A1-20120906-C00142
    Figure US20120226067A1-20120906-C00143
     59
    Figure US20120226067A1-20120906-C00144
    Figure US20120226067A1-20120906-C00145
    Figure US20120226067A1-20120906-C00146
     60
    Figure US20120226067A1-20120906-C00147
    Figure US20120226067A1-20120906-C00148
    Figure US20120226067A1-20120906-C00149
     65
    Figure US20120226067A1-20120906-C00150
    Figure US20120226067A1-20120906-C00151
    Figure US20120226067A1-20120906-C00152
     71
    Figure US20120226067A1-20120906-C00153
    Figure US20120226067A1-20120906-C00154
    Figure US20120226067A1-20120906-C00155
     72
    Figure US20120226067A1-20120906-C00156
    Figure US20120226067A1-20120906-C00157
    Figure US20120226067A1-20120906-C00158
     76
    Figure US20120226067A1-20120906-C00159
    Figure US20120226067A1-20120906-C00160
    Figure US20120226067A1-20120906-C00161
     77
    Figure US20120226067A1-20120906-C00162
    Figure US20120226067A1-20120906-C00163
    Figure US20120226067A1-20120906-C00164
     80
    Figure US20120226067A1-20120906-C00165
    Figure US20120226067A1-20120906-C00166
    Figure US20120226067A1-20120906-C00167
     85
    Figure US20120226067A1-20120906-C00168
    Figure US20120226067A1-20120906-C00169
    H
     87
    Figure US20120226067A1-20120906-C00170
    Figure US20120226067A1-20120906-C00171
    Figure US20120226067A1-20120906-C00172
     88
    Figure US20120226067A1-20120906-C00173
    Figure US20120226067A1-20120906-C00174
    Figure US20120226067A1-20120906-C00175
     89
    Figure US20120226067A1-20120906-C00176
    Figure US20120226067A1-20120906-C00177
    Figure US20120226067A1-20120906-C00178
     90
    Figure US20120226067A1-20120906-C00179
    Figure US20120226067A1-20120906-C00180
    Figure US20120226067A1-20120906-C00181
     91
    Figure US20120226067A1-20120906-C00182
    Figure US20120226067A1-20120906-C00183
    Figure US20120226067A1-20120906-C00184
     92
    Figure US20120226067A1-20120906-C00185
    Figure US20120226067A1-20120906-C00186
    Figure US20120226067A1-20120906-C00187
     96
    Figure US20120226067A1-20120906-C00188
    H
    Figure US20120226067A1-20120906-C00189
     97
    Figure US20120226067A1-20120906-C00190
    Figure US20120226067A1-20120906-C00191
    Figure US20120226067A1-20120906-C00192
     98
    Figure US20120226067A1-20120906-C00193
    Figure US20120226067A1-20120906-C00194
    Figure US20120226067A1-20120906-C00195
     99
    Figure US20120226067A1-20120906-C00196
    Figure US20120226067A1-20120906-C00197
    Figure US20120226067A1-20120906-C00198
    109
    Figure US20120226067A1-20120906-C00199
    Figure US20120226067A1-20120906-C00200
    Figure US20120226067A1-20120906-C00201
    110
    Figure US20120226067A1-20120906-C00202
    Figure US20120226067A1-20120906-C00203
    Figure US20120226067A1-20120906-C00204
    111
    Figure US20120226067A1-20120906-C00205
    Figure US20120226067A1-20120906-C00206
    Figure US20120226067A1-20120906-C00207
    112
    Figure US20120226067A1-20120906-C00208
    Figure US20120226067A1-20120906-C00209
    Figure US20120226067A1-20120906-C00210
    122
    Figure US20120226067A1-20120906-C00211
    Figure US20120226067A1-20120906-C00212
    Figure US20120226067A1-20120906-C00213
    123
    Figure US20120226067A1-20120906-C00214
    Figure US20120226067A1-20120906-C00215
    Figure US20120226067A1-20120906-C00216
    124
    Figure US20120226067A1-20120906-C00217
    Figure US20120226067A1-20120906-C00218
    Figure US20120226067A1-20120906-C00219
    125
    Figure US20120226067A1-20120906-C00220
    Figure US20120226067A1-20120906-C00221
    Figure US20120226067A1-20120906-C00222
    126
    Figure US20120226067A1-20120906-C00223
    Figure US20120226067A1-20120906-C00224
    Figure US20120226067A1-20120906-C00225
    127
    Figure US20120226067A1-20120906-C00226
    Figure US20120226067A1-20120906-C00227
    Figure US20120226067A1-20120906-C00228
    128
    Figure US20120226067A1-20120906-C00229
    Figure US20120226067A1-20120906-C00230
    Figure US20120226067A1-20120906-C00231
    129
    Figure US20120226067A1-20120906-C00232
    Figure US20120226067A1-20120906-C00233
    Figure US20120226067A1-20120906-C00234
    130
    Figure US20120226067A1-20120906-C00235
    Figure US20120226067A1-20120906-C00236
    Figure US20120226067A1-20120906-C00237
    131
    Figure US20120226067A1-20120906-C00238
    Figure US20120226067A1-20120906-C00239
    Figure US20120226067A1-20120906-C00240
    132
    Figure US20120226067A1-20120906-C00241
    Figure US20120226067A1-20120906-C00242
    Figure US20120226067A1-20120906-C00243
    133
    Figure US20120226067A1-20120906-C00244
    Figure US20120226067A1-20120906-C00245
    Figure US20120226067A1-20120906-C00246
    134
    Figure US20120226067A1-20120906-C00247
    Figure US20120226067A1-20120906-C00248
    Figure US20120226067A1-20120906-C00249
    135
    Figure US20120226067A1-20120906-C00250
    Figure US20120226067A1-20120906-C00251
    Figure US20120226067A1-20120906-C00252
    136
    Figure US20120226067A1-20120906-C00253
    Figure US20120226067A1-20120906-C00254
    Figure US20120226067A1-20120906-C00255
    137
    Figure US20120226067A1-20120906-C00256
    Figure US20120226067A1-20120906-C00257
    Figure US20120226067A1-20120906-C00258
    138
    Figure US20120226067A1-20120906-C00259
    Figure US20120226067A1-20120906-C00260
    Figure US20120226067A1-20120906-C00261
    139
    Figure US20120226067A1-20120906-C00262
    Figure US20120226067A1-20120906-C00263
    Figure US20120226067A1-20120906-C00264
    140
    Figure US20120226067A1-20120906-C00265
    Figure US20120226067A1-20120906-C00266
    Figure US20120226067A1-20120906-C00267
    141
    Figure US20120226067A1-20120906-C00268
    Figure US20120226067A1-20120906-C00269
    Figure US20120226067A1-20120906-C00270
    142
    Figure US20120226067A1-20120906-C00271
    Figure US20120226067A1-20120906-C00272
    Figure US20120226067A1-20120906-C00273
    143
    Figure US20120226067A1-20120906-C00274
    Figure US20120226067A1-20120906-C00275
    Figure US20120226067A1-20120906-C00276
    144
    Figure US20120226067A1-20120906-C00277
    Figure US20120226067A1-20120906-C00278
    Figure US20120226067A1-20120906-C00279
    145
    Figure US20120226067A1-20120906-C00280
    Figure US20120226067A1-20120906-C00281
    146
    Figure US20120226067A1-20120906-C00282
    Figure US20120226067A1-20120906-C00283
    Figure US20120226067A1-20120906-C00284
    147
    Figure US20120226067A1-20120906-C00285
    Figure US20120226067A1-20120906-C00286
    Figure US20120226067A1-20120906-C00287
    148
    Figure US20120226067A1-20120906-C00288
    Figure US20120226067A1-20120906-C00289
    Figure US20120226067A1-20120906-C00290
    149
    Figure US20120226067A1-20120906-C00291
    Figure US20120226067A1-20120906-C00292
    Figure US20120226067A1-20120906-C00293
    150
    Figure US20120226067A1-20120906-C00294
    Figure US20120226067A1-20120906-C00295
    Figure US20120226067A1-20120906-C00296
    151
    Figure US20120226067A1-20120906-C00297
    Figure US20120226067A1-20120906-C00298
    Figure US20120226067A1-20120906-C00299
    152
    Figure US20120226067A1-20120906-C00300
    Figure US20120226067A1-20120906-C00301
    Figure US20120226067A1-20120906-C00302
    153
    Figure US20120226067A1-20120906-C00303
    Figure US20120226067A1-20120906-C00304
    Figure US20120226067A1-20120906-C00305
    154
    Figure US20120226067A1-20120906-C00306
    Figure US20120226067A1-20120906-C00307
    Figure US20120226067A1-20120906-C00308
    155
    Figure US20120226067A1-20120906-C00309
    Figure US20120226067A1-20120906-C00310
    Figure US20120226067A1-20120906-C00311
    156
    Figure US20120226067A1-20120906-C00312
    Figure US20120226067A1-20120906-C00313
    Figure US20120226067A1-20120906-C00314
    157
    Figure US20120226067A1-20120906-C00315
    Figure US20120226067A1-20120906-C00316
    Figure US20120226067A1-20120906-C00317
    158
    Figure US20120226067A1-20120906-C00318
    Figure US20120226067A1-20120906-C00319
    Figure US20120226067A1-20120906-C00320
    159
    Figure US20120226067A1-20120906-C00321
    Figure US20120226067A1-20120906-C00322
    Figure US20120226067A1-20120906-C00323
    160
    Figure US20120226067A1-20120906-C00324
    Figure US20120226067A1-20120906-C00325
    Figure US20120226067A1-20120906-C00326
    161
    Figure US20120226067A1-20120906-C00327
    Figure US20120226067A1-20120906-C00328
    Figure US20120226067A1-20120906-C00329
    162
    Figure US20120226067A1-20120906-C00330
    Figure US20120226067A1-20120906-C00331
    Figure US20120226067A1-20120906-C00332
    163
    Figure US20120226067A1-20120906-C00333
    Figure US20120226067A1-20120906-C00334
    Figure US20120226067A1-20120906-C00335
    164
    Figure US20120226067A1-20120906-C00336
    Figure US20120226067A1-20120906-C00337
    Figure US20120226067A1-20120906-C00338
    165
    Figure US20120226067A1-20120906-C00339
    Figure US20120226067A1-20120906-C00340
    Figure US20120226067A1-20120906-C00341
    166
    Figure US20120226067A1-20120906-C00342
    Figure US20120226067A1-20120906-C00343
    Figure US20120226067A1-20120906-C00344
    167
