WO2006009674A1 - Macrocyclic modulators of the ghrelin receptor - Google Patents

Macrocyclic modulators of the ghrelin receptor Download PDF

Info

Publication number
WO2006009674A1
WO2006009674A1 PCT/US2005/020857 US2005020857W WO2006009674A1 WO 2006009674 A1 WO2006009674 A1 WO 2006009674A1 US 2005020857 W US2005020857 W US 2005020857W WO 2006009674 A1 WO2006009674 A1 WO 2006009674A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
ring
alkyl
hydrogen
optionally
Prior art date
Application number
PCT/US2005/020857
Other languages
English (en)
French (fr)
Other versions
WO2006009674A8 (en
Inventor
Hamid Hoveyda
Mark Peterson
Graeme Fraser
Mahesh Ramaseshan
Original Assignee
Tranzyme Pharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/872,142 external-priority patent/US7521420B2/en
Application filed by Tranzyme Pharma, Inc. filed Critical Tranzyme Pharma, Inc.
Priority to JP2007516620A priority Critical patent/JP5363726B2/ja
Priority to ES05785185T priority patent/ES2393498T3/es
Priority to CN2005800280726A priority patent/CN101111512B/zh
Priority to CA2579726A priority patent/CA2579726C/en
Priority to EP05785185A priority patent/EP1773869B9/en
Publication of WO2006009674A1 publication Critical patent/WO2006009674A1/en
Publication of WO2006009674A8 publication Critical patent/WO2006009674A8/en

