CA2528375C - Macrocyclic antagonists of the motilin receptor - Google Patents

Macrocyclic antagonists of the motilin receptor Download PDF

Info

Publication number
CA2528375C
CA2528375C CA2528375A CA2528375A CA2528375C CA 2528375 C CA2528375 C CA 2528375C CA 2528375 A CA2528375 A CA 2528375A CA 2528375 A CA2528375 A CA 2528375A CA 2528375 C CA2528375 C CA 2528375C
Authority
CA
Canada
Prior art keywords
boc
group
hydrogen
bts
ddz
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2528375A
Other languages
French (fr)
Other versions
CA2528375A1 (en
Inventor
Sylvie Beaubien
Kamel Benakli
Graeme Fraser
Hamid Hoveyda
Eric Marsault
Mark Peterson
Robert Deziel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ocera Therapeutics Inc
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
Application filed by Tranzyme Pharma Inc filed Critical Tranzyme Pharma Inc
Publication of CA2528375A1 publication Critical patent/CA2528375A1/en
Application granted granted Critical
Publication of CA2528375C publication Critical patent/CA2528375C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

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

Abstract

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

Description

MACROCYCLIC ANTAGONISTS OF THE MOTILIN RECEPTOR
FIELD OF THE INVENTION
The present invention relates to novel conformationally-defined macrocyclic compounds, pharmaceutical compositions comprising same and intermediates used in their manufacture. More particularly, the invention relates to macrocyclic compounds that have been demonstrated to selectively antagonize the activity of the motilin receptor. The invention further relates to macrocyclic compounds useful as therapeutics for a range of gastrointestinal disorders, in particular those in which malfunction of gastric motility or increased motilin secretion is observed, such as hypermotilinemia, irritable bowel syndrome and dyspepsia.
BACKGROUND OF THE INVENTION
A number of peptide horoones are involved in the control of the different functions in the gastrointestinal (GI) tr4ct, including absorption, secretion, blood flow and motility (Mulvihill, et al. in Basic and Clinical Endocrinology, 4th edition, Greenspan, F.S.;
Baxter, J.D., eds., Appleton & Lange: Norwalk, CT, 1994, pp 551-570). Since interactions between the brain and GI system are critical to the proper modulation of these functions, these peptides can be produced locally in the GI tract or distally in the CNS.
One of these peptide hormones, motilin, a linear 22-amino acid peptide, plays a critical regulatory role in the GI physiological system though governing of fasting gastrointestinal motor activity. As such, the peptide is periodically released from the duodenal mucosa during fasting in mammals, including humans. More precisely, motilin exerts a powerful effect on gastric motility through the contraction of gastrointestinal smooth muscle to stimulate gastric emptying, decrease intestinal transit time and initiate phase III of the migrating motor complex in the small bowel (Itoh, Z., Ed., Motilin, Academic Press: San Diego, CA, 1990, ASIN: 0123757304; Nelson, D.K. Dig. Dis. Sci. 1996, 4/, 2006-2015;
Peeters, Ti.; Vantrappen, G.; Janssens, J. Gastroenterology 1980, 79, 716-719).
Motilin exerts these effects through receptors located predominantly on the human antrum and proximal duodenum, although its receptors are found in other regions of the GI tract as well (Peeters, T.L.; Bormans, V.; Vantrappen, G. ReguL Pept. 1988, 23, 171-182).
Therefore, motilin hormone is involved in motility of both the upper and lower parts of the GI system (Williams et al. Am. J. Physiol. 1992, 262, G50-G55). In addition, motilin and its receptors have been found in the CNS and periphery, suggesting a physiological role in the nervous system that has not yet been definitively elucidated (Depoortere, I.; Peeters, T.L. Am. J. PhysioL 1997, 272, G994-999 and O'Donohue, T.L et al. Peptides 1981, 2, 467-477). For example, motilin receptors in the brain have been suggested to play a regulatory role in a number of CNS functions, including feeding and drinking behavior, micturition reflex, central and brain stem neuronal modulation and pituitary hormone secretion (ltoh, Z. Motilin and Clinical Applications. Peptides 1997, 18, 593-608; Asakawa, A.; Inui, A.; Momose, K.; et al., M. Peptides 1998, /9, 987-990 and Rosenfeld, D.J.;
Garthwaite, T.L. PhysioL Behav. 1987, 39, 753-756). Physiological studies have provided confirmatory evidence that motilin can indeed have an effect on feeding behavior (Rosenfeld, D.J.; Garthwaite, T.L. Phys. Behav. 1987, 39, 735-736).
The recent identification and cloning of the human motilin receptor (WO
99/64436) has simplified and accelerated the search for agents which can modulate its activity for specific therapeutic purposes.
Due to the critical and direct involvement of motilin in control of gastric motility, agents that either diminish (hypomotility) or enhance (hypermotility) the activity at the motilin receptor, are a particularly attractive area for further investigation in the search for new effective pharmaceuticals towards these indications.
Peptidic agonists of the motilin receptor, which have clinical application for the treatment of hypomotility disorders, have been reported (U.S. 5,695,952; 5,721,353;
6,018,037;
6,380,158; 6,420,521, U.S. Appl. 2001/0041791, WO 98/42840; WO 01/00830 and WO

02/059141). Derivatives of erythromycin, commonly referred to as motilides, have also been reported as agonists of the motilin receptor (U.S. 4,920,102; 5,008,249;
5,175,150;
5,418,224; 5,470,961; 5,523,401, 5,554,605; 5,658,888; 5,854,407; 5,912,235;
6,100,239;
6,165,985; 6,403,775).
Antagonists of the motilin receptor are potentially extremely useful as therapeutic treatments for diseases associated with hypermotility and hypermotilinemia, including irritable bowel syndrome, dyspepsia, gastroesophogeal reflux disorders, Crohn's disease, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, diabetes mellitus, obesity, malabsorption syndrome, carcinoid syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome, gastric stasis and eating disorders leading to obesity.
o A variety of peptidic compounds have been described as antagonists of the motilin receptor (Depoortere, I.; Macielag, M.J.; Galdes, A.; Peeters, T.L. Eur. J.
Pharmacol. 1995, 286, 241-247; US 5,470,830; 6,255,285; 6,586,630; 6,720,433; U.S.
2003/0176643; WO
02/64623). These peptidic antagonists suffer from the known limitations of peptides as drug molecules, in particular poor oral bioavailability and degradative metabolism.
Cyclization of peptidic derivatives is a method employed to improve the properties of a linear peptide both with respect to metabolic stability and conformational freedom. Cyclic molecules tend to be more resistant to metabolic enzymes. Such cyclic tetrapeptide motilin antagonists have been reported (Haramura, M. et al J. Med. Chem. 2002, 45, 670-675, U.S. 2003/0191053; WO 02/16404).
Other motilin antagonists, which are non-peptidic and non-cyclic in nature have also been reported (U.S. 5,972,939; 6,384,031; 6,392,040; 6,423,714; 6,511,980;
6,624,165;
6,667,309; U.S. 2002/0111484; 2001/041701; 2002/0103238; 2001/0056106, 2002/0013352; 2003/0203906 and 2002/0002192) The macrocyclic motilin antagonists of the present invention comprise elements of both peptidic and non-peptidic structures in a combination which has not been pursued for this application previously.
Indeed, the structural features of antagonists of the present invention are different. In particular, within the known motilin antagonists which are cyclic peptides, it was found that such derivatives containing D-amino acids were devoid of activity. In contrast, for the tripeptidomimetic compounds of the present invention, the D-stereochemistry is required for two of the three building elements.
The motilin antagonists of the present invention are also distinct from the prior art in that they comprise a tether element to fulfill the dual role of controlling conformations and providing additional sites for interaction either through hydrophobic interactions, hydrogen bonding or dipole-dipole interactions.
SUMMARY OF THE INVENTION
In a first aspect, the present invention is directed to compounds of formula (I):

z(CH2)ni 0 R3""
9zl R6 11111/,(CH)M R5 0 (CH2)p R2 v R1 T __ Z3 (I) and pharmaceutically acceptable salts, hydrates or solvates thereof wherein:
Z1, Z2 and Z3 are independently selected from the group consisting of 0, N and NRio , wherein R10 is selected from the group consisting of hydrogen, lower alkyl, and substituted lower alkyl;
R1 is independently selected from the group consisting of lower alkyl substituted with aryl, lower alkyl substituted with substituted aryl, lower alkyl substituted with heteroaryl and lower alkyl substituted with substituted heteroaryl;
5 R2 is hydrogen;
R3 is independently selected from the group consisting of alkyl and cycloalkyl with the proviso that when Z1 is N, R3 can form a four, five, six or seven-membered heterocyclic ring together with Z1;
R4 is hydrogen;

R6 and R6 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl and substituted heteroaryl, with the proviso that at least one of R5 and R6 is hydrogen;
X is selected from the group consisting of 0, NR8, and N(R9)2+;
is - wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and arnidino; and - R9 is selected from the group consisting of hydrogen, lower alkyl, and substituted lower alkyl;
m, n1 and p are independently selected from 0, 1 or 2; and T is a bivalent radical of formula II:
-U-(CH2)d-W-Y-Z-(CH2)e- (II) wherein d and e are independently selected from 0, 1, 2, 3, 4 or 5;
wherein U is bonded to X of formula (1) and is -CH2- or ¨C(=0)- ;
wherein Y and Z are each optionally present;
W, Y and Z are independently selected from the group consisting of: -0-, -NR28-, -S-, -SO-, -SO2-, -C(=0)-, -C(=0)-0-, -0-C(=0)-, -C(=0)-NH-, -NH-C(=0)-, -CR29R30-, -CH=CH- with a configuration Z or E, and -CEC-, or from a ring structure independently selected from the group:
R32 R.31 k/G2 G2 G1 GiçG2 I ""-= G2 Gi's'* /7 K2) G1,14G G2 R61 G ^^ 1¨R38 R394 '02 G
R35 R37 1 p (CH2)f 2 s.+7 R49 wherein any carbon atom contained within said ring structure, can be replaced by a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
G1 and G2 each independently represent a covalent bond or a bivalent radical selected from the group consisting of -0-, -NR41-, -S-, -SO-, -SO2-, -C(=0)-, -C(=0)-0-, -0-C(=0)-, -C(=0)NH-, -NH-C(=0)-, -S02-NH-, -NH-S02-, -CR42R4(3-, -CH=CH-with a configuration Z or E, and -CC-;with the proviso that 01 is bonded closer to U than 02, K1, K2, K3, 1<4, K6, K16 and K16 are independently selected from the group consisting of 0, NR44 and S;
f is selected from 1, 2, 3, 4, 5 or 6;
R31, R32, R38, R36, R48 and R46 are independently selected from hydrogen, halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido; and R33, R34, R35, R36, R37, R47, R30 and R31 are independently selected from hydrogen, halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido.
The invention also concerns a compound represented by the general formula (I):

\z(CH2)n1 0 Z2.Ny " R
(CH2)m 5 0 (CH2)p R1 T ______________________________ Z3 (I) or pharmaceutically acceptable salts thereof wherein:
Z1, Z2, and Z3 are independently NR10, wherein Rio is selected from the group consisting of hydrogen and lower alkyl;
R1 is -(CH2),,Rii, wherein q is 0, 1 or 2 and Ril is selected from the group consisting of:
F F

, , 7a i I A

I
Ai A
( A2<---/\ F---1 1 ¨,I \ 1 ,...\- --;;--)1-4 -/, k -A2 A3 A ' " A As A4 Al Ai ir-B4 Bi A2(4-----", B2 Al-----<-B3 A2 ______________ I / A4 -1---t.,y i A3 \ A3 I I I
õ--B A A2 Ai A fr-B5 II _eC4 tsi li 4 ii 5 A .. Il B6,..._/(\ B\/ I ________ A8.,..,\ i N X
A2 A2 and N A2' wherein A1, A2, A3, A4 and A5 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, such as trifluoromethyl, hydroxy, alkoxy, and nitro;
B1, B2, B3, B4, B5 and B7 are independently NRiaa, S or 0, wherein Riaa is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; and 136 and B8 are independently N or CH;
R2 is hydrogen;
R3 is selected from the group consisting of: ¨(CH2)5CH3, -CH(CH3)(CH2)tCH3, -(CH2),CH(CH3)2, -C(CH3)3, and -(CH2)rR21, wherein:
s is 0, 1, 2 or 3;
t is 1 or 2;
u is 0 or 1;
y is 0, 1 or 2; and R21 is selected from the group consisting of:
, . , 7b I I
I
400 g-aa 400 )\
(cliDzi E1 and, wherein z1 is 1, 2, 3 or 4 and E1 is optionally present and selected from the group consisting of hydroxy and alkoxy;
R4 and R5 are each hydrogen;
R6 is selected from the group consisting of hydrogen, ¨(CH2)aaCH3, -CH2SCH3, -CH2CH2SCH3, -(CH2)bbCH(CH3)2, -CH(CH3)(CH2)ccCH3, -(CH2)dd-NR22R23, and -(CH2)eeR24, wherein:
aa is 0, 1, 2 or 3;
bb is 0 or 1;
cc is 1 or 2;
dd is 1, 2, 3 or 4;
ee is 0, 1 or 2;
R22 and R23 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, amido, amidino, sulfonyl and sulfonamido;
R24 is selected from the group consisting of hydroxy, alkoxy, ?
I Lew 001 / n õ...--oz2 I
and , 7c wherein E2 is optionally present and is selected from the group consisting of hydroxy and alkoxy; 139 and B10 are independently selected from the group consisting of NRiab, S and 0, wherein Rub is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; B11 is selected from the group consisting of N and CH; and z2 is 1, 2, 3 or 4; and X is NR8, wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and amidino;
with the provisos that when Z1, Z2 and Z3 are all NH, R1 is:
;2?_ \ 40 Ni NRp ORp N or Rp and R2 is:

Or ORp' then R3 is not:
cH3 )1;\ , , ORp , 'NHRp NHRp \ N __ <NRp, H
NHRp )_õ\
or ORp Rp I

, 7d and when Z1, Z2 and Z3 are all NH, R1 is:
--a2- 40 \ [0 / Ol N. NRp or , ORp ' N , Rp and R3 is:
-¨1-H , +C H3 , ->Z1 , T'sS5 , , NORp , sr\sj.r --µ¨ 1¨\
\--\__--NHRp , NHRp 1-1 __ eRP, , NHRp - -i , , ORp , / 0 N,NRp , N or Rp then R2 is not:
CH3 1 =
,/14, _555 , , or ORp ;
and when Z1, Z2 and Z3 are all NH, R2 is:
+ C H 3)11--- --3-15 , , or ORp =
and R3 is:
I

7e , +CH3 'sss p , rcss NHRp NHRp eRP, NHRp ;-zz_ 1101 1\17NRp ORp N or Rp then R1 is not:
-`?z.
/ N .zNRp ORp N or Rp wherein Rp is hydrogen or a protecting group;
m, n1 and p are 0.
T is selected from the group consisting of:
( )U30 ID/\(Z3) 1µ58 I I
\\ /\ (X) U3 (Z3) (X) (Z3) (X) . ' 7f I R55 R57 (X) \,\
L'6))--(x) HO -co i 0 .\N I
(4) S 1 (X) (X) (Z3) I
(Z3) (X) , and wherein U3 is CH2, L2 is CH or N;
1.6 is CH or N;
L6 is CR52R63 or 0;
R46 is H or CH3;
R62, R53, R54, R55, R56 and R57 are independently selected from the group consisting of hydrogen and lower alkyl;
R58 is selected from the group consisting of halogen and amidino; and (X) is the site of a covalent bond to X in formula (I); and (Z3) is the site of a covalent bond to Z3 in formula (I).

7g The invention also concerns a compound represented by the general formula (I):

,(CH)n1 0 R3""

(OH )m" R5 (CHOP

R1 T _____ Z3 (I) or pharmaceutically acceptable salts thereof wherein:
Z1, Z2, and Z3 are independently NR10, wherein R10 is selected from the group consisting of hydrogen and lower alkyl;
R1 is -(CHAPii, wherein q is 0, 1 or 2, and R11 is selected from the group consisting of:
F F

7h eK, Al7 1401401 LL27 IWO
>/4 A

B1 BJv /

N

B8 N and = N
wherein A1, A2 and A3 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, hydroxy, alkoxy and nitro;
B1, B2, B3, 134, 136 and B7 are independently NRiaa, S or 0, wherein Riaa is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido;
B6 and B8 are independently N or CH;
R2 is hydrogen;
R3 is selected from the group consisting of: ¨(CH2)50H3, -CH(0H3)(0H2)10H3, -(CH2),CH(CH3)2, -C(0F13)3, and -(CH2)rR21, wherein:
s is 0, 1, 2 or 3;
t is 1 or 2;
u is 0 or 1;
y is 0, 1 or 2; and R21 is selected from the group consisting of:

7i Laza.
E Os Os (cui2)71 and wherein z1 is 1, 2, 3 or 4 and E1 is optionally present and selected from the group consisting of hydroxy and alkoxy;
R4 and R5 are each hydrogen;
R6 is each independently selected from the group consisting of hydrogen, ¨
(CH2),aCH3, -CH2SCH3, -CH2CH2SCH3, -(CH2)bbCH(C1-13)2, -CH(CH3)(CH2).CH3, -(CH2)dd-NR22R23, and -(CH2).eR24, wherein aa is 0, 1, 2 or 3;
bb is 0 oil;
cc is 1 or 2;
dd is 1, 2, 3 or 4;
ee is 0, 1 or 2;
R22 and R23 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, amido, amidino, sulfonyl and sulfonamido;
R24 is selected from the group consisting of hydroxy, alkoxy B
B9 r! 0 2\,¨ / Bil 2)72 -rsr and 7j wherein E2 is optionally present and is selected from the group consisting of hydroxy and alkoxy; B9 and B10 are independently selected from the group consisting of NRiab, S and 0, wherein Rub is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; B11 is selected from the group consisting of N and CH; and z2 is 1, 2, 3 or 4; and X is NR8, wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and amidino;
with the provisos that when Z1, Z2 and Z3 are all NH, R1 is:
\ N NRp ORp or Rp and R2 is:

+CH 3 or ORp then R3 is not:
¨FH, +CH3 , "-z;\ , _sk/\ , ORp , NH Rp NH Rp eRp, HN
NH Rp =-1z27 ORp or NNRp Rp 7k and when Z1, Z2 and Z3 are all NH, R1 is:

=zz_ 40 \ 0 / 110 or N-7NRp ' ORp ' N
Rp and R3 is:
4--( CH3 ORp , , "-t,'-\ , '555-../\ , , f\sis \-----\___--NHRp , NHRp \ N ____ <
/NRp , H ' NHRp \ ;
- -*--\_ or f\l,N1Rp .
, ORp ' N
Rp then R2 is not:
+CH3 0 40 il,"\ 'iss.
or ORp ;
and when Z1, Z2 and Z3 are all NH, R2 is:
+a-13 x,.. -_,55,..7. X _____ , or ORp and R3 is:

!

, , IX -'11/, -FH , C H3, i-C\ , ''sss-N/\ ,, ORp , y-µ?; ¨k \-----\_--NHRp , NHRp \ N
____ -(c //NRp , H
' NH Rp 01\1õN
.
, ' -ORp N or Rp Rp then R1 is not:
- -i---\
/ \
=az_ 10 \ ' lip / 0 or N zr\IRp ORp ' N .
Rp wherein Rp is hydrogen or a protecting group;
m, n1 and p are 0; and T is selected from the group consisting of:

, 7m /
(x) (Z3) I

(X) N.--- (Z3) (Z3) 10 I o(X) -.-.._.N,,,.-(X) (Z3) I=1 A 40 o,, (X) g3) 1 (x) (z3) (4) o,. 410 o(X) NH
(Z3) 40 H2 N 10 (Z3) (X) (Z3) CI
(Z3) F F (Z3) (Z3) 10 o(X) 0 o(X) 5 o(X) (Z3) (Z3) (Z3) 1 1 o4 g3) 5 o'\,_,AX) 10 o(X) 0\ ___________________________________________________________________ /(X) (X) = (Z3) ,.(X) (Z3) N (Z3) 0)....4.
(X) 0 N (X) 10 I Hd 'OH and , wherein R30 and R31 are selected from the group consisting of hydrogen and methyl;
and (X) is the site of a covalent bond to X in formula (I); and (Z3) is the site of a covalent bond to Z3 in formula (I).

7n The invention also concerns a pharmaceutical composition comprising:
(a) a compound as defined herein; and (b) a pharmaceutically acceptable carrier.
The invention yet further concerns the use of a compound as defined herein or of a pharmaceutical composition as defined above, for treating, or for the making of a medicament for treating a gastrointestinal disorder associated with the motilin receptor or motility dysfunction in humans or other mammals.
The invention yet further concerns a use of a compound as defined herein or of a pharmaceutical composition as defined above, for treating, or for the making of a medicament for treating a gastrointestinal disorder associated with hypermotility or hypermotilinemia in humans or other mammals.
The invention yet further concerns a use of a compound as defined herein or of a pharmaceutical composition as defined above, for treating, or for the making of a medicament for treating irritable bowel syndrome or dyspepsia in humans or other mammals.
The invention yet further concerns a use of a compound as defined herein or of a pharmaceutical composition as defined above, for treating, or for the making of a medicament for treating Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome or celiac disease in humans and other mammals.

In a second aspect, the invention also proposes compounds of formula (1) which are antagonists of the motilin receptor.
In a third aspect, the invention proposes a method of treating a disorder associated with the motilin receptor or motility dysfunction in humans and other mammals , comprising administering a therapeutically effective amount of a compound of formula (1).
DETAILED DESCRIPTION OF THE INVENTION
Preferably in formula (I), as depicted hereinabove, R1 is selected from the group consisting of -(CH2)gRii, and -CHR12R13 wherein q is 0, 1,2 or 3; and R11 and R12 are independently selected from a ring structure from the following group:
F F
A2 .7r=1:\

C-1=
A

A

I I
A2 _______________________ A4 2 A7 _ A3 _3 A4 wherein any carbon atom in said ring structure can be replaced a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
A1, A2, A3, A4 and A5 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido;
B1, B2, B3, and B4 are independently selected from NR14., S or 0, wherein R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, fornwl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido;
R13 is as defined for as R11 and R12 or is selected from the group comprising lower alkyl, substituted lower alkyl, hydroxy, alkoxy, aryloxy, amino, carbw, carboxyalkyl, carboxyaryl, and amido.
wherein A1, A2, A3, A4 and A5 are most preferably selected from halogen, trifluroromethyl, C1.6 alkyl or Ci.6alkoxy.
Preferably, R11, R12 and R13 are selected from the group consisting of:
vw 410 Rb 00 'ac!

s\
S
/ ,114 av,r wherein Ra and Rb are chosen from the group consisting of Cl, F, CF3, OCH3, OH, and C(CH3)3 and CH3.