    Figure US20120226067A1-20120906-C00345
    Figure US20120226067A1-20120906-C00346
    Figure US20120226067A1-20120906-C00347
    168
    Figure US20120226067A1-20120906-C00348
    Figure US20120226067A1-20120906-C00349
    Figure US20120226067A1-20120906-C00350
    169
    Figure US20120226067A1-20120906-C00351
    Figure US20120226067A1-20120906-C00352
    Figure US20120226067A1-20120906-C00353
    170
    Figure US20120226067A1-20120906-C00354
    Figure US20120226067A1-20120906-C00355
    Figure US20120226067A1-20120906-C00356
    171
    Figure US20120226067A1-20120906-C00357
    Figure US20120226067A1-20120906-C00358
    Figure US20120226067A1-20120906-C00359
    172
    Figure US20120226067A1-20120906-C00360
    Figure US20120226067A1-20120906-C00361
    Figure US20120226067A1-20120906-C00362
    173
    Figure US20120226067A1-20120906-C00363
    Figure US20120226067A1-20120906-C00364
    Figure US20120226067A1-20120906-C00365
    174
    Figure US20120226067A1-20120906-C00366
    Figure US20120226067A1-20120906-C00367
    Figure US20120226067A1-20120906-C00368
    175
    Figure US20120226067A1-20120906-C00369
    Figure US20120226067A1-20120906-C00370
    Figure US20120226067A1-20120906-C00371
    176
    Figure US20120226067A1-20120906-C00372
    Figure US20120226067A1-20120906-C00373
    Figure US20120226067A1-20120906-C00374
    177
    Figure US20120226067A1-20120906-C00375
    Figure US20120226067A1-20120906-C00376
    Figure US20120226067A1-20120906-C00377
    178
    Figure US20120226067A1-20120906-C00378
    Figure US20120226067A1-20120906-C00379
    Figure US20120226067A1-20120906-C00380
    179
    Figure US20120226067A1-20120906-C00381
    Figure US20120226067A1-20120906-C00382
    Figure US20120226067A1-20120906-C00383
    180
    Figure US20120226067A1-20120906-C00384
    Figure US20120226067A1-20120906-C00385
    Figure US20120226067A1-20120906-C00386
    181
    Figure US20120226067A1-20120906-C00387
    Figure US20120226067A1-20120906-C00388
    Figure US20120226067A1-20120906-C00389
    182
    Figure US20120226067A1-20120906-C00390
    Figure US20120226067A1-20120906-C00391
    Figure US20120226067A1-20120906-C00392
    183
    Figure US20120226067A1-20120906-C00393
    Figure US20120226067A1-20120906-C00394
    Figure US20120226067A1-20120906-C00395
    184
    Figure US20120226067A1-20120906-C00396
    Figure US20120226067A1-20120906-C00397
    Figure US20120226067A1-20120906-C00398
    185
    Figure US20120226067A1-20120906-C00399
    Figure US20120226067A1-20120906-C00400
    Figure US20120226067A1-20120906-C00401
    186
    Figure US20120226067A1-20120906-C00402
    Figure US20120226067A1-20120906-C00403
    Figure US20120226067A1-20120906-C00404
    187
    Figure US20120226067A1-20120906-C00405
    Figure US20120226067A1-20120906-C00406
    Figure US20120226067A1-20120906-C00407
    188
    Figure US20120226067A1-20120906-C00408
    Figure US20120226067A1-20120906-C00409
    Figure US20120226067A1-20120906-C00410
    189
    Figure US20120226067A1-20120906-C00411
    Figure US20120226067A1-20120906-C00412
    Figure US20120226067A1-20120906-C00413
    190
    Figure US20120226067A1-20120906-C00414
    Figure US20120226067A1-20120906-C00415
    Figure US20120226067A1-20120906-C00416
    191
    Figure US20120226067A1-20120906-C00417
    Figure US20120226067A1-20120906-C00418
    Figure US20120226067A1-20120906-C00419
    192
    Figure US20120226067A1-20120906-C00420
    Figure US20120226067A1-20120906-C00421
    Figure US20120226067A1-20120906-C00422
    193
    Figure US20120226067A1-20120906-C00423
    Figure US20120226067A1-20120906-C00424
    Figure US20120226067A1-20120906-C00425
    194
    Figure US20120226067A1-20120906-C00426
    Figure US20120226067A1-20120906-C00427
    Figure US20120226067A1-20120906-C00428
    195
    Figure US20120226067A1-20120906-C00429
    Figure US20120226067A1-20120906-C00430
    Figure US20120226067A1-20120906-C00431
    196
    Figure US20120226067A1-20120906-C00432
    Figure US20120226067A1-20120906-C00433
    Figure US20120226067A1-20120906-C00434
    197
    Figure US20120226067A1-20120906-C00435
    Figure US20120226067A1-20120906-C00436
    Figure US20120226067A1-20120906-C00437
    198
    Figure US20120226067A1-20120906-C00438
    Figure US20120226067A1-20120906-C00439
    Figure US20120226067A1-20120906-C00440
    199
    Figure US20120226067A1-20120906-C00441
    Figure US20120226067A1-20120906-C00442
    Figure US20120226067A1-20120906-C00443
    200
    Figure US20120226067A1-20120906-C00444
    Figure US20120226067A1-20120906-C00445
    Figure US20120226067A1-20120906-C00446
    201
    Figure US20120226067A1-20120906-C00447
    Figure US20120226067A1-20120906-C00448
    Figure US20120226067A1-20120906-C00449
    202
    Figure US20120226067A1-20120906-C00450
    Figure US20120226067A1-20120906-C00451
    Figure US20120226067A1-20120906-C00452
    203
    Figure US20120226067A1-20120906-C00453
    Figure US20120226067A1-20120906-C00454
    Figure US20120226067A1-20120906-C00455
    204
    Figure US20120226067A1-20120906-C00456
    Figure US20120226067A1-20120906-C00457
    Figure US20120226067A1-20120906-C00458
    205
    Figure US20120226067A1-20120906-C00459
    Figure US20120226067A1-20120906-C00460
    Figure US20120226067A1-20120906-C00461
    206
    Figure US20120226067A1-20120906-C00462
    Figure US20120226067A1-20120906-C00463
    Figure US20120226067A1-20120906-C00464
    207
    Figure US20120226067A1-20120906-C00465
    Figure US20120226067A1-20120906-C00466
    Figure US20120226067A1-20120906-C00467
    208
    Figure US20120226067A1-20120906-C00468
    Figure US20120226067A1-20120906-C00469
    Figure US20120226067A1-20120906-C00470
    209
    Figure US20120226067A1-20120906-C00471
    Figure US20120226067A1-20120906-C00472
    Figure US20120226067A1-20120906-C00473
    210
    Figure US20120226067A1-20120906-C00474
    Figure US20120226067A1-20120906-C00475
    Figure US20120226067A1-20120906-C00476
    211
    Figure US20120226067A1-20120906-C00477
    Figure US20120226067A1-20120906-C00478
    Figure US20120226067A1-20120906-C00479
    212
    Figure US20120226067A1-20120906-C00480
    Figure US20120226067A1-20120906-C00481
    Figure US20120226067A1-20120906-C00482
    213
    Figure US20120226067A1-20120906-C00483
    Figure US20120226067A1-20120906-C00484
    Figure US20120226067A1-20120906-C00485
    214
    Figure US20120226067A1-20120906-C00486
    Figure US20120226067A1-20120906-C00487
    Figure US20120226067A1-20120906-C00488
    215
    Figure US20120226067A1-20120906-C00489
    Figure US20120226067A1-20120906-C00490
    Figure US20120226067A1-20120906-C00491
    216
    Figure US20120226067A1-20120906-C00492
    Figure US20120226067A1-20120906-C00493
    Figure US20120226067A1-20120906-C00494
    217
    Figure US20120226067A1-20120906-C00495
    Figure US20120226067A1-20120906-C00496
    Figure US20120226067A1-20120906-C00497
    218
    Figure US20120226067A1-20120906-C00498
    Figure US20120226067A1-20120906-C00499
    Figure US20120226067A1-20120906-C00500
    219
    Figure US20120226067A1-20120906-C00501
    Figure US20120226067A1-20120906-C00502
    Figure US20120226067A1-20120906-C00503
    220
    Figure US20120226067A1-20120906-C00504
    Figure US20120226067A1-20120906-C00505
    Figure US20120226067A1-20120906-C00506
    221
    Figure US20120226067A1-20120906-C00507
    Figure US20120226067A1-20120906-C00508
    222
    Figure US20120226067A1-20120906-C00509
    Figure US20120226067A1-20120906-C00510
    Figure US20120226067A1-20120906-C00511
    223
    Figure US20120226067A1-20120906-C00512
    Figure US20120226067A1-20120906-C00513
    Figure US20120226067A1-20120906-C00514
    224
    Figure US20120226067A1-20120906-C00515
    Figure US20120226067A1-20120906-C00516
    Figure US20120226067A1-20120906-C00517
    225
    Figure US20120226067A1-20120906-C00518
    Figure US20120226067A1-20120906-C00519
    Figure US20120226067A1-20120906-C00520
    226
    Figure US20120226067A1-20120906-C00521
    Figure US20120226067A1-20120906-C00522
    Figure US20120226067A1-20120906-C00523
    227
    Figure US20120226067A1-20120906-C00524
    Figure US20120226067A1-20120906-C00525
    Figure US20120226067A1-20120906-C00526
    T K i 1,2
     1
    Figure US20120226067A1-20120906-C00527
    B
     2
    Figure US20120226067A1-20120906-C00528
    A
     3
    Figure US20120226067A1-20120906-C00529
    B
     4
    Figure US20120226067A1-20120906-C00530
    A
     5
    Figure US20120226067A1-20120906-C00531
    B
     6
    Figure US20120226067A1-20120906-C00532
    B
     7
    Figure US20120226067A1-20120906-C00533
    B
     8
    Figure US20120226067A1-20120906-C00534
    B
     9
    Figure US20120226067A1-20120906-C00535
    B
     10
    Figure US20120226067A1-20120906-C00536
    A
     11
    Figure US20120226067A1-20120906-C00537
    A
     12
    Figure US20120226067A1-20120906-C00538
    B
     13
    Figure US20120226067A1-20120906-C00539
    B
     14
    Figure US20120226067A1-20120906-C00540
    B
     15
    Figure US20120226067A1-20120906-C00541
    A
     16
    Figure US20120226067A1-20120906-C00542
    A
     17
    Figure US20120226067A1-20120906-C00543
    B
     18