Links

Classifications

    • 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
    • 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
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • 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
    • 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
    • C07D498/08Bridged 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
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Patent Application Serial No. 10/872,142 filed June 18, 2004, currently pending, which claims the benefit under 35 U.S. C. ⁇ 119(e) of United States Provisional Patent Application Serial No. 60/479,223, filed June 18, 2003.
  • This continuation-in-part application also claims the benefit under 35 U.S. C. ⁇ 119(e) of United States Provisional
  • the present invention relates to novel conformationally-defined macrocyclic compounds that bind to and/or are functional modulators of the ghrelin (growth hormone secretagogue) receptor including GHS-RIa and subtypes, isoforms and/or variants thereof.
  • the present invention also relates to intermediates of these compounds, pharmaceutical compositions containing these compounds and methods of using the compounds.
  • These novel macrocyclic compounds are useful as therapeutics for a range of disease indications, hi particular, these compounds are useful for treatment and prevention of gastrointestinal disorders including, but not limited to, post-operative ileus, gastroparesis, including diabetic gastroparesis, opioid bowel dysfunction, chronic intestinal pseudo-obstruction, short bowel syndrome and functional gastrointestinal disorders.
  • ghrelin is a recently characterized 28-amino acid peptide hormone isolated originally from the stomach of rats with the orthologue subsequently identified in humans. (Kojima, M.; Hosoda, H. et al. Nature 1999, 402, 656- 660.) The existence of this peptide in a range of other species suggests a conserved and important role in normal body function.
  • This peptide has been demonstrated to be the endogenous ligand for a previously orphan G protein-coupled receptor (GPCR), type 1 growth hormone secretatogue receptor (hGHS-Rla) (Howard, A.D.; Feighner, S.D.; et al. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science 1996, 273, 974-977.) found predominantly in the brain (arcuate nucleus and ventromedial nucleus in the hypothalamus, hippocampus and substantia nigra) and pituitary. (U.S. Pat. No. 6,242,199; Intl. Pat. Appl. Nos.
  • the receptor has also been detected in other areas of the central nervous system (CNS) and in peripheral tissues, for instance adrenal and thyroid glands, heart, lung, kidney, and skeletal muscles. This receptor was identified and cloned prior to the isolation and characterization of the endogenous peptide ligand and is distinct from other receptors involved in the regulation of growth hormone (GH) secretion, in particular, the growth hormone-releasing hormone (GHRH) receptor.
  • GH growth hormone
  • GHRH growth hormone-releasing hormone
  • a unique characteristic of both the rat and human peptides is the presence of the ra-octanoyl (Oct) moiety on Ser 3 .
  • the des-acyl form predominates in circulation, with approximately 90% of the hormone in this form.
  • This group is derived from a post- translational modification and appears relevant for bioactivity and possibly also for transport into the CNS.
  • Banks, W. A.; Tsch ⁇ p, M.; Robinson, S. M.; Heiman, M.L. Extent and direction of ghrelin transport across the blood-brain barrier is determined by its unique primary structure. J. Pharmacol. Exp. Ther.
  • the des-octanoyl form of the hormone was at least 100-fold less potent than the parent peptide, although it has been suggested that the des-acyl species may be responsible for some of the other biological effects associated with ghrelin.
  • This des-acyl form has also been postulated to be primarily responsible for the cardiovascular and cell proliferation effects attributed to ghrelin, while the acylated form participates in maintenance of energy balance and growth hormone release.
  • Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERKl/2 and PI-3 kinase/AKT. J. Cell Biol. 2002, 159, 1029-1037)
  • Jei r -Gln 14 -ghrelin and its octanoylated derivative have been isolated as endogenous forms of the hormone arising from alternative splicing of the ghrelin gene, but both are found to be inactive in stimulating GH release in vivo.
  • GHS were projected to have utility in the treatment of a variety of other disorders, including wasting conditions (cachexia) as seen in HIV patients and cancer-induced anorexia, musculoskeletal frailty in the elderly, and growth hormone deficient diseases.
  • the hormone has a variety of other endocrine and non-endocrine functions (Broglio, F.; Gottero, C; Arvat, E.; Ghigo, E. Endocrine and non-endocrine actions of ghrelin. Horm. Res. 2003, 59, 109-117) and has been shown to interact with a number of other systems in playing a role in maintaining proper energy balance. (Horvath, T. L.; Diano, S.; Sotonyi, P.; Herman, M.; Tschop, M. Ghrelin and the regulation of energy balance - a hypothalamic perspective.
  • the peptide ghrelin plays a role as an orexigenic signal in the control of feeding, in which it acts to counteract the effects of leptin. Indeed, it was the first gut peptide proven to have such orexigenic properties. (Kojima, M.; Kangawa, K. Ghrelin, an orexigenic signaling molecule from the gastrointestinal tract. Curr. Opin.
  • the hormone also is implicated in the hypothalamic regulation of the synthesis and secretion of a number of other neuropeptides involved in appetite and feeding behavior.
  • Levels of ghrelin are elevated in response to fasting or extended food restriction.
  • Ghrelin also has been implicated in various aspects of reproduction and neonatal development. (Arvat, E.; Gianotti, L.; Giordano, R.; et al. Growth hormone-releasing hormone and growth hormone secretagogue-receptor ligands. Focus on reproductive system. Endocrine 2001, 14, 35-43) Also of significance are the cardiovascular effects of ghrelin, since the peptide is a powerful vasodilator.
  • ghrelin agonists have potential for the treatment of chronic heart failure (Nagaya, N.; Kangawa, K. Ghrelin, a novel growth hormone-relasing peptide, in the treatment of chronic heart failure.
  • Regal. Pept. 2003, 114, 1 ⁇ -11 ⁇ Nagaya, N.; Kangawa, K. Ghrelin improves left ventricular dysfunction and cardiac cachexia in heart failure.
  • ghrelin Cardiac effects of ghrelin and its endogenous derivatives des-octanoyl ghrelin and des- Gln 14 -ghrelin. Eur. J. Pharmacol. 2003, 476, 87-95) Intl. Pat. Appl. Publ. WO 2004/014412 describes the use of ghrelin agonists for the protection of cell death in myocardial cells and as a cardioprotectant treatment for conditions leading to heart failure. Lastly, evidence has been obtained that ghrelin may have implications in anxiety and other CNS disorders as well as the improvement of memory.
  • GHS-RIb GHS-RIb
  • GHS endogenous peptides and synthetic GHS.
  • high affinity binding sites for ghrelin and des-acyl ghrelin have also been found in breast cancer cell lines, cardiomyocytes, and guinea pig heart that are involved in mediating the antiproliferative, cardioprotective and negative cardiac inotropic effects of the peptides.
  • specific GHS binding sites besides GHS-RIa and GHS-RIb have been found in prostate cancer cells.
  • ghrelin and Jes-acyl ghrelin exert different effects on cell proliferation in prostate carcinoma cell lines.
  • BIM- 28163 functions as an antagonist at the GHS-RIa receptor and inhibits receptor activation by native ghrelin.
  • this same molecule is a full agonist with respect to stimulating weight gain and food intake.
  • the existence of a still uncharacterized receptor subtype has been proposed based on binding studies in various tissues that showed differences between peptidic and non-peptidic GHS. (Ong, H.; Menicoll, N.; Escher, F.; Collu, R.; Deghenghi, R.; Locatelli, V.; Ghigo, E.; Muccioli, G.; Boghen, M.; Nilsson, M.
  • the variety of activities associated with the ghrelin receptor could also be due to different agonists activating different signaling pathways as has been shown for ghrelin and adenosine, both of which interact as agonists at GHS-RIa (Carreira, M.C.; Camina, J.P.; Smith, R.G.; Casanueva, F.F. Agonist-specific coupling of growth hormone secretagogue receptor type Ia to different intracellular signaling systems. Role of adenosine. Neuroendocrinology 2004, 79, 13-25.)
  • the activity of the ghrelin receptor might also be at least partially governed by such complexes.
  • GHS-RIa with GHRH
  • GH growth hormone
  • GHRH GH-releasing hormone
  • Endocrinol. 2004, 214, 81-95 maybe involved in modulating the function of the receptor.
  • POI post-operative ileus
  • Ghrelin acts locally in the stomach to stimulate and coordinate the firing of vagal afferent neurons and thereby modulate gut motility.
  • Ghrelin agonists duplicate the effects of ghrelin, thus targeting directly the underlying cause of POI to accelerate normalization of gut function and enable more rapid discharge from the hospital.
  • Intravenous administration is often the preferred route of treatment for POI due to the impaired GI motility in these patients that impedes oral therapy. No agent is currently approved by the U.S. FDA specifically for the treatment of POI.
  • Another major motility disorder is gastroparesis, a particular problem for both type
  • Bowel Dysfunction Drugs 2003, 63, 649-671.
  • Drugs 2003, 63, 649-671. is the term applied to the confluence of symptoms involving the reduced GI motility that results from treatment with opioid analgesics.
  • OBD opioid analgesics
  • a ghrelin agonist can be expected to counteract the dysmotility resulting from opioid use.
  • Short bowel syndrome is a condition that occurs after resection of a substantial portion of small intestine and is characterized by malnutrition. Patients are observed to have decreased ghrelin levels resulting from loss of the ghrelin-producing neuroendocrine cells of the intestine. It is possible the short bowel feeds back on the release of the hormone. (Rrsek, M.; Rosicka, M.; Haluzik, M.; et al. Plasma ghrelin levels in patients with short bowel syndrome. Endocr. Res.
  • Chronic intestinal pseudo-obstruction is a syndrome defined by the presence of chronic intestinal dilation and dysmotility in the absence of mechanical obstruction or inflammation. Both genetic and acquired causes are known to result in this disorder, which affects high numbers of individuals worldwide annually.
  • ghrelin protects against ethanol-induced gastric ulcers in rats: studies on the mechanism of action. Endocrinology 2003, 144, 353-359.) and Crohn's disease.
  • GI dysmotility is a significant problem in other mammals as well.
  • the motility dysfunction termed ileus or colic is the number one cause of mortality among horses.
  • ileus is one of the most common complications of equine intestinal surgery, in other words, post-operative ileus. This condition may also have a non-surgical etiology.
  • Some horses may be predisposed to ileus based upon the anatomy and functioning of their digestive tract. Virtually any horse is susceptible to colic with only minor differences based upon age, sex and breed.
  • ileus may affect other animals, for example canines. (Roussel, AJ.Jr.; Cohen, N.D.; Hooper, R.N.; Rakestraw, P. C.
  • WO 01/00830 reports on short gastrointestinal peptides (SGIP) that secrete growth hormone and also promote GI motility, but these were not shown to be due to action at the ghrelin receptor.
  • U.S. Patent No. 6,548,501 discloses specific compounds, but as GHS, useful for stimulation of GI motility. Moreover, other endogenous factors are known to stimulate secretion of GH, but do not promote GI motility. Indeed, many actually inhibit this physiological function. Specific receptor agonists such as the compounds of the present invention have much better potential to be selective and effective therapeutic agents. Work has continued at the development of potent and selective GHS with a number of small molecule derivatives now being known as has been recently summarized. (Carpino, P. Exp. Opin.
  • simplification of this cyclic derivative led to still potent, linear compounds, whereas, for compounds of the invention, linear analogues have been found to be devoid of ghrelin receptor activity.
  • the macrocyclic compounds of the invention possess agonist activity. As previously mentioned, however, unlike other agonists of the hGHS-Rla receptor, the compounds of the invention unexpectedly have an insignificant stimulatory effect on the release of growth hormone. Accordingly, the compounds of the present invention can exhibit selective action in the GI tract or for metabolic disorders without side effects due to GH release.
  • the present invention provides novel conformationally-defmed macrocyclic compounds. These compounds can function as modulators, in particular agonists, of the ghrelin (growth hormone secretagogue) receptor (GHS-RIa).
  • GHS-RIa growth hormone secretagogue receptor
  • the present invention relates to compounds according to formula I, II and/or III:
  • R 1 is hydrogen or the side chain of an amino acid, or alternatively R 1 and R 2 together form a A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively R 1 and Rg together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 2 is hydrogen or the side chain of an amino acid, or alternatively, R 1 and R 2 together form a 4-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below; or alternatively R 2 and Rg together form a 3-, 4-, 5-, 6- or 7-membered ring, optionally comprising an O, S or additional N
  • R 3 is hydrogen or the side chain of an amino acid, or alternatively R 3 and R 4 together form a 3-, 4-, 5-, 6- or 7-membered ring, optionally comprising an O or S atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively, R 3 and R 7 or R 3 and R 11 together form a 4-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O 5 S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 4 is hydrogen or the side chain of an amino acid, or alternatively R 4 and R 3 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O or S atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively R 4 and R 7 or R 4 and R 11 together form a A-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 5 and R 6 are each independently hydrogen or the side chain of an amino acid or alternatively Rs and R 6 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 7 is hydrogen, lower alkyl, substituted lower alkyl " , cycloalkyl, substituted cycloalkyl, a heterocyclic group, or a substituted heterocyclic group, or alternatively R 3 and R 7 or R 4 and R 7 together form a 3-, 4-, 5-, 6-, 7- or 8-membered heterocyclic ring optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as described below;
  • R 8 is substituted for one or more hydrogen atoms on the 3-, 4-, 5-, 6-, 7- or 8-membered ring structure and is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, 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, or, alternatively, R 8 is a fused cycloalkyl, a substituted fused cycloalkyl, a fused heterocyclic, a substituted fused heterocycl
  • X is O, NR 9 or N(R 1 O) 2 + ; wherein R 9 is hydrogen, lower alkyl, substituted lower alkyl, sulfonyl, sulfonamido or amidino and R 10 is hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 9 and R 1 together form a 3-, A-, 5-, 6- or 7- membered ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined above; Z 1 is O or NR 11 , wherein R 11 is hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 3 and R 11 together or R 4 and R 11 together form a 4-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined above;
  • Z 2 is O or NR 12 , wherein R 12 is hydrogen, lower alkyl, or substituted lower alkyl; m, n and p are each independently 0, 1 or 2; T is a bivalent radical of formula IV:
  • R 21 and R 22 are each independently hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 21 and R 22 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N, wherein the ring is optionally substituted with R 8 as defined above;
  • R 23 , R 39 and R 42 are each independently hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, amidino, sulfonyl or sulfonamido;
  • R 24 and R 25 are each independently hydrogen, lower alkyl, substituted lower alkyl, RAA > wherein R AA is a side chain of an amino acid such as a standard or unusual amino acid, or alternatively R 24 and R 25 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N; or alternatively one of R 24 or R 25 is hydroxy, alkoxy, aryloxy, amino, mercapto, carbamoyl, amidino, ureido or guanidino while the other is hydrogen, lower alkyl or substituted lower alkyl, except when the carbon to which R 24 and R 25 are bonded is also bonded to another heteroatom;
  • Ra 6 , R31, R 35 and R 38 are each optionally present and, when present, are substituted for one or more hydrogen atoms on the indicated ring and each is independently selected from the group consisting of halogen, trifluoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido;
  • R 27 is optionally present and is substituted for one or more hydrogen atoms on the indicated ring and each is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido;
  • R 28 , R29, R 30 , R32, R33, R34, R36 and R 37 are each optionally present and, when no double bond is present to the carbon atom to which it is bonded in the ring, two groups are optionally present, and when present, is substituted for one hydrogen present in the ring, or when no double bond is present to the carbon atom to which it is bonded in the ring, is substituted for one or both of the two hydrogen atoms present on the ring and each is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, foraiyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureid
  • R 50 is -(CH 2 ) SS CH 3 , -CH(CH 3 )(CH 2 ) tt CH 3 , -(CH 2 ) UU CH(CH 3 ) 2 , -C(CH 3 ) 3 ,
  • R 55 is hydrogen or C 1 -C 4 alkyl;
  • R 56 is amino, hydroxy, alkoxy, cycloalkyl or substituted cycloalkyl;
  • R 57 is hydrogen, alkyl, acyl, amino acyl, sulfonyl, carboxyalkyl or carboxyaryl;
  • R 51 is hydrogen, C 1 -C 4 alkyl or C 1 -C 4 alkyl substituted with hydroxy or alkoxy;
  • R 52 is -(CHR 58 )W w R 59 , wherein ww is 0, 1, 2 or 3;
  • R 58 is hydrogen, C 1 -C 4 alkyl, amino, hydroxy or alkoxy;
  • R 59 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl or substituted cycloalkyl;
  • R 53 is hydrogen or C 1 -C 4 alkyl;
  • X 2 is O, NR 9 or N(R 10 ) 2 + ; wherein R 9 is hydrogen, lower alkyl, substituted lower alkyl, sulfonyl, sulfonamido or amidino and R 10 is hydrogen, lower alkyl, or substituted lower alkyl; Z 5 is O or NR 12 , wherein R 12 is hydrogen, lower alkyl, or substituted lower alkyl; and
  • T 2 is a bivalent radical of formula V:
  • G 1 and G 2 are defined above, and wherein any carbon atom in the ring is optionally replaced by N, with the proviso that the aromatic ring cannot contain more than four N atoms and the cycloalkyl ring cannot contain more than two N atoms;
  • R 62 is hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, amidino, sulfonyl or sulfonamido;
  • R 63 and R 64 are each independently hydrogen, lower alkyl, substituted lower alkyl or RAA; or alternatively R 63 and R 64 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N; or alternatively one of R 63 and R 64 is hydroxy, alkoxy, aryloxy, amino, mercapto, carbamoyl, amidino, ureido or guanidino, while the other is hydrogen, lower alkyl or substituted lower alkyl, except when the carbon to which R 63 and R 64 are bonded is also bonded to another heteroatom; and RA A indicates the side chain of an amino acid such as a standard or unusual amino acid;
  • R 65 and R 68 are each optionally present, and, when present are substituted for one or more hydrogen atoms on the ring and each is independently halogen, trifluoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl or sulfonamido; R 66 and R 67 are each optionally present, and when no double bond is present to the carbon atom to which it is bonded in the ring, two groups are optionally present, and, when present, each
  • R 70 is hydrogen, C 1 -C 4 alkyl or alternatively R 70 and R 71 together form a 3-, A-, 5-,
  • 6- or 7-membered ring optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8a as defined below;
  • R 71 is hydrogen, -(CH 2 ) aa CH 3 , -CH(CH 3 )(CH 2 ) bb CH 3 , -(CH 2 ) CC CH(CH 3 ) 2 , -(CH 2 ) dd -R 76 or -CH(OR 77 )CH 3 or, alternatively R 71 and R 70 together form a 3-, 4-, 5-, 6- or 7-membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8a as defined below ; wherein aa is 0, 1, 2, 3, 4 or 5; bb is 1, 2 or 3; cc is 0, 1, 2 or 3; and dd is 0, 1, 2, 3 or 4; R 76 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl or substituted cycloalkyl; R 77 is hydrogen, alkyl, acyl, amino acyl
  • R 73 is hydrogen, or alternatively R 73 and R 72 together form a 3-, A-, 5-, 6- or 7- membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8b as defined below;
  • R 74 is hydrogen or C 1 -C 4 alkyl or alternatively R 74 and R 75 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 80 as defined below;
  • R 75 is -(CHR 78 )R 79 or alternatively R 75 and R 74 together form a 3-, A-, 5-, 6- or 7- membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8c as defined below; wherein R 78 is hydrogen, C 1 -C 4 alkyl, amino, hydroxy or alkoxy, and R 7 9 is selected from the group consisting of the following structures:
  • E 1 , E 2 , E 3 , E 4 and E 5 are each optionally present and when present are each independently selected from the group consisting of halogen, trifiuoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, cyano, sulfinyl, sulfonyl and sulfonamido, and represent substitution at one or more available positions on the monocyclic or bicyclic aromatic ring, wherein said substitution is made with the same or different selected group member, and J 1 and J 2 are each independently O or S;
  • R 8a , R 8b and R 80 are each independently substituted for one or more hydrogen atoms on the 3-, A-, 5-, 6- or 7-membered ring structure and are independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, 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, or, alternatively, R 8a , R 8b and R 8c are each independently a fused cycloalkyl, a substituted fused cycl
  • T 3 is the same as defined for T 2 with the exception that U a is bonded to X 3 of formula III.
  • the compound is a ghrelin receptor agonist or a GHS-Rl a receptor agonist.
  • compositions comprising: (a) a compound of the present invention; and (b) a pharmaceutically acceptable carrier, excipient or diluent.
