WO1994022494A1 - RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS - Google Patents

RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS Download PDF

Info

Publication number
WO1994022494A1
WO1994022494A1 PCT/US1994/003256 US9403256W WO9422494A1 WO 1994022494 A1 WO1994022494 A1 WO 1994022494A1 US 9403256 W US9403256 W US 9403256W WO 9422494 A1 WO9422494 A1 WO 9422494A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
substituted
gly
formula
asp
Prior art date
Application number
PCT/US1994/003256
Other languages
French (fr)
Inventor
William Frank Degrado
Shaker Ahmed Mousa
Michael Sworin
John Andrew Barrett
David Scott Edwards
Thomas David Harris
Milind Rajopadhye
Shuang Liu
Original Assignee
The Du Pont Merck Pharmaceutical Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to RU95118183A priority Critical patent/RU2145608C1/en
Priority to EP94912870A priority patent/EP0692982B1/en
Application filed by The Du Pont Merck Pharmaceutical Company filed Critical The Du Pont Merck Pharmaceutical Company
Priority to BR9406055A priority patent/BR9406055A/en
Priority to DE69425138T priority patent/DE69425138T2/en
Priority to RO95-01701A priority patent/RO114895B1/en
Priority to JP6522205A priority patent/JP3042887B2/en
Priority to AT94912870T priority patent/ATE194293T1/en
Priority to UA95094341A priority patent/UA32577C2/en
Priority to AU65248/94A priority patent/AU6524894A/en
Priority to BR9406820A priority patent/BR9406820A/en
Priority to DK94912870T priority patent/DK0692982T3/en
Publication of WO1994022494A1 publication Critical patent/WO1994022494A1/en
Priority to NO953886A priority patent/NO307568B1/en
Priority to FI954655A priority patent/FI954655A/en
Priority to KR1019950704228A priority patent/KR960701666A/en
Priority to BG100036A priority patent/BG100036A/en
Priority to LVP-95-296A priority patent/LV11106B/en
Priority to GR20000401959T priority patent/GR3034272T3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/75Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0474Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group
    • A61K51/0482Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group chelates from cyclic ligands, e.g. DOTA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/082Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins the peptide being a RGD-containing peptide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/008Peptides; Proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2123/00Preparations for testing in vivo

Definitions

  • Radiolabeled Platelet GPIIb/IIIa Receptor Antagonists As Imaging Agents For The Diagnosis Of Thromboembolic
  • This invention relates to novel
  • radiopharmaceuticals that are radiolabeled cyclic compounds containing carbocyclic or heterocyclic ring systems; to methods of using said radiopharmaceuticals as imaging agents for the diagnosis of arterial and venous thrombi; to novel reagents for the preparation of said radiopharmaceuticals; and to kits comprising said reagents.
  • thromboembolic disorders such as unstable angina, myocardial infarction, transient ischemic attack, stroke, atherosclerosis and diabetes.
  • the contribution of platelets to these disease processes stems from their ability to form aggregates, or platelet thrombi, especially in the arterial wall following injury. See generally, Fuster et al., JACC, Vol. 5, No. 6, pp. 175B- 183B (1985); Rubenstein et al., Am. Heart J., Vol. 102, pp. 363-367 (1981); Hamm et al., J. Am. Coll. Cardiol., Vol. 10, pp. 998-1006 (1987); and Davies et al.,
  • Platelet activation and aggregation is also thought to play a significant role in venous thromboembolic disorders such as venous thrombophlebitis and subsequent pulmonary emboli. It is also known that patients whose blood flows over artificial surfaces, such as prosthetic synthetic cardiac valves, are at risk for the
  • thromboembolic disorders would be highly useful, and several attempts have been made to develop radiolabeled agents targeted to platelets for non-invasive
  • thromoboembolisms are described, for example, in Koblik et al., Semin. Nucl. Med., Vol. 19, pp. 221-237 (1989).
  • This invention provides novel radiopharmaceuticals that are radiolabeled cyclic compounds containing carbocyclic or heterocyclic ring systems which act as antagonists of the platelet glycoprotein Ilb/IIIa complex. It also provides methods of using said
  • radiopharmaceuticals as imaging agents for the diagnosis of arterial and venous thrombi. It further provides novel reagents for the preparation of said
  • FIGURES Figure la. Illustrated are typical images of the radiopharmaceutical compound of Example 12 administered at 1 mCi/Kg,i.v. in a canine deep venous thrombosis model. In this model thrombi were formed in the jugular veins during a period of stasis which was followed by reflow. The compounds were administered beginning at reflow. Depicted is the uptake in a rapidly growing venous thrombus at 15, 60 and 120 min post- administration.
  • Figure lb Illustrated are typical images of the radiopharmaceutical compound of Example 19 administered at 1 mCi/Kg,i.v. in a canine deep venous thrombosis model.
  • thrombi were formed in the jugular veins during a period of stasis which was followed by reflow.
  • the compounds were administered beginning at reflow. Depicted is the uptake in a rapidly growing venous thrombus at 15, 60 and 120 min post- administration. DETAILED DESCRIPTION OF THE INVENTION
  • the present invention is directed to novel reagents for preparing a radiopharmaceutical of formulae: (QL n ) d C h ; (Q) d 'L n -C h , wherein, d is 1-3, d' is 2-20, L n is a linking group, C h is a metal chelator, and Q is a compound of formula (I):
  • R 31 is a C 6 -C 14 saturated, partially
  • R 32 is selected from:
  • Z is S or O; "n" and n' are independently 0-2;
  • R 1 and R 22 are independently selected from the following groups: hydrogen,
  • heterocyclic ring being substituted with 0-2 R 12 ;
  • R 1 and R 21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R 12 ; when n' is 2, R 1 or R 21 can alternatively be taken together with R 1 or R 21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
  • R 21 and R 23 are independently selected from: hydrogen;
  • R 22 and R 23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R 12 ; when n" is 2, R 22 or R 23 can
  • R 1 and R 2 where R 21 is H, can
  • heterocyclic ring being substituted with 0-2 R 12 ;
  • C 1 -C 4 haloalkoxy C 1 -C 4 alkylcarbonyloxy, C 1 -C 4 alkylcarbonyl, C 1 -C 4 alkylcarbonylamino, -OCH 2 CO 2 H, 2- (l-morpholino)ethoxy, C 1 -C 4 alkyl (alkyl being substituted with
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl;
  • R 2 is H or C 1 -C 8 alkyl;
  • R 10 and R 10a are selected independently from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C 1 -C 5 alkyl, C 3 -C 6 cycloalkyl, C 3 -
  • R 4 is H or C 1 -C 3 alkyl;
  • R 5 is selected from:
  • R 11 a bond to L n ; aryl substituted with 0-2 R 12 ; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
  • R 3 and R 4 may also be taken together to form (CH 2 ) n X
  • R 16 is selected from: an amine protecting group
  • K is a D-isomer or L-isomer amino acid of structure
  • R 6 is H or C 1 -C 8 alkyl
  • R 7 is selected from: -(C 1 -C 7 alkyl) X
  • substitution on the phenyl is at the 3 or 4 position; wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
  • X is selected from:
  • R 6 and R 7 can alternatively be taken together to form
  • M is a D-isomer or L-isomer amino acid of
  • R 17 is H, C 1 -C 3 alkyl;
  • R 8 is selected from:
  • R 34 and R 35 are independently selected from:
  • R 34 and R 35 can alternatively be taken together form:
  • a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
  • a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O.
  • R 31 is bonded to (C (R 23 )R 22 ) n" and
  • n" is 0 and n' is 1;
  • n" is 0 and n' is 2;
  • n" is 1 and n' is 0;
  • n" is 2 and n' is 0;
  • n" is 2 and n' is 1;
  • n" is 2 and n' is 2.
  • R 6 is methyl, ethyl, or propyl.
  • R 32 is selected from:
  • R 1 and R 22 are independently selected from the following groups: hydrogen,
  • R 1 and R 21 can alternatively join to form a 5-7 membered carbocyclic ring
  • R 1 or R 21 can alternatively be taken together with R 1 or R 21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
  • R 22 and R 23 can alternatively join to form a
  • R 1 and R 2 where R 21 is H, can alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R 12 ;
  • R 11 is selected from one or more of the
  • heterocyclic ring being substituted with 0-2 R 12 ; is H or CH 3 ; is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 - C 6 cycloalkylmethyl, C 1 -C 6
  • R 7 is selected from:
  • X is selected from:
  • R 6 and R 7 can alternatively be taken together to form
  • M is a D-isomer or L-isomer amino acid of
  • R 17 is H, C 1 -C 3 alkyl
  • R 8 is selected from: -CO 2 R 13 ,-SO 3 R 13 , -SO 2 NHR 14 , -B (R 34 ) (R 35 ),
  • heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
  • R 34 and R 3 5 are independently selected from:
  • R 34 and R 35 can alternatively be taken together form:
  • a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms
  • a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O.
  • R 31 is selected from the group consisting of:
  • R 31 is selected from the group consisting of: (a) a 6 membered saturated, partially saturated, or aromatic carbocyclic ring of formulae:
  • any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted with 0-3 R 10 , and optionally bears a bond to Ln;
  • any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R 10 , and optionally bears a bond to L n ;
  • any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R 10 , and optionally bears a bond to L n .
  • R 31 is selected from (the dashed bond may be a single or double bond):
  • R 31 may be independently substituted with 0-3 R 10 or R 10a , and optionally bears a bond to L n ; n" is 0 or 1; and n' is 0-2.