    Figure US20120226067A1-20120906-C00544
    B
     19
    Figure US20120226067A1-20120906-C00545
    A
     20
    Figure US20120226067A1-20120906-C00546
    B
     21
    Figure US20120226067A1-20120906-C00547
    A
     22
    Figure US20120226067A1-20120906-C00548
    A
     23
    Figure US20120226067A1-20120906-C00549
    A
     24
    Figure US20120226067A1-20120906-C00550
    A
     25
    Figure US20120226067A1-20120906-C00551
    B
     26
    Figure US20120226067A1-20120906-C00552
    A
     27
    Figure US20120226067A1-20120906-C00553
    B
     28
    Figure US20120226067A1-20120906-C00554
    B
     29
    Figure US20120226067A1-20120906-C00555
    B
     30
    Figure US20120226067A1-20120906-C00556
    B
     34
    Figure US20120226067A1-20120906-C00557
    B
     38
    Figure US20120226067A1-20120906-C00558
    B
     39
    Figure US20120226067A1-20120906-C00559
    B
     40
    Figure US20120226067A1-20120906-C00560
    C
     41
    Figure US20120226067A1-20120906-C00561
    C
     52
    Figure US20120226067A1-20120906-C00562
    B
     55
    Figure US20120226067A1-20120906-C00563
    B
     56
    Figure US20120226067A1-20120906-C00564
    B
     57
    Figure US20120226067A1-20120906-C00565
    B
     58
    Figure US20120226067A1-20120906-C00566
    B
     59
    Figure US20120226067A1-20120906-C00567
    B
     60
    Figure US20120226067A1-20120906-C00568
    C
     65
    Figure US20120226067A1-20120906-C00569
    B
     71
    Figure US20120226067A1-20120906-C00570
    B
     72
    Figure US20120226067A1-20120906-C00571
    B
     76
    Figure US20120226067A1-20120906-C00572
    C
     77
    Figure US20120226067A1-20120906-C00573
    C
     80
    Figure US20120226067A1-20120906-C00574
    B
     85
    Figure US20120226067A1-20120906-C00575
    B
     87
    Figure US20120226067A1-20120906-C00576
    B
     88
    Figure US20120226067A1-20120906-C00577
    C
     89
    Figure US20120226067A1-20120906-C00578
    C
     90
    Figure US20120226067A1-20120906-C00579
    C
     91
    Figure US20120226067A1-20120906-C00580
    C
     92
    Figure US20120226067A1-20120906-C00581
    B
     96
    Figure US20120226067A1-20120906-C00582
    C
     97
    Figure US20120226067A1-20120906-C00583
    C
     98
    Figure US20120226067A1-20120906-C00584
    C
     99
    Figure US20120226067A1-20120906-C00585
    C
    109
    Figure US20120226067A1-20120906-C00586
    B
    110
    Figure US20120226067A1-20120906-C00587
    B
    111
    Figure US20120226067A1-20120906-C00588
    B
    112
    Figure US20120226067A1-20120906-C00589
    B
    122
    Figure US20120226067A1-20120906-C00590
    B
    123
    Figure US20120226067A1-20120906-C00591
    B
    124
    Figure US20120226067A1-20120906-C00592
    B
    125
    Figure US20120226067A1-20120906-C00593
    B
    126
    Figure US20120226067A1-20120906-C00594
    B
    127
    Figure US20120226067A1-20120906-C00595
    B
    128
    Figure US20120226067A1-20120906-C00596
    B
    129
    Figure US20120226067A1-20120906-C00597
    A
    130
    Figure US20120226067A1-20120906-C00598
    B
    131
    Figure US20120226067A1-20120906-C00599
    A
    132
    Figure US20120226067A1-20120906-C00600
    A
    133
    Figure US20120226067A1-20120906-C00601
    A
    134
    Figure US20120226067A1-20120906-C00602
    C
    135
    Figure US20120226067A1-20120906-C00603
    B
    136
    Figure US20120226067A1-20120906-C00604
    B
    137
    Figure US20120226067A1-20120906-C00605
    B
    138
    Figure US20120226067A1-20120906-C00606
    B
    139
    Figure US20120226067A1-20120906-C00607
    B
    140
    Figure US20120226067A1-20120906-C00608
    B
    141
    Figure US20120226067A1-20120906-C00609
    A
    142
    Figure US20120226067A1-20120906-C00610
    A
    143
    Figure US20120226067A1-20120906-C00611
    B
    144
    Figure US20120226067A1-20120906-C00612
    A
    145
    Figure US20120226067A1-20120906-C00613
    A
    146
    Figure US20120226067A1-20120906-C00614
    A
    147
    Figure US20120226067A1-20120906-C00615
    B
    148
    Figure US20120226067A1-20120906-C00616
    B
    149
    Figure US20120226067A1-20120906-C00617
    A
    150
    Figure US20120226067A1-20120906-C00618
    B
    151
    Figure US20120226067A1-20120906-C00619
    B
    152
    Figure US20120226067A1-20120906-C00620
    B
    153
    Figure US20120226067A1-20120906-C00621
    B
    154
    Figure US20120226067A1-20120906-C00622
    B
    155
    Figure US20120226067A1-20120906-C00623
    A
    156
    Figure US20120226067A1-20120906-C00624
    A
    157
    Figure US20120226067A1-20120906-C00625
    B
    158
    Figure US20120226067A1-20120906-C00626
    A
    159
    Figure US20120226067A1-20120906-C00627
    B
    160
    Figure US20120226067A1-20120906-C00628
    B
    161
    Figure US20120226067A1-20120906-C00629
    B
    162
    Figure US20120226067A1-20120906-C00630
    B
    163
    Figure US20120226067A1-20120906-C00631
    A
    164
    Figure US20120226067A1-20120906-C00632
    B
    165
    Figure US20120226067A1-20120906-C00633
    A
    166
    Figure US20120226067A1-20120906-C00634
    B
    167
    Figure US20120226067A1-20120906-C00635
    A
    168
    Figure US20120226067A1-20120906-C00636
    A
    169
    Figure US20120226067A1-20120906-C00637
    B
    170
    Figure US20120226067A1-20120906-C00638
    A
    171
    Figure US20120226067A1-20120906-C00639
    A
    172
    Figure US20120226067A1-20120906-C00640
    A
    173
    Figure US20120226067A1-20120906-C00641
    B
    174
    Figure US20120226067A1-20120906-C00642
    B
    175
    Figure US20120226067A1-20120906-C00643
    B
    176
    Figure US20120226067A1-20120906-C00644
    B
    177
    Figure US20120226067A1-20120906-C00645
    B
    178
    Figure US20120226067A1-20120906-C00646
    B
    179
    Figure US20120226067A1-20120906-C00647
    B
    180
    Figure US20120226067A1-20120906-C00648
    B
    181
    Figure US20120226067A1-20120906-C00649
    A
    182
    Figure US20120226067A1-20120906-C00650
    A
    183
    Figure US20120226067A1-20120906-C00651
    B
    184
    Figure US20120226067A1-20120906-C00652
    B
    185
    Figure US20120226067A1-20120906-C00653
    B
    186
    Figure US20120226067A1-20120906-C00654
    B
    187
    Figure US20120226067A1-20120906-C00655
    A
    188
    Figure US20120226067A1-20120906-C00656
    A
    189
    Figure US20120226067A1-20120906-C00657
    B
    190
    Figure US20120226067A1-20120906-C00658
    A
    191
    Figure US20120226067A1-20120906-C00659
    A
    192
    Figure US20120226067A1-20120906-C00660
    A
    193
    Figure US20120226067A1-20120906-C00661
    A
    194
    Figure US20120226067A1-20120906-C00662
    B
    195
    Figure US20120226067A1-20120906-C00663
    A
    196
    Figure US20120226067A1-20120906-C00664
    197
    Figure US20120226067A1-20120906-C00665
    198
    Figure US20120226067A1-20120906-C00666
    A
    199
    Figure US20120226067A1-20120906-C00667
    B
    200
    Figure US20120226067A1-20120906-C00668
    A
    201
    Figure US20120226067A1-20120906-C00669
    B
    202
    Figure US20120226067A1-20120906-C00670
    A
    203
    Figure US20120226067A1-20120906-C00671
    B
    204
    Figure US20120226067A1-20120906-C00672
    A
    205
    Figure US20120226067A1-20120906-C00673
    B
    206
    Figure US20120226067A1-20120906-C00674
    B
    207
    Figure US20120226067A1-20120906-C00675
    B
    208
    Figure US20120226067A1-20120906-C00676
    B
    209
    Figure US20120226067A1-20120906-C00677
    C
    210
    Figure US20120226067A1-20120906-C00678
    211
    Figure US20120226067A1-20120906-C00679
    A
    212
    Figure US20120226067A1-20120906-C00680
    A
    213
    Figure US20120226067A1-20120906-C00681
    B
    214
    Figure US20120226067A1-20120906-C00682
    B
    215
    Figure US20120226067A1-20120906-C00683
    B
    216
    Figure US20120226067A1-20120906-C00684
    A
    217
    Figure US20120226067A1-20120906-C00685
    B
    218
    Figure US20120226067A1-20120906-C00686
    A
    219
    Figure US20120226067A1-20120906-C00687
    B
    220
    Figure US20120226067A1-20120906-C00688
    A
    221
    Figure US20120226067A1-20120906-C00689
    B
    222
    Figure US20120226067A1-20120906-C00690
    A
    223
    Figure US20120226067A1-20120906-C00691
    C
    224
    Figure US20120226067A1-20120906-C00692
    B
    225
    Figure US20120226067A1-20120906-C00693
    B
    226
    Figure US20120226067A1-20120906-C00694
    C
    227
    Figure US20120226067A1-20120906-C00695
    B
    Notes
    Radioligand competitive binding assays performed using Method B1
    Values reported as ranges: A = 0.001-0.100 μM; B = 0.100-1.0 μM; C = 1.0-10.0 μM
    X is NH except for:
    Compound 223 and 225, X is:
    Figure US20120226067A1-20120906-C00696
    Compound 224, X is NMe
    Compound 226, X is:
    Figure US20120226067A1-20120906-C00697
    Compound 227, X is
    Figure US20120226067A1-20120906-C00698
    Z1, Z2 and Z3 are NH except for compounds 30, 173 and 174 and where Z1 is O and compound 111 where Z2 is O.
    R2, R4 and R5 are hydrogen except for compound 85 where it is:
    Figure US20120226067A1-20120906-C00699
    m, n1 and p are zero.