  • kits comprising one or more containers containing pharmaceutical dosage units comprising an effective amount of one or more compounds of the present invention packaged with optional instructions for the use thereof.
  • aspects of the present invention further provide methods of stimulating gastrointestinal motility, modulating GHS-RIa receptor activity in a mammal and/or treating a gastrointestinal disorder comprising administering to a subject in need thereof an effective amount of a modulator that modulates a mammalian GHS-RIa receptor.
  • a modulator that modulates a mammalian GHS-RIa receptor.
  • interaction of the modulator and the GHS-RIa receptor does not result in a significant amount of growth hormone release.
  • the modulator is a compound of formula I, II and/or III.
  • Additional aspects of the present invention provide methods of diagnosing tumors and/or acromegaly, comprising administering compounds of the present invention and a radiolabeled metal binding agent and detecting the binding of the composition to a biological target, and treating tumors and/or acromegaly comprising administering a therapeutically effective amount of a composition comprising a compound of the present invention.
  • aspects of the present invention relate to methods of making the compounds of formula I, II and/or III.
  • aspects of the present invention further relate to methods of preventing and/or treating disorders described herein, in particular, gastrointestinal disorders, including post ⁇ operative ileus, gastroparesis, such as diabetic and post-surgical gastroparesis, opioid- induced bowel dysfunction, chronic intestinal pseudo-obstruction, short bowel syndrome, emesis such as caused by cancer chemotherapy, constipation such as associated with the hypomotirity phase of irritable bowel syndrome (IBS), delayed gastric emptying associated with wasting conditions, gastroesophageal reflux disease (GERD), gastric ulcers, Crohn's disease, gastrointestinal disorders characterized by dysmotility and other diseases and disorders of the gastrointestinal tract.
  • the present invention also relates to compounds of formula I, II and/or III used for the preparation of a medicament for prevention and/or treatment of the disorders described herein.
  • Figure 1 shows a scheme presenting a general synthetic strategy to provide conformationally-defmed macrocycles of the present invention.
  • Figure 2 shows a general thioester strategy for making macrocyclic compounds of the present invention.
  • FIG. 3 shows a general ring-closing metathesis (RCM) strategy for macrocyclic compounds of the present invention.
  • Figure 4 shows competitive binding curves for binding of exemplary compounds of the present invention to the AGHS-RIa receptor.
  • Figure 5 shows concentration-response curves for activation of the ⁇ GHS-R1 a receptor by exemplary compounds of the present invention.
  • Figure 6 shows graphs depicting pharmacokinetic parameters for exemplary compounds of the present invention, specifically after oral administration of 8 mg/kg compound 298 (panel A), after subcutaneous injection of 2 mg/kg compound 298 with cyclodextrin (panel B), after intravenous administration of 2 mg/kg compound 25 with cyclodextrin (panel C) and after intravenous administration of 2 mg/kg compound 298 with cyclodextrin (panel D).
  • Figure 7 panels A and B shows graphs presenting effects on gastric emptying for exemplary compounds of the present invention.
  • Figure 8 shows a graph presenting effects of postoperative ileus for an exemplary compound of the present invention.
  • Figure 9 shows graphs depicting the effect on pulsatile growth hormone release for an exemplary compound of the present invention.
  • Figure 10 shows a competive binding curve for binding of an exemplary compound of the present invention to the AGHS-RIa receptor.
  • Figure 11 shows an activation curve demonstrating the agonism of an exemplary compound of the present invention.
  • Figure 12 shows a graph depicting agonism and lack of growth hormone release for an exemplary compound of the present invention.
  • Figure 13 shows graphs depicting receptor desentization associated with binding of an exemplary compound of the present invention to the AGHS-RIa receptor.
  • Figure 14 (panels A and B) shows graphs presenting effects on gastric emptying for an exemplary compound of the present invention.
  • Figure 15 shows a graph presenting effects on postoperative ileus for an exemplary compound of the present invention.
  • Figure 16 shows graphs depicting reversal of morphine-delayed gastric emptying (panel A) and morphine-delayed gastrointestinal transit (panel B) for an exemplary compound of the present invention.
  • Figure 17 shows graphs depicting effects on gastroparesis for exemplary compounds of the present invention.
  • alkyl refers to straight or branched chain saturated or partially unsaturated hydrocarbon groups having from 1 to 20 carbon atoms, in some instances 1 to 8 carbon atoms.
  • lower alkyl refers to alkyl groups containing 1 to 6 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl, ethyl, isopropyl, tert-butyl, 3-hexenyl, and 2-butynyl.
  • unsaturated is meant the presence of 1, 2 or 3 double or triple bonds, or a combination of the two. Such alkyl groups may also be optionally substituted as described below.
  • cycloalkyl refers to saturated or partially unsaturated cyclic hydrocarbon groups having from 3 to 15 carbon atoms in the ring, in some instances 3 to 7, and to alkyl groups containing said cyclic hydrocarbon groups.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclopropylmethyl, cyclopentyl, 2-(cyclohexyl)ethyl, cycloheptyl, and cyclohexenyl.
  • Cycloalkyl as defined herein also includes groups with multiple carbon rings, each of which may be saturated or partially unsaturated, for example decalinyl, [2.2.1]-bicycloheptanyl or adamantanyl. AU such cycloalkyl groups may also be optionally substituted as described below.
  • aromatic refers to an unsaturated cyclic hydrocarbon group having a conjugated pi electron system that contains 4n+2 electrons where n is an integer greater than or equal to 1.
  • Aromatic molecules are typically stable and are depicted as a planar ring of atoms with resonance structures that consist of alternating double and single bonds, for example benzene or naphthalene.
  • aryl refers to an aromatic group in a single or fused carbocyclic ring system having from 6 to 15 ring atoms, in some instances 6 to 10, and to alkyl groups containing said aromatic groups.
  • aryl groups include, but are not limited to, phenyl, 1-naphthyl, 2-naphthyl and benzyl.
  • Aryl as defined herein also includes groups with multiple aryl rings which may be fused, as in naphthyl and anthracenyl, or unfused, as in biphenyl and terphenyl.
  • Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated or aromatic, for example, indanyl or tetrahydronaphthyl (tetralinyl). AU such aryl groups may also be optionally substituted as described below.
  • heterocycle refers to saturated or partially unsaturated monocyclic, bicyclic or tricyclic groups having from 3 to 15 atoms, in some instances 3 to 7, with at least one heteroatom in at least one of the rings, said heteroatom being selected from O, S or N.
  • Each ring of the heterocyclic group can contain one or two O atoms, one or two S atoms, one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the fused rings completing the bicyclic or tricyclic heterocyclic groups may contain only carbon atoms and may be saturated or partially unsaturated.
  • heterocyclic also refers to alkyl groups containing said monocyclic, bicyclic or tricyclic heterocyclic groups. Examples of heterocyclic rings include, but are not limited to, 2- or 3-piperidinyl, 2- or 3- piperazinyl, 2- or 3-morpholinyl. AU such heterocyclic groups may also be optionally substituted as described below
  • heteroaryl refers to an aromatic group in a single or fused ring system having from 5 to 15 ring atoms, in some instances 5 to 10, which have at least one heteroatom in at least one of the rings, said heteroatom being selected from O, S or N.
  • Each ring of the heteroaryl group can contain one or two O atoms, one or two S atoms, one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the fused rings completing the bicyclic or tricyclic groups may contain only carbon atoms and may be saturated, partially unsaturated or aromatic.
  • the N atoms may optionally be quaternized or oxidized to the N-oxide.
  • Heteroaryl also refers to alkyl groups containing said cyclic groups.
  • Examples of monocyclic heteroaryl groups include, but are not limited to pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, and triazinyl.
  • bicyclic heteroaryl groups include, but are not limited to indolyl, benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzo furanyl, isobenzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, purinyl, pyrrolopyridinyl, furopyridinyl, thienopyridinyl, dihydroisoindolyl, and tetrahydroquinolinyl.
  • tricyclic heteroaryl groups include, but are not limited to carbazolyl, benzindolyl, phenanthrollinyl, acridinyl, phenanthridinyl, and xanthenyl. All such heteroaryl groups may also be optionally substituted as described below.
  • hydroxy refers to the group -OH.
  • alkoxy refers to the group -OR a , wherein R 3 is alkyl, cycloalkyl or heterocyclic. Examples include, but are not limited to methoxy, ethoxy, tert-butoxy, cyclohexyloxy and tetrahydropyranyloxy.
  • aryloxy refers to the group -OR b wherein R b is aryl or heteroaryl.
  • Examples include, but are not limited to phenoxy, benzyloxy and 2-naphthyloxy.
  • acyl refers to the group wherein R 0 is alkyl, cycloalkyl, heterocyclic, aryl or heteroaryl. Examples include, but are not limited to, acetyl, benzoyl and furoyl.
  • amino acyl indicates an acyl group that is derived from an amino acid.
  • amino refers to an -NR d R e group wherein R d and Re are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclic, aryl and heteroaryl. Alternatively, R d and R e together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted
  • heterocyclic unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • R f and R g together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • R; and Rj together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or
  • Carboxy refers to the group -CO 2 H.
  • Carboxyalkyl refers to the group -CO 2 R k , wherein Rk is alkyl, cycloalkyl or heterocyclic.
  • Carboxyaryl refers to the group -CO 2 R m , wherein R m is aryl or heteroaryl.
  • cyano refers to the group -CN.
  • halo refers to fluoro, fluorine or fluoride, chloro, chlorine or chloride, bromo, bromine or bromide, and iodo, iodine or iodide, respectively.
  • mercapto refers to the group -SR n wherein R n is hydrogen, alkyl, cycloalkyl, heterocyclic, aryl or heteroaryl.
  • nitro refers to the group -NO 2 .
  • trifluoromethyl refers to the group -CF 3 .
  • sulfonyl refers to the group wherein R q1 is alkyl, cycloalkyl, heterocyclic, aryl or heteroaryl.
  • R r and R s together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfmyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • R x and R y together form a heterocyclic ring or 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfmyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • R aa and R ⁇ together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • optionally substituted is intended to expressly indicate that the specified group is unsubstituted or substituted by one or more suitable substituents, unless the optional substituents are expressly specified, in which case the term indicates that the group is unsubstituted or substituted with the specified substituents.
  • various groups may be unsubstituted or substituted (i.e., they are optionally substituted) unless indicated otherwise herein (e.g., by indicating that the specified group is unsubstituted).
  • R gg and RM 1 , R y and R kk or R pp and Rq q together form a heterocyclic ring of 3 to 8 members, optionally substituted with unsubstituted alkyl, unsubstituted cycloalkyl, unsubstituted heterocyclic, unsubstituted aryl, unsubstituted heteroaryl, hydroxy, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, carboxyaryl, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, guanidino or ureido, and optionally containing one to three additional heteroatoms selected from O, S or N.
  • substituted for aryl and heteroaryl groups includes as an option having one of the hydrogen atoms of the group replaced by cyano, nitro or trifluor
  • substitution is made provided that any atom's normal valency is not exceeded and that the substitution results in a stable compound.
  • such substituted group is preferably not further substituted or, if substituted, the substituent comprises only a limited number of substituted groups, in some instances 1, 2, 3 or 4 such substituents.
  • stable compound or “stable structure” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity and formulation into an efficacious therapeutic agent.
  • amino acid refers to the common natural (genetically encoded) or synthetic amino acids and common derivatives thereof, known to those skilled in the art.
  • standard or “proteinogenic” refers to the genetically encoded 20 amino acids in their natural configuration.
  • unnatural or “unusual” refers to the wide selection of non-natural, rare or synthetic amino acids such as those described by Hunt, S. in Chemistry and Biochemistry of the Amino Acids, Barrett, G.C., Ed., Chapman and Hall: New York, 1985.
  • residue with reference to an amino acid or amino acid derivative refers to a group of the formula:
  • RA A is an amino acid side chain
  • n 0, 1 or 2 in this instance.
  • fragment with respect to a dipeptide, tripeptide or higher order peptide derivative indicates a group that contains two, three or more, respectively, amino acid residues.
  • amino acid side chain refers to any side chain from a standard or unnatural amino acid, and is denoted R AA -
  • R AA the side chain of alanine
  • valine is isopropyl
  • tryptophan is 3-indolylmethyl.
  • agonist refers to a compound that duplicates at least some of the effect of the endogenous ligand of a protein, receptor, enzyme or the like.
  • antagonist refers to a compound that inhibits at least some of the effect of the endogenous ligand of a protein, receptor, enzyme or the like.
  • growth hormone secretagogue refers to any exogenously administered compound or agent that directly or indirectly stimulates or increases the endogenous release of growth hormone, growth hormone-releasing hormone, or somatostatin in an animal, in particular, a human.
  • a GHS may be peptidic or non-peptidic in nature, in some instances, with an agent that can be administered orally. In some instances, the agent can induce a pulsatile response.
  • modulator refers to a compound that imparts an effect on a biological or chemical process or mechanism.
  • a modulator may increase, facilitate, upregulate, activate, inhibit, decrease, block, prevent, delay, desensitize, deactivate, down regulate, or the like, a biological or chemical process or mechanism.
  • a modulator can be an "agonist” or an "antagonist.”
  • Exemplary biological processes or mechanisms affected by a modulator include, but are not limited to, receptor binding and hormone release or secretion.
  • Exemplary chemical processes or mechanisms affected by a modulator include, but are not limited to, catalysis and hydrolysis.
  • variable when applied to a receptor is meant to include dimers, trimers, tetramers, pentamers and other biological complexes containing multiple components. These components can be the same or different.
  • peptide refers to a chemical compound comprised of two or more amino acids covalently bonded together.
  • peptidomimetic refers to a chemical compound designed to mimic a peptide, but which contains structural differences through the addition or replacement of one of more functional groups of the peptide in order to modulate its activity or other properties, such as solubility, metabolic stability, oral bioavailability, lipophilicity, permeability, etc. This can include replacement of the peptide bond, side chain modifications, truncations, additions of functional groups, etc.
  • non-peptide peptidomimetic When the chemical structure is not derived from the peptide, but mimics its activity, it is often referred to as a "non-peptide peptidomimetic.”
  • protecting group refers to any chemical compound that may be used to prevent a potentially reactive functional group, such as an amine, a hydroxyl or a carboxyl, on a molecule from undergoing a chemical reaction while chemical change occurs elsewhere in the molecule. A number of such protecting groups are known to those skilled in the art and examples can be found in "Protective Groups in Organic Synthesis," Theodora W. Greene and Peter G. Wuts, editors, John Wiley & Sons, New York, 3 rd edition, 1999 [ISBN 0471160199].
  • amino protecting groups include, but are not limited to, phthalimido, trichloroacetyl, benzyloxycarbonyl, tert-butoxycarbonyl, and adamantyloxycarbonyl.
  • amino protecting groups are carbamate amino protecting groups, which are defined as an amino protecting group that when bound to an amino group forms a carbamate.
  • amino carbamate protecting groups are allyloxycarbonyl (Alloc), benzyloxycarbonyl (Cbz), 9-fluorenyknethoxycarbonyl (Fmoc), tert-butoxycarbonyl (Boc) and ⁇ , ⁇ -dimethyl- 3,5-dimethoxybenzyloxycarbonyl (Ddz).
  • hydroxyl protecting groups include, but are not limited to, acetyl, tert-butyldimethylsilyl (TBDMS), trityl (Trt), tert-butyl, and tetrahydropyranyl (THP).
  • carboxyl protecting groups include, but are not limited to methyl ester, tert-butyl ester, benzyl ester, trimethylsilylethyl ester, and 2,2,2-trichloroethyl ester.
  • solid phase chemistry refers to the conduct of chemical reactions where one component of the reaction is covalently bonded to a polymeric material (solid support as defined below). Reaction methods for performing chemistry on solid phase have become more widely known and established outside the traditional fields of peptide and oligonucleotide chemistry.
  • solid support refers to a mechanically and chemically stable polymeric matrix utilized to conduct solid phase ⁇ . chemistry. This is denoted by “Resin,” “P-” or the following symbol: ⁇ *
  • polystyrene examples include, but are not limited to, polystyrene, polyethylene, polyethylene glycol, polyethylene glycol grafted or covalently bonded to polystyrene (also termed PEG-polystyrene, TentaGelTM, Rapp, W.; Zhang, L.; Bayer, E. In Innovations and Persepctives in Solid Phase Synthesis.
  • polystyrene polyethylene
  • polyethylene glycol polyethylene glycol
  • polyethylene glycol grafted or covalently bonded to polystyrene also termed PEG-polystyrene, TentaGelTM, Rapp, W.; Zhang, L.; Bayer, E. In Innovations and Persepctives in Solid Phase Synthesis.
  • These materials can optionally contain additional chemical agents to form cross-linked bonds to mechanically stabilize the structure, for example polystyrene cross-linked with divinylbenezene (DVB, usually 0.1-5%, preferably 0.5-2%).
  • DVD divinylbenezene
  • This solid support can include as non-limiting examples aminomethyl polystyrene, hydroxymethyl polystyrene, benzhydrylamine polystyrene (BHA), methylbenzhydrylamine (MBHA) polystyrene, and other polymeric backbones containing free chemical functional groups, most typically, -NH 2 or -OH, for further derivatization or reaction.
  • BHA benzhydrylamine polystyrene
  • MBHA methylbenzhydrylamine
  • the materials used as resins are insoluble polymers, but certain polymers have differential solubility depending on solvent and can also be employed for solid phase chemistry.
  • polyethylene glycol can be utilized in this manner since it is soluble in many organic solvents in which chemical reactions can be conducted, but it is insoluble in others, such as diethyl ether.
  • reactions can be conducted homogeneously in solution, then the product on the polymer precipitated through the addition of diethyl ether and processed as a solid. This has been termed "liquid-phase" chemistry.
  • linker when used in reference to solid phase chemistry refers to a chemical group that is bonded covalently to a solid support and is attached between the support and the substrate typically in order to permit the release (cleavage) of the substrate from the solid support. However, it can also be used to impart stability to the bond to the solid support or merely as a spacer element. Many solid supports are available commercially with linkers already attached.
  • the term "effective amount” or “effective” is intended to designate a dose that causes a relief of symptoms of a disease or disorder as noted through clinical testing and evaluation, patient observation, and/or the like, and/or a dose that causes a detectable change in biological or chemical activity.
  • the detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process.
  • the dosage will vary depending on the administration routes, symptoms and body weight of the patient but also depending upon the compound being administered.
  • Administration of two or more compounds "in combination” means that the two compounds are administered closely enough in time that the presence of one alters the biological effects of the other.
  • the two compounds can be administered simultaneously (concurrently) or sequentially. Simultaneous administration can be carried out by mixing the compounds prior to administration, or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration.
  • the phrases "concurrent administration”, “administration in combination”, “simultaneous administration” or “administered simultaneously” as used herein, means that the compounds are administered at the same point in time or immediately following one another, hi the latter case, the two compounds are administered at times sufficiently close that the results observed are indistinguishable from those achieved when the compounds are administered at the same point in time.
  • pharmaceutically active metabolite is intended to mean a pharmacologically active product produced through metabolism in the body of a specified compound.
  • solvate is intended to mean a pharmaceutically acceptable solvate form of a specified compound that retains the biological effectiveness of such compound.
  • examples of solvates include compounds of the invention in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