  • R 31 is selected from:
  • R 31 may be independently substituted with 0-3 R 10 or R 10a , and may optionally bear a bond to L n ;
  • R 21 and R 23 are independently H or C 1 -C 4 alkyl
  • R 2 is H or C 1 -C 8 alkyl
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, - (C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl;
  • R 1 0 and R 10a are selected independently from:
  • R 4 is H or C 1 -C 3 alkyl
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 - C 6 cycloalkylmethyl, C 1 -C 6
  • R 16 is selected from:
  • K is an L-isomer amino acid of structure
  • R 6 is H or C 1 -C 8 alkyl
  • R 6 and R 7 can alternatively be taken together to form
  • M is a D-isomer or L-isomer amino acid of
  • R 17 is H, C 1 -C 3 alkyl; R 8 is selected from:
  • R 10 is selected independently from: H, C 1 -C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy;
  • R 1 is H, C 1 -C 4 alkyl, phenyl, benzyl,
  • R 2 is H or methyl
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 3 is H or CH 3 ;
  • R 4 is H or C 1 -C 3 alkyl
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 - C 6 cycloalkylmethyl, C 1 -C 6
  • R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -; or
  • R 16 is selected from:
  • R 6 is H or C 1 -C 8 alkyl
  • R 7 is:
  • R 6 and R 7 are alternatively be taken together to form
  • n 0,1 and X is -NH 2 or -NHC (-NH) (NH 2 );
  • M is a D-isomer or L-isomer amino acid of
  • R 17 is H, C 1 -C 3 alkyl;
  • R 8 is selected from:
  • heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
  • the phenyl ring in formula (II) may be substituted with 0-3 R 10 or R 10a ;
  • R 10 or R 10a are selected independently from: H, C 1 - C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy;
  • R 1 is H, C 1 -C 4 alkyl, phenyl, benzyl, or phenyl- (C 2 - C 4 ) alkyl;
  • R 2 is H or methyl;
  • R 13 is selected independently from:. H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10 alkyl) aryl, or C 3 -C 10 alkoxyalkyl; when two R 13 groups are bonded to a single N, said R 13 groups may alternatively be taken together to form -(CH 2 ) 2-5 - or - (CH 2 )O(CH 2 )-;
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 -C 6
  • cycloalkylmethyl C 1 -C 6 cycloalkylethyl, phenyl, phenylmethyl, CH 2 OH, CH 2 SH, CH 2 OCH 3 , CH 2 SCH 3 , CH 2 CH 2 SCH 3 , (CH 2 ) S NH 2 ,
  • R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -;
  • R 16 is selected from: an amine protecting group
  • K is an L-isomer amino acid of structure
  • R 6 is H or C 3 -C 8 alkyl
  • M is a D-isomer or L-isomer amino acid of structure
  • R 8 is selected from:
  • R 10a are selected independently from: H, C 1 - C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy;
  • R 1 is H
  • R 2 is H
  • R 13 is selected independently from: H, C 1 -C 10
  • alkyl C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10 alkyl) aryl, or C 3 -C 10 alkoxyalkyl;
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, - (C 1 -C 10 alkyl) aryl, or C 3 -C 10 alkoxyalkyl; when two R 13 groups are bonded to a single N, said R 13 groups may alternatively be taken together to form -(CH 2 ) 2-5 - or - (CH 2 )O(CH 2 )-;
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 3 is H and R 5 is H, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 ,
  • R 3 is CH 3 and R 5 is H; or R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -;
  • R 16 is selected from:
  • K is an L-isomer amino acid of formula
  • M is a D-isomer or L-isomer amino acid of structure
  • R 4 is H or CH 3 ;
  • R 17 is H;
  • R 1 and R 2 are independently selected from H,
  • J is selected from D-Val, D-2-aminobutyric acid, D- Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, ⁇ -Ala,
  • L is selected from Gly, ⁇ -Ala, Ala;
  • M is selected from Asp; (XMeAsp; ⁇ MeAsp; NMeAsp; D- Asp.
  • R 31 is a phenyl ring and bears a bond to L n ;
  • R 1 and R 2 are independently selected from H,
  • J is selected from: D-Val, D-2-aminobutyric acid,
  • K is selected from NMeArg
  • L is Gly; M is selected from Asp; OMeAsp; ⁇ MeAsp; NMeAsp;
  • a 1 , A 2 , A 3 , A 4 , A 5 , A 6 , and A 7 are
  • NR 40 R 41 independently selected at each occurrence from the group: NR 40 R 41 , S, SH, S (Pg), O, OH, PR 42 R 43 , P(O)R 42 R 43 , P(S)R 42 R 43 ,
  • W is a bond, CH, or a spacer group selected from the group: C 1 -C 10 alkyl substituted with 0-3 R 52 , aryl substituted with 0-3
  • R 52 cycloaklyl substituted with 0-3 R 52 , heterocycioalkyl substituted with 0-3 R 52 , aralkyl substituted with 0-3 R 52 and alkaryl substituted with 0-3 R 52 ;
  • W a is a C 1 -C 10 alkyl group or a C 3 -C 14 carbocycle;
  • R 40 , R 41 , R 42 , R 43 , and R 44 are each
  • R 52 is independently selected at each
  • R 53 , R 53a , and R 54 are independently selected at each occurrence from the group: a bond to L n , C 1 -C 6 alkyl, phenyl, benzyl, C 1 -C 6 alkoxy, halide, nitro, cyano, and
  • Pg is a thiol protecting group capable of
  • a 1 , A 2 , A 3 , A 4 , A 5 , A 6 , and A 7 are
  • W is a bond, CH, or a spacer group selected from the group: C 1 -C 3 alkyl substituted with 0-3 R 52 ;
  • W a is a methylene group or a C 3 -C 6 carbocycle;
  • R 40 , R 41 , R 42 , R 43 , and R 44 are each
  • R 53 , R 53a , and R 54 are independently selected at each occurrence from the group: a bond to L n , C 1 -C 6 alkyl. [18] Included in the present invention are those reagents in [1]-[15] above, of formula:
  • a 1 and A 4 are SH or SPg
  • a 2 and A 3 are NR 41 ;
  • W is independently selected from the group :
  • R 41 and R 52 are independently selected from hydrogen and a bond to L n , and,
  • W is a bond
  • a 2 is NHR 40 , wherein R 40 is heterocycle substituted with R 52 , wherein the heterocycle is selected from the group: pyridine, pyrazine, proline, furan, thiofuran, thiazole, and diazine, and R 52 is a bond to Ln.
  • W is a bond
  • a 2 is NHR 40 , wherein R 4 0 is heterocycle
  • R 52 substituted with R 52 , wherein the heterocycle is selected from pyridine and thiazole, and R 52 is a bond to L n .
  • M 1 a compound of formula: M 1 -[Y 1 (CR 55 R 56 )h(Z 1 )h"Y 2 ] h '-M 2 wherein: M 1 is -[(CH 2 ) g Z 1 ] g ,-(CR 55 R 56 ) g" -;
  • M 2 is -(CR 55 R 56 ) g" -[Z 1 (CH 2 ) g ] g ,-;
  • g is independently 0-10;
  • g' is independently 0-1;
  • g" is 0-10;
  • h 0-10
  • h' is 0-10
  • Z 1 is independently selected at each
  • R 55 and R 56 are independently selected at each occurrence from: hydrogen;
  • R 57 is independently selected at each
  • R 58 is independently selected at each
  • M 1 is -[(CH 2 ) g Z 1 ] g '-(CR 55 R 56 ) g" - ;
  • M 2 is -(CR 55 R 56 ) g" -[Z 1 (CH 2 ) g ] g' -;
  • g is independently 0-10;
  • g' is independently 0-1;
  • g" is 0-10;
  • h 0-10
  • h' is 0-10
  • Z 1 is independently selected at each
  • R 55 and R 56 are independently selected at each occurrence from: hydrogen;
  • R 57 is independently selected at each
  • R 58 is independently selected at each occurrence from the group:hydrogen; C 1 -C 6 alkyl; benzyl, and phenyl.
  • h 0-10
  • h' is 1-10;
  • R 55 and R 56 are independently selected at each occurrence from: hydrogen;
  • R 58 is independently selected at each occurrence from the group:hydrogen; C 1 -C 6 alkyl; benzyl, and phenyl.
  • h 0-5;
  • h* is 1-5;
  • R 55 and R 56 are independently selected at each occurrence from: hydrogen;
  • h 0-5;
  • h' is 1-5;
  • R 55 and R 56 are independently selected at each occurrence from: hydrogen.
  • kits for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile
  • kits for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile
  • kits for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile
  • radiopharmaceutical comprising a complex of a reagent of [1]-[15] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 4 3 Sc, 45 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 2 01 T1.
  • radiopharmaceutical comprising a complex of a reagent of [16] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 4 5 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 201 Tl.
  • radiopharmaceutical comprising a complex of a reagent of [17] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 4 5 Ti, 6 7 Ga, 6 8 Ga, 97 Ru, 72 As, 82 Rb, and 201 ⁇ l .
  • radiopharmaceutical comprising a complex of a reagent of [18] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 4 5 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 201 Tl.
  • radiopharmaceutical comprising a complex of a reagent of [19] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 4 5 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 201 Tl.
  • radiopharmaceutical comprising a complex of a reagent of [20] and a radionuclide selected from the group 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 4 5 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 201 Tl.
  • a radiopharmaceutical comprising a complex of a reagent of [21] and a radionuclide selected from the group 99m Tc, 111 In, and 62 Cu.
  • radiopharmaceutical comprising a complex of a reagent of [22] and a radionuclide selected from the group 99m Tc, 111 In, and 62 Cu.
  • radiopharmaceutical comprising a complex of a reagent of [23] and a radionuclide selected from the group 99m Tc, 111 In, and 62 Cu.
  • radiopharmaceutical comprising a complex of a reagent of [24] and a radionuclide selected from the group 99m Tc, and 111 In.
  • radiopharmaceuticals of [29] which are:
  • Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
  • radioimaging a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
  • Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
  • Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
  • radioimaging deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [33], and (ii) scanning the mammal using a radioimaging devise.
  • radioimaging deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [34], and (ii) scanning the mammal using a radioimaging devise.
  • radioimaging deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [35], and (ii) scanning the mammal using a radioimaging devise.
  • a method for visualizing sites of platelet deposition in a mammal by radioimaging comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [37], and (ii) scanning the mammal using a radioimaging devise.
  • radioimaging deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [38], and (ii) scanning the mammal using a radioimaging devise.
  • R 31 is a C 6 -C 14 saturated, partially
  • R 32 is selected from:
  • Z is S or O; n" and n' are independently 0-2;
  • R 1 and R 22 are independently selected from the following groups: hydrogen,
  • R 1 and R 21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R 12 ; when n 1 is 2, R 1 or R 21 can alternatively be taken together with R 1 or R 21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
  • R 22 and R 23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R 12 ; when n" is 2, R 22 or R 23 can
  • R 22 or R 23 on an adjacent carbon atom can alternatively be taken together with R 22 or R 23 on an adjacent carbon atom to .form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms;
  • R 1 and R 2 , where R 21 is H, can alternatively be taken together with R 22 or R 23 on an adjacent carbon atom to .form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms;
  • R 11 is selected from one or more of the
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 21 and R 23 are independently selected from: hydrogen;
  • R 2 is H or C 1 -C 8 alkyl
  • alkylcarbonyloxy C 1 -C 4 alkylcarbonyl, C 1 -C 4 alkylcarbonylamino, -OCH 2 CO 2 H,
  • J is ⁇ -Ala or an L-isomer or D-isomer amino acid of structure
  • R 3 is H or C 1 -C 8 alkyl
  • R 4 is H or C 1 -C 3 alkyl
  • R 5 is selected from:
  • R 3 and R 4 may also be taken together to form (CH 2 ) n X
  • R 16 is selected from:
  • K is a D-isomer or L-isomer amino acid of structure
  • R 6 is H or C 1 -C 8 alkyl
  • R 7 is selected from: -(C 1 -C 7 alkyl)X;
  • substitution on the phenyl is at the 3 or 4 position; wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
  • X is selected from:
  • R 6 and R 7 can alternatively be taken together to form
  • M is a D-isomer or L-isomer amino acid of
  • q ' is 0-2 ;
  • R 17 is H, C 1 -C 3 alkyl;
  • R 8 is selected from:
  • R 34 and R 35 are independently selected from:
  • R 34 and R 35 can alternatively be taken together form:
  • a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
  • a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; and wherein the radiolabel is selected from the group: 123 I, 125 I, 131 I, 18 F, 11 C, 13 N, 1 5 O, 75 Br.