Claims (15)

1-39. (canceled)
40. A compound of formula (IV):
Figure US20120226067A1-20120906-C00700
wherein R1 is C1-C4 alkyl; PG5 is hydrogen or an amine protecting group; and PG6 is hydrogen or a hydroxy protecting group.
41. The compound of claim 40, wherein R1 is methyl.
42. The compound of claim 40, wherein PG5 is hydrogen.
43. The compound of claim 40, wherein PG5 is a carbamate protecting group.
44. The compound of claim 40, wherein PG5 is selected from the group consisting of tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc), α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl (Ddz) and allyloxycarbonyl (Alloc).
45. The compound of claim 40, wherein PG6 is hydrogen.
46. The compound of claim 40 represented by the following structures:
Figure US20120226067A1-20120906-C00701
wherein PG5 is hydrogen or an amine protecting group; and PG6 is hydrogen or a hydroxy protecting group.
47. The compound of claim 44, wherein PG5 is α,α-dimethyl-3,5-dimethoxybenzyloxy-carbonyl (Ddz).
48. The compound of claim 46, wherein PG5 is α,α-dimethyl-3,5-dimethoxybenzyloxy-carbonyl (Ddz).
49. The compound of claim 47 represented by the following structures:
Figure US20120226067A1-20120906-C00702
50. The compound of claim 44, wherein PG5 is tert-butoxycarbonyl (Boc).
51. The compound of claim 50 represented by the following structures:
Figure US20120226067A1-20120906-C00703
52. A process of using a compound of claim 40 to make a compound of formula (I);
Figure US20120226067A1-20120906-C00704
wherein R1 is C1-C4 alkyl.
53. A process of using a compound of claim 40 to make a compound of formula (G);
Figure US20120226067A1-20120906-C00705
wherein R1 is C1-C4 alkyl and PG1 is an amine protecting group.
US13/412,148 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds Abandoned US20120226067A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/412,148 US20120226067A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US47922303P 2003-06-18 2003-06-18
PCT/CA2004/000913 WO2004111077A1 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor
US10/872,142 US7521420B2 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor
CAPCT/CA04/00913 2004-06-18
US12/273,648 US8129561B2 (en) 2003-06-18 2008-11-19 Processes for intermediates for macrocyclic compounds
US12/273,638 US9181298B2 (en) 2003-06-18 2008-11-19 Intermediates for macrocyclic compounds
US13/412,148 US20120226067A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/273,648 Division US8129561B2 (en) 2003-06-18 2008-11-19 Processes for intermediates for macrocyclic compounds