  • Novel macrocyclic compounds of the present invention include macrocyclic compounds comprising a building block structure including a tether component that undergoes cyclization to form the macrocyclic compound.
  • the building block structure can comprise amino acids (standard and unnatural), hydroxy acids, hydrazino acids, aza- amino acids, specialized moieties such as those that play a role in the introduction of peptide surrogates and isosteres, and a tether component as described herein.
  • the tether component can be selected from the following:
  • (Z 2 ) is the site of a covalent bond of T to Z 2 , and Z 2 is as defined below for formula I, and wherein (X) is the site of a covalent bond of T to X, and X is as defined below for formula I;
  • L 7 is -CH 2 - or -0-;
  • R 100 is lower alkyl;
  • R 101 and R 102 are each independently hydrogen, lower alkyl or substituted lower alkyl;
  • xx is 2 or 3;
  • yy is 1 or 2; zz is 1 or 2; and
  • aaa is O or 1.
  • Macrocyclic compounds of the present invention further include those of formula I, formula II and/or formula III:
  • R 1 is hydrogen or the side chain of an amino acid, or alternatively R 1 and R 2 together form a A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively R 1 and R 9 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 2 is hydrogen or the side chain of an amino acid, or alternatively, R 1 and R 2 together form a 4-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below; or alternatively R 2 and R 9 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 3 is hydrogen or the side chain of an amino acid, or alternatively R 3 and R 4 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O or S atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively, R 3 and R 7 or R 3 and R 11 together form a A-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 4 is hydrogen or the side chain of an amino acid, or alternatively R 4 and R 3 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O or S atom in the ring, wherein the ring is optionally substituted with R 8 as defined below, or alternatively R 4 and R 7 or R 4 and R 11 together form a A-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 5 and R 6 are each independently hydrogen or the side chain of an amino acid or alternatively R 5 and R 6 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8 as defined below;
  • R 7 is hydrogen, lower alkyl, substituted lower alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, or a substituted heterocyclic group, or alternatively R 3 and R 7 or R 4 and R 7 together form a 3-, A-, 5-, 6-, 7- or 8-membered heterocyclic ring optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as described below;
  • R 8 is substituted for one or more hydrogen atoms on the 3-, A-, 5-, 6-, 7- or 8-membered ring structure and is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfmyl, sulfonyl and sulfonamido, or, alternatively, R 8 is a fused cycloalkyl, a substituted fused cycloalkyl, a fused heterocyclic, a substituted fused heterocycl
  • X is O, NR 9 or N(R 1O ) 2 + ; wherein R 9 is hydrogen, lower alkyl, substituted lower alkyl, sulfonyl, sulfonamido or amidino and R 10 is hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 9 and R 1 together form a 3-, A-, 5-, 6- or 7- membered ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined above; Z 1 is O or NR n , wherein R 11 is hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 3 and R 11 together or R 4 and R 11 together form a A-, 5-, 6-, 7- or 8-membered heterocyclic ring, optionally comprising an O, S or additional N atom in the ring, wherein the ring is optionally substituted with R 8 as defined above;
  • Z 2 is O or NR 12i wherein R 12 is hydrogen, lower alkyl, or substituted lower alkyl; m, n and p are each independently 0, 1 or 2;
  • T is a bivalent radical of formula IV:
  • R 21 and R 22 are each independently hydrogen, lower alkyl, or substituted lower alkyl, or alternatively R 21 and R 22 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N, wherein the ring is optionally substituted with R 8 as defined above;
  • R 23 , R 39 and R 42 are each independently hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, amidino, sulfonyl or sulfonamido;
  • R 24 and R 25 are each independently hydrogen, lower alkyl, substituted lower alkyl, RA A , wherein R AA is a side chain of an amino acid such as a standard or unusual amino acid, or alternatively R 24 and
  • R 25 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N; or alternatively one of R 24 or R 25 is hydroxy, alkoxy, aryloxy, amino, mercapto, carbamoyl, amidino, ureido or guanidino while the other is hydrogen, lower alkyl or substituted lower alkyl, except when the carbon to which R 24 and R 25 are bonded is also bonded to another heteroatom;
  • R 26 , R 3 i, R 35 and R 38 are each optionally present and, when present, are substituted for one or more hydrogen atoms on the indicated ring and each is independently selected from the group consisting of halogen, trifluoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido;
  • R 27 is optionally present and is substituted for one or more hydrogen atoms on the indicated ring and each is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido;
  • R-28, R29, Rao, R32, R33, R34, R36 and R37 are eacn optionally present and, when no double bond is present to the carbon atom to which it is bonded in the ring, two groups are optionally present, and when present, is substituted for one hydrogen present in the ring, or when no double bond is present to the carbon atom to which it is bonded in the ring, is substituted for one or both of the two hydrogen atoms present on the ring and each is independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, urei
  • R 40 and R 41 are each independently hydrogen, lower alkyl, substituted lower alkyl, R AA as defined above, or alternativelyR-w and R 41 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N wherein the ring is optionally substituted with R 8 as defined above, or alternatively one of R 40 and R 41 is hydroxy, alkoxy, aryloxy, amino, mercapto, carbamoyl, amidino, ureido or guanidino, while the other is hydrogen, lower alkyl or substituted lower alkyl, except when the carbon to which R 40 and R 41 are bonded is also bonded to another heteroatom; with the proviso that T is not an amino acid residue, dipeptide fragment, tripeptide fragment or higher order peptide fragment including standard amino acids;
  • R 50 is -(CH 2 ) SS CH 3 , -CH(CH 3 )(CH 2 ) tt CH 3 , -(CH 2 ) UU CH(CH 3 ) 2 , -C(CH 3 ) 3 ,
  • R 55 is hydrogen or C 1 -C 4 alkyl
  • R 56 is amino, hydroxy, alkoxy, cycloalkyl or substituted cycloalkyl
  • R 57 is hydrogen, alkyl, acyl, amino acyl, sulfonyl, carboxyalkyl or carboxyaryl
  • R 51 is hydrogen, C 1 -C 4 alkyl or C 1 -C 4 alkyl substituted with hydroxy or alkoxy;
  • R 52 is -(CHR 58 ) Ww R 59 , wherein ww is 0, 1, 2 or 3;
  • R 58 is hydrogen, C 1 -C 4 alkyl, amino, hydroxy or alkoxy;
  • R 59 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl or substituted cycloalkyl;
  • R 53 is hydrogen or C 1 -C 4 alkyl
  • X 2 is O, NR 9 or N(R 10 ) 2 + ; wherein R 9 is hydrogen, lower alkyl, substituted lower alkyl, sulfonyl, sulfonamido or amidino and R 10 is hydrogen, lower alkyl, or substituted lower alkyl;
  • Z 5 is O or NR 12 , wherein R 12 is hydrogen, lower alkyl, or substituted lower alkyl;
  • T 2 is a bivalent radical of formula V:
  • G 1 and G 2 are defined above, and wherein any carbon atom in the ring is optionally replaced by N, with the proviso that the aromatic ring cannot contain more than four N atoms and the cycloalkyl ring cannot contain more than two N atoms;
  • R 62 is hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, amidino, sulfonyl or sulfonamido;
  • R 63 and R 64 are each independently hydrogen, lower alkyl, substituted lower alkyl or R AA ; or alternatively R 63 and R 64 together form a 3- to 12-membered cyclic ring optionally comprising one or more heteroatoms selected from the group consisting of O, S and N; or alternatively one of R 63 and R 64 is hydroxy, alkoxy, aryloxy, amino, mercapto, carbamoyl, amidino, ureido or guanidino, while the other is hydrogen, lower alkyl or substituted lower alkyl, except when the carbon to which R 63 and R 64 are bonded is also bonded to another heteroatom; and RAA indicates the side chain of a standard or unusual amino acid;
  • R 65 and R 68 are each optionally present, and, when present are substituted for one or more hydrogen atoms on the ring and each is independently halogen, trifluoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfmyl, sulfonyl or sulfonamido;
  • R 66 and R 67 are each optionally present, and when no double bond is present to the carbon atom to which it is bonded in the ring, two groups are optionally present, and, when present, each is substituted for one hydrogen present in the ring, or when no double bond is present to the carbon atom to which it is bonded in the ring, is substituted for one or both of the two hydrogen atoms present on the ring and each is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfmyl, sulfonyl, sulfon
  • R 69 is optionally present, and when present is substituted for one or more hydrogen atoms on the ring and each is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl or sulfonamido; K -6 is O or S; and ff is 1, 2, 3, 4 or 5; with the proviso that T 2 is not an amino acid residue, dipeptide fragment, tripeptide fragment or higher order peptide fragment including standard amino acids; or
  • R 7O is hydrogen, C 1 -C 4 alkyl or alternatively R 7 o and R 71 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8a as defined below;
  • R 7I is hydrogen, -(CH 2 ) aa CH 3 , -CH(CH 3 )(CH 2 ) bb CH 3 , -(CH 2 ) CC CH(CH 3 ) 2 , -(CH 2 ) dd -R 76 or -CH(OR 77 )CH 3 or, alternatively R 71 and R 70 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8a as defined below ; wherein aa is 0, 1, 2, 3, 4 or 5; bb is 1, 2 or 3; cc is 0, 1, 2 or 3; and dd is 0, 1, 2, 3 or 4; R 76 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl or substituted cycloalkyl; R 77 is hydrogen, alkyl, acyl, amino acy
  • R 72 is C 1 -C 4 alkyl; or alternatively R 72 and R 73 together form a 3-, A-, 5-, 6- or 7- membered ring, optionally comprising an O or S atom in the ring, wherein the ring is optionally substituted with R 8b as defined below;
  • R 73 is hydrogen, or alternatively R 73 and R 72 together form a 3-, 4-, 5-, 6- or 7- membered ring, optionally comprising an O, S or N atom in the ring, wherein the ring is optionally substituted with R 8b as defined below;
  • R 74 is hydrogen or C 1 -C 4 alkyl or alternatively R 74 and R 75 together form a 3-, A-, 5-, 6- or 7-membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 80 as defined below;
  • R 75 is -(CHR 78 )R 79 or alternatively R 75 and R 74 together form a 3-, 4-, 5-, 6- or 7- membered ring, optionally comprising an O, N or S atom in the ring, wherein the ring is optionally substituted with R 8c as defined below; wherein R 78 is hydrogen, C 1 -C 4 alkyl, amino, hydroxy or alkoxy, and R 79 is selected from the group consisting of the following structures:
  • E 1 , E 2 , E 3 , E 4 and E 5 are each optionally present and when present are each independently selected from the group consisting of halogen, trifluoromethyl, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, cyano, sulfmyl, sulfonyl and sulfonamido, and represent substitution at one or more available positions on the monocyclic or bicyclic aromatic ring, wherein said substitution is made with the same or different selected group member, and J 1 and J 2 are each independently O or S;
  • R- 8a , R- 8b and R 8c are each independently substituted for one or more hydrogen atoms on the 3-, 4-, 5-, 6- or 7-membered ring structure and are independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, a heterocyclic group, a substituted heterocyclic group, 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, or, alternatively, R 8a , R 8b and R 8c are each independently a fused cycloalkyl, a substituted fused
  • Z 10 is O or NR 12, wherein R 12 is hydrogen, lower alkyl, or substituted lower alkyl; and
  • T 3 is the same as defined for T 2 with the exception that U a is bonded to X 3 of formula III.
  • the compound can have one of the following structures:
  • the present invention includes isolated compounds.
  • An isolated compound refers to a compound that, in some embodiements, comprises at least 10%, at least 25%, at least 50% or at least 70% of the compounds of a mixture.
  • the compound, pharmaceutically acceptable salt thereof or pharmaceutical composition containing the compound exhibits a statistically significant binding and/or antagonist activity when tested in biological assays at the human ghrelin receptor.
  • the compounds of formula I, II and/or III disclosed herein have asymmetric centers.
  • the inventive compounds may exist as single stereoisomers, racemates, and/or mixtures of enantiomers and/or diastereomers. All such single stereoisomers, racemates, and mixtures thereof are intended to be within the scope of the present invention. In particular embodiments, however, the inventive compounds are used in optically pure form.
  • the terms "S” and "R” configuration as used herein are as defined by the IUPAC 1974 Recommendations for Section E, Fundamentals of Stereochemistry (Pure Appl. Chem. 1976, 45, 13-30.)
  • the compounds may be prepared as a single stereoisomer or a mixture of stereoisomers.
  • the non-racemic forms may be obtained by either synthesis or resolution.
  • the compounds may, for example, be resolved into the component enantiomers by standard techniques, for example formation of diastereomeric pairs via salt formation.
  • the compounds also may be resolved by covalently bonding to a chiral moiety.
  • the diastereomers can then be resolved by chromatographic separation and/or crystallographic separation. In the case of a chiral auxiliary moiety, it can then be removed.
  • the compounds can be resolved through the use of chiral chromatography. Enzymatic methods of resolution could also be used in certain cases.
  • an “optically pure” compound is one that contains only a single enantiomer.
  • the term “optically active” is intended to mean a compound comprising at least a sufficient excess of one enantiomer over the other such that the mixture rotates plane polarized light.
  • Optically active compounds have the ability to rotate the plane of polarized light. The excess of one enantiomer over another is typically expressed as enantiomeric excess (e.e.).
  • the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chiral center(s).
  • the prefixes "d” and “1" or (+) and (-) are used to denote the optical rotation of the compound (i.e., the direction in which a plane of polarized light is rotated by the optically active compound).
  • the "1" or (- ) prefix indicates that the compound is levorotatory (i.e., rotates the plane of polarized light to the left or counterclockwise) while the "d” or (+) prefix means that the compound is dextrarotatory (i.e., rotates the plane of polarized light to the right or clockwise).
  • the sign of optical rotation, (-) and (+) is not related to the absolute configuration of the molecule, R and S.
  • Embodiments of the present invention further provide intermediate compounds formed through the synthetic methods described herein to provide the compounds of formula I, II and/or III.
  • the intermediate compounds may possess utiltity as a therapeutic agent for the range of indications described herein and/or a reagent for further synthesis methods and reactions.
  • the compounds of formula I, II and/or II can be synthesized using traditional solution synthesis techniques or solid phase chemistry methods, hi either, the construction involves four phases: first, synthesis of the building blocks 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, the post-cyclization processing fourth stage involving removal of protecting groups and optional purification provides the desired final compounds. Synthetic methods for this general type of macrocyclic structure are described in Ml. Pat. Appls. WO 01/25257, WO 2004/111077, WO 2005/012331 and WO 2005/012332, including purification procedures described in WO 2004/111077 and WO 2005/012331.
  • the macrocyclic compounds of formula I, II and/or III may be synthesized using solid phase chemistry on a soluble or insoluble polymer matrix as previously defined.
  • solid phase chemistry a preliminary stage involving the attachment of the first building block, also termed "loading," to the resin must be performed.
  • the resin utilized for the present invention preferentially has attached to it a linker moiety, L.
  • linkers are attached to an appropriate free chemical functionality, usually an alcohol or amine, although others are also possible, on the base resin through standard reaction methods known in the art, such as any of the large number of reaction conditions developed for the formation of ester or amide bonds.
  • linker moieties for the present invention are designed to allow for simultaneous cleavage from the resin with formation of the macrocycle in a process generally termed "cyclization-release.”
  • the thioester strategy proceeds through a modified route where the tether component is actually assembled during the cyclization step.
  • assembly of the building blocks proceeds sequentially, followed by cyclization (and release from the resin if solid phase).
  • An additional post-cyclization processing step is required to remove particular byproducts of the RCM reaction, but the remaining subsequent processing is done in the same manner as for the thioester or analogous base-mediated cyclization strategy.
  • steps including the methods provided herein may be performed independently or at least two steps may be combined. Additionally, steps including the methods provided herein, when performed independently or combined, may be performed at the same temperature or at different temperatures without departing from the teachings of the present invention.
  • Novel macrocyclic compounds of the present invention include those formed by a novel process including cyclization of a building block structure to form a macrocyclic compound comprising a tether component described herein. Accordingly, the present invention provides methods of manufacturing the compounds of the present invention comprising (a) assembling building block structures, (b) chemically transforming the building block structures, (c) cyclizing the building block structures including a tether component, (d) removing protecting groups from the building block structures, and (e) optionally purifiying the product obtained from step (d). In some embodiments, assembly of the building block structures may be sequential. In further embodiments, the synthesis methods are carried out using traditional solution synthesis techniques or solid phase chemistry techniques. A. Amino acids
  • Amino acids, 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 [for example Advanced ChemTech (Louisville, KY, USA), Bachem (Bubendorf, Switzerland), Chemhnpex (Wood Dale, IL, USA), Novabiochem (subsidiary of Merck KGaA, Darmstadt, Germany), PepTech (Burlington, MA, USA), Synthetech (Albany, OR, USA)] or synthesized through standard methodologies known to those in the art.
  • Ddz-amino acids were either obtained commercially from Orpegen (Heidelberg, Germany) or Advanced ChemTech (Louisville, KY, USA) or synthesized using standard methods utilizing Ddz-OPh or Ddz-N 3 .
  • Bts-amino acids were synthesized by known methods.
  • N-Alkyl amino acids in particular N-methyl amino acids, are commercially available from multiple vendors (Bachem, Novabiochem, Advanced ChemTech, Chemlmpex). In addition, N-alkyl amino acid derivatives were accessed via literature methods. (Hansen, D. W., Jr.; Pilipauskas, D. J. Org. Chem. 1985, 50, 945-950.)
  • Tethers were obtained from the methods previously described in Intl. Pat. Appl.
  • Table IA presents a summary of the synthesis of 228 representative compounds of the present invention.
  • the reaction methodology employed for the construction of the macrocyclic molecule is indicated in Column 2 and relates to the particular scheme of the synthetic strategy, for example, use of the thioester strategy as shown in figure 2 or the RCM approach as shown in figure 3.
  • Column 3 indicates if any substituents are present on N BBI -
  • Columns 4-6 and 8 indicate the individual building blocks employed for each compound, amino acids, hydroxy acids or tether utilizing either standard nomenclature or referring to the building block designations presented elsewhere in this application.
  • Table IB presents a summary of the synthesis of 122 representative compounds of the present invention, and Table 1C presents the synthesis of an additional 15 representative compounds.
  • the reaction methodology employed for the construction of the macrocyclic molecule is indicated in the Column 2 and relates to the particular scheme of the synthetic strategy.
  • Columns 3-6 indicate the individual building blocks employed for each compound, amino acids or tether utilizing either standard nomenclature or referring to the building block designations presented elsewhere in this application.
  • Column 7 indicates the method used for attachment of the tether.
  • the building blocks are listed in the opposite order from which they are added in order to correlate the building block number with standard peptide nomenclature.
  • Table 2A Analytical unaracie ⁇ zaiio ⁇ ⁇ or ⁇ epresem» ⁇ ve uum ⁇ uunus « ⁇ me ⁇ ⁇ ..i ⁇
  • the compounds of the present invention were evaluated for their ability to interact at the human ghrelin receptor utilizing a competitive radioligand binding assay, fluorescence assay or Aequorin functional assay as described below. Such methods can be conducted in a high throughput manner to permit the simultaneous evaluation of many compounds.
  • GHS-RIa human
  • swine and rat GHS-receptors U.S. Pat. No. 6,242,199, Ml. Pat. Appl. Nos. WO 97/21730 and 97/22004
  • canine GHS-receptor U.S. Pat. No. 6,645,726
  • GHS-R/HEK 293 were prepared from HEK-293 cells stably transfected with the human ghrelin receptor (hGHS-Rla). These membranes were provided by PerkinElmer BioSignal (#RBHGHSM, lot#1887) and utilized at a quantity of 0.71 ⁇ g/assay point. 1. [ 125 I] -Ghrelin (PerkinElmer, #NEX-388); final concentration: 0.0070-0.0085 nM
  • Binding Buffer 25 mM Hepes (pH 7.4), 1 mM CaCl 2 , 5 mM MgCl 2 , 2.5 mM EDTA, 0.4% BSA