  • the radiolabel is selected from the group: 123 I, 125 I, 131 I, 18 F, 11 C, 13 N, 1 5 O, 75 Br.
  • R 31 is bonded to (C(R 23 )R 22 ) n" and
  • n" is 0 and n' is 1;
  • n" is 0 and n' is 2;
  • n" is 1 and n' is 0;
  • n" is 1 and n' is 1;
  • n" is 1 and n' is 2;
  • n" is 2 and n' is 0;
  • n" is 2 and n' is 1;
  • n" is 2 and n' is 2.
  • R 31 is selected from the group consisting of: (a) a 6 membered saturated, partially . saturated or aromatic carbocyclic ring substituted with 0-3 R 10 or R 10a ; (b) a 8-11 membered saturated, partially saturated, or aromatic fused bicyclic carbocyclic ring substituted with 0-4 R 10 or R 10a ; or (c) a 14 membered saturated, partially saturated, or aromatic fused tricyclic carbocyclic ring substituted with 0-4 R 10 or R 10a .
  • R 31 is selected from the group consisting of:
  • any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R 10 ;
  • any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R 10 or R 10a ;
  • any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R 10 or R 10a .
  • R 31 is selected from (the dashed bond may be a single or double bond):
  • n" is 0 or 1; and n' is 0-2.
  • R 31 is selected from:
  • R 31 may be substituted
  • R 1 and R 22 are independently selected from H, C 1 -C 4 alkyl, phenyl, benzyl,
  • R 21 and R 23 are independently H or C 1 -C 4 alkyl ;
  • R 2 is H or C 1 -C 8 alkyl;
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl; R 10 and R 10a are selected independently from:
  • J is ⁇ -Ala or an L-isomer or D-isomer amino acid of structure
  • R 3 is H or CH 3 ;
  • R 4 is H or C 1 -C 3 alkyl
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 - C 6 cycloalkylmethyl, C 1 -C 6
  • R 16 is selected from:
  • R 3 and R 5 can alternatively be taken together to form -(CH 2 ) t - (t - 2-4) or
  • K is an L-isomer amino acid of structure
  • R 6 is H or C 1 -C 8 alkyl
  • R 7 is
  • R 6 and R 7 can alternatively be taken together to form
  • M is a D-isomer or L-isomer amino acid of
  • q ' is 0-2 ;
  • R 17 is H, C 1 -C 3 alkyl
  • R 8 is selected from:
  • heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O
  • -SO 2 NHCOR 13 -CONHSO 2 R 13a
  • R 10 is selected independently from: H, C 1 -C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy;
  • R 1 is H, C 1 -C 4 alkyl, phenyl, benzyl, or phenyl-(C 1 - C 4 ) alkyl;
  • R 2 is H or methyl
  • R 13 is selected independently from: H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl;
  • J is ⁇ -Ala or an L-isomer or D-isomer amino acid of structure
  • R 3 is H or CH 3 ;
  • R 4 is H or C 1 -C 3 alkyl;
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 - C 6 cycloalkylmethyl, C 1 -C 6
  • R 16 is selected from:
  • R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -; or
  • K is an L-isomer amino acid of structure
  • R 6 is H or C 1 -C 8 alkyl
  • R 7 is :
  • M is a D-isomer or L-isomer amino acid of
  • R 17 is H, C 1 -C 3 alkyl;
  • R 8 is selected from:
  • the phenyl ring in formula (II) may be further substituted with 0-3 R 10 or R 10a ;
  • R 10 or R 10a are selected independently from: H, C 1 - C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy;
  • R 1 is H, C 1 -C 4 alkyl, phenyl, benzyl, or phenyl- (C 2 - C 4 ) alkyl;
  • R 2 is H or methyl
  • R 13 is selected independently from: H, C 1 -C 10
  • R 13 groups when two R 13 groups are bonded to a single N, said R 13 groups may alternatively be taken together to form -(CH 2 ) 2-5 - or -(CH 2 )O(CH 2 )-;
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl,
  • alkyl aryl, or C 3 -C 10 alkoxyalkyl
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 3 is H or CH 3 ; is H;
  • R 5 is H, C 1 -C 8 alkyl, C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylmethyl, C 1 -C 6 cycloalkylethyl, phenyl, phenylmethyl, CH 2 OH, CH 2 SH, CH 2 OCH 3 , CH 2 SCH 3 , CH 2 CH 2 SCH 3 , (CH 2 ) S NH 2 ,
  • R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -;
  • R 16 is selected from:
  • K is an L-isomer amino acid of structure
  • R 6 is H or C 3 -C 8 alkyl
  • Y is NH or O
  • M is a D-isomer or L-isomer amino acid of structure
  • R 17 is H, C 1 -C 3 alkyl
  • R 8 is selected from:
  • R 10 or R 10a are selected independently from: H, C 1 - C 8 alkyl, phenyl, halogen, or C 1 -C 4 alkoxy; R 1 is H ;
  • R 2 is H; R 13 is selected independently from: H, C 1 -C 10
  • alkyl C 3 -C 10 cycloalkyl, C 4 -C 12
  • R 13a is C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 4 -C 12
  • alkylcycloalkyl aryl, -(C 1 -C 10 alkyl) aryl, or C 3 -C 10 alkoxyalkyl; when two R 13 groups are bonded to a single N, said R 13 groups may alternatively be taken together to form -(C H2 ) 2-5 - or - (CH 2 )O(CH 2 )-;
  • R 14 is OH, H, C 1 -C 4 alkyl, or benzyl
  • R 3 is H and R 5 is H, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 ,
  • R 3 is CH 3 and R 5 is H;
  • R 3 and R 5 can alternatively be taken together to form -CH 2 CH 2 CH 2 -;
  • R 16 is selected from:
  • K is an L-isomer amino acid of formula
  • M is a D-isomer or L-isomer amino acid of structure
  • R 4 is H or CH 3 ;
  • R 17 is H
  • R 1 and R 2 are independently selected from H, methyl;
  • J is selected from D-Val, D-2-aminobutyric acid, D- Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, ⁇ -Ala,
  • L is selected from Gly, ⁇ -Ala, Ala;
  • M is selected from Asp; OMeAsp; ⁇ MeAsp; NMeAsp; D- Asp.
  • R 1 and R 2 are independently selected from H,
  • J is selected from: D-Val, D-2-aminobutyric acid,
  • K is selected from NMeArg
  • L is Gly
  • K is selected from Asp; OMeAsp; ⁇ MeAsp; NMeAsp;
  • radiolabeled compound as in one of [51]- [65] wherein the- radiolabel is selected from the group: 18 F, 11 C, 123 I, and 125 I.
  • the radiolabel is 123 I.
  • radiopharmaceutical composition comprising a radiopharmaceutically acceptable carrier and a radiolabeled compound of any of [51]-[67].
  • radiopharmaceutical composition comprising a compound of any of [51]-[67], and imaging said mammal.
  • [70] Included in the present invention is a method of diagnosing a disorder associated with. platelet deposition in a mammal comprising administering to said mammal a radiopharmaceutical composition comprising a compound of any of [51]-[67], and imaging said mammal.
  • the cyclic compounds of the present invention are radiolabeled.
  • radiolabeled it is meant that the subject cyclic platelet glycoprotein Ilb/IIIa compounds contain a radioisotope which is suitable for administration to a mammalian patient.
  • Radioisotopes are known to those skilled in the art and include, for example, isotopes of halogens (such as chlorine, fluorine, bromine and iodine), and metals including technetium and indium.
  • Preferred radioisotopes include 11C, 18F, 123 I, 125 I, 131 I, 99m Tc, 94m Tc, 95 Tc, 111 In, 62 Cu, 43 Sc, 45 Ti, 67 Ga, 68 Ga, 97 Ru, 72 As, 82 Rb, and 201 Tl. Most preferred are the isoptopes 123 I, 111 ln, and 99m Tc.
  • Radiolabeled compounds of the invention may be prepared using standard radiolabeling procedures well known to those skilled in the art. Suitable synthesis methodology is described in detail below. As discussed below, the cyclic platelet
  • glycoprotein Ilb/IIIa compounds of the invention may be radiolabeled either directly (that is, by incorporating the radiolabel directly into the compounds) or
  • the radiolabeling may be isotopic or nonisotopic.
  • isotopic radiolabeling one group already present in the cyclic compounds described above is substituted with (exchanged for) the radioisotope.
  • nonisotopic radiolabeling the radioisotope is added to the cyclic compounds without substituting with (exchanging for) an already existing group.
  • Direct and indirect radiolabeled compounds, as well as isotopic and nonisotopic radiolabeled compounds are included within the phrase "radiolabeled compounds" as used in
  • radiolabeling should also be reasonably stable, both chemically and metabolically, applying recognized standards in the art. Also, although the compounds of the invention may be labeled in a variety of fashions with a variety of different radioisotopes, as those skilled in the art will recognize, such radiolabeling should be carried out in a manner such that the high binding affinity and specificity of the unlabeled cyclic platelet GPIIb/IIIa compounds of the invention to the GPIIb/IIIa receptor is not significantly affected.
  • binding affinity and specificity is not affected more than about 3 log units, preferably not more than about 2 log units, more preferably not more than about 1 log unit, even more preferably not more than about 500%, and still even more preferably not more than about 250%, and most preferably the binding affinity and specificity is not affected at all.
  • radiolabeled compounds the label may appear at any position on Q.
  • Preferred radiolabeled compounds of the invention are radiolabeled compounds wherein the radiolabel is located on the carbocyclic ring system of R 31 , the R 5 substituent on J, and at R 1 or R 22 .
  • Even more preferred radiolabeled compounds of the invention are those of formula (II), wherein the radiolabel is located on the carbocyclic ring system of R 31 , or the R 5 substituent on J.
  • the preferred radiolabel is a halogen label, especially an iodine radiolabel.
  • the preferred metal nuclides are 99m Tc and 111 ln.
  • Preferred linking groups, Ln, and metal chelators, C h are
  • thromboembolic disorders such as arterial or venous thrombosis, including, for example, unstable angina, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, diabetes, thrombophlebitis, pulmonary emboli, or
  • the radiolabeled compounds of the invention are useful with both newly formed and older thrombi.