Publications (1)

Publication Number Publication Date
US20120226067A1 true US20120226067A1 (en) 2012-09-06

Family

ID=33551871

Family Applications (9)

Application Number Title Priority Date Filing Date
US10/872,142 Active 2024-07-28 US7521420B2 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor
US12/273,648 Expired - Fee Related US8129561B2 (en) 2003-06-18 2008-11-19 Processes for intermediates for macrocyclic compounds
US12/273,638 Active 2027-11-17 US9181298B2 (en) 2003-06-18 2008-11-19 Intermediates for macrocyclic compounds
US13/036,484 Active US8497242B2 (en) 2003-06-18 2011-02-28 Processes for intermediates for macrocyclic compounds
US13/412,148 Abandoned US20120226067A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US13/411,979 Abandoned US20120165566A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US13/412,009 Abandoned US20120226072A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US13/411,959 Abandoned US20120226066A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US14/873,056 Active US10040751B2 (en) 2003-06-18 2015-10-01 Intermediates for macrocyclic compounds

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US10/872,142 Active 2024-07-28 US7521420B2 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor
US12/273,648 Expired - Fee Related US8129561B2 (en) 2003-06-18 2008-11-19 Processes for intermediates for macrocyclic compounds
US12/273,638 Active 2027-11-17 US9181298B2 (en) 2003-06-18 2008-11-19 Intermediates for macrocyclic compounds
US13/036,484 Active US8497242B2 (en) 2003-06-18 2011-02-28 Processes for intermediates for macrocyclic compounds

Family Applications After (4)

Application Number Title Priority Date Filing Date
US13/411,979 Abandoned US20120165566A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US13/412,009 Abandoned US20120226072A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US13/411,959 Abandoned US20120226066A1 (en) 2003-06-18 2012-03-05 Processes for intermediates for macrocyclic compounds
US14/873,056 Active US10040751B2 (en) 2003-06-18 2015-10-01 Intermediates for macrocyclic compounds

Country Status (9)

Country Link
US (9) US7521420B2 (en)
EP (2) EP1633774B1 (en)
JP (1) JP4928261B2 (en)
AT (1) ATE457995T1 (en)
CA (1) CA2528375C (en)
DE (1) DE602004025569D1 (en)
DK (1) DK1633774T3 (en)
ES (1) ES2338789T3 (en)
WO (1) WO2004111077A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10227380B2 (en) 2012-02-15 2019-03-12 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2284459C (en) * 1999-10-04 2012-12-11 Neokimia Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
ATE457995T1 (en) * 2003-06-18 2010-03-15 Tranzyme Pharma Inc MACROCYCLIC MOTILIN RECEPTOR ANTAGONISTS
USRE42624E1 (en) 2003-06-18 2011-08-16 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
US8921521B2 (en) 2003-06-18 2014-12-30 Ocera Therapeutics, Inc. Macrocyclic modulators of the Ghrelin receptor
US20090198050A1 (en) * 2003-06-18 2009-08-06 Tranzyme Pharma Inc. Macrocyclic Modulators of the Ghrelin Receptor
US7476653B2 (en) 2003-06-18 2009-01-13 Tranzyme Pharma, Inc. Macrocyclic modulators of the ghrelin receptor
US7491695B2 (en) * 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
USRE42013E1 (en) 2003-06-18 2010-12-28 Tranzyme Pharma Inc. Macrocyclic modulators of the ghrelin receptor
EP1648923B1 (en) * 2003-07-31 2008-12-31 Tranzyme Pharma Inc. Spatially-defined macrocycles incorporating peptide bond surrogates
PT1648922E (en) * 2003-07-31 2011-01-04 Tranzyme Pharma Inc Spatially-defined macrocyclic compounds useful for drug discovery
JP5739766B2 (en) * 2004-06-18 2015-06-24 オセラ セラピューティクス, インコーポレイテッド Use of macrocyclic modulators of ghrelin receptor
WO2006046977A1 (en) * 2004-10-26 2006-05-04 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and methods of using the same
CA2583345A1 (en) * 2005-06-13 2006-12-28 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US20090275648A1 (en) * 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US8088733B2 (en) 2006-07-06 2012-01-03 Tranzyme Pharma Inc. Methods of using macrocyclic agonists of the ghrelin receptor for treatment of gastrointestinal motility disorders
EP2431380A3 (en) 2006-09-11 2013-07-03 Tranzyme Pharma, Inc. Macrocyclic antagonist of the motilin receptor for treatment of gastrointestinal dysmotility disorders
EP2118123B1 (en) 2007-01-31 2015-10-14 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
AU2015201862B2 (en) * 2007-02-09 2016-10-13 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
AU2008241532A1 (en) * 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
US20080287371A1 (en) * 2007-05-17 2008-11-20 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor for modulation of the migrating motor complex
WO2010022249A2 (en) * 2008-08-20 2010-02-25 Ensemble Discovery Corporation Macrocyclic compounds for inhibition of tumor necrosis factor alpha
EP2462147B8 (en) * 2009-08-05 2015-03-18 Polyphor AG Conformationally constrained, fully synthetic macrocyclic compounds
US20120270769A1 (en) * 2009-10-23 2012-10-25 Marsault Eric Methods of using macrocyclic inhibitors of serine protease enzymes
CN102812037A (en) 2009-10-30 2012-12-05 特兰齐姆制药公司 Macrocyclic Ghrelin Receptor Antagonists And Inverse Agonists And Methods Of Using The Same
US9119832B2 (en) 2014-02-05 2015-09-01 The Regents Of The University Of California Methods of treating mild brain injury
ES2811911T3 (en) * 2015-10-27 2021-03-15 Hoffmann La Roche Peptide macrocycles against Acinetobacter baumannii
WO2017075535A1 (en) 2015-10-28 2017-05-04 Oxeia Biopharmaceuticals, Inc. Methods of treating neurodegenerative conditions
EP3388444A1 (en) 2017-04-10 2018-10-17 F. Hoffmann-La Roche AG Anti-bacterial peptide macrocycles and use thereof
US11505573B2 (en) 2018-03-28 2022-11-22 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
US11819532B2 (en) 2018-04-23 2023-11-21 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
CN108752214A (en) * 2018-07-09 2018-11-06 上海华堇生物技术有限责任公司 A kind of new preparation process of 2- hydroxy-betas-nitrostyrolene

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090240027A1 (en) * 2003-06-18 2009-09-24 Tranzyme Pharma Inc. Intermediates for macrocyclic compounds