Also preferably, R3 in formula (I), is selected from the group consisting of:
¨(CH2)sCH3, -CH(CH3)(CH2)tCH3, -CH(0R15)CH3, -CH2SCH3 -CH2CH2SCH3, -CH2S(==0)CH3, -CH2CH2S(=0)CH3, -CI2S(=0)2CH3, -CF2CH2S(=0)2CH3, -(CH2)CH(CH3)2, -C(CH3)3, and -(CH2)y-R21, wherein:
s and u are independently selected from 0, 1, 2, 3, 4 or 5;
t is independently selected from 1, 2, 3 or 4;
y is selected from 0, 1, 2, 3 or 4;
R15 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl and acyl;
R21 is selected from a ring structure selected from the following group:

(VW rvw 4I c.2)z Ei Ei wherein any carbon atom in said ring structure can be replaced by a nitrogen atom, with the proviso that if said ring structure is a monocyclic ring structure, it does not 5 comprise more than four nitrogen atoms and if said ring structure is a bicyclic ring structure, it does not comprise more than six nitrogen atoms;
z is selected from 1, 2, 3, 4 or 5;
El, E2 and E3 are each optionally present and are independently selected from the lo group consisting of halogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, cyano, nitro, mercapto, sulfinyl, sulfonyl and sulfonamido; and J is optionally present and is selected from the group consisting of alkyl, substituted , alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, amidino, mercapto, sulfinyl, sulfonyl and sulfonamido.
The tether portion (T) of formula (I) is preferably selected from the group consisting of:

1-'2,(z3) \ (z3) (Z3) (X) (X) (X) (z3) (Z3) (X) (X) (X) (X)(Z3) (7 3) (X) (Z3) (X) (Z3) L5 <ThZ3) L6))--(x) wherein L1 is 0, NH or NMe; L2 is CH or N; L3 is CH or N; L4 is 0 or CH2; L5 is CH or N
L6 is CR52R53 or 0; R46 is H or CH3, R52, R53, R54, R55, R56 and R57 are independently selected from hydrogen, lower alkyl, substituted lower alkyl, hydroxy, alkoxy, aryloxy, amino, and oxo; or R52 together with R53 or R54 together with R55 or R56 together with R57 can independently form a three to seven-membered cyclic ring comprising carbon, oxygen, sulfur and /or nitrogen atoms;
(X) is the site of a covalent bond to X in formula (I); and (Z3) is the site of a covalent bond to Z3 in formula (I).
In a particularly preferred embodiment of the invention, there are provided compounds of formula (I) wherein m, n and p are 0, X, Z1, Z2 and Z3 are NH and R2, R4 and R5 are hydrogen, represented by formula (ill):

(III) According to another aspect of the invention, there are provided compounds of formula (I) wherein when Z1 is a nitrogen atom, R3 forms a four, five, six or seven-membered heterocyclic ring together with Z1 ,represented by formula (IV):

N R
Z2,6 (C H2)m R6 0 (CH2)p T _________________________________ Z3 (IV) wherein said heterocyclic ring may contain a second nitrogen atom, or an oxygen, or sulfur atom;
n2 is selected from 0, 1, 2 or 3 R7 is optionally present and is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, hydroxy, alkoxy, aryloxy, oxo, amino, halogen, formyl, acyl, carboxy, carboxyalkyl, carboxyaryl, amido, carbamoyl, guanidino, ureido, is amidino, mercapto, sulfinyl, sulfonyl and sulfonamido.
It is to be understood, that in the context of the present invention, the terms amino, guanidine, ureido and amidino encompass substituted derivatives thereof as well.

Preferably, the invention provides a method of treating a disorder associated with hypermotility or hypermotilinemia in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (1).
DESCRIPTION OF PREFERRED EMBODIMENTS
Specifically preferred compounds of the present invention, include, but are not limited to:

i ¨7, p ,...
NH HN<
HN /<. _ _________________________________________________ c--( \--NH 0 -/---\

* (7 * pH HN S NH HN
c-$ NH HN

.
Ã0 220 ' * o 14 * 24 * \
195 *

NJ\
o --( 0 --( o NH
\-- HN*c--:
NH HN
\o * pH HN 0 NH HN NH HN
(-0 CI \-0 (-0 (-0 212 . HO
202 *
164 * 4 *
¨(. õ0 O ,)---,< _cNH2 ---( 9 ---( 0 \-NH HN 0 ;----1NH2 ---/ 0 "NH

(-0 HN 0 CI * !---NH NH HN-c ID
' HN 0 .. \-NH H
ci -0 HO
ç= NH /<N-C<IHN ,...c-NH
HN
NH HN
( 156 *
155 * HO
, 26 * 10 *
---( 0 O '1-4 --( 0 0 -/*.c---:
\-NH HN < 0 i-- -c0H
---NH HN
-NH HN
* pH HN et.. 0 ¨
= \-0 ¨ CI * /NH HN \ ( \--0 11 *
1 24 * 2 *
81 *
--( 0 -- 9 --( 0 --( 9 O )-4 ___c--cc: __ 0 j i< ______ 0 ) _______ i<
_cNH2 , ---NH HN-- ---col \-NH HN '\H HN-roll \--NH HN
1.0 ii..
(7 tt 0 / = NH HN HN
\--=
*
0 ¨
CI # CI * (_.NH(iHN
CI * NH ¨
200 * 21 * 149 \ / N

O )--4 0 " 0 >--4 < 0 ;/¨=
/

NH
* s\ (_. HN \ NH
0 * K__ HN NH HN
\ NH HN
0 = *
HO
190 * 191 * 126 * 17 *

--( p ¨I o ¨I, o --( o o ")--e< o "j=--4 _ \----NH HN--c: NH HN- 0 )¨HN
c:1 ---INIII
HN-ro<1 1 0 ( NH
* NH HN * pH FIN * (__NH :c--N o * pH HN
F F
HO F F
222 . 193 * 171 * 142 *
--(-, 0 --/-; 0 ---(:. 0 -/ 0 O,- 0._. -/-4 ( 0 )--NH FIN< \-NH HN-r: NH HN - NH 1-IN<

F * pH HN * Co HN \ /'4H HN *7 HN
F
187 * 163 * 167 * 23 *
I

0 ________________________________ <
--NH HN 0 ) i< _c---0<1 -NH
.\ FIN--c: ha. -NH I-IN -NH HN
,..
6 pH HN CI = 7H HN
F
* iNH HN NH HN
\ -0 CI \ -0 (-0 168 # 170 *
220 F * 133 .
---( 0 ___./
-.., P --( 0 .) i<N _c--:11 (-)c\-N-)H SN
-NH HN --NH HN --NH

H
¨ro<
a * iNH HN
CI * pH HN
CI * (_NH HN NH HN
\--0 .-*--..\ --O 0 V (-0 , , CI F
216 * 198 * 192 * 146 *
, , , =

¨i o --( o ¨, p ¨I o o")-- o "j-4 -NH
HN< -NH HN< NH HN--c--: \----NH)-HN-c:-cc.. I,.=
NH HN 0 NH HN \ NH
0 * K_ HN , \ NH
=
* HN
(-0 (-0 = \-0 \ \
19 * 22* 172 * 165 *
0 ---/ 9 HN, 1--NH2 =-( 0 : 0 -)--4 0 ---NHCI HN -c-c<13 .--NIFI HN< --NH HN
too. 0 * / \
NH ' HN * /--0 NH HN NH HN * /NH
HN
\
CI CI CI
-- = (-0 \--0 CI
144 * 131 *
204 * 15 *
--( 0 -- 0 / -. 0 0 )-4 _c=-:. N 0 )- 0 ) __ 'K
_cON
--NH HN (:)\ -NH. HN--c \ \--NH HN--c-c-4:1 \-NFI
HN , 1.= c5". 0 * 71-1 HN NH HN NH HN NH HN
CI
\-0 (-0 (-0 ( -0 CI , CI CI
218 * 211 * 145 * 182 *
---/ --/ 0 ---µ 0 0 ')-e / _______________________________________________________________________ 'S
_ci (:1\ ---NN HN--c NH HN
---NH H -NH HN---cl NH HN NH HN NH HN cc' NH
HN

\
132 * 16 * \
141 * *

, _ In addition to the preferred tethers (T) illustrated previously, other specific tethers employed for compounds of the invention are shown hereinbelow:
NHPG NHPG

OC) H43,-- 0 0.õ,õ..iviripG

I
HO
NHPG

NH
NHPG
NHPG
PG'NH NHPG
*0OH= * 0¨L

OH
T28 T32 T33a [(R)-isomer]
T33b 1(S)-isomer]

NHPG NIIPG NHPG F X
=

T35 T36 (X =
T34 T37 (X = Cl) NHPG OH
ONHPG * NHPG NHPG

*OH

NIRG
0 ipHO

PG and PG' indicate a standard amine protecting group compatible with the synthetic protocol, such as Boc, Ddz, Fmoc, or Alloc In a preferred embodiment, the present invention is directed to a method of treating irritable bowel syndrome, dyspepsia, Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, diabetes mellitus, obesity, postgastroenterectomy syndrome, atrophic colitis or gastritis, gastric stasis, gastrointestinal dumping syndrome, s celiac disease and eating disorders leading to obesity in humans and other mammals comprising administering a therapeutically effective amount of a compound of formula (I).
SYNTHETIC METHODS
A. General Information Reagents and solvents were of reagent quality or better and were used as obtained from various commercial suppliers unless otherwise noted. DMF, DCM and THE
used are of DriSolv (EM Science, E. Merck) or synthesis grade quality except for (i) deprotection, (ii) resin capping reactions and (iii) washing. NMP used for the amino acid (AA) coupling reactions is of analytical grade. DMF was adequately degassed by placing under vacuum for a minimum of 30 min prior to use. Tyr(3tBu) was synthesized following the method reported in JP2000 44595. Cpa was made using literature methods (Tetrahedron: Asymmetry 2003, 14, 3575-3580) or obtained commercially. Boc- and Fmoc-protected amino acids and side chain protected derivatives, including those of N-methyl and unnatural amino acids, were obtained from commercial suppliers or synthesized through standard methodologies known to those in the art. Ddz-amino acids were either synthesized by standard procedures or obtained commercially from Orpegen (Heidelberg, Germany) or Advanced ChemTech (Louisville, KY, USA). Bts-amino acids were synthesized as described in Example 6.
Hydroxy acids were obtained from commercial suppliers or synthesized from the corresponding amino acids by literature methods. Analytical TLC was performed on pre-coated plates of silica gel 60F254 (0.25 mm thickness) containing a fluorescent indicator. The term "concentrated/evaporated under reduced pressure" indicates evaporation utilizing a rotary evaporator under either water aspirator pressure or the stronger vacuum provided by a mechanical oil vacuum pump as appropriate for the solvent being removed. "Dry pack" indicates chromatography on silica gel that has not been pre-treated with solvent, generally applied on larger scales for purifications where a large difference in Rf exists between the desired product and any impurities. For solid phase chemistry processes, "dried in the standard manner" is that the resin is dried first in air (1 h), and subsequently under vacuum (oil pump usually) until full dryness is attained (-30 min to 0/N).
B. Synthetic Methods for Building Blocks of the Invention Example 6: Standard Procedure for the Synthesis of Bts-Amino Acids 0 BtsNH CO2H
H3N CO2 __________ RAA

To a solution of the amino acid or amino acid derivative (0.1 mol, 1.0 eq) in 0.25 N
sodium hydroxide (0.08 mol, 0.8 eq) with an initial pH of approximately 9.5 (pH meter) at rt, solid Bts-CI (0.11 mol, 1.1 eq) was added in one portion. The resulting suspension was stirred vigorously for 2-3 d. The pH of the reaction should be adjusted with 5.0 N sodium hydroxide as required to remain within the range 9.5-10.0 during this time. Typically, the pH has to be adjusted every 20-30 min during the first 5 h. Once the pH stops dropping, it is an indication that the reaction is almost complete. This can be confirmed by TLC (Et0Ac:Me0H, 95:5). Upon completion, the reaction mixture was washed with Et20. Washing is continued until the absence of non-polar impurities in the aqueous layer is confirmed by TLC (typically 3 x 100 mL). The aqueous solution was then cooled to 0 C, acidified to pH 2.0 with 1 N HCI until no additional cloudiness forms, and extracted with Et0Ac (3 x 100 mL). Alternatively, a mixture of DCM
and Et0Ac may be used as the extraction solvent, depending on the solubility of the product obtained from different amino acids or derivatives. Note that DCM
cannot be used solely as solvent because of the emulsion formed during extraction. The combined organic phases were washed with brine (2 x 150 mL), dried over MgSO4, filtered and evaporated under reduced pressure. DCM (1x) and hexanes (2x) were evaporated from the residue in order to ensure complete removal of the Et0Ac and give the desired compound as a solid in 55-98% yield.
The following are modifications that have proven useful for certain amino acids:
Gly, Ala, D-Ala, 6-Ala and GABA: Use 1.5 eq of amino acid per eq of Bts-CI, in order to prevent dibetsylation.

Met: Carry out the reaction under N2 to prevent oxidation.
Gin and Asn: Due to the solubility of Bts-Gin and Bts-Asn, the work-up required is modified from the standard procedure: Upon completion of the reaction, the reaction mixture was washed with diethyl ether. Washing is continued until the absence of non-5 polar impurities in the aqueous layer is confirmed by TLC (typically 3 x 100 mL). The aqueous phase was then cooled to 0 C and acidified to pH 2.0 with 6 N HCI. 6 N
HCI
was employed to minimize the volume of the solution due to the water solubility of Bts-Gln and Bts-Asn. (They are, in contrast, difficult to dissolve in DCM, Et0Ac or chloroform.) The solution was maintained at 0 C for 10 min and the product was 10 collected by filtration as a white precipitate. The solid was washed with cold water (1x), cold brine (2x) and water (lx, 25 C). The pH of this wash was taken, if it is not approximately 4, the solid was washed again with water. Finally, the solid was washed with cold Et0Ac, then with cold Et20 (2x), and finally dried under vacuum (oil pump) (83-85% yield).
C. General Synthetic Strategy to Conformationally-Defined Macrocycles of the Present Invention Scheme 1 E31_22_3..
0¨L¨BB3 H2L43.
L¨BB3.BB2=BBi-X-PIAG
"loading" of 1st sequential building building block* block assembly [X = 0, NH]
(deprotection, coupling) PG-Y 1¨ "EIC17-D-IER -Z deprotection, then cyclizationl release _________________________________________________________ 0¨L¨BB3.13B2aB1-X
r BB3=BB21EIE;) tether attachment \1 --CTETHER
[Y =0, NH] PG-Y
0¨L
O¨L = polymer with linker (L) for solid phase; PG =
protecting group appropriate protecting group for solution phase;
for the latter, the scheme typically starts with the P/AG =
protecting and/or protected first building block activating group BEil building blocks (amino acids, hydroxy acids) Z = reactive group *BBi, numbered in order from C-terminus in analogy to standard peptide nomenclature, NOT in order of addition The compounds of Formula I can be synthesized using traditional solution synthesis techniques or solid phase chemistry methods. In either, the construction involves four phases: first, synthesis of the building blocks, including one to four moieties, comprising recognition elements for the biological target receptor, plus one tether moiety, primarily for control and definition of conformation. These building blocks are assembled together, typically in a sequential fashion, in a second phase employing standard chemical transformations. The precursors from the assembly are then cyclized in the third stage to provide the macrocyclic structures. Finally, a post-cyclization processing stage involving removal of protecting groups and optional purification then provides the desired final compounds (Scheme 1). This method has been previously disclosed in WO 01/25257 and U.S. Patent 7,169,899.
D. Procedures for the Synthesis of Representative Tethers of the Present Invention The important tether component required for compounds of the invention are synthesized as described in W001/25257, or herein.
Example 16: Standard Procedure for the Synthesis of Tether T8 40 CO OH TMSC1, COOMe Me0H K2CO3, K1, DMF, 70 C
OH OH
CoIlidine; DMF; LiCI; then COOMe DIBAL, DCM, OH MsCI 0 C; then NaN3 (1) PPh3;
io3 then H20, 60 C
NH, Ddz-OPh NI1Ddz 0/=,,,...,OTHP (2) IN Ha DMF; 50 C
(T8) Step T8-1: Chlorotrimethylsilane (116 mL, 0.91 mol, 1.5 eq) was added to a suspension of 2-hydroxycinnamic acid (100 g, 0.61 mol, 1.0 eq) in Me0H (500 mL, HPLC
grade) over 30 min at 0 C. The resulting mixture was stirred at rt 0/N. The reaction was monitored by TLC (Et0Ac/MeOH: 98/2). Heating the reaction mixture in a hot water can accelerate the process if necessary. After the reaction was completed, the reaction mixture was evaporated under reduced pressure to afford methyl 2-hydroxycinnamate as a white solid (108.5 g) in quantitative yield. The identity of this intermediate compound is confirmed by NMR. This reaction can be carried out on larger (kg) scale with similar results Step 18-2: 3,4-Dihydro-2H-pyran (DHP, 140 mL, 1.54 mol, 2.52 eq) was added dropwise to 2-bromoethanol (108 mL, 1.51 mol, 2.5 eq) in a 2 L three-neck flask with mechanical stirring at 0 C over 2 h. The resulting mixture was stirred for additional 1 h at it. Methyl 2-hydroxycinnamate from Step T8-1 (108 g, 0.61 mol, 1.0 eq), potassium carbonate (92.2 g, 0.67 mol, 1.1 eq), potassium iodide (20 g, 0.12 mol, 0.2 eq) and DMF
(300 mL, spectrometric grade) were added to the above flask. The reaction mixture was stirred at 70 C (external temperature) for 24 h. The reaction was monitored by TLC
(DCM/Et20: 95/5). The reaction was allowed to coolto it and Et20 (450 mL) was added. The inorganic salts were removed by filtration and washed with Et20 (3 x 50 mL). The filtrate was diluted with hexanes (400 mL) and washed with water (3 x mL), dried over MgSO4, filtered and the filtrate evaporated under reduced pressure.
The crude ester (desired product and excess Br-C2H4-0THP) was used for the subsequent reduction without further purification.
Step 18-3: DIBAL (1.525 L, 1.525 mol, 2.5 eq, 1.0 M in DCM) was added slowly to a solution of the above crude ester from Step T8-2 (0.61 mol based on the theoretical yield) in anhydrous DCM (610 mL) at -35 C with mechanical stirring over 1.5 h.
The resulting mixture was stirred for 1.5 h at -35 C, then 1.5 h at 0 C. The reaction was monitored by TLC (hex/Et0Ac: 50/50). When complete, Na2SO4.10 H20 (100 g, 0.5 eq) was slowly added; hydrogen evolution was observed, when it subsided water was added (100 mL). The mixture was warmed to it and stirred for 10 min, then warmed to 40 C with hot water and stirred under reflux for 20 min. The mixture was cooled to it, diluted with DCM (600 mL), and the upper solution decanted into a filter.
The solid that remained in the flask was washed with dichloromethane (5 x 500 mL) with mechanical stirring and filtered. The filtrate from each wash was checked by TLC, and additional washes performed if necessary to recover additional product. In an alternative work-up procedure, after dilution with DCM (600 mL), the mixture was filtered. The resulting solid was then continuously extracted with 0.5% TEA in dichloromethane using a Soxhlet extractor. Higher yield was typically obtained by this alternative procedure, although it does require more time. The filtrate was concentrated under reduced pressure and the residue purified by dry pack (Et0Ac/hex/Et3N: 20/80/0.5) to give the product alcohol as a yellowish oil (yield: 90%).
The identity and purity were confirmed by NMR.
Step T8-4: To a mixture of the allylic alcohol from Step T8-3 (28 g, 0.100 mol, 1.0 eq) and collidine (0.110 mol, 1.1 eq) in 200 mL of anhydrous DMF under N2 was added anhydrous LiCI (4.26 g, 0.100 mol, 1.0 eq.) dissolved in 100 mL of anhydrous DMF.
The mixture was then cooled to 0 C, and MsCI (12.67 g, 0.110 mol, 1.1 eq., freshly distilled over P205), was added dropwise. The reaction was allowed to warm to rt and monitored by TLC (3:7 Et0Ac/hex). When the reaction was complete, NaN3 (32.7 g, 0.500 mol, 5.0 eq.) was added. The reaction mixture was stirred at rt 0/N with progress followed by NMR. When the reaction was complete, the mixture is poured into an ice-cooled water bath, and extracted with diethyl ether (3x). The combined organic phases were then washed sequentially with citrate buffer (2x), saturated sodium bicarbonate (2x), and finally with brine(1x). The organic layer was dried with MgSO4, filtered and the filtrate concentrated under reduced pressure. The allylic azide was obtained in 90% combined yield, and was of sufficient quality to use as such for the following step.
Step T8-5: PPh3 (25.9 g, 0.099 mol, 1.5 eq) was added at 0 C to a solution of the allylic azide from Step 18-4 (20.0 g, 0.066 mol, 1.0 eq.) in 100 mL of THF. The solution was stirred for 30 min at 0 C and 20 h at rt. Water (12 mL) was then added and the resulting solution was heated at 60 C for 4 h. The solution was cooled to rt, (15 mL) added and the mixture stirred for 90 min at 50 C. The separated organic phase was extracted with 0.05 N HCI (2 x 100 mL). The combined aqueous phase was washed with Et20 (5 x 150 mL) and toluene (4 x 150 mL) (more extraction could be necessary, follow by TLC), which were combined and back-extracted with 0.05 N
HCI
(1 x 100 mL). This acidic aqueous phase from back-extraction was combined with the main aqueous phase and washed with ether (5 x 150 mL) again. The pH of the aqueous phase was then adjusted to 8-9 by the addition of sodium hydroxide (5 N).
Care must be exercised to not adjust the pH above 9 due to the reaction conditions required by the next step. The aqueous phase was concentrated under reduced pressure (aspirator, then oil pump) or lyophilized to dryness. Toluene (2x) was added to the residue and then also evaporated under reduced pressure to remove traces of water. The crude product (desired amino alcohol along with inorgnic salt) was used for the next reaction without further purification.
Step 18-6: A mixture of the crude amino alcohol from Step 18-5 (0.5 mol based on the theoretical yield), Ddz-OPh (174 g, 0.55 mol, 1.1 eq) and Et3N (70 mL, 0.5 mol, 1.0 eq) in DMF (180 mL) was stirred for 24 h at 50 C. Additional DMF is added if required to solubilize all materials. The reaction was monitored by TLC (hex/Et0Ac: 50/50, ninhydrin detection). After the reaction was complete, the reaction mixture was diluted with Et20 (1.5 L) and water (300 mL). The separated aqueous phase was extracted with Et20 (2 x 150 mL). The combined organic phase was washed with water (3 x mL) and brine (1 x 500 mL), dried over MgSO4, filtered and the filtrate concentrated under reduced pressure. The layers were monitored by TLC to ensure no product was lost into the aqueous layer. If so indicated, perform one or more additional extractions with Et20 of the aqueous phase to recover this material. The crude product was purified by dry pack (recommended column conditions: Et0Adhex/Et3N: 35/65/0.5 to 65/35/0.5) to give the tether Ddz-T8 as a pale yellow syrup (yield: ¨40%). The identity and purity of the product was confirmed by NMR.
1H NMR (DMSO-d6): 1.6 ppm (s, 6H, 2 x CH3), 3.6-3.8 ppm (wide s, 10 H, 2 x OCH3, 2 x OCH2), 3.95 ppm (triplet, 2H, CH2N), 6-6.2 ppm (m, 2H, 2 x CH), 6.2-6.5 ppm (m, 3H, 3 x CH, aromatic), 6.6-7.6 ppm (m, 5H, aromatic).
Example 17: Standard Procedure for the Synthesis of Tether 19 I. OH 00 , NaH, DMF,O/N ()OH
100 C, N2 NHDdz [Example 181 Cul, Pd2Cl2(PPh3)2, Ar CH3CN/Et3N (3:1), 4h 1) Pt(IV)0, H2, ECM 0 ()OH 24-48h, r.t. OH
=
NHDdz 2) Scavenger resin NHDdz DCM/Toluene 1:1 The yield of Ddz-T9 from T9-0 on a 65 g scale was 60.9 g (91%) 1H NMR (CDCI3): 0 7.19-7.01, (m, 2H), 6.92-9.83 (m, 2H), 6.53 (bs, 2H), 6.34 (t, 1H), 5.17 (bt, 1H), 4.08 (m, 2H), 3.98 (m, 2H), 3.79 (s, 6H), 3.01 (bd, 2H), 2.66 (t, 3H), 1.26 (bs, 8H);
13C NMR (CDCI3) 0 160.9, 156.8, 155.6, 149.6, 130.4, 127.5, 121.2, 111.7, 103.2, 98.4, 80., 69.7, 61.6, 55.5, 40.3, 30.5, 29.3, 27.4 10 Tether T9 can also be synthesized from T8 by reduction as in step T9-3 or with other appropriate hydrogenation catalysts known to those in the art.
Example 18: Standard Procedure for the Synthesis of Ddz-propargylamine Ddz-N3, DIPEA, TMG
.'NHDdz 15 DMF, 50 C, 0/N, N2 In a dried three-neck flask, a solution of propargylamine (53.7 g, 0.975 mol, 1.5 eq) in degassed DMF (Drisolv, 388 mL) was treated with Ddz-N3 (170.9 g, 0.65 mol, 1.0 eq), tetramethylguanidine (TMG, 81.4 mL, 0.65 mol, 1.0 eq) and DIPEA (113.1 mL, 0.65 mol, 20 1.0 eq) and stirred at 50 C 0/N. The reaction was monitored by TLC
(conditions:25/75 Et0Ac/hex. Rf: 025; detection: UV, ninhydrin). Upon completion, DMF was evaporated under reduced pressure until dryness and the residue dissolved in Et20 (1 L).
The organic solution was washed sequentially with citrate buffer (pH 4.5, 3x), saturated aqueous sodium bicarbonate (2x), and brine (2x), then dried with MgSO4, filtered and the filtrate evaporated under reduced pressure. A pale orange solid was obtained. This solid was triturated with 1% Et0Ac in hex, then collected by filtration and dried under vacuum (oil pump) to provide the desired product (153.4 g, 85.2%).
Example 19: Standard Procedure for the Synthesis of Tether T10 Method A
1) PPh3, DIAD, THF, 4 h, rt Bz0 OH HONHDdz (Example 9] HO O.,.,-,NHDdz 2) KOH, Et0H/H20 (1/1), 0/N, rt (T10-0) (T10-1) 605g 666 g (63%) HOBr TBAI, K2CO3, DMF
85 C, 2 d -=( __________________________________________________________ HO

315 g (42% purified) Two alternative routes to this tether have been developed. The first synthetic approach proceeded starting from the commercially available monobenzoate of resorcinol (110-0).
Mitsunobu reaction under standard conditions with the protected amino alcohol from Example 9, followed by saponification of the benzoate provided T10-1 in good yield after recrystallization. Alkylation of the phenol with 2-bromoethanol using the optimized conditions shown permitted the desired product Ddz-T10 to be obtained after dry pack purification in 42% yield.
TLC (Et0Ac/Hexanes 1:1, detection: UV, ninhydrin; Rf = 0.17) 1H NMR (CDCI3) 67.18, t, 1H,J = 8.2Hz; 6.51, m, 5H; 6.34, t, 1H, J = 2.2Hz;
5.19, s, 1H;
4.05, t, 2H, J = 5.0Hz; 3.94, m, 4H; 3.75, s, 6H; 3.49, d, 2H J = 5.2Hz; 1.73, s, 6H.