  • the radiolabeled compounds of the invention may also be used to diagnose other present or potential conditions where there is overexpression of the GPIIb/IIIa receptors, such as with metastatic cancer cells.
  • the subject compounds may be effectively employed in low doses, thereby minimizing any risk of toxicity.
  • the subject compounds are of a much smaller size than, for example, the radiolabeled 7E3 antibodies known in the art, allowing easier attainment of suitable target/background (T/B) ratio for detecting thrombi.
  • T/B target/background
  • radiolabeled compounds of the invention may also be employed for therapeutic purposes, in addition to the diagnostic usage described above.
  • GPIIb/IIIa mediates the process of platelet activation and aggregation.
  • the radiolabeled compounds of the present invention inhibit the
  • D and L-isomers of a particular amino acid are designated herein using the conventional 3- letter abbreviation of the amino acid, as indicated by the following examples: D-Leu, D-Leu, L-Leu, or L-Leu.
  • any variable for example, R 1 through R 8 , m, n, p, X, Y, etc.
  • its definition on each occurrence is independent of its definition at every other occurrence.
  • said group may optionally be substituted with up to two R 11 and R 11 at each occurrence is selected independently from the defined list of possible R 11 .
  • each of the two R 13 substituents on N is independently selected from the defined list of possible R 13 .
  • substituent may be bonded to any atom on the ring.
  • stable compound or “stable structure” is meant herein a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • substituted means that an one or more hydrogen on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound.
  • 2 hydrogens on the atom are replaced.

Abstract

This invention provides novel radiopharmaceuticals that are radiolabeled cyclic compounds containing carbocyclic or heterocyclic ring systems which act as antagonists of the platelet glycoprotein IIb/IIIa complex; to methods of using said radiopharmaceuticals as imaging agents for the diagnosis of arterial and venous thrombi; to novel reagents for the preparation of said radiopharmaceuticals; and to kits comprising said reagents.

Description

TITLE
Radiolabeled Platelet GPIIb/IIIa Receptor Antagonists As Imaging Agents For The Diagnosis Of Thromboembolic
Disorders
CROSS-REFERENCE TO RELATED APPLICATIONS The present application is a continuation-in-part of our copending application U.S.S.N. 08/040,336 filed March 30, 1993, the disclosure of which is hereby incorporated herein by reference. FIELD OF THE INVENTION
This invention relates to novel
radiopharmaceuticals that are radiolabeled cyclic compounds containing carbocyclic or heterocyclic ring systems; to methods of using said radiopharmaceuticals as imaging agents for the diagnosis of arterial and venous thrombi; to novel reagents for the preparation of said radiopharmaceuticals; and to kits comprising said reagents.
BACKGROUND OF THE INVENTION
The clinical recognition of venous and arterial thromboembolic disorders is unreliable, lacking in both sensitivity and specificity. In light of the
potentially life threatening situation, the need to rapidly diagnose thromboembolic disorders using a non invasive method is an unmet clinical need. Platelet activation and resulting aggregation has been shown to be associated with various pathophysiological conditions including cardiovascular and cerebrovascular
thromboembolic disorders such as unstable angina, myocardial infarction, transient ischemic attack, stroke, atherosclerosis and diabetes. The contribution of platelets to these disease processes stems from their ability to form aggregates, or platelet thrombi, especially in the arterial wall following injury. See generally, Fuster et al., JACC, Vol. 5, No. 6, pp. 175B- 183B (1985); Rubenstein et al., Am. Heart J., Vol. 102, pp. 363-367 (1981); Hamm et al., J. Am. Coll. Cardiol., Vol. 10, pp. 998-1006 (1987); and Davies et al.,
Circulation, Vol. 73, pp. 418-427 (1986). Recently, the platelet glycoprotein Ilb/IIIa complex (GPIIb/IIIa), has been identified as the membrane protein which mediates platelet aggregation by providing a common pathway for the known platelet agonists. See Philips et al., Cell, Vol. 65, pp. 359-362 (1991).
Platelet activation and aggregation is also thought to play a significant role in venous thromboembolic disorders such as venous thrombophlebitis and subsequent pulmonary emboli. It is also known that patients whose blood flows over artificial surfaces, such as prosthetic synthetic cardiac valves, are at risk for the
development of platelet plugs, thrombi and emboli. See generally Fuster et al., JACC, Vol. 5, No. 6, pp. 175B- 183B (1985); Rubenstein et al., Am. Heart J., Vol. 102, pp. 363-367 (1981); Hamm et al., J. Am. Coll. Cardiol., Vol. 10, pp. 998-1006 (1987); and Davies et al.,
Circulation, Vol. 73, pp. 418-427 (1986).
A suitable means for the non-invasive diagnosis and monitoring of patients with such potential
thromboembolic disorders would be highly useful, and several attempts have been made to develop radiolabeled agents targeted to platelets for non-invasive
radionuclide imaging. For example, experimental studies have been carried out with 99mTc monoclonal antifibrin antibody for diagnostic imaging of arterial thrombus. See Cerqueira et al., Circulation, Vol., 85, pp. 298-304 (1992). The authors report the potential utility of such agents in the imaging of freshly formed arterial thrombus. Monoclonal antibodies labeled with 1311 and specific for activated human platelets have also been reported to have potential application in the diagnosis of arterial and venous thrombi. However, a reasonable ratio of thrombus to blood (target/background) was only attainable at 4 hours after the administration of the radiolabeled antibody. See Wu et al., Clin. Med. J., Vol. 105, pp. 533-559 (1992). The use of 1251, 1311, 99mTc, and lllln radiolabeled 7E3 monoclonal
antiplatelet antibody in imaging thrombi has also been recently discussed. Coller et al., PCT Application Publication No. WO 89/11538 (1989). The radiolabeled 7E3 antibody has the disadvantage, however, of being a very large mole.cular weight molecule. Other researchers have employed enzymatically inactivated t-PA
radioiodinated with 1231, 1251 and 1311 for the
detection and the localization of thrombi. See Ordm et al., Circulation, Vol. 85, pp. 288-297 (1992). Still other approaches in the radiologic detection of
thromoboembolisms are described, for example, in Koblik et al., Semin. Nucl. Med., Vol. 19, pp. 221-237 (1989).
Arterial and venous thrombus detection and
localization is of critical importance in accurately diagnosing thromboembolic disorders and determining proper therapy. New and better radiolabeled agents for non-invasive radionuclide imaging to detect thrombi are needed. The present invention is directed to this important end.
SUMMARY OF THE INVENTION
This invention provides novel radiopharmaceuticals that are radiolabeled cyclic compounds containing carbocyclic or heterocyclic ring systems which act as antagonists of the platelet glycoprotein Ilb/IIIa complex. It also provides methods of using said
radiopharmaceuticals as imaging agents for the diagnosis of arterial and venous thrombi. It further provides novel reagents for the preparation of said
radiopharmaceuticals. It further provides kits
comprising said reagents.
BRIEF DESCRIPTION OF THE FIGURES Figure la. Illustrated are typical images of the radiopharmaceutical compound of Example 12 administered at 1 mCi/Kg,i.v. in a canine deep venous thrombosis model. In this model thrombi were formed in the jugular veins during a period of stasis which was followed by reflow. The compounds were administered beginning at reflow. Depicted is the uptake in a rapidly growing venous thrombus at 15, 60 and 120 min post- administration. Figure lb. Illustrated are typical images of the radiopharmaceutical compound of Example 19 administered at 1 mCi/Kg,i.v. in a canine deep venous thrombosis model. In this model thrombi were formed in the jugular veins during a period of stasis which was followed by reflow. The compounds were administered beginning at reflow. Depicted is the uptake in a rapidly growing venous thrombus at 15, 60 and 120 min post- administration. DETAILED DESCRIPTION OF THE INVENTION
[1] The present invention is directed to novel reagents for preparing a radiopharmaceutical of formulae: (QLn)dCh ; (Q)d'Ln-Ch, wherein, d is 1-3, d' is 2-20, Ln is a linking group, Ch is a metal chelator, and Q is a compound of formula (I):
Figure imgf000007_0001
or a pharmaceutically acceptable salt or
prodirug form thereof, wherein:
R31 is a C6-C14 saturated, partially
saturated, or aromatic carbocyclic ring system, substituted with 0-4 R10 or R10a, and optionally bearing a bond to Ln; a heterocyclic ring system, optionally substituted with 0-4 R10 or R10a, and optionally bearing a bond to Ln;
R32 is selected from:
-C(=O)-;
-C(=S)- -S(=O)2-;
-S(=O)-;
-P(=Z) (ZR13)-;
Z is S or O; "n" and n' are independently 0-2;
R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11 ; a bond to Ln; aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12;
=O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O)N (R13) 2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2,
-N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino)ethoxy; R1 and R21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R12; when n' is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R21 and R23 are independently selected from: hydrogen;
C1-C4 alkyl, optionally substituted with
1-6 halogen;
benzyl;
R22 and R23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2, R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms; R1 and R2, where R21 is H, can
alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R12; R11 is selected from one or more of the following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N (R13) 2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2,
-N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy,
C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C3-C6 cycloalkoxy, C1-C4 alkyl (alkyl being substituted with 1-5 groups selected independently from:
-NR13R14, -CF3, NO2, -SO2R13a, or
-S(=O)R13a), aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12;
R12 is selected from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a,
-C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2. -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl,. C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2- (l-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a);
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-; R14 is OH, H, C1-C4 alkyl, or benzyl; R2 is H or C1-C8 alkyl; R10 and R10a are selected independently from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3-
C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)N (R13)2, -C(=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B(R34) (R35), C3-C6 cycloalkoxy,
-OC(=O)R13, -C(=O)R13,-OC(=O)OR13a,
-OR13, -(C1-C4 alkyl) -OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13,
-NR13C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2,
C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl (including -CVFW where v = 1 to 3 and w = 1 to
(2v+1)). C1-C4 haloalkoxy, C1-C4
alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino) ethoxy, C1-C4 alkyl
(alkyl being substituted with -N(R13)2, -CF3, NO2, or -S(=O)R13a); is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3) C (R4) (R5) C (=O) -, wherein: R3 is H or C1-C8 alkyl;
R4 is H or C1-C3 alkyl; R5 is selected from:
hydrogen;
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2
R11; a bond to Ln; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with
0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N (R13)2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13af -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH)NHR13,
=NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, =NOR13, -B (R34) (R35) , -OCH2CO2H, 2-(1-morpholino) ethoxy, -SC(=NH)NHR13, N3, -Si(CH3)3, (C1-C5 alkyl)NHR16;
-(C0-C6 alkyl) X;
Figure imgf000014_0001
independently 0,1;
Figure imgf000014_0002
-(CH2)mS(O)p' (CH2)2X, where m = 1,2 and p' = 0-2; wherein X is defined below; and
R3 and R4 may also be taken together to form (CH2)nX
Figure imgf000014_0004
-CH2CHCH2-, where
n = 0,1 and X is
Figure imgf000014_0003
R3 and R5 can alternatively be taken together to form -(CH2)t- or -CH2S(O)p-C(CH3)2-, where t = 2-4 and p' = 0-2; or R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from: an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is a D-isomer or L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is selected from: -(C1-C7 alkyl) X;
Figure imgf000015_0001
wherein each q is independently 0-2 and
substitution on the phenyl is at the 3 or 4 position;
Figure imgf000015_0002
wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
Figure imgf000015_0003
-(CH2)mO-(C1-C4 alkyl)-X, where m = 1 or 2;
-(CH2)mS(O)p'-(C1-C4 alkyl)-X, where m = 1 or 2 and p' = 0-2; and
X is selected from:
Figure imgf000016_0001
-C(=NH) (NH2); -SC(=NH)-NH2; -NH-
C(-NH) (NHCN); -NH-C(=NCN) (NH2);
-NH-C(=N-OR13) (NH2);
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000016_0003
-(CH2)qCH(CH2)q-, wherein each q is independently 1 or 2 and wherein n = 0 or 1 and X is -NH2 or
Figure imgf000016_0002
L is -Y(CH2)VC(=O)-, wherein: Y is NH, N(C1-C3 alkyl), O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000017_0001
wherein: q' is 0-2; R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R35 are independently selected from:
-OH,
-F,
-N(R13)2, or C1-C8-alkoxy ;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O.