Family Cites Families (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5175150A (en) 1985-08-31 1992-12-29 Kitasato, Kenkyusho Erythromycin derivative
US5008249A (en) 1985-08-31 1991-04-16 Kitasato Kenkyusho Therapeutic method of stimulating digestive tract contractile motion in mammals
US5695952A (en) 1986-09-12 1997-12-09 Kyowa Hakko Kogyo Co., Ltd. Method for producing Leu13 !motilin
GB8728820D0 (en) 1987-12-09 1988-01-27 Fisons Plc Compounds
US4920102A (en) 1988-04-18 1990-04-24 Eli Lilly And Company Method for treating gastrointestinal disorders
EP0358608A3 (en) 1988-09-09 1990-10-31 Gesellschaft für Biotechnologische Forschung mbH (GBF) Macrocyclic lacton derivatives with a microbicidal activity
EP0378078A1 (en) 1989-01-06 1990-07-18 Sanwa Kagaku Kenkyusho Co., Ltd. Motilin-like polypeptide and use thereof
US5459049A (en) 1989-01-06 1995-10-17 Sanwa Kagaku Kenkyushko Co., Ltd. Motilin-like polypeptide and use thereof
EP0413532A3 (en) 1989-08-18 1991-05-15 Fisons Plc Macrocyclic compounds
US5143915A (en) 1990-07-25 1992-09-01 E. R. Squibb & Sons, Inc. Dihydropyrimidine macrocyclic lactones useful as calcium antagonists and agonists
US5196452A (en) 1991-01-29 1993-03-23 Genelabs Incorporated Macrocyclic anti-viral compound and method
US5523418A (en) 1991-04-09 1996-06-04 Abbott Laboratories Macrocyclic lactam prokinetic agents
AU663089B2 (en) 1991-04-09 1995-09-28 Abbott Laboratories Macrocyclic lactam prokinetic agents
US5563172A (en) 1991-09-05 1996-10-08 Abbott Laboratories Macrocyclic amide and urea immunomodulators
US5561137A (en) 1991-09-05 1996-10-01 Abbott Laboratories Thio-heterocyclic macrolactam immunomodulators
US5708002A (en) 1991-09-05 1998-01-13 Abbott Laboratories Macrocyclic immunomodulators
US5604234A (en) 1991-09-05 1997-02-18 Abbott Laboratories Substituted thiol macrolactam immunomodulators
US5457111A (en) 1991-09-05 1995-10-10 Abbott Laboratories Macrocyclic immunomodulators
US5534632A (en) 1991-09-05 1996-07-09 Abbott Laboratories Macrocyclic carbamate immunomodulators
US5561228A (en) 1991-09-05 1996-10-01 Abbott Laboratories Macrocyclic immunomodulators
US5604294A (en) 1991-09-05 1997-02-18 Luly; Jay R. Macrocyclic immunomodulators
US5244889A (en) 1991-11-26 1993-09-14 Ciba-Geigy Corporation Certain macrocyclic lactam derivatives
DE4200145A1 (en) 1992-01-07 1993-07-08 Kali Chemie Pharma Gmbh 7,10-EPOXY-OXACYCLODODANE DERIVATIVES, METHODS AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS
ATE138925T1 (en) 1992-03-19 1996-06-15 Takeda Chemical Industries Ltd ERYTHROMYCIN DERIVATIVES, PRODUCTION AND USE THEREOF
MY113693A (en) 1992-05-26 2002-05-31 Chugai Pharmaceutical Co Ltd Erythromycin derivatives having an enterokinesis stimulating action
US5328902A (en) 1992-08-13 1994-07-12 American Cyanamid Co. 7-(substituted)-9-[(substituted glycyl)amido]-6-demethyl-6-deoxytetracyclines
US5292741A (en) 1992-08-18 1994-03-08 Merck & Co., Inc. Macrocycles incorporating quinazolinones
US5252579A (en) 1993-02-16 1993-10-12 American Home Products Corporation Macrocyclic immunomodulators
WO1994021634A1 (en) 1993-03-17 1994-09-29 Abbott Laboratories Substituted alicyclic-aliphatic amine-containing macrocyclic immunomodulators
CA2156064A1 (en) 1993-03-17 1994-09-29 Megumi Kawai Substituted alicyclic amine-containing macrocyclic immunomodulators
US5457194A (en) 1993-03-17 1995-10-10 Abbott Laboratories Substituted aliphatic amine-containing macrocyclic immunomodulators
US5470830A (en) 1993-08-06 1995-11-28 Ohmeda Pharmaceutical Products Division Inc. Motilin-like polypeptides that inhibit gastrointestinal motor activity
US5422341A (en) 1993-08-06 1995-06-06 Ohmeda Pharmaceutical Products Division Inc. Motilin-like polypeptides with gastrointestinal motor stimulating activity
US5612350A (en) 1993-11-30 1997-03-18 Abbott Laboratories Macrocyclic immunomodulators with novel cyclohexyl ring replacements
US5624949A (en) 1993-12-07 1997-04-29 Eli Lilly And Company Protein kinase C inhibitors
FI108138B (en) 1993-12-07 2001-11-30 Lilly Co Eli Process for the preparation of therapeutically useful 3,4 [(N, N'-1,1 ') - substituted alkylene) bis (3,3'-indolyl)] 1H-pyrrole-2,5-dione derivatives
ATE198602T1 (en) 1994-02-04 2001-01-15 Merrell Pharma Inc MACROCYCLIC DIFLUOROSTATONE DERIVATIVES AS ANTIVIRAL AGENTS
US6100377A (en) 1994-06-10 2000-08-08 The Trustees Of The University Of Pennsylvania Constrained peptides
AUPM982594A0 (en) 1994-12-02 1995-01-05 University Of Queensland, The HIV protease inhibitors
GB9511357D0 (en) 1995-06-06 1995-08-02 Johnson Matthey Plc Improved antiviral compounds
US5811515A (en) 1995-06-12 1998-09-22 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides, and peptidomimetics by catalytic ring closing metathesis
US6124453A (en) 1995-07-04 2000-09-26 Novartis Ag Macrolides
US6281352B1 (en) 1995-11-14 2001-08-28 Dupont Pharmaceuticals Company Macrocyclic compounds as metalloprotease inhibitors
ZA969646B (en) 1995-11-20 1998-05-18 Lilly Co Eli Protein kinase C inhibitor.
US5888971A (en) 1996-02-20 1999-03-30 Ortho Pharmaceutical Corporation, Inc. Macrocyclic peptides useful in the treatment of thrombin related disorders
US5849691A (en) 1996-02-20 1998-12-15 The United States Of America As Represented By The Department Of Health And Human Services Peptidomimetic inhibitors of cathepsin D and plasmepsins I and II
IL117426A (en) * 1996-03-10 2005-09-25 Yissum Res Dev Co Synthetic pseudopeptides having osteogenic activity and pharmaceutical compositions containing the same
US5734012A (en) 1996-05-16 1998-03-31 Ohmeda Pharmaceutical Products Division Inc. Cyclic motilin-like polypeptides with gastrointestinal motor stimulating activity
CA2207137A1 (en) * 1996-06-14 1997-12-14 James Erwin Fritz Scavenger assisted combinatorial process for preparing libraries of secondary amine compounds
US5712253A (en) 1996-06-18 1998-01-27 Abbott Laboratories Macrocyclic 13-membered ring derivatives of erythromycins A and B
US20020168761A1 (en) 2000-01-24 2002-11-14 Gour Barbara J. Peptidomimetic modulators of cell adhesion
DE19644195A1 (en) 1996-10-24 1998-04-30 Solvay Pharm Gmbh 10,13,15-Trioxatricyclo [9.2.1.1. · 9 ·. · 6 ·] -pentadecanone derivatives, processes for their preparation and pharmaceuticals containing these compounds
WO1998023629A1 (en) 1996-11-26 1998-06-04 Chugai Seiyaku Kabushiki Kaisha 13-membered ring macrolide compounds, medicine containing the same, and process for producing the same
US6867305B2 (en) * 1996-12-03 2005-03-15 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
MX9709494A (en) * 1996-12-04 1998-07-31 Pfizer Cyclic peptidic compounds, process for its production, compositions containing them and use of the same.
US5952320A (en) 1997-01-07 1999-09-14 Abbott Laboratories Macrocyclic inhibitors of matrix metalloproteinases and TNFα secretion
US6291653B1 (en) 1997-03-24 2001-09-18 Zymogenetics, Inc. Antibodies to motilin homologs
CN1170933C (en) 1997-03-24 2004-10-13 津莫吉尼蒂克斯公司 Motilin homologs
US6380158B1 (en) 1997-03-24 2002-04-30 Zymogenetics, Inc. Motilin homologs
AU6895598A (en) 1997-04-11 1998-11-11 Eli Lilly And Company Combinatorial libraries of peptidomimetic macrocycles and processes therefor
JP3583928B2 (en) * 1997-08-15 2004-11-04 中外製薬株式会社 Phenethylamine derivative
TW460478B (en) * 1997-08-15 2001-10-21 Chugai Pharmaceutical Co Ltd Phenethylamine derivatives
US5972939A (en) 1997-10-28 1999-10-26 Ortho-Mcneil Pharmaceutical, Inc. Cyclopentene derivatives useful as antagonists of the motilin receptor
US6080838A (en) 1997-11-25 2000-06-27 University Of Virginia Patent Foundation Peptidomimetic of helix-turn-helix or gamma-turn
JP4200393B2 (en) 1997-11-25 2008-12-24 ソニー株式会社 Information processing apparatus and information processing method
US6165985A (en) 1998-02-13 2000-12-26 Solvay Pharmaceuticals Gmbh 11-acetyl-12,13-dioxabicyclo[8.2.1]-tridecenone derivatives, processes for their preparation and pharmaceutical compositions comprising them
US6228986B1 (en) 1998-04-13 2001-05-08 Ortho-Mcneil Pharmaceutical, Inc. Solid-phase synthesis of novel 14-membered macroycles for high throughput screening
SE9802080D0 (en) 1998-06-11 1998-06-11 Hellstroem Pharmaceutical composition for the treatment of functional dyspepsia and / or irritable bowel syndrome and new use of substances therein
JP2002517488A (en) 1998-06-11 2002-06-18 デュポン ファーマシューティカルズ カンパニー Method for producing large ring metalloprotease inhibitors
ATE354582T1 (en) 1998-06-12 2007-03-15 Merck & Co Inc CLONING AND IDENTIFICATION OF THE MOTILIN RECEPTOR
JP2002519427A (en) 1998-07-02 2002-07-02 メルク エンド カムパニー インコーポレーテッド Prenyl protein transferase inhibitors
US6562823B1 (en) 1998-07-02 2003-05-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6358985B1 (en) 1998-07-02 2002-03-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
TW509699B (en) 1998-09-24 2002-11-11 Chugau Pharmaceutical Co Ltd Ethylamine derivatives
AUPP616498A0 (en) * 1998-09-25 1998-10-15 University Of Queensland, The Synthesis of cyclic peptides
CA2347128A1 (en) 1998-10-28 2000-05-04 Kosan Biosciences, Inc. Library of novel "unnatural" natural products