  • the reaction was arrested by filtering samples through Multiscreen Harvest plates (pre-soaked in 0.5% polyethyleneimine) using a Tomtec Harvester, washed 9 times with 500 ⁇ L of cold 50 mM Tris-HCl (pH 7.4, 4°C), and then plates were air-dried in a fumehood for 30 min. A bottom seal was applied to the plates prior to the addition of 25 ⁇ L of MicroScint-0 to each well. Plates were than sealed with TopSeal-A and counted for 30 sec per well on a TopCount Microplate Scintillation and Luminescence Counter (PerkinElmer) using a count delay of 60 sec. Results were expressed as counts per minute (cpm).
  • Binding activity at the gherlin receptor for representative compounds of the present invention is shown below in Table 3A through 3D.
  • Compound structures for Tables 3A, 3B and 3D are presented with the various groups as defined for the general structure of formula I.
  • m, n and p are 0; X, Z 1 and Z 2 are each NH.
  • R 1 is H for all entries.
  • the tethers (T) are illustrated with the bonding to X and Z 2 as indicated.
  • the compounds themselves are shown for Table 3 C.
  • Competitive binding curves for representative compounds 1, 2, 3, 4 and 25 are shown in Figure 4.
  • Table 3 ⁇ Binding Activity at the Human Ghrelin Receptor for Compounds of the Invention
  • RIa receptor can be determined using the method described below which can also be used as a primary screen for ghrelin receptor activity in a high throughput fashion. (LePoul, E.; et al. Adaptation of aequorin functional assay to high throughput screening. J. Biomol.
  • Membranes were prepared using AequoScreenTM (EUROSCREEN, Belgium) cell lines expressing the human ghrelin receptor (cell line ES-410-A; receptor accession #60179). This cell line is typically constructed by transfection of the human ghrelin receptor into CHO-Kl cells co-expressing G ⁇ l6 and the mitochondrially targeted Aequorin
  • Ghrelin reference agonist
  • Assay buffer DMEM (Dulbecco's Modified Eagles Medium) containing 0.1% BSA (bovine serum albumin; pH 7.0).
  • AequoScreenTM cells were collected from culture plates with Ca 2+ and Mg 2+ -free phosphate buffered saline (PBS) supplemented with 5 mM EDTA, pelleted for 2 min at
  • IOOOX g re-suspended in DMEM - Ham's F12 containing 0.1% BSA at a density of 5 x 10 6 cells/mL, and incubated O/N at rt in the presence of 5 ⁇ M coelenterazine. After loading, cells were diluted with assay buffer to a concentration of 5 x 10 5 cells/mL.
  • ghrelin reference agonist
  • 50 ⁇ L of the cell suspension was mixed with 50 ⁇ L of the appropriate concentration of test compound or ghrelin (reference agonist) in 96-well plates (duplicate samples).
  • Ghrelin (reference agonist) was tested at several concentrations concurrently with the test compounds in order to validate the experiment.
  • the emission of light resulting from receptor activation in response to ghrelin or test compounds was recorded using the Hamamatsu FDSS 6000 reader (Hamamatsu Photonics K.K., Japan).
  • RLU Relative Light Units
  • motilin itself as been demonstrated to have some GH-releasing effects. (Samson, W.K.; Lumpkin, M.D.; Nilaver, G.; McCann, S. M. Motilin: a novel growth hormone releasing agent. Brain Res. Bull. 1984, 12, 57-62.)
  • Cell culture assays for determining growth hormone release can be employed as described in Cheng, et al. Endocrinology 1989, 124, 2791-2798.
  • anterior pituitary glands are obtained from male Sprague-Dawley rats and placed in cold culture medium. These pituitaries are sectioned, for example into one-eighth sections, then digested with trypsin. Cells are collected after digestion, pooled, and transferred into 24 well plates (minimum 200,000 cells per well). After a monolayer of cells has formed, generally after at least 4 d in culture, the cells are washed with medium prior to exposure to the test samples and controls.
  • Varying concentrations of the test compounds and of ghrelin as a positive control were added to the medium.
  • the cells are left for 15 min at 37 0 C, then the medium removed and the cells stored frozen.
  • the amount of GH release was measured utilizing a standard radioimmunoassay as known to those in the art.
  • Rats male, Sprague-Dawley ( ⁇ 250g) Rats/Treatment Group: 6 (2 subsets of 3 rats each, alternate bleeds)
  • test compound was sent in solution in a formulation (such as with cyclodextrin) appropriate for dosing. It will be appreciated by one skilled in the art that appropriate modifications to this protocol can be made as required to adequately test the properties of the compound under analysis. Typical Dose
  • Table 5 Representative Intravenous Blood Sampling Schedule.
  • Table 6 Representative Subcutaneous & Oral Blood Sampling Schedule.
  • 0.7 mL of blood were collected from each animal. It is expected that this volume of blood will yield a sample of at least .0.3 mL of plasma.
  • EDTA was used as an anti-coagulant for whole blood collection. Whole blood samples were chilled and immediately processed by centrifugation to obtain plasma.
  • Plasma samples were stored frozen (-70 0 C) until analysis.
  • Analytical detection of parent compound in plasma samples performed by LC-MS after an appropriate preparation protocol: extraction using solid phase extraction (SPE) cartridges (Oasis MCX, Oasis HLB) or liquid-liquid extraction.
  • SPE solid phase extraction
  • the analyte was quantitated based upon a standard curve and the method validated with internal standards.
  • compounds of the invention were evaluated for possible effects on gastric emptying in fasted rats.
  • compounds 25 and 298 at 100 ⁇ g/kg caused a significant increase (>30%) in gastric emptying relative to the vehicle control group.
  • the relative efficacy (39% increase) of compounds 25 and 298 at 100 ⁇ g/kg i.v. was similar to concurrently run positive reference agents GHRP-6 at 20 ⁇ g/kg i.v. (40% increase) and metoclopramide at 10 mg/kg i.v. (41% increase).
  • compounds 25 and 298 at a dose of 100 ⁇ g/kg demonstrated gastrokinetic activity in rats, with efficiency similar to GHRP-6 at 20 ⁇ g/kg and metoclopramide at 10 mg/kg. Further, compound 25 also demonstrated gastric emptying at 30 ⁇ g/kg. This is significantly more potent than other compounds interacting at this receptor previously found to enhance GI motility, which were unable to promote gastric emptying at 100 ⁇ g/kg (U.S. Patent No. 6,548,501).
  • GHRP-6 and test samples were dissolved in vehicle of 9% HPBCD/0.9% NaCl.
  • vehicle 9% HPBCD/0.9% NaCl.
  • test substances or vehicle (9% HPBCD/0.9% NaCl) were each administered intravenously (i.v.) at a dosing volume of 5 mL/kg.
  • 8-well strip (Costar, USA), 96-well plate (Costar, USA), Animal case (ShinTeh, R. O. C), Centrifugal separator (Kokusan, H-107, Japan), Glass syringe (1 mL, 2 mL, Mitsuba, Japan), Hypodermic needle (25G x 1", TOP Corporation, Japan), Microtube (Treff, Switzerland), pH-meter (Hanna, USA), Pipetamam (PlOO, Gilson, France), Pipette tips (Costar, USA), Rat oral needle (Natsume, Japan), Spectra Fluor plus (Austria), Stainless scissors (Klappencker, Germany) and Stainless forceps (Klappencker, Germany).
  • Test substances were each administered intravenously to a group of 5 O/N-fasted Wistar derived male rats weighing 200 ⁇ 20 g immediately after methylcellulose (2%) containing phenol red (0.05%) was administered orally at 2 mL/animal. The animals were then sacrificed 15 minutes later. The stomach was immediately removed, homogenized in 0.1 N NaOH (5 mL) and centrifuged. Following protein precipitation by 20% trichloroacetic acid (0.5 mL) and re-alkalization of the supernatant with 0.1 N NaOH, total phenol red remaining in the stomach was determined by a colorimetric method at 560 nm. A 30 percent or more (>30%) increase in gastric emptying, detected as the decrease in phenol red concentration in the stomach relative to the vehicle control group, is considered significant. Results for two representative compounds of the invention are shown in Figure 7 and in the Examples below.
  • Rat Sprague-Dawley, male, ⁇ 300 g.
  • POI post-operative ileus
  • Intestines were gently replaced into the abdomen and the abdominal wound was stitched closed under sterile conditions.
  • Geometric mean ⁇ (%total radioactivity X number of segment)/100
  • the compounds of the invention likewise can be tested in a number of animal models for their effect on GH release.
  • rats Boers, C.Y.; Momany, F.; Reynolds, G.A.; Chang, D.; Hong, A.; Chang, K. Endocrinology 1980, 106, 663-667
  • dogs Hickey, G.; Jacks, T.; Judith, F.; Taylor, J.; Schoen, W.R.; Krupa, D.; Cunningham, P.; Clark, J.; Smith, R.G.
  • Rats 225-300 g were purchased from Charles River Canada (St. Constant, Canada) and individually housed on a 12-h light, 12-h dark cycle (lights on, time: 0600-1800) in a temperature (22 + 1° C)- and humidity-controlled room.
  • Purina rat chow Rosina Co., St. Louis, MO
  • tap water were freely available.
  • chronic intracerebroventricular (icv) and intracardiac venous cannulas were implanted under sodium pentobarbital (50 mg/kg, ip) anesthesia using known techniques.
  • icv cannula was verified by both a positive drinking response to icv carbachol (100 ng/10 ⁇ ) injection on the day after surgery and methylene blue dye at the time of sacrifice.
  • the rats were placed directly in isolation test chambers with food and water freely available until body weight returned to preoperative levels (usually within 5-7 d). During this time, the rats were handled daily to minimize any stress associated with handling on the day of the experiment.
  • food was removed 1.5 h before the start of sampling and was returned at the end. Free moving rats were iv injected with either test sample at various levels (3, 30, 300, 1000 jug/kg) or normal saline at two different time points during a 6-h sampling period.
  • the times 1100 and 1300 were chosen because they reflect typical peak and trough periods of GH secretion, as previously documented.
  • the human ghrelin peptide (5 ⁇ g, Phoenix Pharmaceuticals, Inc., Belmont, CA) was used as a positive control in the experiments and was diluted in normal saline just before use.
  • a 10-fold lower dose of the test sample or normal saline was administered icv at the same time points, 1100 and 1300.
  • Blood samples (0.35 mL) were withdrawn every 15 min over the 6-h sampling period (time: 1000-1600) from all animals.
  • an additional blood sample was obtained 5 min after each injection.
  • Plasma GH concentrations were measured in duplicate by double antibody RIA using materials supplied by the NIDDK Hormone Distribution Program (Bethesda, MD). The averaged plasma GH values for 5-6 rats per group are reported in terms of the rat GH reference preparation. The standard curve was linear within the range of interest; the least detectable concentration of plasma GH under the conditions used was approximately 1 ng/niL. All samples with values above the range of interest were reassayed at dilutions ranging from 1:2 to 1:10. The intra- and interassay coefficients of variation were acceptable for duplicate samples of pooled plasma containing a known GH concentration.
  • the macrocyclic compounds of the present invention or pharmacologically acceptable salts thereof according to the invention may be formulated into pharmaceutical compositions of various dosage forms.
  • one or more compounds, including optical isomers, enantiomers, diastereomers, racemates or stereochemical mixtures thereof, or pharmaceutically acceptable salts thereof as the active ingredient is intimately mixed with appropriate carriers and additives according to techniques known to those skilled in the art of pharmaceutical formulations.
  • a pharmaceutically acceptable salt refers to a salt form of the compounds of the present invention in order to permit their use or formulation as pharmaceuticals and which retains the biological effectiveness of the free acids and bases of the specified compound and that is not biologically or otherwise undesirable.
  • Examples of such salts are described in Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Wermuth, CG. and Stahl, P.H. (eds.), Wiley-Verlag Helvetica Acta, Zurich, 2002 [ISBN 3-906390-26-8].
  • Examples of such salts include alkali metal salts and addition salts of free acids and bases.
  • Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, xylenesulfonates, phenylacetates, phenylprop
  • a desired salt may be prepared by any suitable method known to those skilled in the art, including treatment of the free base with an inorganic acid, such as, without limitation, hydrochloric acid, hydrobromic acid, hydroiodic, carbonic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with an organic acid, including, without limitation, formic acid, acetic acid, propionic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, stearic acid, ascorbic acid, glycolic acid, salicylic acid, pyranosidyl acid, such as glucuronic acid or galacturonic acid, alpha-hydroxy acid, such as citric acid or tartaric acid, amino acid, such as aspartic acid or glutamic acid, aromatic acid, such as benzoic acid or cinnamic acid, sulfonic acid, such as p-tol
  • an inorganic acid such
  • an inventive compound is an acid
  • a desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth metal hydroxide; or the like.
  • an inorganic or organic base such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth metal hydroxide; or the like.
  • suitable salts include organic salts derived from amino acids such as glycine, lysine and arginine; ammonia; primary, secondary, and tertiary amines such as ethylenediamine, N,N'-dibenzylethylenediamine, diethanolamine, choline, and procaine, and cyclic amines, such as piperidine, morpholine, and piperazine; as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • compositions for oral administration may be, for example, solid preparations such as tablets, sugar- coated tablets, hard capsules, soft capsules, granules, powders and the like, with suitable carriers and additives being starches, sugars, binders, diluents, granulating agents, lubricants, disintegrating agents and the like. Because of their ease of use and higher patient compliance, tablets and capsules represent the most advantageous oral dosage forms for many medical conditions.
  • compositions for liquid preparations include solutions, emulsions, dispersions, suspensions, syrups, elixirs, and the like with suitable carriers and additives being water, alcohols, oils, glycols, preservatives, flavoring agents, coloring agents, suspending agents, and the like.
  • suitable carriers and additives being water, alcohols, oils, glycols, preservatives, flavoring agents, coloring agents, suspending agents, and the like.
  • Typical preparations for parenteral administration comprise the active ingredient with a carrier such as sterile water or parenterally acceptable oil including polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil, with other additives for aiding solubility or preservation may also be included.
  • a carrier such as sterile water or parenterally acceptable oil including polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil, with other additives for aiding solubility or preservation may
  • compositions according to embodiments of the present invention include those suitable for oral, rectal, topical, inhalation (e.g., via an aerosol) buccal (e.g., sub-lingual), vaginal, topical (i.e., both skin and mucosal surfaces, including airway surfaces), transdermal administration and parenteral (e.g., subcutaneous, intramuscular, intradermal, intraarticular, intrapleural, intraperitoneal, intrathecal, intracerebral, intracranially, intraarterial, or intravenous), although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular active agent which is being used.
  • compositions for injection will include the active ingredient together with suitable carriers including propylene glycol-alcohol-water, isotonic water, sterile water for injection (USP), emulPhorTM-alcohol- water, cremophor-ELTM or other suitable carriers known to those skilled in the art. These carriers may be used alone or in combination with other conventional solubilizing agents such as ethanol, propylene glycol, or other agents known to those skilled in the art. Where the macrocyclic compounds of the present invention are to be applied in the form of solutions or injections, the compounds may be used by dissolving or suspending in any conventional diluent.
  • the diluents may include, for example, physiological saline, Ringer's solution, an aqueous glucose solution, an aqueous dextrose solution, an alcohol, a fatty acid ester, glycerol, a glycol, an oil derived from plant or animal sources, a paraffin and the like. These preparations may be prepared according to any conventional method known to those skilled in the art.
  • compositions for nasal administration may be formulated as aerosols, drops, powders and gels.
  • Aerosol formulations typically comprise a solution or fine suspension of the active ingredient in a physiologically acceptable aqueous or non-aqueous solvent.
  • Such formulations are typically presented in single or multidose quantities in a sterile form in a sealed container.
  • the sealed container can be a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device such as a single use nasal inhaler, pump atomizer or an aerosol dispenser fitted with a metering valve set to deliver a therapeutically effective amount, which is intended for disposal once the contents have been completely used.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant such as a compressed gas, air as an example, or an organic propellant including a fluorochlorohydrocarbon or fluorohydrocarbon.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles, wherein the active ingredient is formulated with a carrier such as sugar and acacia, tragacanth or gelatin and glycerin.
  • a carrier such as sugar and acacia, tragacanth or gelatin and glycerin.
  • compositions for rectal administration include suppositories containing a conventional suppository base such as cocoa butter.
  • compositions suitable for transdermal administration include ointments, gels and patches.
  • compositions known to those skilled in the art can also be applied for percutaneous or subcutaneous administration, such as plasters.
  • other conventional pharmacologically acceptable additives may be incorporated, for example, excipients, stabilizers, antiseptics, wetting agents, emulsifying agents, lubricants, sweetening agents, coloring agents, flavoring agents, isotonicity agents, buffering agents, antioxidants and the like.
  • additives there may be mentioned, for example, starch, sucrose, fructose, dextrose, lactose, glucose, mannitol, sorbitol, precipitated calcium carbonate, crystalline cellulose, carboxymethylcellulose, dextrin, gelatin, acacia, EDTA, magnesium stearate, talc, hydroxypropylmethylcellulose, sodium metabisulfite, and the like.
  • the composition is provided in a unit dosage form such as a tablet or capsule.
  • the present invention provides kits including one or more containers comprising pharmaceutical dosage units comprising an effective amount of one or more compounds of the present invention.
  • the present invention further provides prodrugs comprising the compounds described herein.
  • prodrug is intended to mean a compound that is converted under physiological conditions or by solvolysis or metabolically to a specified compound that is pharmaceutically active.
  • the "prodrug” can be a compound of the present invention that has been chemically derivatized such that, (i) it retains some, all or none of the bioactivity of its parent drug compound, and (ii) it is metabolized in a subject to yield the parent drug compound.
  • the prodrug of the present invention may also be a "partial prodrug" in that the compound has been chemically derivatized such that, (i) it retains some, all or none of the bioactivity of its parent drug compound, and (ii) it is metabolized in a subject to yield a biologically active derivative of the compound.
  • Known techniques for derivatizing compounds to provide prodrugs can be employed. Such methods may utilize formation of a hydrolyzable coupling to the compound.
  • the present invention further provides that the compounds of the present invention may be administered in combination with a therapeutic agent used to prevent and/or treat metabolic and/or endocrine disorders, gastrointestinal disorders, cardiovascular disorders, obesity and obesity-associated disorders, central nervous system disorders, genetic disorders, hyperproliferative disorders and inflammatory disorders.
  • agents include analgesics (including opioid analgesics), anesthetics, antifungals, antibiotics, antiinflammatories (including nonsteroidal anti-inflammatory agents), anthelmintics, antiemetics, antihistamines, antihypertensives, antipsychotics, antiarthritics, antitussives, antivirals, cardioactive drugs, cathartics, chemotherapeutic agents (such as DNA- interactive agents, antimetabolites, tubulin-interactive agents, hormonal agents, and agents such as asparaginase or hydroxyurea), corticoids (steroids), antidepressants, depressants, diuretics, hypnotics, minerals, nutritional supplements, parasympathomimetics, hormones (such as corticotrophin releasing hormone, adrenocorticotropin, growth hormone releasing hormone, growth hormone, thyrptropin-releasing hormone and thyroid stimulating hormone), sedatives, sulfonamides, stimulants, sympathomimetics, tranquilizer
  • Subjects suitable to be treated according to the present invention include, but are not limited to, avian and mammalian subjects, and are preferably mammalian.
  • Mammals of the present invention include, but are not limited to, canines, felines, bovines, caprines, equines, ovines, porcines, rodents (e.g. rats and mice), lagomorphs, primates, humans, and the like, and mammals in utero. Any mammalian subject in need of being treated according to the present invention is suitable.
  • Human subjects are preferred. Human subjects of both genders and at any stage of development (i.e., neonate, infant, juvenile, adolescent, adult) can be treated according to the present invention.
  • Illustrative avians according to the present invention include chickens, ducks, turkeys, geese, quail, pheasant, ratites (e.g., ostrich) and domesticated birds (e.g., parrots and canaries), and birds in ovo.
  • ratites e.g., ostrich
  • domesticated birds e.g., parrots and canaries
  • the present invention is primarily concerned with the treatment of human subjects, but the invention can also be carried out on animal subjects, particularly mammalian subjects such as mice, rats, dogs, cats, livestock and horses for veterinary purposes, and for drug screening and drug development purposes.