13C NMR (CDCI3) 6 160.856; 6 160.152; 160.005; 155.410; 149.305; 130.279;
107.438;
107.310; 103.163; 101.877; 98.517; 69.488; 67.382; 61.595; 55.427; 40.420;
29.427.
HPLC (standard gradient) tR: 7.25 min MS: 420 (M+H) Method B
The second synthetic route to T10 is presented in the accompanying scheme.
1) PPh3, DIAD
NHBoc (35-65%) HO 0 OHHO' *
NHBoc 2) PPh3, DIAD
OTBDMS (80%) 3) TBAF (95%) From resorcinol, two successive Mitsunobu reactions are conducted with the appropriate two carbon synthons illustrated, themselves derived from 2-aminoethanol and ethylene glycol, respectively, through known protection methodologies. Lastly, deprotection of the silyl ether, also under standard conditions provided Boc-T10.
Although the yields in the two methods are comparable, the first required less mechanical is manipulation and is preferred for larger scales.
Example 20: Standard Procedure for the Synthesis of Tether T11 Pd(PPh3)4 5 mol%
CCIo (0.98 equiv.) (7)0H Cul 5.2 mol%

Et3N¨DMF (1:3) NBr I ' I
Cs2CO3 (20 mol%) NBr (1 eq) DMF, 130 C, 4 h T11-1 76% NHBoc T11-0 (1.25 eq) I()OH Pt02 (4 mol%) Et3N¨Et0H (1:51 v/v) T11 T H2 (80 psi), rt, overnight NNHBOC

97.8%
> 98%
TLC (15:85 THF/DCM; detection: UV; Rf: 0.33).
1H NMR (DMSO-d6) 68.00, d, 1H; 7.32, d, 1H; 7.15, m, 1H; 6.44, s, 2H; 6.33, s, 1H; 3.99, t, 2H; 3.71, m, 8H; 2.89, m =4, 2H; 2.71, t, 2H; 1.71, m = 5, 2H; 1.61, s, 6H.
13C NMR, solvent DMSO-d6) 6 160.879; 153.275; 151.405; 150.447; 140.773;
122.666;
118.934; 103.347; 98.456; 79.778; 70.449; 60.212; 55.717; 55.599; 29.740;
28.592.
HPLC (standard gradient) tR: 5.4 min MS: 419 (M+H) Example 26: Standard Procedure for the Synthesis of Tether T12 HO N112 110 NHDdz S DdzN3, D1PEA, TMG
DMF, 50 C
S
(12-0) Ddz-T12 In a 3-L flame-dried three-neck flask, a solution of (aminomethyl)phenylthiobenzyl alcohol (12-0, 96 g, 0.39 mol) in degassed DMF (1 L, 0.4 M) was prepared. To this was added DdzN3 (0.95 eq), followed by TMG (0.39 mol, 49 mL). The reaction was stirred for 10 min, then D1PEA (68 mL, 0.39 mol) added. The mixture was heated at 50 C under N2 until TLC

indicated no DdzN3 remained (48 h typically). (TLC eluent: Et0Ac:Hex 50:50;
detection:
ninhydrin). Upon completion, to the reaction mixture was added 3 L citrate buffer and the separated aqueous layer extracted with Et20 (3 x 1500 mL). The combined organic phase was washed sequentially with citrate buffer (2 x 200 mL), water (2 x 200 mL) and brine (2 x 200 mL). The organic layer was dried over MgSO4, filtered and the filtrate evaporated under reduced pressure. A dark orange oil was obtained, which was purified by dry-pack.
For this procedure, the oil was first dissolved in Et0Ac:Hex:DCM:TEA
(20:80:1:0.5, v/v/v/v). At this point, a little extra DCM was sometimes required to ensure complete dissolution. The solution was loaded onto the column, then the column eluted with o Et0Ac:Hex:DCM:Et3N (20:80:1:0.5) until all the impurities were separated out as indicated by TLC, paying particular attention to that closest to the desired product.
The elution was then continued with Et0Ac:Hex:Et3N 30:70:0.5 (v/v/v) and finally with Et0Ac:hexanes:Et3N
(50:50:0.5) to elute the desired product. After removal of the solvent from the fractions containing the product under reduced pressure, the residue was dissolved in the minimum amount of DCM, a three-fold larger volume of hexanes added, then the solvents again evaporated under reduced pressure. This treatment was repeated until an off-white foam was obtained. The latter solidified while drying under vacuum (oil pump).
Alternatively, the material yielded a solid after sequential concentration with DCM (1x) and hexanes (2x).
Tether Ddz-T12 was obtained as an off-white solid (85-90% yield).
Example 29: Standard Procedure for Attachment of Tethers Utilizing the Mitsunobu Reaction Example 29-A: Using PPh3-DIAD Isolated Adduct To a 0.2 M solution of the appropriate tether (1.5 eq) in THF or THF-toluene (1:1) was added the PPh3-DIAD (pre-formed by mixing equivalent amounts of the reagents and isolated by evaporation of solvent, see Example 29-C) adduct (1.0 eq.). The resultant mixture was manually agitated for 10 sec (the solution remained turbid), then added to the resin. Alternatively, the resin was added to the solution. The reaction suspension was agitated 0/N (after ¨5 min the mixture becomes limpid). The resin was filtered and washed 2x DCM, lx toluene, lx Et0H, lx toluene, lx (DCM/Me0H), lx (THF/Me0H), lx (DCM/Me0H), lx (THF/Me0H), 2x DCM, then dried in the standard manner.
Example 29-B: Using "PPh3-DIAD In Situ Procedure"

To a 0.2 M solution of the appropriate tether (4 eq) in THF or THF-toluene (1:1) was added triphenylphosphine (4 eq). The resultant mixture was manually shaken until a homogenous solution was obtained, then added to the resin. Alternatively, the resin (or MiniKans containing resin) was added to the solution. To this suspension was then added DIAD (3.9 5 eq) and the reaction agitated 0/N. Note: Since the reaction is exothermic, for larger scales, the reaction should be cooled in an ice bath. In addition, an appropriate vent must be supplied to allow any pressure build-up to be released. The resin was filtered and washed DCM (2x), toluene (1x), Et0H (1x), toluene (1x), DCM/Me0H (1x), 1x THF/Me0H
(1x), DCM/Me0H (1x), THF/Me0H (1x), 2x DCM, then dried in the standard manner.
Example 29-C: Procedure for Synthesis of PPh3-DIAD Adduct 0".= PPh3 ____________ 30min CD
0 II 4h3 0 ci DIAD (led.) (led.) Adduct 1 DIAD (1 eq) was added dropwise to a well-stirred solution of triphenylphosphine (1 eq) in THF (0.4 M) at 0 C under nitrogen. The mixture was then maintained at 0 C with stirring for 30 min. The white solid obtained was collected by filtration (use medium sized fritted filters), washed with cold anhydrous THF until the washes were colorless, and lastly washed once with anhydrous Et20. The white solid product was then vacuum-dried (oil pump) and stored under nitrogen. (Note: The PPh3-DIAD adduct can be made in larger than immediately required quantity and stored under nitrogen; it is very important to store this reagent under anhydrous conditions.) Example 30: Standard Procedure for Attachment of Tethers via Reductive Amination In certain instances, the Mitsunobu process of Example 29 cannot be applied or is not efficient for incorporation of the tether. Hence, reductive amination has been developed as an alternative that can be employed for tether incorporation as illustrated hereinbelow for one of the preferred tethers. Similar chemistry can be used to incorporate other tethers of the present invention.

'Ddz (30-2) e 0 y 0 0F3002 .3N ,ir [J,J1-0 ___________ 0 TA/10E-Me0H (1:3) BH3epyr (2 eq) RT, 0/N
Ddz (30-1) (30-3) The Tether (30-2) with the amine protected as its Ddz derivative was efficiently oxidized to the corresponding aldehyde 30-2 using S03.pyr in DMSO-Et3N-DCM. This aldehyde (0.14 mmol, 56 mg, 1.5 eq based upon loading of resin support) was dissolved in a 1:3 mixture of TMOF¨Me0H (DriSolv, 4 mt.) at rt. To this was added the resin containing the tripeptide (30-1, as its trifluoroacetic acid salt from the deprotection of the terminal amine), the mixture was agitated briefly to wet the resin, and then borane-pyridine complex (as the commercially available 8 M solution, 23 pL, 2 eq) was introduced to the suspension. The reaction was agitated 0/N, then the resin filtered, washed with DCM (2x), THE
(1x), DCM/Me0H [3:1] (1x), THF/Me0H [3:1] (1x), DCM (2x) and dried in the standard manner.
Care must be taken to ensure that the desired resin bound product 30-3 is not contaminated with the dialkylated material. However, even if the reaction does not proceed to completion or if a small amount of the dialkylation side product is present, the material is of sufficient purity for the macrocyclization reaction.
Example 32: Standard Procedure for the Synthesis of Tether T28 NO2 1, NaBH,4, THF:Me0H 1:1 io CH3NO2, AcOH
- 40 ¨ 2. H2, Pd/C
, THF:H20 ____________________________________________________ , NHBoc OH NH40Ac, 110 C, o/n 3. (Boc)20 OH OH
(79%) (90%) TBDMSO(CH2)2Br, K2CO3, KI
DMF, 75 C
NHBoc TBAF NHBoc (74%) OH Y
THF ______________________________ 101 0,0TBDMS
C
(95%) Boc-T28 Henry reaction of 2-hydroxybenzaldehyde 28-0 provided 28-1 in 79% yield. This was followed by reduction first with sodium borohydride, then with catalytic hydrogenation, to give the amine, which was then protected as its Boc derivative, 28-2. Yields of these first two steps were lower on larger scales. Alkylation of 28-2 with the TBDMS ether of 2-bromoethanol, itself synthesized by standard methods, gave 28-3 in 74% yield.
Deprotection of the silyl ether under standard conditions yielded the desired protected tether, Boc-T28. Alternative use of ethylene carbonate for the phenol alkylation to avoid the protection/deprotection steps, gave 73% yield.
Example 36: Standard Procedure for the Synthesis of Tether T32 OTBDMS
OH OH Br,-,OTBDMS 0 NBS, TfOH
CH3CN el Br (32-A) Br K2CO3, KI, DMF' (94%) 70 C
CN CN (100%) CN

c,,,OTBDMS
o/\/OTBDMS
1) LiHMDS, THF (Boc)20, 1 N NaOH
Br 40 Br 2) 1M citric acid dioxane (67%, 2 steps) H2N NH BocHN NH

NHDdz OTBDMS OH
(32-B) 0 //

I) H2, PtO2 Pda2(PPh3)2, PPh3 NHDdz 95% Et0H

NHDdz HN(iPr)2, 70 C 2) TBAF, THF
(100%) (88%, 2 steps) BocHN NH BocHN NH
32-5 Ddz-T32(Boc) Overall yield 55%
(7 steps) TLC (100% Et0Ac; detection: UV, CMA; Rf = 0.24).
1H NMR (CDCI3, ppm): 7.74 (1H, dd), 7.35 (1H, d), 6.72 (1H, d), 6.53-6.49 (2H, m), 3.61-3.29 (1H, m), 5.06 (1H, t), 4.25-4.01 (2H, m), 3.91-3.89 (2H, m), 3.73 (3H, s), 2.99 (2H, dd), 2.63 (2H, t), 1.71 (8H, broad), 1.53 (9H, s).
13C NMR (CDCI3, ppm): 163.8, 162.2, 161.0, 159.7, 155.9, 149.4, 130.0, 129.1, 128.0, 126.8, 110.8, 98.1, 80.9, 79.3, 69.7, 61.3, 55.5, 39.1, 29.3, 28.5, 26.7.
Example 37: Standard Procedure for the Synthesis of Tether T33a and T33b HO OMe ly 4.6 OH
_________________________________________________________________ 1111 OilLOMe DIBAL 1.0 M oy,OH
I DIAD, PPh3 =I CH2Cl2, -78 C
=
THF, rt, o/n (99%) 33-0 (96%) 33-1 33-2 NHDciz Cul, Et3N/CH3CN (3:1) PdC12(PPh3)2, Ar, o/n (88%) =

NHDdz 41) H2, 95% Et0H , Pt02, oirl or'OH
2) PS-TMT, CH2Cl2 (93%) NHalz Ddz-T33a 33-3 Overall yield 77.2%, 4 steps The construction to the (R)-isomer of this tether (T33a) was accomplished from 2-iodophenol (33-0) and (S)-methyl lactate (33-A). Mitsunobu reaction of 33-0 and 33-A
proceeded with inversion of configuration in excellent yield to give 33-1.
Reduction of the ester to the corresponding alcohol (33-2) also occurred in high yield and was followed by Sonagashira reaction with Ddz-propargylamine. The alkyne in the resulting coupling product, 33-3, was reduced with catalytic hydrogenation. Workup with scavenger resin provided the desired product, Ddz-T33a.
1H NMR (CDCI3) 8 (ppm) 7.18-7.11 (m, 2H), 6.90 (m, 2H), 6.52 (m, 2H), 6.33(m, 1H), 5.09 (bt, 1H), 4.52 (m, 1H), 3.77 (s, 6H), 3.08 (bq, 2H), 2.64 (bt, 2H), 1.75 (m, 8H), 1.27 (bd, 3H), 13C NMR (CDCI3) 8 160.8, 155.5, 149.5, 131.2, 130.6, 127.4, 121.2, 113.3, 103.2, 98,4, 80.7, 74.8, 66.5, 55,4, 40.2, 30.6, 29.3, 29.2, 27.4, 16.1 HPLC (standard gradient): tR: 7.93 min The synthesis of the (S)-enantiomer (Ddz-T33b) was carried out in an identical manner in comparable yield starting from (R)-methyl lactate (33-B) '44"r0I-1 OMe , > 0 0, + .0-:,,, , 0 NHDdz 33-0 33-B Ddz-T33b is Example 38:
Standard Procedure for the Synthesis of Tether T34 NMe2SO4 Me'N'IL LiHMDS, oxirane -1µ1 Ac20 HO N
)'),,. \, Me NaOH,Na0H, H2OH20 0 N Me THF, rt, BF3:0Et2 crNN OH pyr, rt crN
OM
(83%) Me (60%) I 3 (98%) I 3 12, ' DCM, AgOCOCF2 (75%) -'-N 3 NHDdz H2 N is ..,,NHDdz 0 I
, , Ddz-propargylamine (32-B) ., A.,/.1 I I N
Cr¨ N OM Pt02, Et0H e.,N,--õ*.r0Ac PdC12(1313h3)2, Cul, Bu4NI, 0 OAc.,..,. õ.......*.i., I 3 (90%) I 3 Et3N. K2CO3 (80%) Me0Na, Me0H 0 (85%)y,ji¨NHDdz Ce''N OH
Ddz-T34 i TLC (100% Et0Ac; detection: CMA, Rf '7: 0.5).

MW Calc. for C24H35N307, 477.55; MS Found (M+H)+ 478.
1H NMR (CDCI3) M.62 (m, 2H), 1.70 (m, 8H), 2.43 (m, 2H), 2.67 (m, 2H), 3.07 (m, 2H), 3.34 (s, 3H), 3.43 (s, 3H), 3.61 (m, 2H), 3.75 (s, 6H), 5.40 (sb, 1H), 6.31 (s, 1H), 6.49 (s, 2H) 5 13C NMR (CDCI3) .623.25 (CH2), 25.97 (CH2), 28.56 (CH3), 39.31 (CH3), 30.09 (CH3), 31.25 (CH2), 32.19 (QH2), 40.16 (QH2), 55.47 (CH3), 61.38 (QH2), 80.65 (Cq), 99.38 (Cq), 103.17 (Cq), 111.01(Cq), 149,60 (Cq), 151.33 (Cq), 152.46 (Cq), 160.80 (Cq).
HPLC (standard gradient) tR: 6.68 min.
Example 39: Standard Procedure for the Synthesis of Tether T35 Br F OH

Br K2CO3, Kl, DMF Br T35-O55 C, 55 C, 0/N, N2 100% NHDdz Pd(PhCN)2C12, dioxane Cul, P(Bu)310% hexanes i-Pr2NH, 60 C, 0/N
4 steps 75.2%
Overall yield: 56%
FOH 1) H2, Pt02, Et0H F AlbOTBDMS
NHDdz 2) TBAF 1.0 M, THF, 1 h 74.5% (2 steps) NHDdz Ddz-T35 T35-2 TLC (25/75 Et0Ac/Hex; detection: UV, ninhydrin; Rf = 0.03) 1H NMR (CDCI3): 67.06-7.00 (bt, 1H), 6.61-6.52 (m, 4H), 6.35 (m, 1H), 5.12 (bt, 1H), 4.03 (m, 2H), 3.95 (m, 2H), 3.77 (s, 6H), 3.11-3.04 (bq, 2H), 2.60 (bt, 2H), 1.75 (m, 8H) 13C NMR (CDCI3): 8 163.9, 160.9, 160.6, 157.6, 157.5, 155.6, 149.5, 130.8, 130.6, 125.9, 107.26, 106.9, 103.2, 98,4, 80.8, 77.5, 69.9, 61,3, 60.9, 60.6, 55,4, 40.3, 30.4, 29.3, 26.9, HPLC (standard gradient): tR = 8.37 min Example 40: Standard Procedure for the Synthesis of Tether T36 74.5% (2 steps) Ati OH TBDMSO Br T35-A 1::X'-OTBDMS
F igr Br K2CO3, KI, DMF F Br T36-0 55 C, 0/N, N2 T36-1 96.7%
.NHDdz Pd(PhCN)2Cl2, dioxane Cul, P(Bu)3 10% hexanes i-Pr2NH, 60 C, 0/N
4 steps 75.2%
Overall yield: 54%
OH 1) H2, Pt02, Et0H (3-0TBDMS
NHDdz F 2) TBAF 1.0 M, THF, 1 h F
NHDclz Ddz-T36 74.5% (2 steps) TLC: (25/75 Et0Ac/Hex; detection: UV, ninhydrin; Rf = 0.03) 1H NMR (CDCI3) 8 (ppm): 6.84-6.75 (m, 3H), 6.52 (bs, 2H), 6.34 (m, 1H), 5.17 (bt, 1H), 4.01 (m, 2H), 3.93 (m, 2H), 3.77 (s, 6H), 3.10 (bq, 2H), 2.63 (bt, 2H), 1.74 (m, 8H) 13C NMR (CDCI3) 5 160.9, 158.9, 155.8, 155.6, 152.9, 152.9, 149.5, 132.4, 132.3, 117.1, 116.8, 112.7, 112.6, 103.2, 98.4, 80.8, 70.4, 61.6, 55.5, 40.2, 30.3, 29.3, 27.4.
HPLC (standard gradient): tR = 8.29 min Example 41: Standard Procedure for the Synthesis of Tether T37 OH TBDMS0 Br CI IV Br K2CO3, KI, DMF CI IWP Br T37-O55 C, T37-1 C, 0/N, N2 86.2%
NHDdz Pd(PPh3)2Cl2 Cul, PPh3, Argon 4 steps i-Pr2NH, 55 C, 0/N
Overall yield: 43% 61.2%
'OH 1) H2, Pt02, Et0H .,i0TBDMS
NHDdz 2) TBAF 1.0 M, THF, 1 h CI .4W- CI 411' 82 /0 ( ste NHDdz 2 ps) Ddz-T37 T37-2 TLC (25/75 Et0Ac/Hex; detection: UV, ninhydrin; Rf = 0.03) 1H NMR (CDCI3): 67.12-7.08 (bd, 2H), 6.76-6.73 (d, 1H), 6.52 (m, 2H), 6.33 (bs, 1H), 6:15 (bt, 1H), 4.02 (m, 2H), 3.95 (m, 2H), 3.79 (s, 6H), 3.09 (bq, 2H), 2.61 (bt, 2H), 1.74 (m, 8H). 13C NMR (CDCI3) 8 160.8, 155.6, 155.4, 149.5, 132.4, 130.1, 127.0, 126.0, 112.8, 103.2, 98.4, 80.8, 70.0, 61.4, 55.5, 40.3, 30.2, 29.3, 24.5, 27.4 HPLC (standard gradient): tR = 9.60 min Example 42: Standard Procedure for the Synthesis of Tether T38 \