[2] Included in the present invention are those reagents in [1] above, wherein:
R31 is bonded to (C (R23)R22)n" and
(C(R21)R1)n' at 2 different atoms on said carbocyclic ring.
[3] Included in the present invention are those reagents in [1] above, wherein: n" is 0 and n' is 0;
n" is 0 and n' is 1;
n" is 0 and n' is 2;
n" is 1 and n' is 0;
n" is 1 and n' is 1; n" is 1 and n' is 2;
n" is 2 and n' is 0;
n" is 2 and n' is 1; or
n" is 2 and n' is 2.
[4] Included in the present invention are those reagents in [1] above, wherein:
wherein R6 is methyl, ethyl, or propyl.
[5] Included in the present invention are those reagents in [1] above, wherein:
R32 is selected from:
-C(-O)-;
-C(=S)'- -S(=O)2-; R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11, C2-C8 alkenyl substituted with 0-2 R11, C2-C8 alkynyl substituted with 0-2 R11, C3-C8 cycloalkyl substituted with 0-2
R11,
C6-C10 bicycloalkyl substituted with 0-2 R11; a bond to Ln; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with
0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -CH2N(R13)2, -N(R13)2, -NHC(=NH)NHR13,
-C(=NH)NHR13, NO2;
R1 and R21 can alternatively join to form a 5-7 membered carbocyclic ring
substituted with 0-2 R12; when n' is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R22 and R23 can alternatively join to form a
3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2 , R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R1 and R2, where R21 is H, can alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R12;
R11 is selected from one or more of the
following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -CH2N(R13)2, -N(R13)2, -NHC (=NH) NHR13, -C(=NH)NHR13, =NOR13, NO2; C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C1-C4 alkyl (substituted with -NR13R14, -CF3, NO2, -SO2R13, or -S(=O)R13a) aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with 0-2 R12; is H or CH3; is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C 6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SNHR16, where s = 3-5; a bond to Ln;
R3 and R5 can alternatively be taken together to form -(CH2)t- (t = 2-4) or
-CH2SC(CH3)2-; or
R7 is selected from:
-(C1-C7 alkyl)X;
wherein
Figure imgf000022_0001
each q is
independently 0-2 and substitution on the phenyl is at the 3 or 4 position;
wherein each q
Figure imgf000022_0002
is
independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position; 1
Figure imgf000023_0001
-(CH2)mO-(C1-C4 alkyl)-X, where m = 1 or 2;
- (CH2)mS-(C1-C4 alkyl)-X, where m = 1 or
2; and
X is selected from:
-NH-C(=NH) (NH2), -NHR13, -C (=NH) (NH2),
-SC(NH)-NH2;
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000023_0002
-CH2CHCH2-, where
n = 0 or 1 and X is -NH2 or -NH- C(=NH) (NH2);
L is -Y(CH2)vC(=O)-, wherein:
Y is NH, N(C1-C3 alkyl), O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000024_0001
R8
wherein: q' is 0-2;
R17 is H, C1-C3 alkyl;
R8 is selected from: -CO2R13,-SO3R13, -SO2NHR14, -B (R34) (R35),
-NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl
(said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R3 5 are independently selected from:
-OH,
-F,
-NR13R14, or
C1-C8-alkoxy;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms
independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O.
[6] Included in the present invention are those reagents in [1] above, wherein:
R31 is selected from the group consisting of:
(a) a 6 membered saturated, partially saturated or aromatic carbocyclic ring substituted with 0-3 R10 or R10a' and optionally bearing a bond to Ln;
(b) a 8-11 membered saturated, partially saturated, or aromatic fused bicyclic carbocyclic ring substituted with 0-3 R10 or R10a' and optionally bearing a bond to Ln; or
(c) a 14 membered saturated, partially saturated, or aromatic fused tricyclic carbocyclic ring substituted with 0-3 R10 or R10a' and optionally bearing a bond to Ln.
[7] Included in the present invention are those reagents in [1] above, wherein:
R31 is selected from the group consisting of: (a) a 6 membered saturated, partially saturated, or aromatic carbocyclic ring of formulae:
Figure imgf000026_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted with 0-3 R10, and optionally bears a bond to Ln;
(b) a 10 membered saturated, partially saturated, or aromatic bicyclic carbocyclic ring of formula:
Figure imgf000026_0002
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R10, and optionally bears a bond to Ln;
(c) a 9 membered saturated, partially saturated, or aromatic bicyclic carbocyclic ring of formula:
Figure imgf000027_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R10, and optionally bears a bond to Ln.
[8] Included in the present invention are those reagents in [1] above, wherein:
R31 is selected from (the dashed bond may be a single or double bond):
Figure imgf000027_0002
; or
Figure imgf000028_0001
wherein R31 may be independently substituted with 0-3 R10 or R10a, and optionally bears a bond to Ln; n" is 0 or 1; and n' is 0-2.
[9] Included in the present invention are those reagents in [1] above, wherein:
R1 and R22 are independently selected from: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl) -OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino)ethqxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a).
[10] Included in the present invention are those reagents in [1] above, wherein:
R31 is selected from:
Figure imgf000029_0001
wherein R31 may be independently substituted with 0-3 R10 or R10a, and may optionally bear a bond to Ln;
R32 is -C(=O)-; n" is 0 or 1; n' is 0-2; R1 and R22 are independently selected from H, C1-C4 alkyl, phenyl, benzyl,
phenyl-(C2-C4) alkyl, C1-C4 alkoxy; and a bond to Ln;
R21 and R23 are independently H or C1-C4 alkyl;
R2 is H or C1-C8 alkyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, - (C1-C10
alkyl)aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-; R14 is OH, H, C1-C4 alkyl, or benzyl;
R1 0 and R10a are selected independently from:
H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkqxy; J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein: R3 is H or CH3;
R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)SNHR16, where s = 3-5; and a bond to Ln; or
R3 and R5 can alternatively be taken together to form -(CH2)t- (t = 2-4) or
-CH2SC(CH3)2-; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is
Figure imgf000031_0001
Figure imgf000032_0001
= 0 or 1;
-(CH2)rX, where r = 3-6;
Figure imgf000032_0002
/
-(CH2)mS(CH2)2X, where m = 1 or 2; -(C3-C7 alkyl)-NH-(C1-C6 alkyl);
Figure imgf000032_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2); or
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000032_0004
-CH2CHCH2-, where n = 0 or 1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y(CH2) VC (=O) -, wherein: Y is NH, O, or S ; and v = 1 or 2 ;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000033_0001
wherein : q' is 0-2;
R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a, -NHCONHSO2R13a, -SO2NHCONHR13.
[11] Included in the present invention are those reagents in [1] above, wherein Q is a 1,3- disubstituted phenyl compound of the formula (Il) :
Figure imgf000034_0001
wherein; the shown phenyl ring in formula (II) may be substituted with 0-3 R10, and may optionally bear a bond to Ln;
R10 is selected independently from: H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl,
phenyl-(C1-C4) alkyl, or a bond to Ln;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, - (C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may .
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein:
R3 is H or CH3;
R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
- (CH2)SNHC(=NH)(NH2), -(CH2)SNHR16, where s = 3-5, or a bond to Ln;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of structure -N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is:
Figure imgf000036_0001
q
= 0 or 1;
-(CH2)rX, where r = 3-6;
-
Figure imgf000036_0002
-(CH2)mS(CH2)2X, where m = 1 or 2; -(C3-C7 alkyl)-NH-(C1-C6 alkyl) l )X
Figure imgf000036_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; -(CH2)m-S-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC (=NH) (NH2), provided that X is not -NH2 when r = 4; or
R6 and R7 are alternatively be taken together to form
(CH2)nX
Figure imgf000037_0002
-CH2CHCH2-, where n = 0,1 and X is -NH2 or -NHC (-NH) (NH2);
L is -Y(CH2)VC(=O)-, wherein:
Y is NH, O, or S; and v = 1,2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000037_0001
wherein: q' is 0-2; R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl
(said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13.
[12] Included in the present invention are those
reagents in [1] above, wherein Q is 1,3- disubstituted phenyl compound of the formula (II)
Figure imgf000038_0001
wherein: the phenyl ring in formula (II) may be substituted with 0-3 R10 or R10a;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy; R1 is H, C1-C4 alkyl, phenyl, benzyl, or phenyl- (C2- C4) alkyl;
R2 is H or methyl; R13 is selected independently from:. H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl; R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein: R3 is H or CH3; R4 is H;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3-C6
cycloalkylmethyl, C1-C6 cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SR16, where s = 3-5; or a bond to Ln;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-;
R16 is selected from: an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C3-C8 alkyl;
R7 is
Figure imgf000040_0001
1;
-(CH2)rX, where r = 3-6;
Figure imgf000040_0002
-(CH2)mS(CH2)2X, where m = 1 or 2;
-(C4-C7 alkyl)-NH-(C1-C6 alkyl)
1
Figure imgf000040_0003
-(CH2)m-O-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2), provided that X is not -NH2 when r = 4; or L is -YCH2C(=O)-, wherein; Y is NH or O;
M is a D-isomer or L-isomer amino acid of structure
Figure imgf000041_0001
wherein : q ' is 1 ; R17 is H, C1-C3 alkyl ;
R8 is selected from:
-CO2H or -SO3R13.