  • animal subjects particularly mammalian subjects such as mice, rats, dogs, cats, livestock and horses for veterinary purposes, and for drug screening and drug development purposes.
  • a modulator such as an agonist, of the ghrelin receptor
  • the compounds of the present invention or an appropriate pharmaceutical composition thereof may be administered in an effective amount. Since the activity of the compounds and the degree of the therapeutic effect vary, the actual dosage administered will be determined based upon generally recognized factors such as age, condition of the subject, route of delivery and body weight of the subject.
  • the dosage can be from about 0.1 to about 100 mg/kg, administered orally 1-4 times per day.
  • compounds can be administered by injection at approximately 0.01 - 20 mg/kg per dose, with administration 1-4 times per day. Treatment could continue for weeks, months or longer. Determination of optimal dosages for a particular situation is within the capabilities of those skilled in the art. 5. Methods of Use
  • the compounds of formula I, II and/or III of the present invention can be used for the prevention and treatment of a range of medical conditions including, but not limited to, metabolic and/or endocrine disorders, gastrointestinal disorders, cardiovascular disorders, obesity and obesity-associated disorders, central nervous system disorders, genetic disorders, hyperproliferative disorders, inflammatory disorders and combinations thereof where the disorder may be the result of multiple underlying maladies.
  • the disease or disorder is irritable bowel syndrome (IBS), non-ulcer dyspepsia, Crohn's disease, gastroesophogeal reflux disorders, constipation, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, diabetes including diabetes mellitus (type II diabetes), obesity, atrophic colitis, gastritis, gastric stasis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome, celiac disease, an eating disorder or obesity.
  • the disease or disorder is congestive heart failure, ischemic heart disease or chronic heart disease.
  • the disease or disorder is osteoporosis and/or frailty, congestive heart failure, accelerating bone fracture repair, metabolic syndrome, attenuating protein catabolic response, cachexia, protein loss, impaired or risk of impaired wound healing, impaired or risk of impaired recovery from burns, impaired or risk of impaired recovery from surgery, impaired or risk of impaired muscle strength, impaired or risk of impaired mobility, alterted or risk of altered skin thickness, impaired or risk of impaired metabolic homeostasis or impaired or risk of impaired renal homeostasis.
  • the disease or disorder involves facilitating neonatal development, stimulating growth hormone release in humans, maintenance of muscle strength and function in humans, reversal or prevention of frailty in humans, prevention of catabolic side effects of glucocorticoids, treatment of osteoporosis, stimulation and increase in muscle mass and muscle strength, stimulation of the immune system, acceleration of wound healing, acceleration of bone fracture repair, treatment of renal failure or insufficiency resulting in growth retardation, treatment of short stature, treatment of obesity and growth retardation, accelerating the recovery and reducing hospitalization of burn patients, treatment of intrauterine growth retardation, treatment of skeletal dysplasia, treatment of hypercortisolism, treatment of Cushing's syndrome, induction of pulsatile growth hormone release, replacement of growth hormone in stressed patients, treatment of osteochondrodysplasias, treatment of Noonans syndrome, treatment of schizophrenia, treatment of depression, treatment of Alzheimer's disease, treatment of emesis, treatment of memory loss, treatment of reproduction disorders, treatment of delayed wound healing, treatment of
  • a method for the treatment of post-operative ileus, cachexia (wasting syndrome), such as that caused by cancer, AIDS, cardiac disease and renal disease, gastroparesis, such as that resulting from type I or type II diabetes, other gastrointestinal disorders, growth hormone deficiency, bone loss, and other age-related disorders in a human or animal patient suffering therefrom comprises administering to said patient an effective amount of at least one member selected from the compounds disclosed herein having the ability to modulate the ghrelin receptor.
  • diseases and disorders treated by the compounds disclosed herein include short bowel syndrome, gastrointestinal dumping syndrome, postgastroenterectomy syndrome, celiac disease, and hyperproliferative disorders such as tumors, cancers, and neoplastic disorders, as well as premalignant and non-neoplastic or non-malignant hyperproliferative disorders.
  • tumors, cancers, and neoplastic tissue that can be treated by the present invention include, but are not limited to, malignant disorders such as breast cancers, osteosarcomas, angiosarcomas, fibrosarcomas and other sarcomas, leukemias, lymphomas, sinus tumors, ovarian, uretal, bladder, prostate and other genitourinary cancers, colon, esophageal and stomach cancers and other gastrointestinal cancers, lung cancers, myelomas, pancreatic cancers, liver cancers, kidney cancers, endocrine cancers, skin cancers and brain or central and peripheral nervous (CNS) system tumors, malignant or benign, including gliomas and neuroblastomas.
  • malignant disorders such as breast cancers, osteosarcomas, angiosarcomas, fibrosarcomas and other sarcomas
  • leukemias lymphomas
  • sinus tumors ovarian, uretal, bladder, prostate
  • the macrocyclic compounds of the present invention can be used to treat post-operative ileus, hi other embodiments, the compounds of the present invention can be used to treat gastroparesis. In still other embodiments, the compounds of the present invention can be used to treat diabetic gastroparesis. hi another embodiment, the compounds of the present invention can be used to treat opioid-induced bowel dysfunction. In further embodiments, the compounds of the present invention can be used to treat chronic intestinal pseudoobstruction.
  • the present invention further provides methods of treating a horse or canine for a gastrointestinal disorder comprising administering a therapeutically effective amount of a modulator having the structure of formula I, II and/or III.
  • a modulator having the structure of formula I, II and/or III.
  • the gastrointestinal disorder is ileus or colic.
  • treatment is not necessarily meant to imply cure or complete abolition of the disorder or symptoms associated therewith.
  • the compounds of the present invention can further be utilized for the preparation of a medicament for the treatment of a range of medical conditions including, but not limited to, metabolic and/or endocrine disorders, gastrointestinal disorders, cardiovascular disorders, obesity and obesity-associated disorders, genetic disorders, hyperproliferative disorders and inflammatory disorders.
  • Step T9-l To a solution of 2-iodophenol (T9-0, 200 g, 0.91 mol, 1.0 eq) in DMF (DriSolv ® , 560 niL) is added sodium hydride 60% in mineral oil (3.64 g, 0.091 mol, 0.1 eq) by portions (hydrogen is seen to evolve). The reaction is heated for 1 h at 100 0 C under nitrogen, then ethylene carbonate is added and the reaction mixture heated O/N at 100 0 C. The reaction is monitored by TLC (conditions: 25/75 EtO Ac/hex; R f : 0.15, detection: UV, CMA). The reaction mixture is allowed to cool, then the solvent evaporated under reduced pressure.
  • Step T9-2 A solution of T9-1 (45.1 g, 0.171 mol, 1.0 eq) and Ddz-propargylamine (synthesized by standard protection procedures, 59.3 g, 0.214 mol, 1.25 eq) in acetonitrile (DriSolv ® , 257 mL) was degassed by passing argon through the solution for 10-15 min. To this was added Et 3 N (85.5 mL, stirred O/N with CaH 2 , then distilled) and the mixture was again purged by bubbling with argon, this time for 5 min.
  • Et 3 N 85.5 mL, stirred O/N with CaH 2 , then distilled
  • Recrystallized copper (I) iodide (1.14 g, 0.006 mol, 0.035 eq) and tra «5-dichloro-bis(triphenylphosphine) palladium (II) (Strem Chemicals, 3.6 g, 0.0051 mol, 0.03 eq) are added and the reaction mixture stirred for 4 h under argon at rt. After 5-10 min, the reaction mixture turned black. The reaction was monitored by TLC (conditions: 55/45 EtO Ac/hex). When complete, the solvent was removed under reduced pressure until dryness, then the residual oil diluted with 1 L of a 15% DCM in Et 2 O solution.
  • Step T9-3 To a solution of Ddz-amino-alcohol T9-2 (65.8 g, 0.159 mol, 1.0 eq) in
  • Tether T9 can also be synthesized from another tether molecule by reduction as in step T9-3 or with other appropriate hydrogenation catalysts known to those in the art.
  • Step Al-I To a solution of diol Al-O (50 g, 567 mmol, 1.0 eq) in CH 2 Cl 2 (1.5 L) were added Et 3 N (34.5 mL, 341 mmol, 0.6 eq) and DMAP (1.73 g, 14.2 mmol, 0.025 eq). TBDMSCl (42.8 g, 284 mmol, 0.5 eq) in CH 2 Cl 2 (100 mL) was added to this mixture at rt over 4 h with a syringe pump.
  • Step Al -2 To a solution of alcohol Al-I (26.5 g, 131 mmol, 1.0 eq) in THF (130 mL) at O 0 C was added PPh 3 (44.7 g, 170 mmol, 1.3 eq). A freshly prepared and titrated 1.3 M solution OfHN 3 (149 mL, 157 mmol, 1.5 eq) was added slowly to this mixture, then DIAD
  • Step Al -4 For the next transformation, THF was evaporated under reduced pressure from the above reaction mixture and the remaining aqueous phase extracted with Et 2 O (5 x 150 mL) and CHCl 3 (3 x 150 mL). The organic phases were monitored by TLC and if any Al -3 was observed, the organic phase was then extracted with 2 N HCl. The aqueous phase was neutralized cautiously to pH 8 with IO N NaOH. CH 3 CN (400 mL) was added to this aqueous solution and Fmoc-OSu (41.9 g, 124 mmol, 0.95 eq) in CH 3 CN (400 mL) added slowly over 50 min. The solution was stirred at rt O/N.
  • the aqueous phase was extracted with Et 2 O, then the combined organic phase dried over MgSO 4 and concentrated under reduced pressure.
  • the solid residue obtained was mixed with H 2 O (120 mL), stirred 30 min, filtered (to remove succinimide byproduct) and dried O/N under vacuum (oil pump).
  • Step Bl-I To 2-bromobenzyl alcohol (Bl-O, 30 g, 160 mmol) in DCM (DriSolv ® , 530 mL) as an approximately 0.3 M solution, was added dihydropyran (Bl-A, 22 mL, 241 mmol). Pyridinium /?-toluenesulfonate (PPTS, 4.0 g, 16 mmol) was added and the reaction mixture stirred vigorously at rt O/N. A saturated solution of Na 2 CO 3 (aq, 200 mL) was then added and the mixture stirred for 30 min.
  • PPTS Pyridinium /?-toluenesulfonate
  • Step B 1-2 Magnesium turnings (2.21 g, 90 mmol) were added to an approximately 0.8 M solution of Bl-I (from which several portions of toluene were evaporated to remove traces of water, 22.14 g, 81.8 mmol) in anhydrous THF (distilled from sodium benzopheneone ketyl, 100 mL) under an atmosphere of nitrogen.
  • the reaction was initiated by adding iodine chips (50 mg, 0.002 equiv).
  • the reaction mixture was heated to reflux for 2 h, during which time most of the Mg turnings disappeared.
  • the reaction was allowed to cool to rt.
  • Step B 1-3 2-(2-Propenyl)benzyl alcohol (T B1 ).
  • the crude THP ether Bl-2 (18.54 g, 80 mmol) was dissolved in MeOH (160 mL) andp-toluenesulfonic acid monohydrate (PTSA, 1.52 g, 8 mmol) added.
  • PTSA p-toluenesulfonic acid monohydrate
  • the resulting mixture was stirred at rt O/N, then concentrated under reduced pressure and the residue diluted with Et 2 O (100 mL).
  • the organic layer was sequentially washed with 5% NaHCO 3 (aq) solution (3 x 50 mL) and brine (1 x 50 mL), then dried over MgSO 4 .
  • Step B2-2 To a solution of alcohol B2-1 (2.0 g, 16.7 mmol, 1.0 eq) in DMF at O 0 C was added cesium carbonate (1.1 g, 3.34 mmol, 0.2 eq) and the mixture stirred at O 0 C for 15 min.
  • Step B4-2 The combined filtrate and washes were evaporated under reduced pressure to give B4-2.
  • a 1 H NMR was taken to check the purity of the resulting compound, which typically contained small amounts of impurities. However, this was sufficiently pure for use in the next step, which was preferably performed on the same day as this step since the aldehyde product (B4-2) had limited stability.
  • Step B4-3 To a suspension of MePPh 3 Br (30.2 g, 84.5 mmol, 2.2 eq) in THF (200 mL) was added t-BuOK in portions (9.5 g, 84.5 mmol, 2.2 eq) and the resulting mixture stirred at rt for 2 h during which time the solution became yellow.
  • Step T76-1 3-Bromo-2-hydroxy-benzaldehyde.
  • 2-bromophenol (76-0, 3.5 g, 20 mmol) and paraformaldehyde (8.1 g, 270 mmol) in 100 mL of dry acetonitrile at room temperature was treated with MgCl 2 (2.85 g, 30 mmol) and triethylamine (TEA, 10.45 ml, 75 mmol).
  • TEA triethylamine
  • Step 76-2 2-Bromo-6-vinyl-phenol.
  • CH 3 PPh 3 Br 72 g, 0.033 mol
  • tBuOK 4.1g, 0.03 mol
  • Step 76-6 (8-Bromo-2J3-chromen-2-yl)-methanol.
  • 76-5 5.5 g, 0.023 mol
  • MeOH 150 mL
  • sodium metal in a catalytic amount under an argon atmosphere.
  • the solution was then stirred at room temperature for 60 min.
  • the resin was removed by filtration and the filtrate evaporated in vacuo. Pure compound 76-6 was recovered as a colorless oil (4.5 g, 98%).
  • Step 76-7 76-6 (4.5 g, 18 mmol) and Ddz-propargyl amine (76-B, 15.16 g, 55.8 mmol) were dissolved in dioxane (150 mL) and diisopropylamine (27 mL). The reaction mixture was degassed by bubbling argon through the solution. PdCl 2 (PhCN) 2 (430 mg, 1.11 mmol, 0.06 eq), CuI (220 mg, 1.11 mmol, 0.06 eq) and tributylphosphine (10% in hexane, 4.4 niL, 2.23 mmol) were added and the mixture was warmed to 70 0 C and stirred O/N. The solvent was removed under high vacuum and the residue purified by flash column chromatography to obtain 76-7 as a pale brown liquid (3.2 g, 80%).
  • Step 76-8 The acetylene 76-7 (4.5 g, 0.2 mol) was dissolved in EtOH (150 mL), then purged with nitrogen for 10 min. PtO 2 (10 mol%, 450 mg) was added, and the mixture purged with a balloon full of hydrogen gas. The mixture was then charged into a Parr bomb, flushed with hydrogen (simply fill with hydrogen at 60 psi, then release and refill, repeat this fill — release — refill cycle 3x), and reacted with hydrogen at 60 psi at room temperature O/N.
  • Step T77-1 3-Bromo-pyridin-2-ol.
  • Step T77-3 The protected alcohol 77-2 (3 g, 9.1 mmol) was dissolved in diisopropylamine (50 mL) and the reaction mixture degassed by bubbling argon through the solution. PdCl 2 (PPh 3 ) 2 (410 mg, 0.61 nimol, 0.06 eq), CuI (74 mg, 0.4 mmol, 0.04 eq) and triphenylphosphine (310 mg, 1.12 mmol) were added, then the mixture was warmed to 70 0 C and stirred O/N. The solvent was removed under high vacuum and the residue was purified by flash chromatography to obtain 77-3 as a pale brown liquid (3.36 g, 70%) [Org. Lett.
  • Step T77-4 The acetylene 77-3 (3 g, 5.67 mmol) was dissolved in EtOH (30 mL) and purged with nitrogen for 10 min. PtO 2 (10 mol%, 300 mg) was added and the mixture purged with a balloon full of hydrogen gas. The mixture was then charged into a Parr bomb, flushed with hydrogen (fill with hydrogen at 80 psi then release and refill, repeat this fill — release — refill cycle 3x), and maintained with hydrogen at 80 psi at room temperature O/N.
  • the reaction mixture was filtered through a pad of Celite (use methanol for washing the residue on the Celite) and the filtrate plus washings was concentrated under reduced pressure to afford a practically pure (clean 1 H NMR), but colored sample of 77-4 in a quantitative yield. Further purification was achieved by subjecting this material to flash chromatography.
  • the product 77-4 has the same R f as the starting material (77-3), hence, 1 H NMR is the best way to distinguish them.
  • Step T77-5 77-4 (3 g, 5.6 mmol) was dissolved in anhydrous THF (200 mL). To the clear solution was added TBAF (6.7 mmol, 7 mL) and the mixture stirred for 2 h at room temperature. The solution was then poured into ice water. The aqueous solution was extracted with dichloromethane (3 x 200 mL). The organic layer was washed sequentially with saturated citrate buffer (1 x 200 mL), water (200 mL) and brine (200 mL). The washed organic extract was dried over anhydrous sodium sulfate, filtered and evaporated to dryness under reduced pressure to give an oily residue.
  • HPLC data In the HPLC procedure, ELSD and CLND data (not listed) were also procured to further assess purity of the final products, and for quantification (CLND). All compounds were analyzed using the same HPLC conditions. The details for the HPLC procedure used was as follows: Column: XTerra MS C18 4.6 x 50 mm, 3.5 /xm, from Waters, HPLC: Alliance 2695 from Waters; MS: Platform LC from Micromass/Waters; CLND: 8060 from Antek; PDA: 996 from Waters; Gradient_B4: (i) 0 to 50% MeOH : 0.1% aqueous TFA in 6 min, (ii) 3 min at 50% MeOH: 0.1% aqueous TFA; (iii) 50 to 90% MeOH : 0.1% aqueous TFA in 5 min; (iv) 3 min at 90% MeOH : 0.1% aqueous TFA. Retention time (IR) for the compound is listed. Modifications were made to the standard methods for compounds 58
  • LS1-10 a. NBS, PPh 3 , DCM (91%); b. H-Nva-OMe, Na 2 CO 3 , DMF 11O 0 C (85%); LS1-7 c. (BoC) 2 O, Na 2 CO 3 , THF:H 2 O; d. LiOH, THF:H 2 O (83% 2 steps)
  • Step LSl-Bl Synthesis of LSl-IO
  • H-Nva-OMe was recovered in 90% yield. It is important to perform the alkylation with the free amine (H-Nva-OMe) to eliminate chloride formation (OTs to Cl) as a side reaction, hi a dried round-bottomed flask, bromide LSl-8a (740 mg, 1.83 mmol, 1.0 eq) and H-Nva-OMe (479 mg, 3.60 mmol, 2.0 eq) were added.
  • Step LS1-B2 Alternative synthesis of LSl-IO
  • Step LS1-C2 Divergent Synthetic Route (no amine protection)
  • H-Nva-OtBu ⁇ Cl was dissolved in an aqueous solution Of Na 2 CO 3 (1 M) and saturated with NaCl to ensure extraction of all of the free amine.
  • This aqueous solution was extracted with AcOEt (3x).
  • the combined organic phases were extracted with brine, dried over MgSO 4 , flitered and concentrated under reduced pressure. About 90% of the free amine, H-Nva-OtBu, was recovered. It is important to perform the alkylation with the free amine (H-Nva-OtBu) to eliminate chloride side product formation (OTs -> Cl).
  • Step LSl-D Synthesis of dipeptide LS1-6
  • the tosylate salt of H-(D)Phe-OBn was dissolved in an aqueous solution of 1 M Na 2 CO 3 and the aqueous solution extracted with AcOEt (3x). The combined organic phases were extracted with brine, dried over MgSO 4 , filtered and concentrated under reduced pressure. The free amine H-(D)Phe-OBn was recovered in 90% yield.
  • the dipeptide was dissolved in a solution of HCl/dioxane (4 M, 30 mL, 10 eq), 50 mL of dioxane were then added to facilitate the agitation and the mixture stirred for 1 h at room temperature; a heterogeneous solution was obtained. The mixture was concentrated under reduced pressure and dried further on mechanical vacuum pump. The dipeptide hydrochloride salt LS1-6 was obtained as pale yellow solid (4.4 g, 100%).
  • Step LSl-E Synthesis of amino acid LS1-5
  • the organic phase was dried over MgSO 4 , filtered and concentrated under reduced pressure.
  • the residue was purified by flash chromatography (gradient: 20% AcOEt, 80% hexanes to 30% AcOEt, 70% hexanes) to give the desired fully protected tripeptide as a pale yellow gummy foam (1.6 g, 73%).
  • Step LSl-F Macrocvchzation and final deprotection
  • Step LS2-A Synthesis of dipeptide LS2-21
  • Dipeptide LS2-24 (6.9 g, 0.015 mol) was dissolved in AcOEt (100 mL), then purged with nitrogen for 10 min. 10% Pd-C (690 mg) was added and the mixture purged with a balloon full of hydrogen gas. The mixture was then hydrogenated under atmospheric pressure using a H 2 balloon. After 12 h, the reaction mixture was filtered through a short pad of Celite, and the filter cake washed with AcOEt. The combined filtrate and washings were concentrated under reduced pressure to afford practically pure (clean NMR), colorless, solid compound LS2-21 (4.30 g, 90%) which was used directly in the next step without further purification.
  • Step LS2-B Synthesis of tripeptide LS2-22
  • Step LS2-C Synthesis of LS2- 23
  • LS2-9 (0.5 g, 1.32 mmol, synthesized as in Step LSl-A for the corresponding Cbz derivative) in 1.33 mL of anhydrous DMF at room temperature was treated with KI (0.12 g, 0.66 mmol) and K 2 CO 3 (0.185 g, 1.32 mmol). The mixture was stirred vigorously at 8O 0 C for 24 hours. After this period of time, this mixture was cooled to room temperature, then 20 ml of water was added and the product extracted with Et 2 O (3 x 30 mL). The combined organic layer was washed with brine (2 x 30 mL), dried over magnesium sulfate and concentrated under vacuum. The residue was purified by flash chromatography [hexanes/ethyl acetate (1:2)] to afford LS2-25 as a white solid (70%).
  • the crude compound LS2-23 was dried using vacuum pump for 1 h and used directly in the next step without further purification.
  • Step LS2-D Synthesis of LS2-26 (Macrolactamization)
  • Step LS2-E Synthesis of Compound 410
  • Step LS3-1 Synthesis of cyclopropylglycine methyl ester hydrochloride salt.
  • LS3-A 20.0 g, 174 mmol, 1.0 eq
  • MeOH 350 niL
  • acetyl chloride 185 mL, 2.6 mol, 15 eq
  • the mixture was then concentrated under vacuum, azeotroped with toluene (3x) and dried under high vacuum 16-18 h to give LS3-1 as a pale yellow solid (30.0 g, > 100% crude yield).
  • Step LS3-2 Synthesis of tether bromide.
  • NBS 12.8 g, 72.0 mmol, 1.15 eq, larger amounts of NBS lead to dibrominated side product
  • PPh 3 18.9 g, 72.0 mmol, 1.15 eq.
  • Step LS3-4 In a dried round-bottom flask, bromide LS3-2 (47.2 g, 117 mmol, 1.0 eq) and freshly prepared LS3-3 (19.1 g, 148 mmol, 1.2 eq) were added. Degassed anhydrous DMF (117 mL), anhydrous Na 2 CO 3 (14.8 g, 140 mmol, 1.2 eq) and KI (19.4 g, 117 mmol, 1.0 eq) were added and the mixture was stirred at 100 0 C under a nitrogen atmosphere for 16- 18 h. Reaction progress was monitored by LC-MS and/or TLC.
PCT/US2005/020857 2004-06-18 2005-06-13 Macrocyclic modulators of the ghrelin receptor WO2006009674A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2007516620A JP5363726B2 (ja) 2004-06-18 2005-06-13 グレリン受容体の大環状モジュレーターの使用方法
ES05785185T ES2393498T3 (es) 2004-06-18 2005-06-13 Moduladores macrocíclicos del receptor de ghrelina
CN2005800280726A CN101111512B (zh) 2004-06-18 2005-06-13 生长素释放肽受体的大环调节剂
CA2579726A CA2579726C (en) 2004-06-18 2005-06-13 Macrocyclic modulators of the ghrelin receptor
EP05785185A EP1773869B9 (en) 2004-06-18 2005-06-13 Macrocyclic modulators of the ghrelin receptor