OH
Acetone, 75 C, 0/N
93%

NHDdz Cut, Pd2Cl2(PPh3)2, Ar CH3CN/Et3N (3:1), 0/N
100%
OH Raney Ni, H2 OH
NHDdz rt, 0/N
100% NHDdz Ddz-T38 T38-2 1H NMR (CDCI3): 8 7.20-7.10, (m, 2H), 6.95-6.80 (m, 2H), 6.55 (bs, 2H), 6.35 (s, 1H), 5.18 (bt, 1H), 4.12 (m, 1H), 3.95 (m, 2H), 3.80 (s, 6H), 3.15 (bq, 2H), 2.65 (t, 2H), 1.98 (bs, 2H), 1.65 (bs, 6H), 1.25 (m, 3H).
13C NMR (CDC13): !3160.8, 156.6, 155.8, 149.6, 130.4, 127.5, 121.3, 111.7, 103.2, 98.4, 80.7, 73.5, 66.6, 55.5, 40.2, 30.5, 29.3, 29.1, 27.3, 19.5.
Chiral T38 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
HPLC (standard gradient) tR = 8.46 min Chiral material can be accessed by starting with the chiral epoxide. For example, the (S)-isomer of 138 was constructed in 89% overall yield from (S)-propylene oxide.
Example 43: Standard Procedure for the Synthesis of Tether T39 OH OH
CO2H TMSCI, Me0H CO2Me Br OTBDMS
99% K2CO3, KI, DMF
70 C, 55%

OTBDMS
crOTBDMS f CO2Me Cul, MeLi, Et20 LiAIH4, THF -0 C, 76% 0Me 0 C, 100%

OTBDMS OH
of 1) MsCI, Et3N
of 28%, 8 steps I
CH2Cl2 40 OH 2) NaN3, DMF, 50 C
NHBoc 3) Pd/C, H2, (Boc)2Cr Et0Ac T39-4 4) TBAF, THF Boc-T39 67%, 4 steps TLC (50% Et0Ac, 50% Hex; detection: UV and CMA; Rf = 0.25) 1H NMR (CDCI3, ppm): 7.11-7.08 (2H, m), 6.86 (1H, t), 6.76 (1H, d), 5.05 (1H, broad), 4.26-3.85 (4H, m), 3.22-3.07 (2H, m), 2.71 (1H, broad), 1.66-1.60 (2H, m), 1.33 (9H, s), 1.17 (3H, d).
13C NMR (CDCI3, ppm): 156.1, 135.0, 127.1, 127.0, 121.4, 111.7, 69.9, 61.5, 39.8, 38.4, 28.7, 20.7.
Chiral T39 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
Example 44: Standard Procedure for the Synthesis of Tether T40 NaH
H BrOTBDMS
Ph3PCH(CH3)CO2Me K2CO3, KI, DMF 40 0.,..0-rBs OH 90-100%
70 C, 100%

CO2Me DIBAL, DCM OH Pd/C, Et3N
O 87-90% 0 L_zOTBS 80-90%

OH
1) MsCI, Et3N f 41%, 8 steps 40 OH CH2Cl2 2) NaN3, DMF, 50 C
40 NHBoc 0 3) Pd/C, H2, (BOC)20 L....y0TBS Et0Ac T40-4 4) TBAF, THF Boc-T40 67%, 4 steps = TLC (50% Et0Ac, 50% Hex; detection: UV and CMA; Rf = 0.25) 1H NMR (CDCI3, ppm): 7.11-7.08 (2H, m), 6.86 (1H, t), 6.76 (1H, d), 5.05 (1H, broad), 5 4.26-3.85 (4H, m), 3.22-3.07 (2H, m), 2.71 (1H, broad), 1.66-1.60 (2H, m), 1.33 (9H, s), 1.17 (3H, d).
13C NMR (CDCI3, ppm): 156.1, 135.0, 127.1, 127.0, 121.4, 111.7, 69.9, 61.5, 39.8, 38.4, 28.7, 20.7.
10 Chiral T40 can be accessed through the use of asymmetric synthesis methods, resolution or chiral chromatography techniques available in the literature.
Example 45: Standard Procedure for the Synthesis of Tether T41 x.071:7.54-:
7,777:3_7.1A7.40'177.47.".7-1:i.7"-.4r"'.770Tciii:',::PliSPCRCOECII.firiP77"7177,77.1-777177.7W77,, .!
= = ' == ....'::-f=-= '''Ivon:::-::',44,0 - ''''' = =
- ').- A....,.-',...,:'"*.--".... .. yq!'-= ...
HO'' ' =;'= = ''..: = :4=1v=ii' . lu ....\=_..j . -...-. = = - = . "'if?' -..1-... = ' ' . =_,,,,µ . ...1:.t.:
- :'-' -!'::== ,..1%;13. '''''''' H= SO = rt 24.fir ' = l'='... ..-'-= - -~ --:'':==
2 4, , ; = , :g .. =4.11' ,Y,'!=-.!'= '=N,.... = ..,tv, = = F., . C1713CN, 70 C,: 24.h .'-',"'' "
= %); 1 = = kx-01, ..., .)=== .,,,, :.;,- ,õ , . r = ifI
..fiCr'. v.:'=bEiliA'lliir: ..= - i: = . . , ;',:=:!.= = ' =J!f4.-,F.i..... :- :=:p. . ' .: .: 64 ,IP'qi..,, i.:1.!, ,4S,.-b = -4'.'! iit 3 .
ill --...';.õ'....'191'ilr-r. ';,4 =
=;'..41,7*. =:"..:,..,: = .i4iWilt=':' õ==..:,..... - 'likt...L.:17 = ,.. ,p.:
rtw .. 'N4ir,.. = !:,,..,..i,,,-. . = = ....,f:J...i, ,,,.... ., ..,;,,-.: '. .1'- =,= .A.f.; = -, . ,,,i!, ,,,,õ,. = = . >1..1 rs'I' '.'1'. i'::.1. , .: . = ;A-41] "(2, !!, ':f I ='ll=!i .', - = .: i.1.0,,.1,!11 ',14141, ._,i. - = = .
ic.'.. = , ' = .. 4,47,,p,, . =i',1.;-:.1.0 , . , c.p, 3.1i!=.Th."..iii-, . ¨ ===';=iii5q;:.=,t.
.'; n =.:1,1i, A '%;=,. = - ' ' '' N.-.
, . . .!: ',.:? - .-t=vilA õ = ...;..,.==.fiN,..-r;,; ' _.=.tp,: f7,4,1 ,L! . = !!'= = ! = ...,IPi: ,,,yE. ' .)'... = : = 1,11 ..,..= :4.1ivr ...,,.- = ,,:t.,;;;ri,.-=:===;Ai.,?..rrii.,-:, .:.?Ni.i.h, :,i, '''', 1.4.'1.:õ 'i;!!:, ,,,I. = :i... a.11 .,=71.: i r2. - . ,,, it. ,-,2.. , . It A
. , ,:.,Ai:õ. :',':1:74.11,11r..4.. ,;,,,==
.r.:',''',:.....dht= - A* ,.....,:; !!';:i.=: .:'14:1=;-:.:?1, =="4: .
":õ.=j: = = = .. tiiitir ;.,. :', = .44,.. 'N-- ip. = !.7:10t...,., = ..-..q?,.,.:.'=
,'.., r..1.1!'''':Ir . i.-...1: -':=:- . .... :,',.:,..,,,iir . . . , ..,::?=== : .. vyr if 24-ti.:-:1194%
::=..:1: . ., 4t,- a-,f:.:-..- . - p.,..:- .],.. . -0.õ, = ,, ,,,,..., ., =.
-...õ- = 40}..,-.:.,=-1.,,i= = :or . = . ;...(:.i:
. .i ! :.4:1=.:,: = ,...s.: ii . =,,:z, rõi, : ..
,,, - - ,-: =,, -4. - = , .. .
. .
= = !.. = ,,Z. . fl' !,^1:e ='''4 = =' =e." = '.i ;' :'ii':.=,.'i" .> Ai.
.,.. ,,_,õ. ...õ .7 . : .
. ;,:õ.....-õ.= ..:.:. ..J . : ..
..:, ..,....,:=..==..,- ..:-.7., .'=:.gi '-' =-.'4: ,41-.:
=-.,u. - :. ,'.;!,:... .._,..4,.z.:C: ,==0'10%,... -:, -...1.
. -,,.' = P- ''' :.3,464õ. ....====2` = .: . g. ;; ..¨
...:..t4r1 ,f Fl 1\17.71->.:'..- ='CtOtif ::=== :'``---4. kr-.' 1:.
tdOile = - -- =-= = ''.. , -'. = i* - t.'-':-' 0),..,õ,,,,,, =._==-: -, = .. . -. -,, ',.=-=::=...i:],r,;,..,..
,...,,,,,',õ4,,..,,,,..,,,=, õ. ,.... DMF õ,.. ..= 4. ,,,.:
...:,, .. .. .,4,41 = = is.: .i:h.
:.,:.........,...p! -,.;.t.rdic,. Etoti) ..!..,,, ,.P.,i.43...._16. ;.".s.''.
r :'.-= ... ::':;`,,='05 C, 24:411i = .,'-:,= 1,1';VX,17).; =
'111'':.11 ,=2.:FsT"-==:...'",.',...f7i:-W2-..1. .... .- = 4.;,,,,=,'::r! =
=::=1:-:''.i-e=N--'..)1=:=.: = ..!=-, '',.',1'.' .:'''' = . =
''''. HI .= = . ,Zh'y..:.'.'.... .=.=,,. :. = 0, .!.ii :. . -1,NI!..:=ptvpli .A.:= . "'Li =48%11.;'.".';:.=Z i Vi .
, . . :.: .. " ''' = = = = = ..'1' .' ... 'i. ,!*94,..ig .
"! I t = . = ''''": ..' :.;1.
= 'Sici., , : -.ii 1 ! ' ''' .. . .: rrõ,õ- ;2::. -:=!,'..!=!.i ,i, =:',4:
' - .... 't= . ....... -,V, . . . = . - --,...:- = .... - .T. -11=,;p1i'jaWietl! ' ,:...õ.i...,-:,. .- ::!, =
....'.,...õ=,A,.:....11.,,,,,...' . A
=41 4 .. . . . .1414 e = . = =Tt, : 56% -..1., = .'::. 9#70Ph .,..-, ...irg -r, .. =="...=
.= .1:=,,, 41:,, .. . = '4' 1...."'' r: A
= : .,..."4; 411- Hi' = ''= ::õ ':;'... 'i..= ''.µ = = ''' ,r= = qj, ,., Dr*, =TEA,, 507Q.,:;711*1 .I..;1 . . :1' = . :.
4i.l.,,=, ..: = .::v.. iii-,-. ,õ . . ,:.i.:1;:-.; 'c' -.11'4: ...=-=P'= .-N".= - ==
, g..,'= .= :: ='..!;,g.... = . .:,=:;;;= =,..v, ,,,,:= =,'Ai:-=
" !ii.H . . .q: . = = = ie= ,...4y = . . = = - - . L':' = .. ,e .?7,!.! ., '".=:.i.p i, 1.. , :17-',. "1.;P'f'A. ',.;:- .:.";;' = : = =E.-?'-'4, =
iivir.: = .. - . = '= = -fK.=,4 '= '' . :=. -;21- .=
.'.'i ',.::. =:,- ;=:,, .,-4? = : . = . .
%= ..V==== '':..#11V,... = ' ' - . k = = . . ..1'illi:==
,, ;4=,; , . -k, -I4._ .. . .44- . . = ;A: =
= . ,,, = .'.4,' .'= . = eij,,,, . ti2, 4: k.O. i'., = .. . !..1.. . . =
:5`'..1(:,:''..õ '21.4,:. : ... ' = :: : . ii4i= ..,;,;., ...i . = !q =IP . .:Y. .. = '' . ==1 ,ip,' .A-4 .=:,,'.i'-1-. .= = . ..
, .. = ' ;1 ' ''' '.4`i:W: ,51:4'' '''Fi ' '.i!=!' ' .. a" .'!= . -1.2 T-j'''. . =
:.' - , = .C. 0 ' i t.. ' = ': . :;..'''''' = =
tiSH4:11:1eUrt , i = = .Y .i'r, ,' ='1,t,.: .... ;=?,=;' nu .. = .=
.= ' = = . ,:: ,i,_.' ',.
- - ., = . :. - -....,... 44 . 14r4,-.. \'''' q., s.,. . .". " '...,1.," :P -.1: = = '' ... ' .1 ti?::
..dzHN,7:(. 440, . ,.. ,., :. . = . . ..
0,4_4. .Rq ,.Jdz . .. ,f : :..,t,' ii!.'.: .'i.,.!... .A;= .
'..,,,..,. .; ,= Jc- H J.:.v.,.. , , .1. = . === = . .= ...= ...,. :: %.1õ:"...',I..:=.e.. , - .=
= :- :=A ., : THF,=rt, 2411:=:=.:4",:t-'11"-:'-=.:==144.,- .
'A!!6C1.?=tif=I,.,..I=;:fAika..61A, .,...: ::.101,...ta.. ..,:µ,õiikm.i.,4 ........rd----(Vti:IiiRA, . :.:= :=::.. : ..... . : r. :. ==:.. ..
:. ,. ::,:l.:1 = = :.=!;t1.0,-...õ¨= .b.tiiiii517'!'"IF.4*I';ifikkliV, --i',0Pv=
95% = \ .4.4,1. .. r,:. . = :., ;=]..:,,A-:.,:.-.3 =:,;:-..: ,ti ::==== ; .../:. \ :742." ;....; =
: . :=0.1:p .'NO-4-4,,r,i,k4.1t r Pk= -irtirt:Vte.:::
. 4:44 il -::7i'4 A.1 141.-4-t:441411P4V.7-*...,Iglan.4i;;Z!.:iergitraktlik.f4 ill VI
raP,Likt;q.l.LT::74=tiaril,,,=::,4,L.
, TLC (100% Et0Ac; detection: CMA; Rf = 0.5) ..
1H NMR (CDCI3) .8.1.23 (s, 3H), 1.49 (s, 3H), 1.69 (s, 3H), 1.74 (s, 3H), 1.90 (m, 2H), 2.35 (m, 1H), 3.35 (m, 2H), 3.76 (s, 6H), 3.92 (m, 2H), 4.40 (m, 2H), 5.10 (m, 1H), 6.15 (s, 1H), 6.25 (s, 2H).
13C NMR (CDCI3).825.52 (CH3), 27.53 (CH3), 28.88 (QH3), 29.61 (CH3), 35.92 (CH2), 42.62 (CH2), 55.43 (CH3), 60.60 (CH2), 82.38 (CH), 83.33 (CH), 83.68 (CH), 84.96 (CH), 98.26 (CH), 103.23 (CH), 118.3 (Cq), 149.50 (Cq), 156.20 (Cq), 160, 02 (Cq) , HPLC (standard gradient): tR = 6.64 min MS: M+H found: 439 , Example 46: Standard Procedure for the Synthesis of Tether T42 /C/, OTs OH OH OH OTs 0 401 Br H2C0 .----,,,,, , OHC Br CH3PPh3Br 11 Br HO , ......, 40 Br MgC12, TEA io t-BuOK, THE, ' IP DEAD, PPh3, THF
CH3CN, reflux -78 C-> rt it 6h T42-3 95% 75% 88%
, [C12(CY3P)2Ru=CHPhl DON1(0.2M), rt, 12 h 70%
O
NHDdz , _____________________ OH
Me0Na 0 0 0 ' Cs0Ac io , Cul, Pd(PhCN)2C12 0 OH µ OAc =
1 1 P(Bu)3, HN(iPI-)2, Br Me0H, RT
Br 0 DMF, 50 C, 0/N 0 OTs T42-7 dioxane, 70 C, 0/N 98% Br 142-6 142-5 70%

NHOdz 80%
H2, F102 EtON, rt, 0/N
. 95%

Ddz-T42 NHDdz 1H NMR (300 MHz, CDCI3) 66.82-6.98 (m, 2H); 6.80-6.75 (m, 1H); 6.53 (s, 2H);
6.35 (t, 1H, 2 Hz); 5.23 (b, 1H); 4.08 (m, 1H); 3.90-3.68 (m, 8H); 3.20-2.97 (m, 2H);
2.95-53 (m, 4H); 2.0-1.63 (m, 10H).
13C NMR (75.5 MHz, CDCI3) !3160.85; 155.56; 152.55; 149.56; 128.13; 127.77;
120.28;
103.22; 98.43; 80.72; 76.80; 65.76; 55.46; 40.23; 30.45; 29.34; 29.22; 27.10;
24.97; 23.94.
E. Examples of Synthetic Strategies for the Macrocyclic Compounds of the Invention Scheme 2: Thioester Strategy for Macrocyclic Compounds of the Present Invention , ' I

, 1. TrtSCH2CH2CO2H
DIC, DMAP, 18 h 0 P3-AA3-0H
CireNH2 DMF:DCM (1:1), RW cy.Thsri., ¨ (PG = Ddz or Boc) 2. Ac20/DIP EA/DCM H SH ___________ capping, 1 h, RW ii 1 %__....õ_.= PyBOP
3. 10%TFA, Et3SiH O¨SH "Cr DIPEA-Linker NMP, 18 h 3 x 15min, RW RW
RW = Resin Wash 1. 1% TFA, 3x 15mln (Ddz) 1. 2% TFA, 15min (Ddz) or 33% TFA, lh (Boc) or 33% TFA, 1h (Boc) Et3SIH, DCM, RW Et3SIH, DCM, RW
2. Bts-AA1-0H
I
HBTU, DIPEA, NMP HBTU, DIPEA, NMP
18h, RW 2. PG-AA2-0H
18h, RW ______________________________________________ 0¨SAAA3NHPG

¨S)(AA3¨AA2¨Arti-NHBts Ph3P-DIAD preformed betaine, Tether iii r% r¨SI AA3--AA2¨AA1--NBts 0 _____________________________________________ ' THF or THF-toluene (1:1), 18h, RW
DdzNI-I-Tether¨) Ag-assisted macrocyclization Method A Method B
Better yields but may require special purification to remove 1. 2% TFA, Et3SiH, DCM 1. 2% TFA, Et3SiH, DCM
Ag impurities to trace levels 1 h, RW
\_ lh, RW
2. 1 eq Ag(CF3CO2), DIPEA 2. DIPEA, THF, MP-carbonate, THF, 24 h MP-carbonate, 24 h Purification: -- 1. PS-thiophenol resin -CombiFlash ., H k AA3--AA2--AA1--1H KOTMS, THF/Et0H (1:1) , H k AA3--AA2¨AAI¨NiBts 2. 50% TFA, DCM
Ag-scavenger ISCO
etc. -Tether ---1 Et3SIH or iPr3SIH
Tether ---1 2h One or more of the amino acids indicated can be replaced by corresponding hydroxy acids and coupled to the next building block utilizing methods known to those in the art.
Example 47: Standard Procedure for Macrocyclization with Thioester Linker The resin containing the cyclization precursor is combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, Foster City, CA, commercially supplied MP-carbonate resin was treated with 3x THF (1 L per 400 g) and dried 0/N at 30 C in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin).
A 0.2 M DIPEA solution in THF was then added to the combined resins (1 mL /60 mg MP-carbonate resin) and the suspension agitated 0/N at rt. Subsequently, the resin was filtered and rinsed 2x THF. The combined filtrates are collected together in an appropriate vessel, then the volatile contents evaporated under reduced pressure [in addition to the standard methods, solvent can also be removed in vacuo using centrifugal evaporation 43a (ThermoSavant Discovery, SpeedVac* or comparable)] to provide the crude macrocycles.
Example 48: Standard Procedure for Silver-Assisted Macrocyclization with Thioester Linker Except for the cyclization itself and subsequent work-up, this procedure is identical to that * trademark of Example 47. The resin containing the cyclization precursor was combined in an appropriate vessel with pre-washed MP-carbonate resin [Argonaut Technologies, commercially supplied MP-carbonate resin was treated with THF (3x, 1 L per 400 g) and dried 0/N at 30 C in a vacuum oven] (1.4 to 1.6 eq relative to the initial loading of the synthesis resin). To this was added THF (1 mL per 100 mg resin) and silver trifluoroacetate (1 eq relative to the initial loading of the resin). Finally, an amount of DIPEA sufficient to obtain a 0.2 M solution was added. The reaction mixture was agitated at it 0/N. The solution was then filtered and the resins washed 2x THF. The filtrates are collected together in an appropriate vessel, then evaporated under reduced pressure [(the volatile contents could also be removed in vacuo using centrifugal evaporation (ThermoSavant Discovery, SpeedVac or comparable)] to provide the crude macrocycles.
For this procedure, silver trifluoroacetate should be stored in a dessicator between uses.
In addition, it is recommended to use a new bottle of THF (or a bottle that has been recently opened under N2 or Ar) to minimize formation of silver oxide.
Additionally, a ring-closing metathesis (RCM) strategy, as developed by Grubbs et al. can also be used to access some of the macrocyclic compounds of the invention (see for example US 5,811,515; Grubbs, R.H. et al. J. Org. Chem. 2001, 66, 5291-5300;
Furstner, A. Angew. Chem. Int. Ed. 2000, 39, 3012-3043).
To access certain derivatives of compounds of the present invention, additional reactions from those in the general scheme were required. For some, it was advantageous to react the functionality to be derivatized prior to the formation of the macrocyclic ring. The cyclic structure can restrict access of reagents to that functionality. For example, in the synthesis of N-methyl and N-acyl derivatives of macrocycles, where the secondary nitrogen atom of the ring is the site of derivatization, the reaction is preferred to be performed prior to the application of the appropriate cyclization protocol.
In other cases, for example the derivatization of side chain functionality, the reaction was best performed after formation of the macrocyclic ring. For example, further reaction of amino moieties on side chains examples was typically efficiently done by reaction of the partially protected macrocycle. In this manner, acylation, sulfonylation, alkylation (via reductive amination), guanidine and urea formation were performed via standard methods.

Table 1, hereinbelow, shows a representative, but by no means exclusive, summary of the chemical synthesis of several representative compounds of the invention.