[13] Included in the present invention are those
reagents in [1] above, wherein: the phenyl ring in formula (II) bears a bond to Ln, and may be further substituted with 0-2 R10 or
R10a. R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H;
R2 is H;
R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, - (C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of formula -N(R3)CH(R5)C(=O)-, wherein:
R3 is H and R5 is H, CH3, CH2CH3, CH(CH3)2,
CH(CH3)CH2CH3, CH2CH2CH3, CH2CH2CH2CH3,
CH2CH2SCH3, CH2CH(CH3)2, (CH2)4NH2, (C3-C5 alkyl) NHR16;
or
R3 is CH3 and R5 is H; or R3 and R5 can alternatively be taken together to form -CH2CH2CH2-;
R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of formula
-N(CH3)CH(R7)C(=O)-, wherein:
R7 is -(CH2)3NHC(=NH) (NH2); L is -NHCH2C(=O)-; and
M is a D-isomer or L-isomer amino acid of structure
wherein:
Figure imgf000043_0001
q' is 1;
R4 is H or CH3; R17 is H;
R8 is
-CO2H;
-SO3H. [14] Included in the present invention are those reagents in [1] above, wherein: the phenyl ring in formula (II) bears a bond to Ln;
R1 and R2 are independently selected from H,
methyl;
J is selected from D-Val, D-2-aminobutyric acid, D- Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, β-Ala,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala, Nε-p-azidobenzoyl-D-Lys, Nε-p- benzoylbenzoyl-D-Lys, Nε-tryptophanyl-D-Lys, Nε-o-benzylbenzoyl-D-Lys, Nε-p-acetylbenzoyl- D-Lys, Nε-dansyl-D-Lys, Nε-glycyl-D-Lys, Nε- glycyl-p-benzoylbenzoyl-D-Lys, Nε-p- phenylbenzoyl-D-Lys, Nε-m-benzoylbenzoyl-D- Lys, Nε-o-benzoylbenzoyl-D-Lys; K is selected from NMeArg, Arg;
L is selected from Gly, β-Ala, Ala;
M is selected from Asp; (XMeAsp; βMeAsp; NMeAsp; D- Asp.
[15] Included in the present invention are those
reagents in [1] above, wherein: R31 is a phenyl ring and bears a bond to Ln;
R1 and R2 are independently selected from H,
methyl; J is selected from: D-Val, D-2-aminobutyric acid,
D-Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, β-Ala,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala;
K is selected from NMeArg;
L is Gly; M is selected from Asp; OMeAsp; βMeAsp; NMeAsp;
D-Asp.
[16] Included in the present invention are those
reagents in [1]-[15] above, wherein Ch is
selected from the group:
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
wherein :
A1, A2, A3, A4, A5, A6, and A7 are
independently selected at each occurrence from the group: NR40R41, S, SH, S (Pg), O, OH, PR42R43, P(O)R42R43, P(S)R42R43,
P(NR44)R42R43;
W is a bond, CH, or a spacer group selected from the group: C1-C10 alkyl substituted with 0-3 R52, aryl substituted with 0-3
R52 , cycloaklyl substituted with 0-3 R52, heterocycioalkyl substituted with 0-3 R52, aralkyl substituted with 0-3 R52 and alkaryl substituted with 0-3 R52; Wa is a C1-C10 alkyl group or a C3-C14 carbocycle; R40, R41, R42, R43, and R44 are each
independently selected from the group: a bond to Ln, hydrogen, C1-C10 alkyl substituted with 0-3 R52, aryl
substituted with 0-3 R52, cycloaklyl substituted with 0-3 R52,
heterocycloalkyl substituted with 0-3 R52, aralkyl substituted with 0-3 R52' alkaryl substituted with 0-3
R52substituted with 0-3 R52 and an electron, provided that when one of R40 or R41 is an electron, then the other is also an electron, and provided that when one of R42 or R43 is an electron, then the other is also an electron; additionally, R40 and R41 may combine to form =C(C1-C3 alkyl) (C1-C3 alkyl);
R52 is independently selected at each
occurrence from the group: a bond to Ln, =O, F, Cl, Br, I, -CF3, -CN, -CO2R53, -C(=O)R53, -C(=O)N(R53)2, -CHO, -CH2OR53, -OC(=O)R53, -OC(=O)OR53a, -OR53,
-OC(=O)N(R53)2, -NR53C(=O)R53,
-NR54C(=O)OR53a, -NR53C(=O)N(R53)2,
-NR54SO2N(R53)2, -NR54SO2R53a, -SO3H, -SO2R53a, -SR53, -S(=O)R53a, -SO2N(R53)2, -N(R53)2, -NHC(=NH)NHR53, -C (=NH) NHR53, =NOR53, NO2, -C(=O)NHOR53, -C(=O)NHNR53R53a, -OCH2CO2H,
2-(1-morpholino) ethoxy, C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, aryl substituted with 0-2 R53, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O;
R53, R53a, and R54 are independently selected at each occurrence from the group: a bond to Ln, C1-C6 alkyl, phenyl, benzyl, C1-C6 alkoxy, halide, nitro, cyano, and
trifluoromethyl; and Pg is a thiol protecting group capable of
being displaced upon reaction with a radionuclide.
[17] Included in the present invention are those reagents in [1]-[15] above, wherein Ch is selected from the group:
Figure imgf000050_0001
Figure imgf000051_0001
wherein:
A1, A2, A3, A4, A5, A6, and A7 are
independently selected at each occurrence from the group: NR40R41, S, SH, S (Pg), OH;
W is a bond, CH, or a spacer group selected from the group: C1-C3 alkyl substituted with 0-3 R52;
Wa is a methylene group or a C3-C6 carbocycle; R40, R41, R42, R43, and R44 are each
independently selected from the group: a bond to Ln, hydrogen, C1-C10 alkyl
substituted with 0-3 R52, and an
electron, provided that when one of R40 or R41 is an electron, then the other is also an electron, and provided that when one of R42 or R43 is an electron, then the other is also an electron; additionally, R40 and R41 may combine to form, =C(C1-C3 alkyl) (C1-C3 alkyl); R52 is independently selected at each
occurrence from the group: a bond to Ln, =O, F, Cl, Br, I, -CF3, -CN, -CO2R53,
-C(=O)R53, -C(=O)N(R53)2, -CHO, -CH2OR53, -OC(=O)R53, -OC(=O)OR53a, -OR53,
-OC(=O)N(R53)2, -NR53C(=O)R53,
-NR54C (=O)OR53a, -NR53C(=O)N(R53)2,
-NR54SO2N(R53)2, -NR54SO2R53a, -SO3H,
-SO2R53a, -SR53, -S(=O)R53a, -SO2N(R53)2, -N(R53)2, -NHC(=NH)NHR53, -C (=NH) NHR53, =NOR53, NO2, -C(=O)NHOR53,
-C(=O)NHNR53R53a, -OCH2CO2H,
2-(1-morpholino) ethoxy.
R53, R53a, and R54 are independently selected at each occurrence from the group: a bond to Ln, C1-C6 alkyl. [18] Included in the present invention are those reagents in [1]-[15] above, of formula:
(QLn)dCh, wherein d is 1; and Ch is selected from:
y
Figure imgf000053_0001
wherein : A1 and A4 are SH or SPg;
A2 and A3 are NR41;
W is independently selected from the group :
CHR52, CH2CHR52, CH2CH2CHR52 and CHR52C=O; and
R41 and R52 are independently selected from hydrogen and a bond to Ln, and,
Figure imgf000053_0002
wherein: A1 is NH2 or N=C (C1-C3 alkyl) (C1-C3 alkyl);
W is a bond; A2 is NHR40, wherein R40 is heterocycle substituted with R52, wherein the heterocycle is selected from the group: pyridine, pyrazine, proline, furan, thiofuran, thiazole, and diazine, and R52 is a bond to Ln.
[19] Included in the present invention are those
reagents in [1]-[15] above, of formula:
(QLn)dCh, wherein d is 1; and wherein Ch is:
,
Figure imgf000054_0001
wherein:
A1 is NH2 or N=C (C1-C3 alkyl) (C1-C3 alkyl);
W is a bond;
A2 is NHR40, wherein R4 0 is heterocycle
substituted with R52, wherein the heterocycle is selected from pyridine and thiazole, and R52 is a bond to Ln.
[20] Included in the present invention are those
reagents in [1]-[15] above, wherein Ln is: a bond between Q and Ch; or,
a compound of formula: M1-[Y1(CR55R56)h(Z1)h"Y2]h'-M2 wherein: M1 is -[(CH2)gZ1]g,-(CR55R56)g"-;
M2 is -(CR55R56)g"-[Z1(CH2)g]g,-;
g is independently 0-10;
g' is independently 0-1;
g" is 0-10;
h is 0-10;
h' is 0-10;
h" is 0-1
Y1 and Y2, at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C(=O)O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3,
NHC(=O), (NH)2C(=O), (NH)2C=S;
Z1 is independently selected at each
occurrence from a C6-C14 saturated, partially saturated, or aromatic carbocyclic ring system, substituted with 0-4 R57; a heterocyclic ring system, optionally substituted with 0-4 R57;
R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5 R57; (C1-C10 alkyl) aryl wherein the aryl is substituted with 0-5 R57;
R57 is independently selected at each
occurrence from the group: hydrogen,
OH, NHR58, C(=O)R58, OC(=O)R58, OC(=O)OR58, C(=O)OR58, C(=O)NR58-, C=N, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively, when attached to an additional molecule Q, R57 is independently selected at each occurrence from the group: O, NR58, C=O, C(=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO, SO2, SO3, NHC(=O), (NH)2C(=O), (NH)2C=S; and,
R58 is independently selected at each
occurrence from the group:hydrogen;
C1-C6 alkyl; benzyl, and phenyl.