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US10/872,142 2004-06-18
US10/872,142 US7521420B2 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor
US62164204P 2004-10-26 2004-10-26
US60/621,642 2004-10-26
US62200504P 2004-10-27 2004-10-27
US60/622,005 2004-10-27
US60/622,055 2004-10-27
US64227105P 2005-01-07 2005-01-07
US60/642,271 2005-01-07
US11/149,731 2005-06-10
US11/149,731 US7476653B2 (en) 2003-06-18 2005-06-10 Macrocyclic modulators of the ghrelin receptor

Publications (2)

Publication Number Publication Date
WO2006009674A1 true WO2006009674A1 (en) 2006-01-26
WO2006009674A8 WO2006009674A8 (en) 2009-11-26

Family

ID=35427667

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/020857 WO2006009674A1 (en) 2004-06-18 2005-06-13 Macrocyclic modulators of the ghrelin receptor

Country Status (7)

Country Link
US (2) US7476653B2 (US20090221689A1-20090903-C00050.png)
EP (3) EP2457893B1 (US20090221689A1-20090903-C00050.png)
JP (1) JP5363726B2 (US20090221689A1-20090903-C00050.png)
CN (1) CN101111512B (US20090221689A1-20090903-C00050.png)
CA (1) CA2579726C (US20090221689A1-20090903-C00050.png)
ES (1) ES2393498T3 (US20090221689A1-20090903-C00050.png)
WO (1) WO2006009674A1 (US20090221689A1-20090903-C00050.png)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006137974A2 (en) * 2005-06-13 2006-12-28 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2008033328A2 (en) 2006-09-11 2008-03-20 Tranzyme Pharma, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
WO2008130464A1 (en) * 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
WO2010084050A2 (en) 2009-01-13 2010-07-29 Novartis Ag Quinazolinone derivatives useful as vanilloid antagonists
WO2011053821A1 (en) 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2011092290A1 (en) 2010-02-01 2011-08-04 Novartis Ag Pyrazolo[5,1b]oxazole derivatives as crf-1 receptor antagonists
WO2011092293A2 (en) 2010-02-01 2011-08-04 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
WO2011095450A1 (en) 2010-02-02 2011-08-11 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
US20110237785A1 (en) * 2003-07-31 2011-09-29 Marsault Eric Spatially-defined macrocyclic compounds useful for drug discovery
EP2491020A1 (en) * 2009-10-23 2012-08-29 Tranzyme Pharma, Inc. Methods of using macrocyclic inhibitors of serine protease enzymes
JP2013040206A (ja) * 2007-12-21 2013-02-28 Helsinn Therapeutics (Us) Inc 消化器系の運動をイパモレリンを用いて刺激する方法
US8921521B2 (en) 2003-06-18 2014-12-30 Ocera Therapeutics, Inc. Macrocyclic modulators of the Ghrelin receptor
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
US10030047B2 (en) 2015-10-27 2018-07-24 Hoffmann-La Roche Inc. Peptide macrocycles against acinetobacter baumannii
US10092621B2 (en) 2014-11-12 2018-10-09 Lyric Pharmaceuticals Inc. Treatment of enteral feeding intolerance
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
US11066443B2 (en) 2017-04-10 2021-07-20 Hoffmann-La Roche Inc. 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

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7476653B2 (en) * 2003-06-18 2009-01-13 Tranzyme Pharma, Inc. Macrocyclic modulators of the ghrelin receptor
US20090198050A1 (en) * 2003-06-18 2009-08-06 Tranzyme Pharma Inc. Macrocyclic Modulators of the Ghrelin Receptor
JP4928261B2 (ja) * 2003-06-18 2012-05-09 トランザイム・ファーマ・インコーポレイテッド モチリン受容体の大環状拮抗薬
US7491695B2 (en) 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
US7550431B2 (en) * 2003-07-31 2009-06-23 Tranzyme Pharma Inc. Spatially-defined macrocycles incorporating peptide bond surrogates
JP5739766B2 (ja) * 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 グレリン受容体の大環状モジュレーターの使用方法
JP5730835B2 (ja) * 2004-06-18 2015-06-10 オセラ セラピューティクス, インコーポレイテッド グレリン受容体の大環状モジュレーターの使用方法
US20090275648A1 (en) * 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
HUE044391T2 (hu) 2005-09-29 2019-10-28 Ipsen Pharma Készítmény gasztrointesztinális motilitászavar kezelésére
CU23558A1 (es) 2006-02-28 2010-07-20 Ct Ingenieria Genetica Biotech Compuestos análogos a los secretagogos peptidicos de la hormona de crecimiento
WO2007106385A2 (en) * 2006-03-10 2007-09-20 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Use of a ghrelin agonist to improve the catabolic effects of glucocorticoid treatment
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
PT2118123E (pt) 2007-01-31 2016-02-10 Harvard College Péptidos de p53 estabilizados e suas utilizações
US20100216706A1 (en) * 2007-05-15 2010-08-26 Horvath Tamas L Ghrelin Protects Substantia Nigra Dopamine Neurons
US9724381B2 (en) 2009-05-12 2017-08-08 The Administrators Of The Tulane Educational Fund Methods of inhibiting the ghrelin/growth hormone secretatogue receptor pathway and uses thereof
EP2483407A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-enhancing gene products
MX2012003912A (es) * 2009-09-30 2012-08-17 Tranzyme Pharma Inc Sales, solvatos y composiciones farmaceuticas de los agonistas macrociclicos del receptor de ghrelina y metodos para su uso.
WO2011146845A1 (en) * 2010-05-21 2011-11-24 Tranzyme Pharma, Inc. Modified macrocyclic ghrelin receptor modulators and methods of using the same
EP2582715B1 (en) * 2010-06-16 2018-11-28 The Administrators of the Tulane Educational Fund Growth hormone secretatogue receptor antagonists and uses thereof
WO2012016186A1 (en) * 2010-07-29 2012-02-02 President And Fellows Of Harvard College Macrocyclic kinase inhibitors and uses thereof
WO2013006451A2 (en) 2011-07-01 2013-01-10 President And Fellows Of Harvard College Macrocyclic insulin-degrading enzyme (ide) inhibitors and uses thereof
CN102557989B (zh) * 2011-12-19 2013-12-25 深圳翰宇药业股份有限公司 优利莫瑞林中间体以及优利莫瑞林制备方法
US10039813B2 (en) 2012-02-07 2018-08-07 Massachusetts Institute Of Technology Use of antagonists of ghrelin or ghrelin receptor to prevent or treat stress-sensitive psychiatric illness
CN105072909A (zh) * 2012-09-27 2015-11-18 阿莱塔纳治疗学股份有限公司 应用控制食欲不振的化合物的组合物和方法
RU2655811C2 (ru) 2012-10-24 2018-05-29 Дайити Санкио Компани, Лимитед Терапевтическое средство для бокового амиотрофического склероза
US9724396B2 (en) 2013-03-15 2017-08-08 Massachusetts Institute Of Technology Use of antagonists of growth hormone or growth hormone receptor to prevent or treat stress-sensitive psychiatric illness
US9119832B2 (en) 2014-02-05 2015-09-01 The Regents Of The University Of California Methods of treating mild brain injury
CN103864588B (zh) * 2014-03-25 2015-09-09 河北工业大学 一种2,3-二甲氧基苯甲醛的制备方法
ES2902795T3 (es) 2015-01-28 2022-03-29 Elanco Animal Health Incorporated Composición para uso crónico como compuesto para aumentar de peso
US10317418B2 (en) 2015-02-24 2019-06-11 Massachusetts Institute Of Technology Use of ghrelin or functional ghrelin receptor agonists to prevent and treat stress-sensitive psychiatric illness
US11040976B2 (en) 2015-04-24 2021-06-22 President And Fellows Of Harvard College Substrate selective inhibitors of insulin-degrading enzyme (IDE) and uses thereof
WO2017075535A1 (en) 2015-10-28 2017-05-04 Oxeia Biopharmaceuticals, Inc. Methods of treating neurodegenerative conditions
CN107759660A (zh) * 2017-12-05 2018-03-06 陕西慧康生物科技有限责任公司 一种三肽‑29的液‑固相合成方法
US11674136B2 (en) 2018-02-09 2023-06-13 President And Fellows Of Harvard College DNA-templated macrocycle library

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025257A2 (en) * 1999-10-04 2001-04-12 Neokimia, Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
WO2001092292A2 (en) * 2000-05-30 2001-12-06 Merck & Co., Inc. Ghrelin analogs
WO2004111077A1 (en) * 2003-06-18 2004-12-23 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor
WO2005012332A1 (en) * 2003-07-31 2005-02-10 Tranzyme Pharma Spatially-defined macrocycles incorporating peptide bond surrogates
WO2005012331A1 (en) * 2003-07-31 2005-02-10 Tranzyme Pharma Spatially-defined macrocyclic compounds useful for drug discovery