Table 1: Synthesis of Representative Compounds of the Present Invention Afiki AA2 AA3 Tether Tether Additional _ Attachment Steps 1 Bts-D- Boc-D-Val Boc-Nva Ddz-T8 Example 29 none Tyr(tBu) 2 Bts-D- Boc-D-Val Boc-Nva Boc-T8 Example 29 none Phe 3 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe 4 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 5 Bts-D- Boc-D-Ala Boc-Nva Ddz-T8 Example 29 none Tyr(tBu) 6 Bts-D- Boc-D-Val Boc-Met Ddz-T8 Example 29 none Tyr(tBu) 7 Bts-D- Boc-D-Val Boc-Nle Ddz-T8 Example 29 none Tyr(tBu) 8 Bts-D- Boc-D-Val Boc-Phe Ddz-T8 Example 29 none Tyr(tBu) 9 Bts-D- Boc-D-Val Boc-Val Ddz-T8 Example 29 none Tyr(tBu) lo Bts-D- Boc-D-Val Boc-Leu Ddz-T9 Example29 none Tyr(tBu) 11 Bts-D-2- Boc-D-Val Boc-Nva Boc-T8 Example 29 none Nal 12 Bts-D- Boc-D-Val Boc-Abu Ddz-T8 Example 29 none Tyr(tBu) 13 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Phe 14 Bts-D-2- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Nal 15 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3CI
16 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(4CI
17 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none Trp(Boc 18 Bts-D- Boc-D-2- Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) Abu 19 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(4F) 20 Bts-D- Boc-D-Val Boc-Leu Boc-T8 Example 29 none Phe 21 Bts-D-2- Boc-D-Val Boc-Leu Boc-T8 Example 29 none Nat 22 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Tyr(OM
e) 23 Bts-D-1- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Nal 24 Bts-D-2- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Thi 28 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(2CI
26 Bts-D- Boc-D-Val Boc-Cpa Ddz-T9 Example 29 none Tyr(tBu) 27 Bts-D-4- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Thz 28 Bts-D-3- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Pal Tyr(tBu) Dap(Boc) 38 Bts-D- Hnva(THP) Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 34 Bts-D- Ddz-D- Boc-Nva Ddz-T8 Example 29 None Tyr(tBu) Tyr(tBu) 38 Bts-D- Boc-D-Val Boc-Ala Ddz-T8 Example 29 none Tyr(tBu) 39 Bts-D- Boc-D-Val Boc-E-Ala Ddz-T8 Example 29 none Tyr(tBu) 40 Bts-D- Boc-D-Val Boc-Gly Ddz-T8 Example 29 none Tyr(tBu) 41 Bts-D- Boc-DPhe Boc-Nva Ddz-T8 Example 29 none Tyr(tBu) 82 Bts-D- Boc-D-Val Boc-Phg Ddz-T8 Example 29 none Tyr(tBu) Bts-D- Ddz-D-Val Ddz- Ddz-T8 Example 29 none Tyr(tBu) Lys(Boc) Tyr(tBu) Orn(Boc) Bts-D- Ddz-D-Val Ddz- Ddz-T8 Example 29 none Tyr(tBu) Ser(tBu) Tyr(tBu) Tyr(tBu) Bts-D- Ddz--D-Val Ddz- Ddz-T8 Example 29 none Tyr(tBu) Trp(Boc) 60 Bts-D- Boc-D-Val Boc- Ddz-T8 Example 29 none Tyr(tBu) Tyr(OMe) 65 Bts-D- Boc-D-Val Boc-Nva Ddz-T2 Example 29 none Tyr(tBu) 71 Bts-D- Boc-D-Val Boc-Nva Ddz-T10 Example 29 none Tyr(tBu) 72 Bts-D- Boc-D-Val Boc-2-Nal Ddz-T8 Example 29 none Tyr(tBu) 76 Bts-D- Boc-D-2-Nal Boc-Nva Ddz-T8 Example 29 none Tyr(tBu) Bts-D- Boc-D-Nle Boc-Nva Ddz-T8 Example 29 none Tyr(tBu) so Bts-D- Boc-D-Val Boc-Ile Ddz-T8 Example 29 none _Tyr(tBu) 85 Bts-D- Boc-D-Val Boc-D-Nva Ddz-T8 Example 29 none Tyr(tBu) 87 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Bip 88 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 89 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Hfe 9 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Dip 91 Bts-D- Boc-D-Nva Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 92 Bts-D- Boc-D-Tle Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 96 Bts-D- Boc-I3-Ala Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) Bts-D- Boc-D-Chg Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 98 Bts-D- Boc-D-Val Boc-Nva Ddz-T18 Example 29 none Tyr(tBu)_ 99 Bts-D- Boc-D-Val Boc-Nva Ddz-T15 Example 29 none Tyr(tBu) 109 Bts-D- Boc-D-Val Ddz- Ddz-T9 Example 29 none _Tyr(tBu) Dab(Boc) 110 Bts-D- Boc-D-Val Boc-Nva Ddz-T11 Example 29 none Tyr(tBu) 111 Bts-D- Boc-D-Val Hval(THP) Ddz-T9 Example 29 none Tyr(tBu) 112 Bts-D- Boc-D-Val Boc-Nva Ddz-T9 Example 29 none Tyr(tBu) 120 Bts-D- Boc-D-Pro Boc-Nva Ddz-T8 Example 29 none , Tyr(tBu) 121 Bts-D- Boc-D-Val Boc-Nva Ac-T8-NH2 Example 29 none Tyr(tBu) 122 Boc-D- Boc-D-Val Boc-Nva Boc-T9 Example 30 none 3-Pal 123 Boc-D- Boc-D-Val Boc-Nva Boc-T9 Example 30 none 2-Pal 124 Boc-D- Boc-D-Val Boc-Nva Boc-T9 Example 30 none 4-Pal 125 Bts-D- Boc-D-Cpg Boc-Nva Boc-T9 Example 29 none Tyr(tBu) 126 Bts-D- Boc-D-Val Boc- Boc-T9 Example 29 none Tyr(tBu) NMeLeu 127 Boc-D- Boc-D-Val Boc-Nva Boc-T12 Example 30 none His(Mts) 128 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Tyr(OM
e) 125 Bts-D-1- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Nal 135 Bts-D-2- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Thi 131 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Phe(3CI
) 132 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Phe(4CI
) 133 Bts-D- Boc-D-Val Boc-Leu Boc-T9 Example 29 none Phe(4F) 134 Bts-D- Boc-D-Val Boc-Leu Boc-T2 Example 29 none Phe(3CI
) 135 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none Tyr(OM
e) 136 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none 1Nal 137 Bts-D-2- Boc-D-Val Boc-Leu Boc-T11 Example 29 none Thi 138 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none Phe(3CI
) 135 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none Phe(4CI
) 140 Bts-D- Boc-D-Val Boc-Leu Boc-T11 Example 29 none _ Phe(4F) 141 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Tyr(OM
e) 142 Bts-D-1- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Nal 143 Bts-D-2- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Thi 144 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Phe(3CI
) 145 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Phe(4CI
) 146 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Phe(4F) 147 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none Tyr(OM
e) 148 Bts-D-1- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none Nal 149 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none Phe(3CI
) 158 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none Phe(4CI
) 151 Bts-D- Boc-D-Val Boc-Cpa Boc-T11 Example 29 none Phe(4F) 152 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Tyr(OM Dap(Boc) e) 153 Bts-D-1- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Nal Dap(Boc) 154 Bts-D-2- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Thi Dap(Boc) 155 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(3CI Dap(Boc) ) 156 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(4CI Dap(Boc) ) 157 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(4F) Dap(Boc) 158 Bts-D- Ddz-D-Val Ddz- Ddz-T11 Example 29 none Phe(3CI Dap(Boc) ) 159 Bts-D- Boc-D-Ile Boc-Nva Boc-T9 Example 29 none -Tyr(But) 160 Bts-D- Boc-D- Boc-Nva Boc-T9 Example 29 none Tyr(But) allolle 161 Boc-D- Boc-D-Val Boc-Nva Boc-T9 Example 30 none Phe(4C
=

moc) 162 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(2M
e) 163 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3M
e) 164 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(4M
e) 166 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(30 Me) 166 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(20 Me) 167 Bts-D-3- Boc-D-Val Boc-Nva Boc-T9 Example 29 none benzothi enyl 168 Bts-D-3- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Thi 169 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none D-HomoP
he(3CI) 170 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3,4 diCI) 171 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3,4 diF) 172 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3,4 di0Me) 173 Bts-D- Hnva(THP) Boc-Nva Boc-T9 Example 29 none 1Nal 174 Bts-D- Hnva(THP) Boc-Nva Boc-T9 Example 29 none Tyr(OM
e) 176 Bts-D- Boc-D-Val Boc-Nva Boc-T33b Example 29 none Tyr(tBu) 176 Bts-D- Boc-D-Val Boc-Nva Boc-T33a Example 29 none Tyr(tBu) 177 Bts-D- Boc-D-Val Boc-Nva Boc-T28 Example 29 none Tyr(tBu) 178 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Tyr(OM Ser(tBu) e) 179 Bts-D-1- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Nat Ser(tBu) 189 Bts-D-2- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Thi Ser(tBu) 181 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(3CI Ser(tBu) 182 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(4CI Ser(tBu) 183 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 none Phe(4F) Ser(tBu) 184 Bts-D-1- Ddz-D-Val Ddz- Ddz-T11 Example 29 none Nal Dap(Boc) 185 Bts-D- Ddz-D-Val Ddz- Ddz-T11 Example 29 none Phe(4CI Dap(Boc) 186 Ddz-D- Ddz-D-Val Ddz- Ddz-T9 Example 30 none Tyr(tBu) His(Mts) 187 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3C
F3) 188 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(3F) 189 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Phe(4N
02) 199 Bts-D-3- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none benzothi enyl 191 Bts-D- Boc-D-Val BOG-Cpa Boc-T9 Example 29 none Phe(30 Me) 192 Bts-D- Boc-D-Val Boo-Cpa Boc-T9 Example 29 none Phe(3,4 diCI) 193 Bts-D- Boc-D-Val Boc-Cpa Boc-T9 Example 29 none Phe(3,4 diF) 194 Bts-D- Boc-D-Val Boc-Nva Boc-T34 Example 29 none Tyr(OM
e) 195 Bts-D- Boc-D-Val Boc-Nva Boc-T38 Example 29 none Tyr(OM

e) 196 Bts-D- Boc-D-Val Boc-Cpa Ddz- Example 29 none Phe(3CI T32(Boc) ) 1 197 Bts-D- Boc-D-Val Boc-Cpa Boc-T34 Example 29 none Phe(3CI
) 198 Bts-D- Boc-D-Val Boc-Cpa Boc-T38 Example 29 none Phe(3CI
) 199 Bts-D- Boc-D-Val Boc-Cpa Boc-T41 Example 29 none Phe(3CI
) 200 Bts-D- Boc-D-Val Boc-Cpa Boc-T8 Example 29 none Phe(3CI
) 201 Bts-D-1- Boc-D-Val Boc-Nva Boc-T8 Example 29 none Nal 202 Bts-D- Boc-D-Val Boc-Nva Boc-T8 Example 29 none Phe(30 Me) 203 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 acetylation Phe(4CI Dap(Boc) ) 204 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 guanidinylati Phe(4CI Dap(Boc) on ) 205 Bts-D- Boc-D-Val Boo- Boc-T9 Example 29 none Phe(3CI NMeLeu ) 206 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 mesylation Phe(4CI Dap(Boc) ) 207 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 TMS-Phe(4CI Dap(Boc) isocyanate ) followed by dilute acid 208 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 guanidinylati Tyr(tBu) Dap(Boc) on 209 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 acetylation Tyr(tBu) Dap(Boc) 210 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 reductive Tyr(tBu) Dap(Boc) amination with acetone 211 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 reductive Phe(4CI Dap(Boc) amination ) with excess formaldehyd 212 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 reductive Phe(4CI Dap(Boc) amination ) with acetone 213 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Tyr(3,5d il) 214 Bts-D- Boc-D-Val Boc- Boc-T9 Example 29 hydrogenoly Tyr(OM Hse(BzI) sis for e) protecting group removal 215 Bts-D- Ddz-D-Val Ddz- Ddz-T9 Example 29 reductive Tyr(tBu) Dap(Boc) amination with excess formaldehyd 216 Bts-D- Boc-D-Val Boc-Cpa Boc-T40 Example 29 none Phe(3CI
) 217 Bts-D- Boc-D-Val Boc-Cpa Boc-T36 Example 29 none Phe(3CI
) 218 Bts-D- Boc-D-Val Boc-Nva Boc-T39 Example 29 none Phe(3CI
) 219 Bts-D- Boc-D-Val Boc-Nva Boc-T37 Example 29 none Phe(3CI
) 220 Bts-D- Boc-D-Val Boc-Nva Boc-T39 Example 29 none Phe(3CI
) 221 Bts-D- Boc-D-Val Boc-Nva Boc-T35 Example 29 none Phe(3CI
) 222 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 none Tyr(3tBu ) 223 Bts-D- Boc-D-Val Boc-Nva Boc-T9 Example 29 acetylation Tyr(But) 224 Bts-D-1- Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive Nal amination with formaldehyd 225 -Bts-D-1- Boc-D-Val Boc-Leu Boc-T9 Example 29 acetylation Nal 226 ¨
Bts-D-1- Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive Nat amination with aldehyde 227 Bts-D-1- Boc-D-Val Boc-Leu Boc-T9 Example 29 reductive Nat amination with benzaldehy de Notes Any amino acid or tether designated as the Boo derivative could be substituted with the corresponding Ddz derivative.
D. Analytical Data for Selected Compounds of the Invention 1H and 13C NMR spectra were recorded on a Varian Mercury 300 MHz spectrometer and are referenced internally with respect to the residual proton signals of the solvent.
Information about the conformation of the molecules in solution can be determined utilizing appropriate two-dimensional NMR techniques known to those skilled in the art.
HPLC
purifications were run on a Waters XTerra* MS C18 column, using the Waters FractionLynx system. Automated medium pressure chromatographic purifications were performed on an Isco CombiFlash* 16x system with disposable silica or C18 cartridges that permitted up to sixteen (16) samples to be run simultaneously.
MS
spectra were recorded on a Waters Micromass* Platform II or ZQ system. HRMS
spectra were recorded with a VG Micromass ZAB-ZF spectrometer. Chemical and biological information were stored and analyzed utilizing the ActivityBase database software (IDBS, Guildford, Surrey, UK).
General Methods for Analyticl HPLC Analyses HPLC analyses are performed on a Waters Alliance system 2695 running at 1 mL/min using an Xterra* MS C18 column 4.6 x 50 mm (3.5 pm). A Waters 996 PDA
provided UV data for purity assessment. An LCPackings splitter (50:40:10) allowed the flow to be separated in three parts. The first part (50%) went to a Micromass Platform II MS equipped with an APCI probe for identity confirmation. The second part (40%) went to an evaporative light scattering detector (ELSD, Polymer * trademarks Laboratories, PL-ELS-1000) for purity assessment and the last portion (10%) to a chemiluminescence nitrogen detector (CLND, Antek* Model 8060) for quantitation and purity assessment. Data was captured and processed utilizing the most recent version of the Waters Millenium software package.
An example LC method suitable for compounds of the present invention uses Me0H
as solvent A, H20 as solvent B and 1%TFA/ H20 as solvent D. Initial mobile-phase composition is 5% A, 85% B and 10% D. Details of the standard gradient method are shown below:
Time A% B% D% Curve 0.00 5 85 10 6 10 1.00 5 85 10 6 6.00 50 40 10 6 9.00 50 40 10 a 14.00 90 0 1.0 6 17.00 90 0 10 6 17.50 5 85 10 6 20.00 5 85 10 6 Compounds 2-6, 8-10, 56, 65 and 144 are as defined in Table (3), hereinbelow.
Compound 2 Yield: 12 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300MHz, DMSO-d6) 6 8.83 (m,1H); 8.53 (m, 1H); 7.63 (m, 1H); 7.4-7.08 (m, 7H);
20 7.00-6.84 (m, 2H); 6.60 (d, 15Hz, 1H); 6.41 (dt, 15Hz, 5.4 Hz, 1H); 4.35 (m, 1H); 4.25-4.05 (m, 3H); 3.94 (dt, 1H, 6Hz, 15Hz); 3.79 (dd, 1H, 3.6Hz, 8.4 Hz); 3.60 (m, 1H);
3.52-3.40 (bd, 1H); 3.22-3.06 (m, 4H); 1.88 (m, 2H); 1.54-1.28 (m, 2H); '1.25 (d, 3H, 4.8Hz); 1.22 (d, 3H, 2.7 Hz); 0.92-0.80 (m, 6H).
HRMS calc. for C301-140N404: 520.3049; found 520.3057 0.0016 HPLC [standard gradient method (refers to that presented in General Methods for Analytical HPLC Analyses)] tR = 9.55 min.
Compound 4 Yield: 12 mg pure macrocycle was obtained (CLND quantification).
* trademark 1H NMR (300 MHz, DMSO-d6) 6 9.35 (b, 1H); 8.98 (b, 1H); 5.52 (d, 1H, 8.4Hz);
8.38 (b, 1H); 7.25 (b, 1H); 7.13-7.07 (m, 4H); 6.86 (t, 2H, 7.5Hz); 6.57 (d, 2H, 8.7 Hz); 4.33 (b, 1,H);
4.21-4.02 (m, 3H); 3.78 (dd, 1H, 3.3Hz; 8.1Hz); 3.65-3.54 (m, 1H); 3.31-3.23 (m, 11-I); 3.13-3.02 (m, 4H); 2.78-2.2.28-2.18 (m, 1H); 2.0-1.80 (m, 2H); 1.50-1.30 (m, 3H);
1.25 (d, 3H, 4.5Hz); 1.22 (d, 3H, 4.5Hz); 1.01 (d, 3H, 6.6Hz); 0.90 (d, 3H, 6.6Hz); (t, 3H, 7.5Hz).
13C NMR (75.5MHz, DMSO-d6) 6 172.22; 171.37; 157.77; 157.44; 156.04; 131.76;
130.80;
130.70; 127.88; 121.82; 115.83; 111.71; 62.13; 60.62; 54.21; 52.81; 47.13;
42.47; 33.31;
29.69; 29.30; 28.61; 20.36; 19.44; 18.72; 17.60; 13.97.
HRMS calc. for C30H42N405: 538.3155; found: 538.3145 0.0016 io HPLC (standard gradient) tR = 8.12 min.
Compound 5 Yield: 17 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300 MHz, DMSO-d6) 69.02 (b, 1H); 8.47 (d, 1H, 8.4Hz); 7.7 (b, 1H);
7.58 (d, 1H, 5.4Hz); 7.28 (dd, 1H, 7.8Hz, 0.8Hz); 7.20 (t, 1H, 9.0Hz, 0.8Hz); 7.14 (d, 2H, 8.4Hz); 6.98-6.91 (m, 3H); 6.66 (d, 8.7Hz); 6.63 (d, 1H, 15.0Hz); 6.43 (dt, 1H, 6.0Hz, 15.0Hz); 4.28-3.86 (m, 6H); 3.60-3.40 (m, 2H); 3.22-3.12 (m, 1H0; 3.05 (d, 2H, 5.4Hz); 1.92-1.80 (m, 1H);
1.56-1.40 (m, 1H); 1.36-1.20 (m, 2H); 1.25 (d, 3H, 6.6Hz); 0.84 (t, 3H, 7.2Hz).
13C NMR (75.5MHz, DMSO-d6) 6 172.54; 171.86; 158.97; 158.56; 127.39; 155.84;
131.62;
129.73; 129.20; 129.02; 128.43; 126.30; 124.51; 122.01; 115.85; 112.88; 61.23;
52.90;
51.23; 47.08; 42.66; 36.13; 33.30; 21.14; 19.57; 17.07; 14.14; 11.49.
HRMS calc. for C281-136N405: 508.2685; found: 508.2681 0.0015 HPLC (standard gradient) tR = 7.67 min.
Compound 6 Yield: 16 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300MHz, DMSO-d6) 6 9.37 (b, 1H); 8.87 (b, 1H); 8.61 (d, 1H, 8.7Hz);
7.62 (b, 1H); 7.27 (d, 1H, 7.8Hz); 7.21 (t, 1H, 8.4Hz); 7.14 (d, 2H, 8.4Hz); 6.98-6.87 (m, 3H); 6.64 (d, 2H, 8.1Hz); 6.70 (d, 1H, 15.6Hz); 6.39 (dt, 1H, 6.3Hz, 15.6Hz); 4.44-4.36 (m, 1H); 4.34-4.08 (m, 2Hz); 4.45-3.92 (dt, 1H, 6.9Hz, 15.6Hz); 3.74 (dd, 1H, 3.6Hz, 8.4Hz);
3.54-3.26 (m, 3H); 3.22-3.02 (m, 3H); 2.60-2.36 (m, 4H); 2.24-2.14 (m, 1H); 2.02 (s, 3H); 1.96-1.89 (m, 1H); 1.80-1.66 (m, 1H); 1.01 (d, 3H, 6.3Hz); 0.90 (d, 3H, 6.6Hz).

13C NMR (75.5MHz, DMSO-d6) 6 171.51; 171.26; 158.90; 158.49; 157.38; 155.86;
131.63;
129.82; 129.21; 128.86; 128.63; 126.21; 121.98; 115.83; 11'2.83; 62.11; 61.06;
51.97;
47.10; 42.78; 30.91; 30.67; 29.34; 20.37; 19.39; 15.06.
HRMS calc. for C301-140N406S: 568.2719; found: 568.2711 0.0017 HPLC Rt (general method) 7.92 min.
Compound 8 Yield: 27 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300MHz, DMSO-d6) 6 9.05 (b, 1H); 8.43 (b, 1H); 8.34 (d, 1H, 9.3Hz);
7.40 (b, 1H); 6.97 (d, 1H, 7.5Hz); 6.92-6.74 (m, 9H); 6.67-6.54 (m, 2H); 6.33-6.25 (m, 3H); 6.10 (dt, 1H, 5.7Hz, 16.2Hz); 4.22 (dt, 1H, 0.9Hz, 12Hz); 3.94-6.66 (m, 4H); 3.30 (dd, 1H, 3.6Hz, 7.8Hz); 3.24 (m, 1H); 3.18 (m, 1H); 2.85-2.68 (m, 3H); 2.44-2.23 (m, 2H); 1.32 (o, 1H, 7.5Hz); 0.97-0.89 (m, 1H); 0.42 (d, 3H, 6.6Hz); 0.01 (d, 3H, 6.6Hz).
.13C NMR (75.5MHz, DMSO-d6) 6 171.20; 157.35; 155.88; 139.12; 131.61; 130.87;
129.74;
129.21; 128.77; 128.88; 126.85; 126.19; 121.97; 115.82; 112.84; 62.04; 61.10;
55.07;
50.01; 47.09; 42.85; 37.42; 29.11.
HRMS calc. For C34F14.2N4.06 : 586.3155; found: 586.3145 0.0017 HPLC Rt (general method) 9.34 min.
Compound 9 Yield: 17 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300MHz, DMSO-d6) 6 9.39 (b, 1H); 8.83 (b, 1H); 8.29 (d, 1H, 9.3Hz);
7.62 (b, 1H); 7.28 (d, 1H, 6.6Hz); 7.20 (t, 1H, 6.9Hz); 7.12 (d, 2H, 7.8Hz); 6.98-6.91 (m, 2H); 6.63 (d, 2H, 8.4Hz); 6.58 (d, 1H, 16.2Hz); 6.40 (dt, 1H, 5.7Hz, 16.2Hz); 4.29-4.13 (m, 3H); 4.03-2 3.92 (m, 2H); 3.52 (m, 1H); 3.15-3.05 (m, 3H); 2.45-2.37 (m, 1H);
1.96-1.88 (m, 1H); 1.25 (dd, 2H, 4.5Hz; 6Hz); 1.01 (d, 3H, 6.3Hz); 0.91 (d, 3H, 6.6Hz); 0.86 (d, 3H, 7.2Hz); 0.81 (d, 3H, 6.6Hz).
130 NMR (75.5MHz, DMSO-d6) 6 171.85; 171.17; 157.37; 155.87; 131.59; 129.88;
129.18;
128.97; 128.78; 128.51; 126.16; 121.97; 115.83; 112.85; 61.55; 61.18; 58.15;
54.22;
47.08; 42.89; 36.32; 29.35; 29.00; 20.34; 19.56; 18.73; 17.44.
HRMS calc. for C301-140N406 536.2998; found: 536.2990 0.0017.