[21] Included in the present invention are those reagents in [1]-[15] above, wherein Ln is: a compound of formula:
M1-[Y1(CR55R56)h(Z1)h"Y2]h,-M2 wherein:
M1 is -[(CH2)gZ1]g'-(CR55R56)g"- ; M2 is -(CR55R56)g"-[Z1(CH2)g]g'-; g is independently 0-10; g' is independently 0-1;
g" is 0-10;
h is 0-10;
h' is 0-10;
h" is 0-1
Y1 and Y2 , at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C(=O)O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3, NHC(=O), (NH)2C(=O), (NH)2C=S;
Z1 is independently selected at each
occurrence from a C6-C14 saturated, partially saturated, or aromatic carbocyclic ring system, substituted with 0-4 R57; a heterocyclic ring system, optionally substituted with 0-4 R57;
R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5
R57;
(C1-C10 alkyl) aryl wherein the aryl is substituted with 0-5 R57;
R57 is independently selected at each
occurrence from the group: hydrogen, OH, NHR58, C(=O)R58, OC(=O)R58, OC(=O)OR58, C(=O)OR58, C(=O)NR58-, C≡N, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively,
when attached to an additional
molecule Q, R57 is independently
selected at each occurrence from the group: O, NR58, C=O, C(=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO,
SO2, SO3, NHC(=O), (NH)2C(=O),
(NH)2C=S, and R57 is attached to an additional molecule Q; and,
R58 is independently selected at each occurrence from the group:hydrogen; C1-C6 alkyl; benzyl, and phenyl.
[22] Included in the present invention are those
reagents in [1]-[15] above, wherein Ln is: -(CR55R56)g"-[Y1(CR55R56)hY2]h'-(CR55R56)g"-, wherein: g" is 1-10;
h is 0-10;
h' is 1-10;
Y1 and Y2, at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C(=O)O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3,
NHC(=O), (NH)2C(=O), (NH)2C=S; R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5
R57;
(C1-C10 alkyl) aryl wherein the aryl
is substituted with 0-5 R57; R57 is independently selected at each occurrence from the group: hydrogen, OH, NHR58, C(=O)R58, OC(=O)R58,
OC(=O)OR58, C(=O)OR58, C(=O)NR58-,
C≡N, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively,
when attached to an additional
molecule Q, R57 is independently
selected at each occurrence from the group: O, NR58, C=O, C(=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO,
SO2, SO3, NHC(=O), (NH)2C(=O),
(NH)2C=S, and R57 is attached to an additional molecule Q; and.
R58 is independently selected at each occurrence from the group:hydrogen; C1-C6 alkyl; benzyl, and phenyl.
[23] Included in the present invention are those
reagents in [1]-[15] above, wherein Ln is:
-(CR55R56)g"-[Y1(CR55R56)hY2]h,-(CR55R56)g"-, Wherein: . g" is 1-5;
h is 0-5;
h* is 1-5;
Y1 and Y2, at each occurrence, are
independently selected from: O, NR56, C=O, C(=O)O, OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3, NHC(=O), (NH)2C(=O), (NH)2C=S;
R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl;
(C1-C10 alkyl) aryl.
[24] Included in the present invention are those reagents in [1]-[15] above, wherein Ln is:
-(CR55R56)g"- [Y1(CR55R56)hY2]h"-(CR55R56)g"- , wherein: g" is 1-5;
h is 0-5;
h' is 1-5;
Y1 and Y2, at each occurrence, are
independently selected from:
O, NR56, C=O, C(=O)O, OC(=O)O, C(=O)NH-, C=NR56, S,
NHC(=O), (NH)2C(=O), (NH)2C=S;
R55 and R56 are independently selected at each occurrence from: hydrogen.
[25] Included in the present invention are those reagents in [1] above, which are:
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
[26] Also included in the present invention is a kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of [23].
[27] Also included in the present invention is a kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of [24].
[28] Also included in the present invention is a kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of [25].
[29] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [1]-[15] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201T1.
[30] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [16] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
[31] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [17] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201τl.
[32] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [18] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
[33] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [19] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
[34] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [20] and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl. [35] Also included in the present invention is a radiopharmaceutical comprising a complex of a reagent of [21] and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
[36] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [22] and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
[37] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [23] and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
[38] Also included in the present invention is a
radiopharmaceutical comprising a complex of a reagent of [24] and a radionuclide selected from the group 99mTc, and 111In.
[39] Also included in the present invention are the radiopharmaceuticals of [29] which are:
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
[40] Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
administering to said mammal an effective amount of a radiopharmaceutical of [29], and (ii) scanning the mammal using a radioimaging devise. [41] Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
administering to said mammal an effective amount of a radiopharmaceutical of [30], and (ii) scanning the mammal using a radioimaging devise.
[42] Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
administering to said mammal an effective amount of a radiopharmaceutical of [31], and (ii) scanning the mammal using a radioimaging devise.
[43] Also included in the present invention is a method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i)
administering to said mammal an effective amount of a radiopharmaceutical of [32], and (ii) scanning the mammal using a radioimaging devise.
[44] A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [33], and (ii) scanning the mammal using a radioimaging devise.
[45] A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [34], and (ii) scanning the mammal using a radioimaging devise.
[46] A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [35], and (ii) scanning the mammal using a radioimaging devise.
[47] A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [36], and (ii) scanning the mammal using a radioimaging devise. [48] A method for visualizing sites of platelet deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [37], and (ii) scanning the mammal using a radioimaging devise.
[49] A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of [38], and (ii) scanning the mammal using a radioimaging devise.
[50] A method for visualizing sites of platelet
deposition in a mammal by radioimagin.g, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 39, and (ii) scanning the mammal using a radioimaging devise. [51] The present invention is also directed to
direct radiolabeled compounds of formula (I):
Figure imgf000074_0001
or a pharmaceutically acceptable salt or
prodrug form thereof wherein: R31 is a C6-C14 saturated, partially
saturated, or aromatic carbocyclic ring system substituted with 0-4 R10 or R10a; R32 is selected from:
-C(=O)-;
-C(=S)- -S(=O)2-;
-S(=O)-;
-P(=Z) (ZR13)-;
Z is S or O; n" and n' are independently 0-2;
R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO,, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C(=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy;
R1 and R21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R12; when n1 is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R22 and R23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2, R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to .form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms; R1 and R2, where R21 is H, can
. alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R12;
R11 is selected from one or more of the
following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CH0, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N (R13) 2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2,
-N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy, C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C3-C6 cycloalkoxy, C1-C4 alkyl (alkyl being substituted with 1-5 groups selected independently from:
-NR13R14, -CF3, NO2, -SO2R13a, or
-S(=O)R13a), aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said heterocyclic ring being substituted with
0-2 R12;
R12 is selected from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C (=O) R13,
-NR13C (=O)OR13a, -NR13C (=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S (=O)R13a);
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
R21 and R23 are independently selected from: hydrogen;
C1-C4 alkyl, optionally substituted with
1-6 halogen;
benzyl;
R2 is H or C1-C8 alkyl;
R10 and R10a are selected independently from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)N(R13)2,
-C(=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B(R34) (R35), C3-C6 cycloalkoxy,
-OC (=O) R13, -C (=O) R13, -OC (=O) OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13,
-NR13C (=O)OR13a, -NR13C (=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl (including -CVFW where v = 1 to 3 and w = 1 to
(2v+1)), C1-C4 haloalkoxy, C1-C4
alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H,
2-(1-morphrlino) ethoxy, C1-C4 alkyl
(alkyl being substituted with -N(R13)2, -CF3, NO2, or -S(=O)R13a); J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or C1-C8 alkyl; R4 is H or C1-C3 alkyl;
R5 is selected from:
hydrogen;
C1-C8 alkyl substituted with 0-2 R11;
C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11; aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, =NOR13, -B (R34) (R35),
-OCH2CO2H, 2-(1-morpholino)ethoxy,
-SC(=NH)NHR13, N3, -Si(CH3)3, (C1-C5 alkyl) NHR16; - (C0-C6 alkyl ) X;
Figure imgf000081_0001
independently 0 , 1 ;
Figure imgf000081_0002
-(CH2)mS(O)p' (CH2)2X, where m = 1,2 and p' = 0-2; wherein X is defined below; and
R3 and R4 may also be taken together to form (CH2)nX
Figure imgf000081_0003
-CH2CHCH2- , where n = 0 , 1 and X is
Figure imgf000082_0001
R3 and R5 can alternatively be taken together to form -(CH2)t- or -CH2S (O)p'C(CH3)2-, where t = 2-4 and p' = 0-2; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5; R16 is selected from:
an amine protecting group;
1-2 amino acids;
4-2 amino acids substituted with an amine protecting group;
K is a D-isomer or L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl;
R7 is selected from: -(C1-C7 alkyl)X;
Figure imgf000082_0002
wherein each q is independently 0-2 and
substitution on the phenyl is at the 3 or 4 position;
Figure imgf000083_0001
wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
Figure imgf000083_0002
-(CH2)mO-(C1-C4 alkyl)-X, where m = 1 or 2;
-(CH2)mS(O)p'-(C1-C4 alkyl)-X, where m = 1 or 2 and p' = 0-2; and
X is selected from:
-N(R13)R13;
Figure imgf000083_0003
-C(=NH) (NH2); -SC (=NH) -NH2; -NH- C (=NH) (NHCN); -NH-C (=NCN) (NH2);
-NH-C (=N-OR13) (NH2);
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000083_0004
-(CH2)qCH(CH2)q- wherein each q is independently 1 or 2 and wherein n = 0 or 1 and X is -NH2 or
Figure imgf000084_0001
L is -Y(CH2)vC(=O)-, wherein:
Y is NH, N(C1-C3 alkyl), O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
wherein:
Figure imgf000084_0002
q ' is 0-2 ; R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R35 are independently selected from:
-OH,
-F,
-N(R13)2, or
C1-C8-alkoxy;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; and wherein the radiolabel is selected from the group: 123I, 125I, 131I, 18F, 11C, 13N, 15 O, 75Br. [52] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein:
R31 is bonded to (C(R23)R22)n" and
(C(R21)R1)n' at 2 different atoms on said carbocyclic ring. [53] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein: n" is 0 and n' is 0;
n" is 0 and n' is 1;
n" is 0 and n' is 2;
n" is 1 and n' is 0;
n" is 1 and n' is 1;
n" is 1 and n' is 2;
n" is 2 and n' is 0;
n" is 2 and n' is 1; or
n" is 2 and n' is 2.
[54] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein R6 is methyl, ethyl, or propyl.
[55] Included ih the present invention are those direct radiolabeled compounds in [51] above, wherein:
R31 is selected from the group consisting of: (a) a 6 membered saturated, partially . saturated or aromatic carbocyclic ring substituted with 0-3 R10 or R10a; (b) a 8-11 membered saturated, partially saturated, or aromatic fused bicyclic carbocyclic ring substituted with 0-4 R10 or R10a; or (c) a 14 membered saturated, partially saturated, or aromatic fused tricyclic carbocyclic ring substituted with 0-4 R10 or R10a.
[56] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein: R31 is selected from the group consisting of:
(a) a 6 membered saturated, partially saturated, or aromatic carbocyclic ring of formula:
Figure imgf000087_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10;
(b) a 10 membered saturated, partially saturated, or aromatic bicyclic
carbocyclic ring of formula:
Figure imgf000088_0001
, wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10 or R10a;
(c) a 9 membered saturated, partially saturated, or aromatic bicyclic
carbocyclic ring of formula:
Figure imgf000088_0002
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10 or R10a. [57] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein:
R31 is selected from (the dashed bond may be a single or double bond):
Figure imgf000089_0001
n" is 0 or 1; and n' is 0-2.