US6329376B1 (en) 1998-10-29 2001-12-11 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2000044770A1 (en) * 1999-01-28 2000-08-03 Chugai Seiyaku Kabushiki Kaisha Substituted phenethylamine derivatives
US20020058286A1 (en) 1999-02-24 2002-05-16 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto and analogues thereof
JP4269041B2 (en) * 1999-03-02 2009-05-27 国立大学法人九州工業大学 Novel cyclic tetrapeptide derivatives and their pharmaceutical uses
US6121257A (en) 1999-03-31 2000-09-19 Abbott Laboratories Sulfamate containing macrocyclic immunomodulators
US6608027B1 (en) * 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus
JP2003503055A (en) 1999-06-30 2003-01-28 ザイモジェネティクス,インコーポレイティド SGIP peptide
US6420521B1 (en) 1999-06-30 2002-07-16 Zymogenetics, Inc. Short gastrointestinal peptides
DE19933701A1 (en) 1999-07-19 2001-01-25 Wilex Biotechnology Gmbh Cyclic peptidomimetic urokinase receptor antagonists
US6660832B1 (en) 1999-08-20 2003-12-09 Isis Pharmaceuticals, Inc. Macrocyclic compounds and preparation methods thereof
DE19942624A1 (en) 1999-08-28 2001-03-08 Chemotopix Gmbh Process for the preparation of cyclic peptidomimetics
WO2001018006A1 (en) 1999-09-09 2001-03-15 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
CA2284459C (en) * 1999-10-04 2012-12-11 Neokimia Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
GB9929318D0 (en) 1999-12-10 2000-02-02 Prometic Biosciences Limited Macrocyclic compounds and their use
US6632818B2 (en) 2000-01-12 2003-10-14 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
AU2001227756A1 (en) 2000-01-12 2001-07-24 Merck And Co., Inc. Inhibitors of prenyl-protein transferase
AU2001227755A1 (en) 2000-01-12 2001-07-24 Merck And Co., Inc. Inhibitors of prenyl-protein transferase
US6525074B2 (en) 2000-01-12 2003-02-25 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
AU2001227757A1 (en) 2000-01-12 2001-07-24 Merck And Co., Inc. Inhibitors of prenyl-protein transferase
ATE340571T1 (en) 2000-01-18 2006-10-15 Univ Mcgill PHARMACEUTICAL PREPARATIONS CONTAINING BETA-TURN PEPTIDOMIMMETIC CYCLIC SUBSTANCES
WO2001068622A1 (en) 2000-03-13 2001-09-20 Ortho-Mcneil Pharmaceutical, Inc. Novel cyclobutene derivatives useful as antagonists of the motilin receptor
US6423714B2 (en) 2000-03-13 2002-07-23 Ortho Mcneil-Pharmaceutical, Inc.. Cyclohexene derivatives useful as antagonists of the motilin receptor
AU2001249144A1 (en) 2000-03-13 2001-09-24 Ortho-Mcneil Pharmaceutical, Inc. Novel cyclopentene derivatives useful as antagonists of the motilin receptor
RU2002129564A (en) 2000-04-05 2004-05-10 Шеринг Корпорейшн (US) MACROCYCLIC INHIBITORS OF SERINE NS3-PROTEASE, INCLUDING N-CYCLIC P2 STRUCTURAL ELEMENTS OF HEPATITIS C VIRUS
US6380228B1 (en) 2000-04-10 2002-04-30 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6610722B2 (en) 2000-04-10 2003-08-26 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2001077116A1 (en) 2000-04-10 2001-10-18 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
CA2406532A1 (en) 2000-04-19 2001-11-01 Schering Corporation Macrocyclic ns-3 serine protease inhibitors of hepatitis c virus compri sing alkyl and aryl alanine p2 moieties
US6511980B2 (en) 2000-05-05 2003-01-28 Ortho Mcneil Pharmaceutical, Inc. Substituted diamine derivatives useful as motilin antagonists
ES2333097T3 (en) 2000-05-31 2010-02-17 Raqualia Pharma Inc USE OF SECRETAGOGS OF GROWTH HORMONE TO STIMULATE GASTROINTESTINAL MOTILITY.
US6721353B1 (en) 2000-06-21 2004-04-13 Cisco Technology, Inc. Network compatibility
US7018981B2 (en) 2000-08-24 2006-03-28 Chugai Seiyaku Kabushiki Kaisha Cyclic motilin receptor antagonists
ATE328888T1 (en) * 2000-08-25 2006-06-15 Sloan Kettering Inst Cancer RADICICOL AND MONOCILLIN AND THEIR ANALOGUES AND THEIR APPLICATIONS
ATE312088T1 (en) 2000-09-05 2005-12-15 Centelion ACID SENSITIVE COMPOUNDS, THEIR PRODUCTION AND USES
WO2002040463A1 (en) 2000-11-20 2002-05-23 Toyama-Ken Macrocyclic compound
US7279304B2 (en) 2000-12-18 2007-10-09 President And Fellows Of Harvard College Methods for preparation of macrocyclic molecules and macrocyclic molecules prepared thereby
AU2002218902A1 (en) 2000-12-27 2002-07-08 Shire Biochem Inc. Macrocyclic anti-viral compounds
WO2002059141A1 (en) 2001-01-25 2002-08-01 Chugai Seiyaku Kabushiki Kaisha Peptide derivatives
KR20020066476A (en) 2001-02-12 2002-08-19 쥬가이 세이야쿠 가부시키가이샤 Preparation of peptide derivatives
BR0207502A (en) 2001-02-23 2004-03-09 Polyphor Ltd Compounds, enantiomers, pharmaceutical composition, use of compounds, and process for manufacturing compounds
DE50204789D1 (en) 2001-05-10 2005-12-08 Solvay Pharm Gmbh NEW 1-AMIDOMETHYL CARBONYL-PIPERIDINE DERIVATIVES, PROCESS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS
WO2002100399A1 (en) 2001-06-12 2002-12-19 Elan Pharmaceuticals, Inc. Macrocycles useful in the treatment of alzheimer's disease
BR0210392A (en) 2001-06-12 2004-10-13 Elan Pharm Inc Compound, methods of treating a patient who has or prevent a patient from contracting a disease or condition and preparing a compound, and, using a compound
DE10146104A1 (en) 2001-09-19 2003-04-03 Bayer Ag Antibacterial marrow cycles
US20030158377A1 (en) * 2001-11-06 2003-08-21 President And Fellows Of Harvard College Methods for preparation of macrocyclic molecules macrocyclic molecules prepared thereby and substrates and solid supports for use therein
CN101157924A (en) 2001-12-11 2008-04-09 人体基因组科学有限公司 Neutrophil leukocyte gene alpha
US6867185B2 (en) 2001-12-20 2005-03-15 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
CA2369711A1 (en) * 2002-01-30 2003-07-30 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
US7119072B2 (en) 2002-01-30 2006-10-10 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis C virus
US6972281B2 (en) * 2002-02-07 2005-12-06 University Of Ottawa Histogranin-like peptides and non-peptides, processes for their preparation and uses thereof
WO2003068168A2 (en) 2002-02-14 2003-08-21 The Brigham And Women's Hospital, Inc. Helical peptidomimetics and use b-amyloid-associated diseases
US7799827B2 (en) 2002-03-08 2010-09-21 Eisai Co., Ltd. Macrocyclic compounds useful as pharmaceuticals
US20030221432A1 (en) 2002-06-03 2003-12-04 Tucker Ronald M. Solid fuel combustion method and apparatus for the conversion of waste into useful energy
DE10226921A1 (en) 2002-06-17 2003-12-24 Bayer Ag Antibacterial amide macrocycles
SI1515921T1 (en) 2002-06-24 2006-08-31 Rockwool Int Process and apparatus for controlling mineral wool production using a cascade rotor
DE10232094A1 (en) 2002-07-15 2004-02-05 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) 5-thiaepothilones and 15-disubstituted epothilones
DE10234422A1 (en) 2002-07-29 2004-02-12 Bayer Ag Antibacterial ester macrocycles
CA2496215C (en) 2002-08-20 2013-07-16 Polyphor Ltd. Template-fixed peptidomimetics with antibacterial activity
DE10239042A1 (en) 2002-08-21 2004-03-04 Schering Ag New fused macrocyclic pyrimidine derivatives, useful as e.g. cyclin-dependent kinase inhibitors for treating e.g. cancer, autoimmune, cardiovascular or neurodegenerative diseases or viral infections
US6747009B2 (en) * 2002-09-19 2004-06-08 Universiteit Leiden Peptidomimetic glutathione analogs
US6653334B1 (en) * 2002-12-27 2003-11-25 Kowa Co., Ltd. Benzoxazole compound and pharmaceutical composition containing the same
US20090198050A1 (en) * 2003-06-18 2009-08-06 Tranzyme Pharma Inc. Macrocyclic Modulators of the Ghrelin Receptor
USRE42013E1 (en) 2003-06-18 2010-12-28 Tranzyme Pharma Inc. Macrocyclic modulators of the ghrelin receptor
US7476653B2 (en) * 2003-06-18 2009-01-13 Tranzyme Pharma, Inc. Macrocyclic modulators of the ghrelin receptor
US7491695B2 (en) * 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
EP1648923B1 (en) * 2003-07-31 2008-12-31 Tranzyme Pharma Inc. Spatially-defined macrocycles incorporating peptide bond surrogates
PT1648922E (en) * 2003-07-31 2011-01-04 Tranzyme Pharma Inc Spatially-defined macrocyclic compounds useful for drug discovery
EA009374B1 (en) * 2003-09-03 2007-12-28 Кова Ко., Лтд. Ppar-activating compound and pharmaceutical composition containing same
US20090275648A1 (en) * 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
EP2431380A3 (en) * 2006-09-11 2013-07-03 Tranzyme Pharma, Inc. Macrocyclic antagonist of the motilin receptor for treatment of gastrointestinal dysmotility disorders
AU2008241532A1 (en) * 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
US20080287371A1 (en) * 2007-05-17 2008-11-20 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor for modulation of the migrating motor complex
US8018037B2 (en) 2009-04-16 2011-09-13 Mediatek Inc. Semiconductor chip package
KR20120081166A (en) 2009-09-30 2012-07-18 트랜자임 파르마 인크 Salts, solvates, and pharmaceutical compositions of macrocyclic ghrelin receptor agonists and methods of using the same
CN102812037A (en) * 2009-10-30 2012-12-05 特兰齐姆制药公司 Macrocyclic Ghrelin Receptor Antagonists And Inverse Agonists And Methods Of Using The Same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090240027A1 (en) * 2003-06-18 2009-09-24 Tranzyme Pharma Inc. Intermediates for macrocyclic compounds