HPLC (standard gradient) tR = 8.15 min.
Compound 10 Yield: 24 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300 MHz, DMSO-d6) 6 9.33 (b, 1H); 8.82 (b, 1H); 8.56 (d, 1H, 8.3Hz);
7.60 (b, 1H); 7.27 (d, 2H, 7.8Hz); 7.20 (t, 1H, 7.8Hz); 7.13 (d, 2H, 8.4Hz); 6.95 (t, 2H, 7.8Hz); 6.64 (d, 211, 8.4Hz); 6.57 (d, 1H, 15.4Hz); 6.38 (dt, 1H, 15.4Hz, 5.8Hz); 4.26-4.10 (m, 3H); 3.96 (dt, 1H, 5.4Hz, 8.4Hz); 3.77 (dd, 1H, 3.7Hz, 7.8Hz); 3.51-3.24 (m, 3H); 3.18-3.02 (m, 3H);
1.90 (h, 1H, 6.4Hz); 1.73-1.54 (m, 2H); 1.45 (dt, 1H, 6.7Hz, 0.9Hz); 0.99 (d, 3H, 6.6Hz);
0.89 (d, 3H, 6.3Hz); 0.87 (d, 3H, 6.0Hz); 0.80 (d, 3H, 6.3Hz).
13C NMR (75.5MHz, DMSO-d6) 6 172.23; 171.17; 157.37; 155.88; 131.62; 129.82;
129.19;
128.95; 128.59; 126.24; 121.99; 115.84; 112.88; 64.23; 61.98; 61.14; 51.43;
61.14; 51.43;
47.07; 42.81; 29.38; 24.85; 24.11; 21.00; 20.32; 19.30.
HRMS calc. for C3+142N406 550.3155; found: 550.3150 0.0016.
HPLC (standard gradient) tR = 8.91 min.
Compound 56 Yield: 16 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300 MHz, DMSO-d6) 6 9.39 (b, 1H); 8.90 (b, 1H); 8.67 (d, 1H, 8.4Hz);
7.74 (b, 411); 7.29-7.08 (m, 4H); 6.99-6.87 (m, 2H); 6.64 (d, 2H, 8.1Hz); 6.61 (d, 1H, 16.5Hz); 6.40 (dt, 1H, 5.7Hz, 16.5Hz); 4.40-4.06 (m, 4H); 4.02-3.95 (m, 11-1); 3.79 (dd, 1H, 3.6Hz, 7.8Hz);
3.55-3.30 (m, 211); 3.16-3.05 (m, 3H); 2.82-2.69 (m, 2H); 2.02-1.85 (m, 2H);
1.64-1.43 (m, 3H); 1.29-1.23 (m, 1H); 1.01 (d, 3H, 6.3Hz); 0.91 (d, 3H, 6.3Hz); 0.86-0.84 (m, 2H).
HPLC (standard gradient) tR = 5.71 min.
Compound 65 Yield: 17 mg pure macrocycle was obtained (CLND quantification).
1H NMR (300 MHz, DMSO-d6) 6 9.60 (b, 1H); 9.39 (b, 1H); 8.88 (b, 1H); 8.70 (d, 1H, 7.5Hz); 8.57 (d, 1H, 4.2Hz); 7.27 (t, 6Hz); 6.96 (d, 2H, 8.4Hz); 6.66 (d, 2H, 8.4Hz); 5.78-5.68 (m, 1H); 5.42-5.33 (m, 111); 3.96-3.89 (m, 1H); 3.80-3.57 (m, 511); 3.41-3.34 (m, 1H);
3.10-2.90 (m, 1H); 2.78-2.66 (m, 1H); 2.21-2.10 (m, 1H); 2.06-1.93 (m, 1H);
1.70-1.60 (m, 1H); 1.52-1.41 (m, 111); 1.39-1.26 (m, 1H); 1.25 (d, 3H, 4.8Hz); 1.23 (d, 3H, 4.5Hz); 0.83 (dd, 3H, 3Hz, 4.5Hz).
13C NMR (75.5MHz, DMSO-d6) 6 172.68; 172.63; 159.15; 158.73; 157.38; 157.25;
130.89;
124.99; 116.03; 62.51; 62.12; 54.29; 49.27; 42.47; 32.77; 30.43; 28.85; 20.46;
19.59;
18.72; 17.39; 13.90; 13.09.
HRMS calc. for C24H36N404: 444.2736; found: 444.2726 0.0013 HPLC (standard gradient) tR = 6.80 min.
Compound 144 1H NMR (300 MHz, CD30D) 67.4 (m, 1H); 7.27 (dt, 1H, 1.5 Hz, 6.6 Hz); 7.22-7.14 (m, o 2H); 7.08-6.98 (m, 2H); 6/8 9t, 2H, 6.6 Hz); 4.45-4.39 (m, 2H); 4.15 (d, 2H, 8.1 Hz);
7.74 (d, 1H, 9.3 Hz); 3.54 (d, 1H, 10.8 Hz); 3.35-3.22 (m, 2H); 3.20 (q, 1H, 1.5 Hz);
2.82-2.71 (m, 1H); 2.61-2.55 (m, 1H); 2.21-2.11 (m, 1h); 2.02-1.94 (m, 1H);
1.74-1.40 (m, 5H); 1.04 (d, 3H, 6.6 Hz); 0.93 (d, 3H, 6.6 Hz); 0.74-0.64 9m, 1H); 0.45-0.28 (m, 2H); 0.15-0.08 (m, 1H); 0.06-0.02 (m, 1H).
13c NMR (75.5 MHz, CD30D) 8 17329; 172.14; 167.51; 155.47; 134.86; 134.81;
130.38; 130.31; 128.81; 128.25; 127.44; 121.63; 110.39; 107.71; 105.02; 67.10;
66.66;
62.81; 62.06; 60.10; 53.99; 41.44; 36.07; 31.91; 30.01; 29.18; 28.94; 27.79;
23.68;
23.15; 19.08; 18.25; 8.17; 4.98; 3.16.
HRMS: calc. for C311141N404C1568.2816; found 568.2802 0.0017 F. Mass Spectral Data for Selected Compounds of the Invention Table 2: Analysis of selected compounds of the invention Molecular Formula Molecular Weight Monoisotopic M+H
(calculated) Mass Found 1 C30H40N405 536.7 536 2 C30H40N404 520.7 520 521 3 C30H42N404 522.7 522 4 C30H42N405 538.7 538 C28H36N405 508.6 508 6 C30H40N405S 568.7 568 7 C31H42N405 550.7 550 __________ 551 8 C34H42N405 586.7 586 587 9 C30H40N405 536.7 536 537 10 C31H42N405 550.7 550 551 11 C34H44N404 572.7 572 573 12 C29H38N405 522.6 522 523 13 C31H44N404 536.7 536 537 14 C35H46N404 586.8 586 587 15 C30H41N404C1 ' 557.1 556 557 16 C30H41N404C1 557.1 556 557 17 C32H43N504 561.7 561 562 18 C29H40N405 524.7 524 525 19 C30H41N404F 540.7 540 541 20 C31H42N404 534.7 534 535 21 C35H44N404 584.7 584 585 22 C31H44N405 552.7 552 553 23 C34H44N404 572.7 572 573 24 C28H40N404S 528.7 528 529 25 C30H41N404C1 557.1 556 557 26 C31H42N405 550.7 550 551 27 C27H39N504S 529.7 529 530 28 C29H41N504 523.7 523 524 29 C28H39N505 525.6 525 526 30 C30H41N306 539.7 539 540 34 C34H40N406 600.7 600 601 38 C28H36N405 508.6 508 509 39 C28H36N405 508.6 508 509 40 C27H34N405 494.6 494 495 41 C34H40N405 584.7 584 585 52 C33H38N405 570.7 570 571 55 C31H43N505 565.7 565 566 ' 56 C30H41N505 551.7 551 552 57 C28H36N406 524.6 524 525 58 C34H40N406 600.7 600 601 59 C36H41N505 623.7 623 624 60 C35H42N406 614.7 614 615 65 C24H36N404 444.6 444 445 71 C29H40N406 540.7 540 541 72 C38H42N405 634.8 634 635 76 C38H42N405 634.8 634 635 77 C31H42N405 550.7 550 551 80 C31H42N405 550.7 550 551 85 C30H40N405 536.7 536 537 87 C36H46N404 598.8 598 599 88 C34H50N405 594.8 594 595 89 C31H44N404 536.7 536 537 90 C36H46N404 598.8 598 599 91 C30H42N405 538.7 538 539 92 C31H44N405 552.7 552 553 96 C28H38N405 510.6 510 511 97 C33H46N405 578.7 578 579 98 C24H39N504 461.6 461 462 99 C24H39N504 461.6 461 462 109 C29H41N505 539.7 539 540 110 C29H41N505 539.7 539 540 111 C30H41N306 539.7 539 540 112 C31H44N405 552.7 552 553 120 C30H38N405 534.6 534 535 121 C32H45N506 595.7 595 596 122 C31H43N404C1 571.2 570 571 _ 123 C29H41N504 523.7 523 524 124 C29H41N504 523.7 523 524 125 C30H40N405 536.7 536 537 -126 C32H46N405 566.7 566 567 127 C30H38N603S 562.7 562 563 128 C32H46N405 566.7 566 567 129 C35H46N404 586.8 586 587 130 C29H42N404S 542.7 542 543 131 C31H43N404C1 571.2 ' 570 571 132 C31H43N404C1 571.2 570 571 133 C31H43N404F 554.7 554 555 , 134 C251-137N403C1 477.0 476 477 135 C31H45N505 567.7 567 568 136 C34H45N504 587.8 587 588 137 C28H41N504S 543.7 543 544 138 C30H42N504C1 572.1 571 572 139 C30H42N504C1 572.1 571 572 140 C30H42N504F 555.7 555 556 141 C32H44N405 564.7 564 565 142 C35H44N404 584.7 584 585 143 C29H40N404S 540.7 540 541 144 C31H41N404C1 569.1 568 569 145 C31H41N404C1 569.1 568 569 146 C31H41N404F 552.7 552 553 147 C31H43N505 565.7 565 566 148 C34H43N504 585.7 585 586 149 C30H40N504C1 570.1 569 570 150 C30H40N504C1 570.1 569 570 151 C30H40N504F 553.7 553 554 152 C29H41N505 539.7 539 540 153 C32H41N504 559.7 559 560 _ 154 C26H37N504S 515.7 515 516 155 C28H38N504C1 544.1 543 544 156 C28H38N504C1 544.1 543 544 157 C28H38N504F 527.6 527 528 158 C27H37N604C1 545.1 544 545 159 C31H44N405 552.7 552 553 160 C31H44N405 552.7 552 553 161 C31H45N504 551.7 551 552 _ 162 C31H44N404 536.7 536 ______ 537 163 C31H44N404 - 536.7 536 537 164 C31H44N404 536.7 536 537 -_ 165 C31H44N405 552.7 552 553 166 C31H44N405 552.7 552 553 _ 167 C32H42N404S 578.8 578 579 _ 168 C28H40N404S 528.7 528 529 169 C31H43N404C1 571.2 570 571 170 C30H40N404C12 591.6 590 591 171 C30H40N404F2 558.7 558 559 172 C32H46N406 582.7 582 583 173 C34H43N305 573.7 573 574 . 174 C31H43N306 553.7 553 554 ' 175 C31H44N405 552.7 552 553 176 C31H44N405 552.7 552 553 177 C29H40N405 524.7 524 525 178 C29H40N406 540.7 540 541 179 C32H40N405 560.7 560 561 180 C26H36N405S 516.7 516 517 181 C28H37N405C1 545.1 544 545 182 C28H37N405C1 545.1 544 545 . 183 C28H37N405F 528.6 528 529 184 C31H40N604 560.7 560 561 185 C27H37N604C1 545.1 544 545 . 186 ' C31H40N605 576.7 576 577 187 C31H41N404F3 590.7 590 591 188 C30H41N404F 540.7 540 541 189 C30H41N506 567.7 567 568 190 C33H42N404S 590.8 590 591 191 C32H44N405 564.7 564 565 192 C31H40N404C12 603.6 602 603 193 C31H40N404F2 570.7 570 571 194 C32H48N606 612.8 612 613 195 C32H46N405 566.7 566 ______ 567 196 C32H43N604C1 611.2 610 611 197 C32H45N605C1 629.2 628 629 , 198 C32H43N404C1 583.2 582 583 199 C27H39N406C1 551.1 550 551 200 C31H39N404C1 567.1 566 567 201 C34H42N404 570.7 570 571 202 C31H42N405 550.7 550 551 203 C30H40N505C1 586.1 585 586 204 C29H40N704C1 586.1 585 586 205 C32H45N404C1 585.2 584 585 206 C29H40N506SC1 622.2 621 622 207 C29H39N605C1 587.1 586 587 208 C29H41N705 567.7 567 568 209 C30H41N506 567.7 567 568 210 C31H45N505 567.7 567 568 211 C30H42N504C1 572.1 571 572 212 C31H44N504C1 586.2 585 586 213 C30H40N40512 790.5 790 791 214 C30H42N406 554.7 554 555 215 C30H43N505 553.7 553 554 216 C32H43N404C1 583.2 582 583 , 217 C31H4ON404FCI 587.1 586 587 , 218 C31H43N404C1 571.2 570 571 .-219 C30H40N404C12 591.6 590 591 220 C31H43N404F 554.7 554 555 221 C30H4ON404FC1 575.1 574 575 222 C34H50N405 594.8 594 595 223 C32H44N406 580.7 580 581 224 C36H48N404 600.8 600 601 225 C37H48N405 628.8 628 629 226 C39H49N504S 683.9 683 684 227 C42H52N404 676.9 676 677 Notes 1. Molecular formulas and molecular weights (MW) are calculated automatically from the structure via ActivityBase software (IDBS, Guildford, Surrey, UK) or, for MW only, from the freeware program Molecular Weight Calculator v. 6.32 2. M+H obtained from LC-MS analysis using the General Method as described 3. All analyses conducted on material after preparative HPLC purification BIOLOGICAL METHODS AND RESULTS
s The compounds of the present invention were evaluated for their ability to interact at the human motilin receptor utilizing a competitive radioligand binding assay as described in Method B1. Further characterization of the interaction can be performed utilizing the functional assays described in Methods B2, B3 and B4. Some of these methods can be conducted, if so desired, in a high throughput manner to permit the simultaneous 10 evaluation of many compounds. Other assays have also been described that are suitable for HIS, such as that based upon the stable expression of a synthetic gene for the human motilin receptor.
Results for the examination of representative compounds of the present invention using 15 Method B1 are presented in Table 3. The binding activity is listed as ranges with the following levels: A = 0.001-0.10 pM; B = 0.10-1.0 pM; C = 1.0-10.0 pM. In addition, the assay results of two additional compounds using this Method are shown below.
As can be observed, this demonstrates the activity of a representative bicyclic compound of Formula IV of the invention, which resulted from incorporation of D-proline as the second 20 recognition building block. Significantly, the lack of binding activity obtained with compound 121, which is the linear analogue of compound 1 (K1= level B), illustrates the critical importance of the cyclic structure to attaining the desired interaction.
(HN
7"))1 * 1'44" ONH * /1'4" ONH2 HO =HO
HN
NHV.4' =

=
Compound 120 Compound 121 IC; =level B Ki > 10 ptM

Competitive binding curves for two representative compounds of the invention (Compounds 8 and 11) are presented hereinbelow:
Target: hMTL-R
Radioligand: [1251]rnotilin ,,t/ 4/11 OH
H

100- f 0 0 ..unnH
c Ni 1=%., HN\

NH

ci" 25-K =185 nM
----Compound 8 Log M of Compound 8 Target: hlIATL-R
Radioligand: [1251imotilin '03 75-I

411*
(.) 0.
u) 25- = 79.3 nM =

Compound 11 Log M of Compound 11 For determination of functional significance of the binding, the compounds are preferably o tested in the Aequorin assay as described in Method B2, although the procedure of Method B3 is also applicable. As can be seen from the data presented in Table 4, the representative compounds examined act as antagonists at the motilin receptor and are devoid of agonist activity at the concentrations studied. The functional activity is listed as ranges with the following levels: A = 0.001-0.10 pM, B = 0.10-1.0 pM. The higher sensitivity of the assay of Method B2, almost 100 times that of Method C, makes it the preferred one for this assessment. This is evident in the EC50 values obtained in each for the positive agonist standard, motilin. Additionally, Method B2 measures the actual signaling event, which makes it more relevant to the effect that is desired, whereas the assay of Method B3 simply measures GTP turnover.
Table 4: Demonstration of Antagonist Activity at the Motilin Receptor Aequorin (Method B2)1 Compound Binding (Ki) 1050 Motilin 0.6 not applicable (human, porcine)2 "Activity is listed as ranges with the following levels: A = 0.001-0.10 pM; B
= 0.10-1.0 pM
2Human and porcine motilin are the same peptide.
In addition, a common and scientifically-accepted ex vivo assay for the measurement of agonist or antagonist activity at the motilin receptor is the contraction of rabbit duodenum or other gastrointestinal smooth muscle tissue. A2-A4 Agonists are defined as compounds that induce >50% contraction relative to the motilin peptide, whereas antagonists are defined as compounds that cause >50% inhibition of the response to motilin.
Compounds of the present invention have shown significant antagonist activity in this assay. For example, compound 144 exhibited a pA2 = 6.95, while compound 165 had a pA2 =
7.17, as calculated from the Schild plots of the response obtained at various concentrations as described in Method B4.
Gastric motility is generally measured in the clinical setting as the time required for gastric emptying and subsequent transit time through the GI tract. Gastric emptying scans are well known to those skilled in the art an, briefly, comprise use of an oral contrast agent, such as barium, or a radiolabeled meal. Solid and liquids can be measured independently.

A test food or liquid is radiolabeled with an isotope (9gniTC) and after ingestion or administration, transit time through the GI tract and gastric emptying are measured by visualization using gamma cameras. These studies are performed before and after the administration of the therapeutic agent to quantify the efficacy of the compound.
Example Method B1: Competitive Radioligand Binding Assay (Motilin Receptor) Materials:
= Membranes were prepared from CHO cells stably transfected with the human motilin receptor and utilized at a quantity of 1.5 pg/assay point. [PerkinElmerTM
SignalScreen Product #6110544]
= [125Q-Motilin (PerkinElmer, #NEX-378); final concentration: 0.04-0.06 nM
= Motilin (BachemTM, #H-4385); final concentration: 1 M
= Multiscreen Harvest plates-GF/B (Millipore-rm, #MAHFB1H60) = Deep-well polypropylene titer plate (Beckman CoulterTM, #267006) = TopSeal-A* (PerkinElmer, #6005185) = Bottom seal (Millipore, #MATAHOPOO) = MicroScint-0* (PerkinElmer, #6013611) Assay Volumes:
= 150 i.tL of membranes diluted in binding buffer = 10 AL of compound diluted in binding buffer = 10 p,L of radioligand (112511-Motilin) diluted in binding buffer Final Test Concentrations (N=11) for Compounds:
10, 5, 2, 1, 0.5, 0.2, 0.1, 0.05, 0.02, 0:01, 0.005 pM.
Compound Handling:
Compounds were provided frozen on dry ice at a stock concentration of 10 mM
diluted in 100% DMS0 and stored at -20 C until the day of testing. On the test day, compounds = trademarks were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO
concentration in the assay was 0.5%.
Assay Protocol:
In deep-well plates, diluted cell membranes (1.5 t.tg/mL) are combined with 10 pL of either binding buffer (total binding, N=5), 1 p,M motilin (non-specific binding, N=3) or the appropriate concentration of test compound. The reaction is initiated by addition of 10 ill of [1251]-motilin (final conc. 0.04 ¨ 0.06 nM) to each well. Plates are sealed with TopSeal-A, vortexed gently and incubated at room temperature for 2 hours. The reaction is arrested by filtering samples through pre-soaked (0.3% polyethyleneimine, 2 h) Multiscreen Harvest plates using a Tomtec Harvester, washed 9 times with 500 pL of cold 50 mM Tris-HCI (pH
7.4), and than plates are air-dried in a fumehood for 30 minutes. A bottom seal is applied to the plates prior to the addition of 25 pt of MicroScint-0 to each well.
Plates are than sealed with TopSeal-A and counted for 30 sec per well on a TopCount Microplate Scintillation and Luminescence Counter (PerkinElmer) where results are expressed as counts per minute (cpm).
Data are analyzed by GraphPadTM Prism (GraphPad Software, San Diego, CA) using a variable slope non-linear regression analysis. K1 values were calculated using a Kd value of 0.16 nM for [125I]-motilin (previously determined during membrane characterization).
Dmax = I - test concentration with maximal displacement ¨ non-specific binding x 100 total binding ¨ non-specific binding where total and non-specific binding represent the cpm obtained in the absence or presence of 1pM motilin, respectively.
Example Method B2: Aequorin Functional Assay (Motilin Receptor) Materials:
= Membranes were prepared using AequoScreen TM (EUROSCREEN, Belgium) cell lines expressing the human motilin receptor (cell line ES-380-A; receptor accession #AF034632). This cell line is constructed by transfection of the human motilin receptor into CHO-K1 cells co-expressing Ga16 and the mitochondrially targeted Aequorin (Ref #ES-WT-A5).
= Motilin (Bachem, #H-4385) = Assay buffer: DMEM-F12 (Dulbeccoe's Modified Eagles Medium) with 15 mM
HEPES
5 and 0.1% BSA (pH 7.0) = Coelenterazine (Molecular ProbesTM, Leiden, The Netherlands) Final Test Concentrations (N=5) for Compounds:
10, 3.16, 1, 0.316, 0.1 pM.
Compound Handling:
Compounds were provided as dry films at a quantity of approximately 1.2 mol in pre-formatted 96-well plates. Compounds were dissolved in 100% DMSO at a concentration of 10 mM and stored at -20 C until further use. Daughter plates were prepared at a concentration of 500 M in 30% DMSO with 0.1% BSA and stored at -20 C until testing.
On the test day, compounds were allowed to thaw at room temperature and than diluted in assay buffer according to the desired test concentrations. Under these conditions, the maximum final DMSO concentration in the assay was 0.6%.
Cell Preparation:
Cells are collected from culture plates with Ca2+ and Mg2+-free phosphate buffered saline (PBS) supplemented with 5 mM EDTA, pelleted for 2 minutes at 1000 x g, resuspended in assay buffer (see above) at a density of 5 x 106 cells/mL and incubated overnight in the presence of 5 M coelenterazine. After loading, cells were diluted with assay buffer to a concentration of 5x 105 cells/mL.
Assay Protocol:
For agonist testing, 50 I of the cell suspension was mixed with 50 I of the appropriate concentration of test compound or motilin (reference agonist) in 96-well plates (duplicate samples). The emission of light resulting from receptor activation was recorded using the Functional Drug Screening System 6000 'FDSS 6000' (Hamamatsu Photonics K.K., Japan).

For antagonist testing, an approximate EC80 concentration of motilin (i.e. 0.5 nM, 100 iAL) was injected onto the cell suspension containing the test compounds (duplicate samples) 15-30 minutes after the end of agonist testing and the consequent emission of light Results are expressed as Relative Light Units (RLU). Concentration response curves were analyzed using GraphPad Prism (GraphPad Software, San Diego, CA) by non-linear regression analysis (sigmoidal dose-response) based on the equation E=Ema(1+EC50/C)n Example Method B3: FlashPlate Motilin [35S]-GTPyS Functional Assay 20 Materials:
= Membranes were prepared from CHO cells stably transfected with the human motilin receptor and utilized at a quantity of 1.5 pg/assay point.
[PerkinElmer SignalScreen Product #61105441 = GTP7S (Sigma, #G-8634) = Motilin (Bachem, #H-4385) = 96-well FlashPlate microplates (PerkinElmer, #SMP200) = Deep-well polypropylene titer plate (Beckman Coulter, #267006) = TopSeal-A (PerkinElmer, #6005185) GDP, 0.1% BSA

Assay Volumes:
= 25 I_ of compound diluted in assay buffer = 25 I_ of assay buffer (agonist assay) or 0.6 FM motilin (0.1 M final concentration) diluted in assay buffer (antagonist assay) = 100 I_ of [35S]-GTPyS diluted in assay buffer Final Test Concentrations (N=12) for Compounds:
50, 20, 10, 5, 2, 1, 0.5, 0.2, 0.1, 0.05, 0.02, 0.01 pM.
Compound Handling:
Compounds were provided frozen on dry ice at a stock concentration of 10 mM
diluted in 100% DMSO and stored at -20 C until the day of testing. On the test day, compounds Were allowed to thaw at room temperature and than diluted in assay buffer according to is the desired test concentrations. Under these conditions, the maximum final DMSO
concentration in the assay was 0.5%.
Assay Protocol:
CHO membranes were immobilized into 96-well FlashPlate microplates. Test compound, GTPyS, motilin and [35S]-GTP7S were combined in each well according to the Assay Volumes described above.
For the assay to measure agonist activity, an additional 25 I of buffer was added to each well in addition to 25 1._ of either buffer (basal value, N=4), 1 M (final conc.) motilin (Emax value, N=3), 25 M (final conc.) GTPyS (non-specific value, N=4), or the appropriate concentration of test compound (N=3).
For the assay to measure antagonist activity, an additional 25 L. of either buffer (unstimulated control) or motilin (0.1 M final conc.) is added to each well, in addition to either 25 L. of buffer (basal value, N=3), 1 M (final conc.) motilin (Emax value, N=3), 25 M (final conc.) GTPyS (non-specific value, N=4), or the appropriate concentration of test compound (N=3).