Family Cites Families (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3239345A (en) 1965-02-15 1966-03-08 Estrogenic compounds and animal growth promoters
US4411890A (en) 1981-04-14 1983-10-25 Beckman Instruments, Inc. Synthetic peptides having pituitary growth hormone releasing activity
US4036979A (en) 1974-01-25 1977-07-19 American Cyanamid Company Compositions containing 4,5,6,7-tetrahydrobenz[b]thien-4-yl-ureas or derivatives and methods of enhancing growth rate
NZ188418A (en) 1977-10-03 1981-02-11 Mobil Oil Corp Lubricants containing oil and an oxazoline
WO1989007110A1 (en) 1988-01-28 1989-08-10 Eastman Kodak Company Polypeptide compounds having growth hormone releasing activity
AU628322B2 (en) 1988-01-28 1992-09-17 Eastman Kodak Company Polypeptide compounds having growth hormone releasing activity
DK0554381T3 (da) 1990-10-25 1996-04-09 Genentech Inc Anvendelse af beskyttende midler mod reaktive oxygenforbindelser
US5663146A (en) 1991-08-22 1997-09-02 Administrators Of The Tulane Educational Fund Polypeptide analogues having growth hormone releasing activity
US5726319A (en) 1992-11-06 1998-03-10 Merck & Co., Inc. Biphenyl substituted dipeptide analogs promote release of growth hormone
WO1994011012A1 (en) 1992-11-06 1994-05-26 Merck & Co., Inc. Substituted dipeptide analogs promote release of growth hormone
US5536716A (en) 1992-12-11 1996-07-16 Merck & Co., Inc. Spiro piperidines and homologs which promote release of growth hormone
CZ151495A3 (en) 1992-12-11 1995-12-13 Merck & Co Inc Spiropiperidine derivatives, process of their preparation and a pharmaceutical composition containing thereof
PL314003A1 (en) 1993-10-19 1996-08-05 Merck & Co Inc Combination of bisulphonates and substances enhancing secretion of growth hormone
SK56296A3 (en) 1993-11-09 1997-02-05 Merck & Co Inc Piperidinal, pyrrolidinal and hexahydro-1h-azepinal derivatives, manufacturing process thereof and pharmaceutical compositions containing them
US5494919A (en) 1993-11-09 1996-02-27 Merck & Co., Inc. 2-substituted piperidines, pyrrolidines and hexahydro-1H-azepines promote release of growth hormone
US5492916A (en) 1993-12-23 1996-02-20 Merck & Co., Inc. Di- and tri-substituted piperidines, pyrrolidines and hexahydro-1H-azepines promote release of growth hormone
EP0730578A4 (en) 1993-11-24 1997-10-08 Merck & Co Inc COMPOUNDS CONTAINING AN INDOLYL GROUP AND THEIR USE FOR PROMOTING THE RELEASE OF GROWTH HORMONES
US5721251A (en) 1993-12-10 1998-02-24 Merck & Co., Inc. Piperidine, pyrrolidine and hexahydro-1H-azepines promote release of growth hormone
EP0736038B1 (en) 1993-12-23 2001-07-04 Novo Nordisk A/S Compounds with growth hormone releasing properties
EP0736039B1 (en) 1993-12-23 2000-10-25 Novo Nordisk A/S Compounds with growth hormone releasing properties
US5721250A (en) 1993-12-23 1998-02-24 Merck & Co. Inc. Di-and tri-substituted piperidines, pyrrolidines and hexahydro-1H-azepines promote release of growth hormone
WO1995034311A1 (en) 1994-06-13 1995-12-21 Merck & Co., Inc. Piperazine compounds promote release of growth hormone
WO1996002530A1 (en) 1994-07-20 1996-02-01 Merck & Co., Inc. Piperidines and hexahydro-1h-azepines spiro substituted at the 4-position promote release of growth hormone
US5798337A (en) 1994-11-16 1998-08-25 Genentech, Inc. Low molecular weight peptidomimetic growth hormone secretagogues
AU4495996A (en) 1995-01-25 1996-08-14 Nippon Chemiphar Co. Ltd. Dihydrophenazine derivatives
UA55381C2 (uk) 1995-01-27 2003-04-15 Ново Нордіск А/С Похідні пептиду (варіанти), що здатні вивільнювати гормон росту, фармацевтична композиція (варіанти), спосіб стимулювання вивільнення гормону росту з гіпофіза, способи підвищення швидкості і темпу росту та виробництва молока або вовни, спосіб лікування нездужань
WO1996024587A1 (en) 1995-02-09 1996-08-15 Novo Nordisk A/S Compounds with growth hormone releasing properties
WO1996024580A1 (en) 1995-02-09 1996-08-15 Novo Nordisk A/S Compounds with growth hormone releasing properties
US5559128A (en) 1995-04-18 1996-09-24 Merck & Co., Inc. 3-substituted piperidines promote release of growth hormone
WO1996033189A1 (en) 1995-04-19 1996-10-24 Merck & Co., Inc. Process for the preparation of spiroindolines
WO1996035713A1 (en) 1995-05-08 1996-11-14 Pfizer, Inc. Dipeptides which promote release of growth hormone
US5936089A (en) 1995-05-29 1999-08-10 Pfizer Inc Dipeptides which promote release of growth hormone
BR9608909A (pt) 1995-06-22 1999-03-02 Novo Nordisk As Composto composição farmacêutica processos para estimular a liberação do hormônio do crescimento da patuitária e aumentar a velocidade e extensão do crescimento dos animais para aumentar a produção de leite ou lã de animais ou para para doenças e uso do composto
ZA966891B (en) 1995-08-21 1998-02-16 Lilly Co Eli 2-acylaminopropanamines as growth hormone secretagogues.
WO1997006803A1 (en) 1995-08-21 1997-02-27 Eli Lilly And Company 2-acylaminopropanamides as growth hormone secretagogues
EP0766966A3 (en) 1995-09-08 2001-02-28 Eli Lilly And Company Method of treating insulin resistance
WO1997011697A1 (en) 1995-09-26 1997-04-03 Merck & Co., Inc. 3-spirolactam, 3-spiroamino, 3-spirolactone and 3-spirobenzopyran piperidines and pyrrolidines promote release of growth hormone
US5723616A (en) 1995-10-27 1998-03-03 Merck & Co., Inc. Process for the preparation of a growth hormone secretagogue
WO1997015191A1 (en) 1995-10-27 1997-05-01 Merck & Co., Inc. Wet granulation formulation of a growth hormone secretagogue
HUP9902095A3 (en) 1995-10-27 2001-05-28 Merck & Co Inc New process for the preparation of a growth hormone secretagogue
US5767124A (en) 1995-10-27 1998-06-16 Merck & Co., Inc. Polymorphic forms of a growth hormone secretagogue
DE69636394T2 (de) 1995-12-13 2007-07-19 Merck & Co., Inc. Testverfahren für die sekretionsrezeptoren von wachstumshormonen
CA2240427C (en) 1995-12-13 2007-08-14 Merck & Co., Inc. Growth hormone secretagogue receptor family
WO1997022367A1 (en) 1995-12-20 1997-06-26 Merck & Co., Inc. Radiolabeled growth hormone secretagogue
IT1277113B1 (it) 1995-12-20 1997-11-04 Romano Deghenghi Composti oligopeptidici contenenti d-2-alchiltriptofano in grado di promuovere la liberazione dell'ormone della crescita
AU715856B2 (en) 1995-12-22 2000-02-10 Novo Nordisk A/S Compounds with growth hormone releasing properties
TW432073B (en) 1995-12-28 2001-05-01 Pfizer Pyrazolopyridine compounds
WO1997034604A1 (en) 1996-03-21 1997-09-25 Merck & Co., Inc. 4-spiroindoline piperidines promote release of growth hormone
AU2722297A (en) 1996-04-03 1997-10-22 Merck & Co., Inc. Piperidines, pyrrolidines and hexahydro-1h-azepines promote release of growth hormone
SE9601397D0 (sv) 1996-04-12 1996-04-12 Pharmacia Ab Use of growth hormone
AU2382097A (en) 1996-04-19 1997-11-12 Novo Nordisk A/S Compounds with growth hormone releasing properties
AU722421B2 (en) 1996-04-24 2000-08-03 Novo Nordisk A/S Compounds with growth hormone releasing properties
US5919777A (en) 1996-04-24 1999-07-06 Novo Nordisk A/S Compounds with growth hormone releasing properties
WO1997041878A1 (en) 1996-05-07 1997-11-13 Merck & Co., Inc. Treatment of mood disorders with a growth hormone secretagogue
EP0900086A4 (en) 1996-05-07 2000-01-12 Merck & Co Inc IMPROVING THE SLEEP BY MEANS OF A SUBSTANCE PROMOTING THE SECRETION OF GROWTH HORMONES
AU2764797A (en) 1996-05-14 1997-12-05 Novo Nordisk A/S Somatostatin agonists and antagonists
AU3220097A (en) 1996-05-22 1997-12-09 Arch Development Corporation Sleep quality improvement using a growth hormone secretagogue
IL126983A0 (en) 1996-05-31 1999-09-22 Novo Nordisk As Growth hormone components and bone anti-resorptive agent in cyclic (coherence) treatment of osteoporosis
WO1998003473A1 (en) 1996-07-22 1998-01-29 Novo Nordisk A/S Compounds with growth hormone releasing properties
AU4342097A (en) 1996-09-13 1998-04-02 Merck & Co., Inc. Piperidines, pyrrolidines and hexahydro-1h-azepines promote release of growth hormone
WO1998018815A1 (en) 1996-10-25 1998-05-07 Merck & Co., Inc. Convergent process for the preparation of a growth hormone secretagogue
SE9703929D0 (sv) 1996-11-22 1997-10-28 Pharmacia & Upjohn Ab Therapeutical use and method
WO1998046569A1 (fr) 1997-04-11 1998-10-22 Sumitomo Pharmaceuticals Co., Ltd. Derives de benzene
JP2000514838A (ja) 1997-05-14 2000-11-07 藤沢薬品工業株式会社 成長ホルモンの放出を促進するピペリジノ誘導体
WO1998058950A1 (en) 1997-06-20 1998-12-30 Novo Nordisk A/S Compounds with growth hormone releasing properties
UA64751C2 (uk) 1997-06-25 2004-03-15 Пфайзер Продактс Інк. Спосіб лікування інсулінової толерантності речовинами, які посилюють секрецію гормону росту (варіанти) та фармацевтична композиція (варіанти)
US6251902B1 (en) 1997-06-25 2001-06-26 Pfizer Inc. Dipeptide derivatives as growth hormone secretagogues
UA53716C2 (uk) 1997-06-25 2003-02-17 Пфайзер Продактс Інк. Тартратна сіль заміщеного дипептиду, спосіб її одержання, проміжні сполуки та спосіб їх одержання, фармацевтична композиція (варіанти), спосіб підвищення рівнів ендогенного гормону росту та спосіб лікування або профілактики захворювань (варіанти)
ZA987383B (en) 1997-08-19 2000-02-17 Lilly Co Eli Treatment of congestive heart failure with growth hormone secretagogues.
US6864250B1 (en) 1997-08-22 2005-03-08 Kaken Pharmaceutical Co., Ltd. N-acylated lipophilic amino acid derivatives
US6737407B1 (en) 1997-09-08 2004-05-18 Metabolic Pharmaceuticals, Ltd. Treatment of obesity
IL137007A0 (en) 1998-01-16 2001-06-14 Novo Nordisk As Compounds with growth hormone releasing properties
WO1999039730A1 (fr) 1998-02-09 1999-08-12 Kaken Pharmaceutical Co., Ltd. Preparation a administration orale contenant des peptides favorisant la secretion d'hormone de croissance
HUP0100998A3 (en) 1998-03-03 2001-12-28 Novo Nordisk As (2e)-5-amino-5-methylhex-2-enoic acid n-methyl-n-((1r)-1-(n-methyl-n-((1r)-1-(methylcarbamoyl)-2-phenylethyl)carbamoyl)-2-(2-naphtyl)ethyl)amide salts, methods for their preparation, pharmaceutical compositions comprising thereof and their use
MA26618A1 (fr) * 1998-04-09 2004-12-20 Smithkline Beecham Corp Composes et compositions pharmaceutiques pour le traitement du paludisme
IL139090A0 (en) 1998-05-11 2001-11-25 Novo Nordisk As Compounds with growth hormone releasing properties
WO1999064456A1 (en) 1998-06-09 1999-12-16 Novo Nordisk A/S A method for preparing a compound with growth hormone releasing properties
PA8475901A1 (es) 1998-06-16 2000-05-24 Pfizer Prod Inc Terapia de combinacion para la fragilidad musculoesqueletica
AU3342099A (en) 1998-06-16 2000-01-05 Pfizer Products Inc. Therapeutic combinations of (selective) estrogen receptor modulators (serm) and growth hormone secretagogues (ghs) for treating musculoskeletal frailty
KR100907638B1 (ko) 1998-06-30 2009-07-14 노보 노르디스크 에이/에스 성장 호르몬 방출성을 가지는 화합물
DE19831217A1 (de) * 1998-07-03 2000-01-05 Schering Ag Neue Porphyrinderivate, diese enthaltende pharmazeutische Mittel und ihre Verwendung in der photodynamischen Therapie und MRI-Diagnostik
EP1112282A4 (en) 1998-08-10 2002-10-31 Merck & Co Inc RECEPTOR OF THE GROWTH HORMONE SECRETION PROMOTER FROM THE DOG
ATE288895T1 (de) 1998-08-20 2005-02-15 Sumitomo Pharma Wachstumshormon-freisetzende oxindolderivate
US6358951B1 (en) 1998-08-21 2002-03-19 Pfizer Inc. Growth hormone secretagogues
CA2343214A1 (en) * 1998-09-03 2000-03-16 Chris Edward Williams Neuroprotection
JP2003503055A (ja) 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ペプチド
WO2001047558A1 (fr) 1999-12-28 2001-07-05 Kaken Pharmaceutical Co., Ltd. Medicaments protegeant les nerfs
CN1935833A (zh) * 2000-04-19 2007-03-28 先灵公司 含有烷基和芳基丙氨酸p2部分的丙型肝炎病毒的大环ns3-丝氨酸蛋白酶抑制剂
DE60140285D1 (de) * 2000-05-31 2009-12-10 Pfizer Prod Inc Verwendung von Wachstumshormonsekretagoga zur Förderung der Beweglichkeit des Verdauungstrakts
HU229233B1 (en) 2000-06-13 2013-09-30 Aeterna Zentaris Gmbh Growth hormone secretagogues
WO2001097831A1 (fr) 2000-06-23 2001-12-27 Kaken Pharmaceutical Co., Ltd. Agents therapeutiques et prophylactiques destines a l'insuffisance cardiaque
US6576686B1 (en) 2000-06-27 2003-06-10 Exxonmobil Chemical Patents Inc. Road marking compound comprising linear tetrablock copolymers
US6635784B2 (en) * 2000-09-29 2003-10-21 Eastman Chemical Company Process for the preparation of enantiomerically-enriched cyclopropylalanine derivates
AU2002250081A1 (en) 2001-02-14 2002-08-28 Uab Research Foundation Combined transductional and transcriptional targeting system for improved gene delivery
ITMI20011445A1 (it) 2001-07-06 2003-01-06 Europ Geie Analoghi della cortistantina capaci di legarsi selettivamente ai recettori dei secretagoghi dell'ormone della crescita
JPWO2004014412A1 (ja) 2002-08-09 2005-12-02 科研製薬株式会社 心筋細胞保護剤
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
BRPI0807046A2 (pt) * 2007-02-09 2015-05-26 Tranzyme Pharma Inc Composto, composição farmacêutica, métodos de tratar um distúrbio, uma doença cardiovascular e um paciente que sofre de motilidade gastrointestinal reduzida ou disfuncional e, kit.
MX2012003912A (es) 2009-09-30 2012-08-17 Tranzyme Pharma Inc Sales, solvatos y composiciones farmaceuticas de los agonistas macrociclicos del receptor de ghrelina y metodos para su uso.

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025257A2 (en) * 1999-10-04 2001-04-12 Neokimia, Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
WO2001092292A2 (en) * 2000-05-30 2001-12-06 Merck & Co., Inc. Ghrelin analogs
WO2004111077A1 (en) * 2003-06-18 2004-12-23 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor
WO2005012332A1 (en) * 2003-07-31 2005-02-10 Tranzyme Pharma Spatially-defined macrocycles incorporating peptide bond surrogates
WO2005012331A1 (en) * 2003-07-31 2005-02-10 Tranzyme Pharma Spatially-defined macrocyclic compounds useful for drug discovery

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9493505B2 (en) 2003-06-18 2016-11-15 Ocera Therapeutics, Inc. Macrocyclic modulators of the ghrelin receptor
US8921521B2 (en) 2003-06-18 2014-12-30 Ocera Therapeutics, Inc. Macrocyclic modulators of the Ghrelin receptor
US8440851B2 (en) * 2003-07-31 2013-05-14 Tranzyme Pharma, Inc. Spatially-defined macrocyclic compounds useful for drug discovery
US20110237785A1 (en) * 2003-07-31 2011-09-29 Marsault Eric Spatially-defined macrocyclic compounds useful for drug discovery
WO2006137974A3 (en) * 2005-06-13 2007-04-12 Tranzyme Pharma Inc Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
JP2009508805A (ja) * 2005-06-13 2009-03-05 トランザイム・ファーマ,インコーポレイテッド 大環状グレリン受容体アンタゴニストおよびインバースアゴニストならびにその使用方法
WO2006137974A2 (en) * 2005-06-13 2006-12-28 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2008033328A2 (en) 2006-09-11 2008-03-20 Tranzyme Pharma, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
WO2008033328A3 (en) * 2006-09-11 2008-07-24 Tranzyme Pharma Inc Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
US9133235B2 (en) 2006-09-11 2015-09-15 Ocera Therapeutics, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
JP2010503620A (ja) * 2006-09-11 2010-02-04 トランザイム・ファーマ,インコーポレイテッド 胃腸の運動障害疾患の治療のためのモチリン受容体の大環状アンタゴニスト
EP2431380A2 (en) 2006-09-11 2012-03-21 Tranzyme Pharma, Inc. Macrocyclic antagonist of the motilin receptor for treatment of gastrointestinal dysmotility disorders
US10258602B2 (en) 2007-02-09 2019-04-16 Ocera Therapeutics, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
EP2644618A1 (en) 2007-02-09 2013-10-02 Tranzyme Pharma, Inc. tether intermediates for the synthesis of macrocyclic ghrelin receptor modulators
US9371297B2 (en) 2007-02-09 2016-06-21 Ocera Therapeutics, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
WO2008130464A1 (en) * 2007-02-09 2008-10-30 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
US9949949B2 (en) 2007-02-09 2018-04-24 Ocera Therapeutics, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
JP2010518090A (ja) * 2007-02-09 2010-05-27 トランザイム・ファーマ,インコーポレイテッド 大環状グレリン受容体修飾因子およびその使用方法
JP2014141507A (ja) * 2007-02-09 2014-08-07 Tranzyme Pharma Inc 大環状グレリン受容体修飾因子およびその使用方法
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
JP2013040206A (ja) * 2007-12-21 2013-02-28 Helsinn Therapeutics (Us) Inc 消化器系の運動をイパモレリンを用いて刺激する方法
WO2010084050A2 (en) 2009-01-13 2010-07-29 Novartis Ag Quinazolinone derivatives useful as vanilloid antagonists
EP2491004A4 (en) * 2009-10-23 2013-07-03 Tranzyme Pharma Inc MACROCYCLIC INHIBITORS OF SERINE PROTEASE CYMS
EP2491020A4 (en) * 2009-10-23 2013-04-24 Tranzyme Pharma Inc METHODS OF USING MACROCYCLIC INHIBITORS OF SERINE PROTEASE ENZYMES
EP2491004A2 (en) * 2009-10-23 2012-08-29 Tranzyme Pharma, Inc. Macrocyclic inhibitors of serine protease enzymes
EP2491020A1 (en) * 2009-10-23 2012-08-29 Tranzyme Pharma, Inc. Methods of using macrocyclic inhibitors of serine protease enzymes
WO2011053821A1 (en) 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2011092293A2 (en) 2010-02-01 2011-08-04 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
WO2011092290A1 (en) 2010-02-01 2011-08-04 Novartis Ag Pyrazolo[5,1b]oxazole derivatives as crf-1 receptor antagonists
WO2011095450A1 (en) 2010-02-02 2011-08-11 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 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
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
US10092621B2 (en) 2014-11-12 2018-10-09 Lyric Pharmaceuticals Inc. Treatment of enteral feeding intolerance
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10030047B2 (en) 2015-10-27 2018-07-24 Hoffmann-La Roche Inc. Peptide macrocycles against acinetobacter baumannii
US11098080B2 (en) 2015-10-27 2021-08-24 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
US11066443B2 (en) 2017-04-10 2021-07-20 Hoffmann-La Roche Inc. 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

Also Published As

Publication number Publication date
US20060025566A1 (en) 2006-02-02
US8334256B2 (en) 2012-12-18
JP5363726B2 (ja) 2013-12-11
CA2579726A1 (en) 2006-01-26
WO2006009674A8 (en) 2009-11-26
EP1773869B9 (en) 2013-01-23
CN101111512A (zh) 2008-01-23
ES2393498T3 (es) 2012-12-21
EP1773869B1 (en) 2012-08-22
EP2457893A1 (en) 2012-05-30
US20090221689A1 (en) 2009-09-03
EP1773869A1 (en) 2007-04-18
EP2457893B1 (en) 2017-08-16
US7476653B2 (en) 2009-01-13
CN101111512B (zh) 2013-05-01
JP2008504238A (ja) 2008-02-14
EP2457925A1 (en) 2012-05-30
CA2579726C (en) 2015-01-13

Similar Documents

Publication Publication Date Title
EP2457893B1 (en) Intermediates for macrocyclic modulators of the ghrelin receptor
US8349887B2 (en) Methods of treating cachexia with macrocyclic modulators of the ghrelin receptor
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
EP2644618B1 (en) tether intermediates for the synthesis of macrocyclic ghrelin receptor modulators
US9493505B2 (en) Macrocyclic modulators of the ghrelin receptor
US20180110824A1 (en) Macrocyclic Modulators of the Ghrelin Receptor
WO2006046977A1 (en) Macrocyclic ghrelin receptor antagonists and methods of using the same
JP5730835B2 (ja) グレリン受容体の大環状モジュレーターの使用方法
JP5739766B2 (ja) グレリン受容体の大環状モジュレーターの使用方法
AU2015201862B2 (en) Macrocyclic ghrelin receptor modulators and methods of using the same
ES2646887T3 (es) Compuestos intermedios para moduladores macrocíclicos del receptor de ghrelina

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2579726

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2007516620

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 7763/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2005785185

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200580028072.6

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005785185

Country of ref document: EP