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10703780B2 (en) 2010-08-13 2020-07-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US11008366B2 (en) 2010-08-13 2021-05-18 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9522947B2 (en) 2011-10-18 2016-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10308699B2 (en) 2011-10-18 2019-06-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10227380B2 (en) 2012-02-15 2019-03-12 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10669230B2 (en) 2012-11-01 2020-06-02 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Also Published As

Publication number Publication date
CA2528375C (en) 2013-11-19
ATE457995T1 (en) 2010-03-15
EP2210612A3 (en) 2010-11-24
US10040751B2 (en) 2018-08-07
JP2007523853A (en) 2007-08-23
CA2528375A1 (en) 2004-12-23
US20090137835A1 (en) 2009-05-28
US20110245459A1 (en) 2011-10-06
US20160221927A1 (en) 2016-08-04
US20120226072A1 (en) 2012-09-06
EP2210612B1 (en) 2016-10-05
US7521420B2 (en) 2009-04-21
ES2338789T3 (en) 2010-05-12
US8129561B2 (en) 2012-03-06
US20120165566A1 (en) 2012-06-28
DK1633774T3 (en) 2010-05-25
DE602004025569D1 (en) 2010-04-01
US8497242B2 (en) 2013-07-30
JP4928261B2 (en) 2012-05-09
US20050054562A1 (en) 2005-03-10
EP2210612A2 (en) 2010-07-28
US20090240027A1 (en) 2009-09-24
WO2004111077A1 (en) 2004-12-23
EP1633774B1 (en) 2010-02-17
EP1633774A1 (en) 2006-03-15
US9181298B2 (en) 2015-11-10
US20120226066A1 (en) 2012-09-06

Similar Documents

Publication Publication Date Title
US10040751B2 (en) Intermediates for macrocyclic compounds
US9133235B2 (en) Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
US8334256B2 (en) Pharmaceutical salts of macrocyclic modulators of the ghrelin receptor
EP2644618B1 (en) tether intermediates for the synthesis of macrocyclic ghrelin receptor modulators
US7491695B2 (en) Methods of using macrocyclic modulators of the ghrelin receptor
US8022252B2 (en) Spatially-defined macrocyclic compounds useful for drug discovery
USRE42013E1 (en) Macrocyclic modulators of the ghrelin receptor
USRE42624E1 (en) Methods of using macrocyclic modulators of the ghrelin receptor
US20090198050A1 (en) Macrocyclic Modulators of the Ghrelin Receptor
US20180110824A1 (en) Macrocyclic Modulators of the Ghrelin Receptor
US8921521B2 (en) Macrocyclic modulators of the Ghrelin receptor

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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