The reaction is initiated by addition of 100 mL of [35q-GTPyS to each well.
Each plate is sealed (TopSeal-A) and incubated in the dark at room temperature for 150 min.
Then, plates are counted for 30 seconds per well on the TopCount NXT.
Data were analyzed by GraphPad Prism 3.0 (GraphPad Software, San Diego, CA) using non-linear regression analysis (sigmoidal dose-response) for the calculation of IC50/EC50 values.
Emu (agonist) or Dm" (antagonist) = Top ¨ Bottom X 100 Bottom Where Top and Bottom correspond to the top and bottom values of the dose-response curve calculated by GraphPad Prism).
Example Method B4: Rabbit Duodenum Contractility Assay Duodenal segments were vertically suspended in organ chambers of 10 mL filled with Krebs buffer and connected to an isotonic force transducer, with a preload of 1 g. After a stabilization period, the muscle strips were challenged with 104 M
acetylcholine and washed. This was repeated until a stable maximal contraction was obtained (2-3 times), with an interval of at least 20 minutes.
After a stable base line was reached, test compounds were added to the bath.
After 15 min incubation, a dose response to motilin was recorded by adding logarithmically increasing concentrations of motilin to the bath (final concentration le to 10-6 M). A blank experiment (no test compound present) was also performed. At the end of the dose response curve, a supramaximal dose of acetylcholine (104 M) was given and this response was used as a reference (100% contraction).
The results of experiments at different concentrations of test compound were combined and analyzed to derive the pA2 value from the Schild plot.

, Table 3: Binding activity of selected compounds R1 R3 Re T KI1,2 I

B
_ A
_ _V
z, B

--f< ___V 23 A
iw/ =
* x ' L/
--- - $ 0):3 -6 jj il OH 1"--( ;2121.S 11023 B

B
7 >`' . OH
-1--( -1-\
\ 1110 8 >' *
OH ¨IX / = B

II B
9 >rj --IX -IX

, --)----- ',,, 10 >õ .
oH -IX A $1 Ox , 11 +// 0 0/"-x A
/
z3 B
12 >j = oH
--1- e,,,,,,_, X
B
>1/4 .
-1-( --1-1)--- Z313 0 , "C-K ____ AX -' 15 x =
--1¨/ 0 Z3 A
1--( ci 16 >,, * ci --1- -1-7- 11101 : A
17 -1 ,Q Z3 / Wy, --I---( N
+/-- e.,,,,,......õ,õõA
B
H
18 \ . B
_,/ Z3 19 11 F -1¨/ - Z3 A
/
1 ox 20 N. 41 ¨i---( ---,..,., 0 oxz3 B

L/\------ Z3 A
X----K 2 ---1- 1 x \
->" *
\ -1--- ¨1.¨/ 110 0.....õ...õ:3 I: 111 111111 ill 0 :3 A
sz 01 A
I NCI 011 1111110 0:
A
1141 OH!'" AP x 111111 0,xZ3 Cs>

11111 xZ3 30 µ O.

II >4 11 õ 1111 X
=
111 411111 µOH 1111 IP 0 X 3 OH 4111 11110 o I
40 11, OH
-111111 L,0 I
OH >1, 411 11),0 .111 4.

* 1111 * OH OX
= OH a NH2 ell 0 I

OH
-1---( -k 1 B
68 , ii oF +.< >, __OH 110 c,..)3 ,r.,44 . Z3 B

o-1 0/'')( N
H
60 .r-ri = Z3 C
OA 1¨( >j . 0\
$ OX
t, 66 >' . B
OF
¨1¨.---...- Z3 71 >, .
OH
¨1---( ¨1¨/ X. * '''''''23 B
= >'' --F( .111 OH /
I OX
}
',.
. 76 rj 11 Z3 C
, OH
O'C

l: +/---Y
80 >sJ it -1---( Z3 B
-, oi :o 4 .
--IX H B
>

, 1 1 x \ Z3 B
I ox 88 >4 11 Z3 ¨I C
0\ X
, / \ +// I.1 0 X

-1- +// Z3 C

i< ¨VZ3 C

91 >j . ¨1¨/ Z3 C
0 -I ¨1--/
* 0 X

92 >4 11 B

1 ( ¨V 23 * OX

),, = H ¨V
OH
* OX
97 >' .

* 0 X
98 )') =
OH
¨IXXNz3 C
99 >j = OH
C

, 109 \,,, . -IX 17 OH NH2 1110 0.....,,,,x23 B
110 >4 = OH V
¨1¨( ¨

ox 111 .
OH ¨1¨( +/ (Z3 B
'Clx . , >j OH
---F( -1--/
__ 122 , B
' I' 123 , B

0,.,.. X
124, B
)=

-1- +// Z3 ...,N
125 ____________________________________________________________________ B
+///

H - --<
IP
126 ____________________________________________________________________ B

H
-1- _ ) 127 ____________________________________________________________________ B

X
j1 -IX -V 0 I ,\ ________________________ -1-( - i 10 __________________________________________________________________________ _ A

\\!
00 -F.< -) . _____________________________________________________________________ B

0_S
. Z3 '`,...,.....
--f<
_______________________________________________________________________ A

I
-1---( _ ) CI
, _________________________________________________________________________ , 133 ' A
>( .
' I -1--( i C
CI
134 '. \
) , , Oct ¨IX _ _______________________________________________________________________ B
135''.7:'=,, '''''', I W z3 -IX _ ) 1 136 == _______ "
) 77.c....õ...: I B
r 00 -1---( _ 0/' \.,. x ox rsIzx3 z3 X
CI

(¨x A

j>
ox cx A
144 \s/

X

) 85 ., , ¨I
1 X _ _________________________________ A Z3 * 0 X
, , , 147 \ B
)., w X
-\
I

,>\ 0/
(D -1-( -ox 150 __________________________________________________________ B
\
j> rN z3 . ' 10 ¨IX _ x ci j 151 >. 0 B
F 1*--( .,,Nwõ z3 1 > .,,,,,,,,,, X
----.

1 --i--( ¨ _.-/NH2 , , , -../ Z3 -1- C) x I
154 ___________________________________________________________ B

i ___/NFI2 ,), a . NH2 Z3 -1---( - -/
156µ(....

ci -___/ 1101 ----( o x \
/ \ 0 NH2 lei 0 X
F .
.

¨

¨1- --/
I
x .?
_l___// Z3 --\
OH

- -( OH
-, ¨1---( -1-7--- Z3 ¨IX

\
¨1¨( 7 \ 410 ___ 7Z3 _ Z3 166 -,.
_ B
\

0 crõ,õõ,õ,,,, X

0 / ¨1---( --/
-7 '0 X

a 100 ________________________________________________________________ -IX ____ _.1 Z3 el crõ,..,,..,...õ,õ.,= X
170 , \ a - e 1-( ___/ Z3 CI -0 ,,,,,, X

\ F
"i<

.I K
\
-F

o V. -1---( - / 1110 c).,,,, x 173 ;/' B
/

I C) x _ 40 0,x OH
. 176 B

-1---( OH
, OH

) O

I o x : Z3 jOH
x 4 -- ¨1----(/OH Z3 I /
¨/_ 01 ,,,.....õ,,,,,..õ......x ..)\
11101 a¨I OH
c).,,,, ZX3 I
, X ____ 7 182 \ A

CI "¨IX 2s( . OH
183\ B
.i'., _ 7 Z3 I

'-.." NH2 ,,N.',õõ,=,. z3 , 185 \ B
/-Nz3 ca 186 te/7''''NH B
, µ=\ 0 c*1 F
, s , 10 ¨IX
¨ 7 10 0õ,.. XZ3 , , F

õ F
i \ ----( /0 1 _ Z3 )\
¨1- ...._.7 Z3 r ..._ I> $ 0 N
_ 191 ' A

192 2' \ ci A
=
a -1---( ¨7 _ , ¨1¨
¨ I> Z3 F 0 ./x 194 B ' i .õ
c--1--( ¨ z3 .
0 N, I

i'\ 410 0- ¨1- _ / Z3 $ 0 X

_ 197 µ a o \ .z3 I
ON X
¨

- 198 . A -0 a ,-, 1.-- j> 11 0 X
¨
199¨ B
¨7 \ a , Her '50H
200 .
ci 0 x 23 A .

201 "-V --õ.

i 10.4 1 , x 11¨ 40 73 111 i oV'-'''-7 203 ' \ Z3 I
Ot4---(\ ,3 x 0 a 11.ji NH
204 , \ 7...,...., )1õ, all N NH2 0 cz3 x I
= H

206 , 0 1111 )----2\ ioi 1 0 0z-vx _____,1 , 206 , 11 Z3 I
Htsi---S--- 0 \ 1 j x I

1101 a , JO

;3x I
, jjm¨ \ \NN2 I

208 r ,)L=-=
I

OH
, ¨1--( HN
( 23 OH

õ
= 0 ¨IX N ( m le ox OH ¨i-I

\

j ¨1---( , N Z3 CI 1 10 x Aõ, ' H 1110 v=,.,,,,, X
CI
213 , B

___ $ 0 X
OH .
I .

OH

V
-1--( 0 crõ.....,.....õ,,,,,õ,x \ Z3 I-_ .. ct ¨1---( _____? 1 1 ozx3 -1--( \ a ¨IX
_ CI
- -./ a n 1---( 11101 o x \ 0 F

lei 0- X

2 '`\ Z3 A

OH -1- - /14.1 0 X

OH -/

/
) Z3 0 ,x ,.
_i_<
00 _ , .'r ) Z3 /

) Z3 00 , .//
/
) o 7 5 Notes Radioligand competitive binding assays performed using Method B1 Values reported as ranges: A = 0.001-0.100 M; B = 0.100-1.0 ,M;
10 C = 1.0-10.0 [LM
X is NH except for:
Compound 223 and 225, X is:

---( \
N---c----71--- ' Compound 224, X is NMe Compound 226, X is:
=õ/
)c c/S

Compound 227, X is Z2 and Z3are NH except for compounds 30, 173 and 174 and where Z1 is 0 and compound 111 where Z2 is O.
=
R2, R4 and R5 are hydrogen except for compound 85 where it is:
m, ni and p are zero.
The scope of the claims should not be limited by the preferred embodiments set forth in the examples, but should be given the broadest interpretation consistent with the description as a whole.

Claims (16)

1. A compound represented by the general formula (l):
or pharmaceutically acceptable salts thereof wherein:
Z1, Z2, and Z3 are independently NR10, wherein R10 is selected from the group consisting of hydrogen and lower alkyl;
R1 is -(CH2)q R11l, wherein q is 0, 1 or 2 and R11 is selected from the group consisting of:
wherein A1, A2, A3, A4 and A5 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, hydroxy, alkoxy, and nitro;
B1, B2, B3, B4, B5 and B7 are independently NR14a, S or O, wherein R14a is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; and 66 and B8 are independently N or CH;
R2 iS hydrogen;
R3 is selected from the group consisting of: ¨(CH2)s CH3, -CH(CH3)(CH2)t CH3, -(CH2)u CH(CH3)2, -C(CH3)3, and -(CH2)y-R21, wherein:
s is 0, 1, 2 or 3;
t is 1 or 2;
u is 0 or 1;
y is 0, 1 or 2; and R21 is selected from the group consisting of:

wherein z1 is 1, 2, 3 or 4 and E1 is optionally present and selected from the group consisting of hydroxy and alkoxy;
R4 and R5 are each hydrogen;
R6 is selected from the group consisting of hydrogen, ¨(CH2)aa CH3, -CH2SCH3, -CH2CH2SCH3, -(CH2)bb CH(CH3)2, -CH(CH3)(CH2)cc CH3, -(CH2)dd-NR22R23, and -(CH2)ee R24, wherein:
aa is 0, 1, 2 or 3;
bb is 0 or 1;
cc is 1 or 2;
dd is 1, 2, 3 or 4;
ee is 0, 1 or 2;
R22 and R23 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, amido, amidino, sulfonyl and sulfonamido;
R24 is selected from the group consisting of hydroxy, alkoxy, and wherein E2 is optionally present and is selected from the group consisting of hydroxy and alkoxy; B9 and B10 are independently selected from the group consisting of NR14b, S and O, wherein R 14b is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; B11 is selected from the group consisting of N and CH; and z2 is 1, 2, 3 or 4; and X is NR8, wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and amidino;
with the provisos that when Z1, Z2 and Z3 are all NH, R1 is:
and when Z1, Z2 and Z3 are all NH, R1 is:
then R2 is not:
and when Z1, Z2 and Z3 are all NH, R2 iS:
and R3 is:

then R1 is not:
wherein Rp is hydrogen or a protecting group;
m, n1 and p are 0; and T is selected from the group consisting of:
wherein U3 is CH2, L2 is CH or N;
L5 is CH or N;
L6 is CR52R53 or O;
R46 is H or CH3, R52, R53, R54, R55, R56 and R57 are independently selected from the group consisting of hydrogen and lower alkyl;
RH iS selected from the group consisting of halogen and amidino; and (X) is the site of a covalent bond to X in formula (l); and (Z3) is the site of a covalent bond to Z3 in formula (l).
2. The compound of claim 1, wherein the substituted alkyl in the definition of A1, A2, A3, A4 and A5, is trifluoromethyl.
3. The compound of claim 1 , wherein R11 is selected from the group consisting of:
wherein Ra and Rb are independently selected from the group consisting of CI, F, CF3, OCH3, OH, CH3 and C(CH3)3.
4. The compound of claim 1, wherein m, n, and p are 0; X, Z1, Z2 and Z3 are NH; and R2, R4 and R5 are hydrogen, represented by formula (III):
wherein R1, R3, R6 and T are as defined in claim 1 .
5. The compound of claim 1 selected from the group consisting of:

6. A compound represented by the general formula (l):

or pharmaceutically acceptable salts thereof wherein:
Z1. Z2, and Z3 are independently NR10, wherein R10 is selected from the group consisting of hydrogen and lower alkyl;
R1 is -(CH2)q R11, wherein q is 0, 1 or 2, and R11 is selected from the group consisting of:
wherein A1 , A2 and A3 are each optionally present and are independently selected from the group consisting of halogen, alkyl, substituted alkyl, hydroxy, alkoxy and nitro;
B1 , B2, B3, B4, B5 and B7 are independently NR14a, S or O wherein R14a is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido;
B6 and B8 are independently N or CH;
R2 is hydrogen;
R3 is selected from the group consisting of: ¨(CH2)s CH3, -CH(CH3)(CH2)t CH3, -(CH2)u CH(CH3)2, -C(CH3)3, and -(CH2)y-R21, wherein:
s is 0, 1, 2 or 3;
t is 1 or 2;
u is 0 or 1;
y is 0, 1 or 2; and R21 is selected from the group consisting of:

wherein z1 is 1, 2, 3 or 4 and E1 is optionally present and selected from the group consisting of hydroxy and alkoxy;
R4 and R5 are each hydrogen;
R6 is independently selected from the group consisting of hydrogen, ¨(CH2)aa CH3, -CH2SCH3, -CH2CH2SCH3, -(CH2)bb CH(CH3)2, -CH(CH3)(CH2)CH3, -(CH2)dd-NR22R23, and -(CH2)ee R24, wherein aa is 0, 1, 2 or 3;
bb is 0 or 1;
cc is 1 or 2;
dd is 1, 2, 3 or 4;
ee is 0, 1 or 2;
R22 and R23 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, amido, amidino, sulfonyl and sulfonamido;
R24 is selected from the group consisting of hydroxy, alkoxy, wherein E2 is optionally present and is selected from the group consisting of hydroxy and alkoxy; B9 and B10 are independently selected from the group consisting of NR14b, S and O, wherein R14b is selected from the group consisting of hydrogen, alkyl, substituted alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl and sulfonamido; B11 is selected from the group consisting of N and CH; and z2 is 1 , 2, 3 or 4; and X is NR8, wherein R8 is selected from the group consisting of hydrogen, lower alkyl, substituted lower alkyl, formyl, acyl, carboxyalkyl, carboxyaryl, amido, sulfonyl, sulfonamido and amidino;
with the provisos that when Z1, Z2 and Z3 are all NH, R1 is:
and when Z1, Z2 and Z3 are all NH, R1 is:
then R2 is not:
and when Z1, Z2 and Z3 are all NH, R2 is:
and R3 is:

then R1 is not:
wherein Rp is hydrogen or a protecting group;
m, n1 and p are 0; and T is selected from the group consisting of:

wherein R30 and R31 are selected from the group consisting of hydrogen and methyl;
and (X) is the site of a covalent bond to X in formula (l); and (Z3) is the site of a covalent bond to Z3 in formula (l).
7. A compound as claimed in claim 6, wherein the substituted alkyl in the definition of A1, A2 or A3, is trifluoromethyl.
8. A pharmaceutical composition comprising:
(a) a compound as defined in any one of claims 1 to 7; and (b) a pharmaceutically acceptable carrier.
9. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for treating a gastrointestinal disorder associated with the motilin receptor or motility dysfunction in humans or other mammals.
10. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for treating a gastrointestinal disorder associated with hypermotility or hypermotilinemia in humans or other mammals.
11. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for treating irritable bowel syndrome or dyspepsia in humans or other mammals.
12. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for treating Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome or celiac disease in humans and other mammals.
13. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for the making of a medicament for treating a gastrointestinal disorder associated with the motilin receptor or motility dysfunction in humans or other mammals.
14. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for the making of a medicament for treating a gastrointestinal disorder associated with hypermotility or hypermotilinemia in humans or other mammals.
15. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for the making of a medicament for treating irritable bowel syndrome or dyspepsia in humans or other mammals.
16. Use of a compound as defined in any one of claims 1 to 7 or of a pharmaceutical composition as defined in claim 8, for the making of a medicament for treating Crohn's disease, gastroesophogeal reflux disorders, ulcerative colitis, pancreatitis, infantile hypertrophic pyloric stenosis, carcinoid syndrome, malabsorption syndrome, diarrhea, atrophic colitis or gastritis, gastrointestinal dumping syndrome, postgastroenterectomy syndrome or celiac disease in humans and other mammals.
CA2528375A 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor Expired - Fee Related CA2528375C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US47922303P 2003-06-18 2003-06-18
US60/479,223 2003-06-18
PCT/CA2004/000913 WO2004111077A1 (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor

Publications (2)

Publication Number Publication Date
CA2528375A1 CA2528375A1 (en) 2004-12-23
CA2528375C true CA2528375C (en) 2013-11-19

Family

ID=33551871

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2528375A Expired - Fee Related CA2528375C (en) 2003-06-18 2004-06-18 Macrocyclic antagonists of the motilin receptor

Country Status (9)

Country Link
US (9) US7521420B2 (en)
EP (2) EP2210612B1 (en)
JP (1) JP4928261B2 (en)
AT (1) ATE457995T1 (en)
CA (1) CA2528375C (en)
DE (1) DE602004025569D1 (en)
DK (1) DK1633774T3 (en)
ES (1) ES2338789T3 (en)
WO (1) WO2004111077A1 (en)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2284459C (en) * 1999-10-04 2012-12-11 Neokimia Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
US7491695B2 (en) * 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
US8921521B2 (en) 2003-06-18 2014-12-30 Ocera Therapeutics, Inc. Macrocyclic modulators of the Ghrelin receptor
JP4928261B2 (en) * 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 Macrocyclic antagonist of motilin receptor
US20090198050A1 (en) * 2003-06-18 2009-08-06 Tranzyme Pharma Inc. Macrocyclic Modulators of the Ghrelin Receptor
US7476653B2 (en) 2003-06-18 2009-01-13 Tranzyme Pharma, Inc. Macrocyclic modulators of the ghrelin receptor
USRE42624E1 (en) 2003-06-18 2011-08-16 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
USRE42013E1 (en) 2003-06-18 2010-12-28 Tranzyme Pharma Inc. Macrocyclic modulators of the ghrelin receptor
ATE419267T1 (en) * 2003-07-31 2009-01-15 Tranzyme Pharma Inc SPACIALLY DEFINED MACROCYCLES CONTAINING PEPTIDE BINDING SURROGATES
ATE490264T1 (en) * 2003-07-31 2010-12-15 Tranzyme Pharma Inc SPACIOUSLY DEFINED MACROCYCLIC COMPOUNDS SUITABLE FOR DRUG DISCOVERY
JP5739766B2 (en) * 2004-06-18 2015-06-24 オセラ セラピューティクス, インコーポレイテッド Use of macrocyclic modulators of ghrelin receptor
WO2006046977A1 (en) * 2004-10-26 2006-05-04 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and methods of using the same
CA2583345A1 (en) * 2005-06-13 2006-12-28 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US20090275648A1 (en) * 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US8088733B2 (en) 2006-07-06 2012-01-03 Tranzyme Pharma Inc. Methods of using macrocyclic agonists of the ghrelin receptor for treatment of gastrointestinal motility disorders
US9133235B2 (en) 2006-09-11 2015-09-15 Ocera Therapeutics, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
US8889632B2 (en) 2007-01-31 2014-11-18 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
CN101657436A (en) 2007-02-09 2010-02-24 特兰齐姆制药公司 Macrocyclic ghrelin receptor modulators and using method thereof
AU2015201862B2 (en) * 2007-02-09 2016-10-13 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor modulators and methods of using the same
ES2430067T3 (en) 2007-03-28 2013-11-18 President And Fellows Of Harvard College Sewn polypeptides
US20080287371A1 (en) * 2007-05-17 2008-11-20 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor for modulation of the migrating motor complex
EP2350031A2 (en) * 2008-08-20 2011-08-03 Ensemble Therapeutics Corporation Macrocyclic compounds for inhibition of tumor necrosis factor alpha
AU2009350857A1 (en) * 2009-08-05 2012-02-02 Polyphor Ag Conformationally constrained, fully synthetic macrocyclic compounds
WO2011050270A2 (en) * 2009-10-23 2011-04-28 Tranzyme Pharma, Inc. 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
KR20170058446A (en) 2010-08-13 2017-05-26 에일러론 테라퓨틱스 인코포레이티드 Peptidomimetic macrocycles
TW201806968A (en) 2011-10-18 2018-03-01 艾利倫治療公司 Peptidomimetic macrocycles
AU2013221432B2 (en) 2012-02-15 2018-01-18 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
AU2013221433B2 (en) 2012-02-15 2018-01-18 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
MX2015005244A (en) 2012-11-01 2015-07-14 Aileron Therapeutics Inc Disubstituted amino acids and methods of preparation and use thereof.
US9119832B2 (en) 2014-02-05 2015-09-01 The Regents Of The University Of California Methods of treating mild brain injury
CN112245565A (en) 2014-09-24 2021-01-22 艾瑞朗医疗公司 Peptidomimetic macrocycles and uses thereof
CA2979847A1 (en) 2015-03-20 2016-09-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
KR20180063138A (en) 2015-10-27 2018-06-11 에프. 호프만-라 로슈 아게 Ascenote Burter Baumann's Peptide Giant Ring against Nee
US20170121385A1 (en) 2015-10-28 2017-05-04 Oxeia Biopharmaceuticals, Inc. Methods of treating neurodegenerative conditions
EP3388444A1 (en) * 2017-04-10 2018-10-17 F. Hoffmann-La Roche AG Anti-bacterial peptide macrocycles and use thereof
US11505573B2 (en) 2018-03-28 2022-11-22 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
US11819532B2 (en) 2018-04-23 2023-11-21 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
CN108752214A (en) * 2018-07-09 2018-11-06 上海华堇生物技术有限责任公司 A kind of new preparation process of 2- hydroxy-betas-nitrostyrolene

Family Cites Families (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5008249A (en) 1985-08-31 1991-04-16 Kitasato Kenkyusho Therapeutic method of stimulating digestive tract contractile motion in mammals
US5175150A (en) 1985-08-31 1992-12-29 Kitasato, Kenkyusho Erythromycin derivative
US5695952A (en) 1986-09-12 1997-12-09 Kyowa Hakko Kogyo Co., Ltd. Method for producing Leu13 !motilin
GB8728820D0 (en) 1987-12-09 1988-01-27 Fisons Plc Compounds
US4920102A (en) 1988-04-18 1990-04-24 Eli Lilly And Company Method for treating gastrointestinal disorders
EP0358608A3 (en) * 1988-09-09 1990-10-31 Gesellschaft für Biotechnologische Forschung mbH (GBF) Macrocyclic lacton derivatives with a microbicidal activity
US5459049A (en) * 1989-01-06 1995-10-17 Sanwa Kagaku Kenkyushko Co., Ltd. Motilin-like polypeptide and use thereof
EP0378078A1 (en) * 1989-01-06 1990-07-18 Sanwa Kagaku Kenkyusho Co., Ltd. Motilin-like polypeptide and use thereof
EP0413532A3 (en) * 1989-08-18 1991-05-15 Fisons Plc Macrocyclic compounds
US5143915A (en) * 1990-07-25 1992-09-01 E. R. Squibb & Sons, Inc. Dihydropyrimidine macrocyclic lactones useful as calcium antagonists and agonists
US5196452A (en) * 1991-01-29 1993-03-23 Genelabs Incorporated Macrocyclic anti-viral compound and method
JPH06509326A (en) * 1991-04-09 1994-10-20 アボツト・ラボラトリーズ Macrocyclic lactam prokinetic agents
US5523401A (en) 1991-04-09 1996-06-04 Abbott Laboratories Macrocyclic lactam prokinetic agents
US5604234A (en) * 1991-09-05 1997-02-18 Abbott Laboratories Substituted thiol macrolactam immunomodulators
US5563172A (en) * 1991-09-05 1996-10-08 Abbott Laboratories Macrocyclic amide and urea immunomodulators
US5604294A (en) * 1991-09-05 1997-02-18 Luly; Jay R. Macrocyclic immunomodulators
WO1993004680A1 (en) * 1991-09-05 1993-03-18 Abbott Laboratories Macrocyclic immunomodulators
US5561137A (en) * 1991-09-05 1996-10-01 Abbott Laboratories Thio-heterocyclic macrolactam immunomodulators
US5534632A (en) 1991-09-05 1996-07-09 Abbott Laboratories Macrocyclic carbamate immunomodulators
US5457111A (en) * 1991-09-05 1995-10-10 Abbott Laboratories Macrocyclic immunomodulators
US5708002A (en) * 1991-09-05 1998-01-13 Abbott Laboratories Macrocyclic immunomodulators
US5244889A (en) * 1991-11-26 1993-09-14 Ciba-Geigy Corporation Certain macrocyclic lactam derivatives
DE4200145A1 (en) 1992-01-07 1993-07-08 Kali Chemie Pharma Gmbh 7,10-EPOXY-OXACYCLODODANE DERIVATIVES, METHODS AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS
US5470961A (en) 1992-03-19 1995-11-28 Takeda Chemical Ind., Ltd. 14 and/or 15-hydroxy 6,9 hemiacetal erythromycin derivatives
MY113693A (en) 1992-05-26 2002-05-31 Chugai Pharmaceutical Co Ltd Erythromycin derivatives having an enterokinesis stimulating action
US5328902A (en) * 1992-08-13 1994-07-12 American Cyanamid Co. 7-(substituted)-9-[(substituted glycyl)amido]-6-demethyl-6-deoxytetracyclines
US5292741A (en) * 1992-08-18 1994-03-08 Merck & Co., Inc. Macrocycles incorporating quinazolinones
US5252579A (en) * 1993-02-16 1993-10-12 American Home Products Corporation Macrocyclic immunomodulators
WO1994021634A1 (en) * 1993-03-17 1994-09-29 Abbott Laboratories Substituted alicyclic-aliphatic amine-containing macrocyclic immunomodulators
US5457194A (en) * 1993-03-17 1995-10-10 Abbott Laboratories Substituted aliphatic amine-containing macrocyclic immunomodulators
JPH08507788A (en) * 1993-03-17 1996-08-20 アボツト・ラボラトリーズ Macrocyclic immunomodulator containing substituted alicyclic amine
US5422341A (en) * 1993-08-06 1995-06-06 Ohmeda Pharmaceutical Products Division Inc. Motilin-like polypeptides with gastrointestinal motor stimulating activity
US5470830A (en) 1993-08-06 1995-11-28 Ohmeda Pharmaceutical Products Division Inc. Motilin-like polypeptides that inhibit gastrointestinal motor activity
US5612350A (en) * 1993-11-30 1997-03-18 Abbott Laboratories Macrocyclic immunomodulators with novel cyclohexyl ring replacements
US5624949A (en) 1993-12-07 1997-04-29 Eli Lilly And Company Protein kinase C inhibitors
JP3581731B2 (en) 1993-12-07 2004-10-27 イーライ・リリー・アンド・カンパニー Protein kinase C inhibitor
CA2182476C (en) 1994-02-04 2000-12-26 Robert A. Farr Macrocyclic difluorostatone derivatives useful as antiviral agents
US6100377A (en) * 1994-06-10 2000-08-08 The Trustees Of The University Of Pennsylvania Constrained peptides
AUPM982594A0 (en) * 1994-12-02 1995-01-05 University Of Queensland, The HIV protease inhibitors
GB9511357D0 (en) * 1995-06-06 1995-08-02 Johnson Matthey Plc Improved antiviral compounds
US5811515A (en) 1995-06-12 1998-09-22 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides, and peptidomimetics by catalytic ring closing metathesis
US6124453A (en) * 1995-07-04 2000-09-26 Novartis Ag Macrolides
US6281352B1 (en) * 1995-11-14 2001-08-28 Dupont Pharmaceuticals Company Macrocyclic compounds as metalloprotease inhibitors
DE69600784T2 (en) * 1995-11-20 1999-04-08 Lilly Co Eli Protein kinase C inhibitor
US5888971A (en) * 1996-02-20 1999-03-30 Ortho Pharmaceutical Corporation, Inc. Macrocyclic peptides useful in the treatment of thrombin related disorders
US5849691A (en) 1996-02-20 1998-12-15 The United States Of America As Represented By The Department Of Health And Human Services Peptidomimetic inhibitors of cathepsin D and plasmepsins I and II
IL117426A (en) * 1996-03-10 2005-09-25 Yissum Res Dev Co Synthetic pseudopeptides having osteogenic activity and pharmaceutical compositions containing the same
US5734012A (en) * 1996-05-16 1998-03-31 Ohmeda Pharmaceutical Products Division Inc. Cyclic motilin-like polypeptides with gastrointestinal motor stimulating activity
CA2207137A1 (en) * 1996-06-14 1997-12-14 James Erwin Fritz Scavenger assisted combinatorial process for preparing libraries of secondary amine compounds
US5712253A (en) * 1996-06-18 1998-01-27 Abbott Laboratories Macrocyclic 13-membered ring derivatives of erythromycins A and B
US20020168761A1 (en) 2000-01-24 2002-11-14 Gour Barbara J. Peptidomimetic modulators of cell adhesion
DE19644195A1 (en) * 1996-10-24 1998-04-30 Solvay Pharm Gmbh 10,13,15-Trioxatricyclo [9.2.1.1. · 9 ·. · 6 ·] -pentadecanone derivatives, processes for their preparation and pharmaceuticals containing these compounds
US6100239A (en) * 1996-11-26 2000-08-08 Chugai Seiyaku Kabushiki Kaisha 13-membered ring macrolide compound, medicine containing the same, and process for producing the same
US6867305B2 (en) * 1996-12-03 2005-03-15 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
MX9709494A (en) * 1996-12-04 1998-07-31 Pfizer Cyclic peptidic compounds, process for its production, compositions containing them and use of the same.
US5952320A (en) * 1997-01-07 1999-09-14 Abbott Laboratories Macrocyclic inhibitors of matrix metalloproteinases and TNFα secretion
US6380158B1 (en) 1997-03-24 2002-04-30 Zymogenetics, Inc. Motilin homologs
US6291653B1 (en) 1997-03-24 2001-09-18 Zymogenetics, Inc. Antibodies to motilin homologs
AU726423B2 (en) 1997-03-24 2000-11-09 Zymogenetics Inc. Motilin homologs
EP1015479A4 (en) 1997-04-11 2002-07-24 Lilly Co Eli Combinatorial libraries of peptidomimetic macrocycles and processes therefor
JP3583928B2 (en) 1997-08-15 2004-11-04 中外製薬株式会社 Phenethylamine derivative
TW460478B (en) * 1997-08-15 2001-10-21 Chugai Pharmaceutical Co Ltd Phenethylamine derivatives
US5972939A (en) 1997-10-28 1999-10-26 Ortho-Mcneil Pharmaceutical, Inc. Cyclopentene derivatives useful as antagonists of the motilin receptor
JP4200393B2 (en) 1997-11-25 2008-12-24 ソニー株式会社 Information processing apparatus and information processing method
US6080838A (en) * 1997-11-25 2000-06-27 University Of Virginia Patent Foundation Peptidomimetic of helix-turn-helix or gamma-turn
US6165985A (en) 1998-02-13 2000-12-26 Solvay Pharmaceuticals Gmbh 11-acetyl-12,13-dioxabicyclo[8.2.1]-tridecenone derivatives, processes for their preparation and pharmaceutical compositions comprising them
US6228986B1 (en) * 1998-04-13 2001-05-08 Ortho-Mcneil Pharmaceutical, Inc. Solid-phase synthesis of novel 14-membered macroycles for high throughput screening
SE9802080D0 (en) * 1998-06-11 1998-06-11 Hellstroem Pharmaceutical composition for the treatment of functional dyspepsia and / or irritable bowel syndrome and new use of substances therein
JP2002517488A (en) * 1998-06-11 2002-06-18 デュポン ファーマシューティカルズ カンパニー Method for producing large ring metalloprotease inhibitors
DE69935219D1 (en) 1998-06-12 2007-04-05 Merck & Co Inc CLONING AND IDENTIFICATION OF THE MOTILIN RECEPTOR
WO2000001701A1 (en) * 1998-07-02 2000-01-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
EP1091736A4 (en) * 1998-07-02 2001-10-24 Merck & Co Inc Inhibitors of prenyl-protein transferase
US6562823B1 (en) * 1998-07-02 2003-05-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
TW509699B (en) * 1998-09-24 2002-11-11 Chugau Pharmaceutical Co Ltd Ethylamine derivatives
AUPP616498A0 (en) * 1998-09-25 1998-10-15 University Of Queensland, The Synthesis of cyclic peptides
WO2000024907A2 (en) 1998-10-28 2000-05-04 Kosan Biosciences, Inc. Library of novel "unnatural" natural products
US6329376B1 (en) 1998-10-29 2001-12-11 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
JP3715202B2 (en) * 1999-01-28 2005-11-09 中外製薬株式会社 Substituted phenethylamine derivatives
US20020058286A1 (en) * 1999-02-24 2002-05-16 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto and analogues thereof
JP4269041B2 (en) * 1999-03-02 2009-05-27 国立大学法人九州工業大学 Novel cyclic tetrapeptide derivatives and their pharmaceutical uses
US6121257A (en) * 1999-03-31 2000-09-19 Abbott Laboratories Sulfamate containing macrocyclic immunomodulators
US6608027B1 (en) * 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus
US6420521B1 (en) 1999-06-30 2002-07-16 Zymogenetics, Inc. Short gastrointestinal peptides
EP1190059A1 (en) 1999-06-30 2002-03-27 ZymoGenetics, Inc. Sgip peptides
DE19933701A1 (en) 1999-07-19 2001-01-25 Wilex Biotechnology Gmbh Cyclic peptidomimetic urokinase receptor antagonists
US6660832B1 (en) 1999-08-20 2003-12-09 Isis Pharmaceuticals, Inc. Macrocyclic compounds and preparation methods thereof
DE19942624A1 (en) 1999-08-28 2001-03-08 Chemotopix Gmbh Process for the preparation of cyclic peptidomimetics
JP2003508535A (en) * 1999-09-09 2003-03-04 メルク エンド カムパニー インコーポレーテッド Inhibitors of prenyl-protein transferase
CA2284459C (en) * 1999-10-04 2012-12-11 Neokimia Inc. Combinatorial synthesis of libraries of macrocyclic compounds useful in drug discovery
GB9929318D0 (en) 1999-12-10 2000-02-02 Prometic Biosciences Limited Macrocyclic compounds and their use
US6566385B2 (en) * 2000-01-12 2003-05-20 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2001051128A1 (en) * 2000-01-12 2001-07-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6525074B2 (en) * 2000-01-12 2003-02-25 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
WO2001051494A1 (en) * 2000-01-12 2001-07-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6413964B1 (en) * 2000-01-12 2002-07-02 Desolms S. Jane Inhibitors of prenyl-protein transferase
DK1265605T3 (en) 2000-01-18 2007-02-12 Univ Mcgill Pharmaceutical compositions comprising peptide mimetic cyclic beta rotation compound
AU2001249144A1 (en) 2000-03-13 2001-09-24 Ortho-Mcneil Pharmaceutical, Inc. Novel cyclopentene derivatives useful as antagonists of the motilin receptor
US6384031B2 (en) * 2000-03-13 2002-05-07 Ortho-Mcneil Pharmaceutical, Inc. Cyclobutene derivatives useful as antagonists of the motilin receptor
WO2001068621A1 (en) * 2000-03-13 2001-09-20 Ortho-Mcneil Pharmaceutical, Inc. Novel cyclohexene derivatives useful as antagonists of the motilin receptor
AR029903A1 (en) 2000-04-05 2003-07-23 Schering Corp MACROCICLIC INHIBITORS OF THE NS3-SERINA PROTEASA, OF THE VIRUS OF HEPATITIS C, WHICH INCLUDE P2 N-CYCLING PARTS, PHARMACEUTICAL COMPOSITIONS AND USE OF THE SAME FOR THE MANUFACTURE OF A MEDICINAL PRODUCT
AU2001251444A1 (en) * 2000-04-10 2001-10-23 Merck And Co., Inc. Inhibitors of prenyl-protein transferase
WO2001077116A1 (en) * 2000-04-10 2001-10-18 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6610722B2 (en) * 2000-04-10 2003-08-26 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
JP4748912B2 (en) 2000-04-19 2011-08-17 シェーリング コーポレイション Macrocyclic NS3-serine protease inhibitors against hepatitis C virus containing alkyl and arylalanine P2 moieties
US6511980B2 (en) 2000-05-05 2003-01-28 Ortho Mcneil Pharmaceutical, Inc. Substituted diamine derivatives useful as motilin antagonists
ES2333097T3 (en) * 2000-05-31 2010-02-17 Raqualia Pharma Inc USE OF SECRETAGOGS OF GROWTH HORMONE TO STIMULATE GASTROINTESTINAL MOTILITY.
US6721353B1 (en) 2000-06-21 2004-04-13 Cisco Technology, Inc. Network compatibility
WO2002016404A1 (en) 2000-08-24 2002-02-28 Chugai Seiyaku Kabushiki Kaisha Cyclic peptide derivative
AU2001286768A1 (en) * 2000-08-25 2002-03-04 Sloan-Kettering Institute For Cancer Research Novel macrocycles and uses thereof
CN1452617A (en) 2000-09-05 2003-10-29 甘瑟尔股份有限公司 Acid-sensitive compounds, preparation and uses thereof
WO2002040463A1 (en) 2000-11-20 2002-05-23 Toyama-Ken Macrocyclic compound
US7279304B2 (en) 2000-12-18 2007-10-09 President And Fellows Of Harvard College Methods for preparation of macrocyclic molecules and macrocyclic molecules prepared thereby
AU2002218902A1 (en) 2000-12-27 2002-07-08 Shire Biochem Inc. Macrocyclic anti-viral compounds
JPWO2002059141A1 (en) 2001-01-25 2004-05-27 中外製薬株式会社 Peptide derivatives
KR20020066476A (en) 2001-02-12 2002-08-19 쥬가이 세이야쿠 가부시키가이샤 Preparation of peptide derivatives
CN1255428C (en) 2001-02-23 2006-05-10 波利弗尔有限公司 Template-fixed peptidomimetics with antimicrobial activity
WO2002092592A1 (en) 2001-05-10 2002-11-21 Solvay Pharmaceuticals Gmbh Novel 1-amidomethylcarbonyl-piperidine derivatives, method and intermediate products for the production thereof and medicament containing said compounds
JP2005505506A (en) 2001-06-12 2005-02-24 イーラン ファーマスーティカルズ、インコーポレイテッド Macrocycles useful for the treatment of Alzheimer's disease
ES2261699T3 (en) 2001-06-12 2006-11-16 Elan Pharmaceuticals, Inc. USEFUL MACROCICLES IN THE TREATMENT OF ALZHEIMER'S DISEASE.
DE10146104A1 (en) 2001-09-19 2003-04-03 Bayer Ag Antibacterial marrow cycles
AU2002363328A1 (en) * 2001-11-06 2003-05-19 President And Fellows Of Harvard College Methods for preparation of macrocyclic molecules macrocyclic molecules prepared thereby and substrates and solid supports for use therein
CN101157924A (en) 2001-12-11 2008-04-09 人体基因组科学有限公司 Neutrophil leukocyte gene alpha
US6867185B2 (en) * 2001-12-20 2005-03-15 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
US7119072B2 (en) 2002-01-30 2006-10-10 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis C virus
CA2369711A1 (en) 2002-01-30 2003-07-30 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
US6972281B2 (en) * 2002-02-07 2005-12-06 University Of Ottawa Histogranin-like peptides and non-peptides, processes for their preparation and uses thereof
EP1572076A3 (en) 2002-02-14 2005-12-21 The Brigham And Women's Hospital, Inc. Helical peptidomimetics
AU2003224672B2 (en) 2002-03-08 2010-02-04 Eisai R&D Management Co., Ltd. Macrocyclic compounds useful as pharmaceuticals
US20030221432A1 (en) 2002-06-03 2003-12-04 Tucker Ronald M. Solid fuel combustion method and apparatus for the conversion of waste into useful energy
DE10226921A1 (en) 2002-06-17 2003-12-24 Bayer Ag Antibacterial amide macrocycles
PL203763B1 (en) 2002-06-24 2009-11-30 Rockwool Int Process and apparatus for making mineral wool
DE10232094A1 (en) 2002-07-15 2004-02-05 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) 5-thiaepothilones and 15-disubstituted epothilones
DE10234422A1 (en) 2002-07-29 2004-02-12 Bayer Ag Antibacterial ester macrocycles
AU2002342618C1 (en) 2002-08-20 2008-10-30 Polyphor Ltd. Template-fixed peptidomimetics with antibacterial activity
DE10239042A1 (en) 2002-08-21 2004-03-04 Schering Ag New fused macrocyclic pyrimidine derivatives, useful as e.g. cyclin-dependent kinase inhibitors for treating e.g. cancer, autoimmune, cardiovascular or neurodegenerative diseases or viral infections
US6747009B2 (en) * 2002-09-19 2004-06-08 Universiteit Leiden Peptidomimetic glutathione analogs
US6653334B1 (en) * 2002-12-27 2003-11-25 Kowa Co., Ltd. Benzoxazole compound and pharmaceutical composition containing the same
JP4928261B2 (en) * 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 Macrocyclic antagonist of motilin receptor
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
US7491695B2 (en) * 2003-06-18 2009-02-17 Tranzyme Pharma Inc. Methods of using macrocyclic modulators of the ghrelin receptor
USRE42013E1 (en) 2003-06-18 2010-12-28 Tranzyme Pharma Inc. Macrocyclic modulators of the ghrelin receptor
ATE490264T1 (en) * 2003-07-31 2010-12-15 Tranzyme Pharma Inc SPACIOUSLY DEFINED MACROCYCLIC COMPOUNDS SUITABLE FOR DRUG DISCOVERY
ATE419267T1 (en) * 2003-07-31 2009-01-15 Tranzyme Pharma Inc SPACIALLY DEFINED MACROCYCLES CONTAINING PEPTIDE BINDING SURROGATES
EP1661890B1 (en) * 2003-09-03 2011-01-05 Kowa Co., Ltd. Ppar-activating compound and pharmaceutical composition containing same
US20090275648A1 (en) * 2005-06-13 2009-11-05 Fraser Graeme L Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
US9133235B2 (en) * 2006-09-11 2015-09-15 Ocera Therapeutics, Inc. Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
CN101657436A (en) * 2007-02-09 2010-02-24 特兰齐姆制药公司 Macrocyclic ghrelin receptor modulators and using method thereof
US20080287371A1 (en) * 2007-05-17 2008-11-20 Tranzyme Pharma Inc. Macrocyclic antagonists of the motilin receptor for modulation of the migrating motor complex
US8018037B2 (en) 2009-04-16 2011-09-13 Mediatek Inc. Semiconductor chip package
BR112012007183A2 (en) * 2009-09-30 2017-06-20 Tranzyme Pharma Inc solvate of a salt of a macrocyclic compound, polymorphic form, processes for preparing the polymorphic form and a pharmaceutical composition, pharmaceutical composition, salt of a macrocyclic compound, and methods for stimulating gastrointestinal motility, for treating a gastrointestinal disorder and for treating an individual and kit
WO2011053821A1 (en) * 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same

Also Published As

Publication number Publication date
ES2338789T3 (en) 2010-05-12
JP2007523853A (en) 2007-08-23
ATE457995T1 (en) 2010-03-15
US20120226067A1 (en) 2012-09-06
US9181298B2 (en) 2015-11-10
JP4928261B2 (en) 2012-05-09
CA2528375A1 (en) 2004-12-23
US20090240027A1 (en) 2009-09-24
US10040751B2 (en) 2018-08-07
EP2210612A3 (en) 2010-11-24
EP1633774B1 (en) 2010-02-17
EP1633774A1 (en) 2006-03-15
DK1633774T3 (en) 2010-05-25
US20120226072A1 (en) 2012-09-06
WO2004111077A1 (en) 2004-12-23
EP2210612A2 (en) 2010-07-28
US20160221927A1 (en) 2016-08-04
EP2210612B1 (en) 2016-10-05
US7521420B2 (en) 2009-04-21
US20120226066A1 (en) 2012-09-06
US20090137835A1 (en) 2009-05-28
US8129561B2 (en) 2012-03-06
US20120165566A1 (en) 2012-06-28
US8497242B2 (en) 2013-07-30
DE602004025569D1 (en) 2010-04-01
US20110245459A1 (en) 2011-10-06
US20050054562A1 (en) 2005-03-10

Similar Documents

Publication Publication Date Title
CA2528375C (en) Macrocyclic antagonists of the motilin receptor
CA2579726C (en) Macrocyclic modulators of the ghrelin receptor
CA2677399C (en) Macrocyclic ghrelin receptor modulators and methods of using the same
AU2005264907B2 (en) Methods of using macrocyclic modulators of the ghrelin receptor
CA2662897C (en) Macrocyclic antagonists of the motilin receptor for treatment of gastrointestinal dysmotility disorders
USRE42624E1 (en) Methods of using macrocyclic modulators of the ghrelin receptor
US20090198050A1 (en) Macrocyclic Modulators of the Ghrelin Receptor
US8921521B2 (en) Macrocyclic modulators of the Ghrelin receptor
JP5739766B2 (en) Use of macrocyclic modulators of ghrelin receptor
JP5730835B2 (en) Use of macrocyclic modulators of ghrelin receptor

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20220301

MKLA Lapsed

Effective date: 20200831