[58] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein:
R1 and R22 are independently selected from: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a,
-C(=O)NHN(R13)2, =NOR13, -B (R34) (R35) , C3- C6 cycloalkoxy, -OC (=O) R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a).
[59] Included in the present invention are those direct radiolabeled compounds in [51] above, wherein:
R31 is selected from:
Figure imgf000091_0001
wherein R31 may be substituted
independently with 0-3 R10 or R10a;
R32 is -C(=O)-; n" is 0 or 1; n' is 0-2;
R1 and R22 are independently selected from H, C1-C4 alkyl, phenyl, benzyl,
phenyl-(C2-C4) alkyl, C1-C4 alkoxy;
R21 and R23 are independently H or C1-C4 alkyl ; R2 is H or C1-C8 alkyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form
-(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl; R10 and R10a are selected independently from:
H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or CH3;
R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)sNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)SNHR16, where s = 3-5; or
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
R3 and R5 can alternatively be taken together to form -(CH2)t- (t - 2-4) or
-CH2SC(CH3)2-; or R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein: R6 is H or C1-C8 alkyl; R7 is
Figure imgf000093_0001
= 0 or 1;
-(CH2)rX, where r = 3-6;
Figure imgf000094_0001
-(CH2)mS(CH2)2X, where m = 1 or 2;
-(C3-C7 alkyl)-NH-(C1-C6 alkyl)
Figure imgf000094_0002
-(CH2)m-O-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and X is -NH2 or -NHC(=NH) (NH2); or
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000094_0003
-CH2CHCH2-, where n = 0 or 1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y(CH2)vC(=O)-, wherein:
Y is NH, O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000095_0001
wherein :
q ' is 0-2 ;
R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35),
-NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl Osaid heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl
(said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13.
[60] Included in the present invention are those direct radiolabeled compounds in [51] above, that are radiolabeled 1,3- disubstituted phenyl compounds of the formula (II):
Figure imgf000096_0001
wherein: the shown phenyl ring in formula (II) may be further substituted with 0-3 R10;
R10 is selected independently from: H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl, or phenyl-(C1- C4) alkyl;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-; R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or CH3; R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)SNHR16, where s = 3-5; or R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is :
Figure imgf000098_0001
= 0 or 1;
-(CH2)rX, where r = 3-6;
Figure imgf000098_0002
-(CH2)mS(CH2)2X, where m = 1 or 2;
-(C3-C7 alkyl)-ΝH-(C1-C6 alkyl)
Figure imgf000098_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2), provided that X is not -NH2 when r = 4; or R6 and R7 are alternatively be taken, together to form
(CH2)nX
Figure imgf000099_0002
-CH2CHCH2-, where n = 0,1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y(CH2)vC(=O)-, wherein:
Y is NH, O, or S; and v = 1,2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000099_0001
wherein: q' is 0-2; R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13) R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13.
[61] Included in the present invention are those
direct radiolabeled compounds in [51] above, that are radiolabeled 1,3-disubstituted phenyl compounds of the formula (II):
Figure imgf000100_0001
wherein: the phenyl ring in formula (II) may be further substituted with 0-3 R10 or R10a;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl, or phenyl- (C2- C4) alkyl;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or CH3; is H;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C1-C6 cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SR16, where s = 3-5;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C3-C8 alkyl;
R7 is
1;
Figure imgf000102_0001
-(CH2)rX, where r = 3-6;
Figure imgf000102_0002
- (CH2)mS(CH2)2X, where m = 1 or 2;
-(C4-C7 alkyl)-NH-(C1-C6 alkyl)
Figure imgf000102_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; -(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2), provided that X is not -NH2 when r = 4; or
L is -YCH2C(=O)-, wherein:
Y is NH or O;
M is a D-isomer or L-isomer amino acid of structure
wherein:
Figure imgf000103_0001
q' is 1;
R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2H or -SO3R13.
[62] Included in the present invention are those
direct radiolabeled compounds in of formula
(II) above, wherein: the phenyl ring in formula (II) may be further
substituted with 0-2 R10 or R10a; R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy; R1 is H ;
R2 is H; R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; R13a is C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of formula -N(R3)CH(R5)C(=O)-, wherein:
R3 is H and R5 is H, CH3, CH2CH3, CH(CH3)2,
CH(CH3)CH2CH3, CH2CH2CH3, CH2CH2CH2CH3,
CH2CH2SCH3, CH2CH(CH3)2, (CH2)4NH2, (C3-C5 alkyl)NHR16;
or R3 is CH3 and R5 is H; or
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of formula
-N(CH3)CH(R7)C(=O)-, wherein: R7 is -(CH2)3NHC(=NH) (NH2);
L is -NHCH2C(=O)-; and
M is a D-isomer or L-isomer amino acid of structure
wherein:
Figure imgf000105_0001
q' is 1;
R4 is H or CH3;
R17 is H;
R8 is
-CO2H;
-SO3H .
[63] Included in the present invention are those
direct radiolabeled compounds in of formula
(II) above, wherein: R1 and R2 are independently selected from H, methyl; ,
J is selected from D-Val, D-2-aminobutyric acid, D- Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, β-Ala,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala, Nε-p-azidobenzoyl-D-Lys, Nε-p- benzoylbenzoyl-D-Lys, Nε-tryptophanyl-D-Lys, Nε-o-benzylbenzoyl-D-Lys, Nε-p-acetylbenzoyl- D-Lys, N-ε-dansyl-D-Lys, Nε-glycyl-D-Lys, Nε- glycyl-p-benzoylbenzoyl-D-Lys, Nε-p- phenylbenzoyl-D-Lys, Nε-m-benzoylbenzoyl-D- Lys, Nε-o-benzoylbenzoyl-D-Lys; K is selected from NMeArg, Arg;
L is selected from Gly, β-Ala, Ala;
M is selected from Asp; OMeAsp; βMeAsp; NMeAsp; D- Asp.
[64] Included in the present invention are those
direct radiolabeled compounds in of formula
(II) above, wherein:
R1 and R2 are independently selected from H,
methyl; J is selected from: D-Val, D-2-aminobutyric acid,
D-Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, β-Ala,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala; K is selected from NMeArg; L is Gly; K is selected from Asp; OMeAsp; βMeAsp; NMeAsp;
D-Asp.
[65] Included in the present invention are those
direct radiolabeled compounds of [51] that
are: the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-2-aminobutyric acid; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Leu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Ala; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Gly; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Pro; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Lys; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is β-Ala; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is NMeGly; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is methyl (isomer 1); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is methyl (isomer 2); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is phenyl (isomer 1); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein J = D-Met, K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H; the radiolabeled compound of formula (II) wherein J = D-Abu, K = diNMe-guanidinyl-Orn , L = Gly, M = Asp, R1 - H, R2 = H; the radiolabeled compound of formula (II) wherein J = D-Abu, K = diNMe-Lys, L = Gly, M = Asp, R1 = H, R2 = H; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- azidobenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-tryptophanyl-
D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-o- benzylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp.
The radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- acetylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-dansyl-D-
Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-glycyl-D-
Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-glycyl-p- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- phenylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-m- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-o- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (III) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000111_0001
the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is D- NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Nle; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Phg; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Phe; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (V) wherein R1 and R2 are H; J is D-Ile; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000112_0001
the radiolabeled compound of formula (V) wherein n"=1; R1, R2, and R22 are H; J is D- Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (V) wherein n"=0; R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000112_0002
the radiolabeled compound of formula (VI) wherein R2 and R22 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000113_0001
the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is Cl; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is l; j is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is I; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R1 0 are H; R10a is Me; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R1 0 a are H; R1 0 is Cl; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10a are H; R10 is MeO; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10a are H; R10 is Me; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is Cl; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1, R2, and R10 are H; R10a is I; J is
D-Abu; K is NMeArg; L is Gly; and M is Asp.
The radiolabeled compound of formula (VII) wherein R1, R2, and R10 are H; R10a is Me; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Tyr; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is NMeAmf; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is βMeAsp; the radiolabeled compound of formula (II) wherein R1 is H; R2 is CH3; J is D-Val; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (III) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula
(VIII) wherein J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000115_0001
[66] Included in the present invention are those
radiolabeled compound as in one of [51]- [65] wherein the- radiolabel is selected from the group: 18F, 11C, 123I, and 125I.
[67] Included in the present invention are those
radiolabeled compounds of [66] wherein the radiolabel is 123I. [68] Included in the present invention is a
radiopharmaceutical composition comprising a radiopharmaceutically acceptable carrier and a radiolabeled compound of any of [51]-[67].
[69] Included in the present invention is a method
of determining platelet deposition in a mammal comprising administering to said mammal a
radiopharmaceutical composition comprising a compound of any of [51]-[67], and imaging said mammal.
[70] Included in the present invention is a method of diagnosing a disorder associated with. platelet deposition in a mammal comprising administering to said mammal a radiopharmaceutical composition comprising a compound of any of [51]-[67], and imaging said mammal. As noted above, the cyclic compounds of the present invention are radiolabeled. By "radiolabeled", it is meant that the subject cyclic platelet glycoprotein Ilb/IIIa compounds contain a radioisotope which is suitable for administration to a mammalian patient.
Suitable radioisotopes are known to those skilled in the art and include, for example, isotopes of halogens (such as chlorine, fluorine, bromine and iodine), and metals including technetium and indium. Preferred radioisotopes include 11C, 18F, 123I, 125I, 131I, 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl. Most preferred are the isoptopes 123I, 111ln, and 99mTc. Radiolabeled compounds of the invention may be prepared using standard radiolabeling procedures well known to those skilled in the art. Suitable synthesis methodology is described in detail below. As discussed below, the cyclic platelet
glycoprotein Ilb/IIIa compounds of the invention may be radiolabeled either directly (that is, by incorporating the radiolabel directly into the compounds) or
indirectly (that is, by incorporating the radiolabel into the compounds through a chelating agent, where the chelating agent has been incorporated into the
compounds). Also, the radiolabeling may be isotopic or nonisotopic. With isotopic radiolabeling, one group already present in the cyclic compounds described above is substituted with (exchanged for) the radioisotope. With nonisotopic radiolabeling, the radioisotope is added to the cyclic compounds without substituting with (exchanging for) an already existing group. Direct and indirect radiolabeled compounds, as well as isotopic and nonisotopic radiolabeled compounds are included within the phrase "radiolabeled compounds" as used in
connection with the present invention. Such
radiolabeling should also be reasonably stable, both chemically and metabolically, applying recognized standards in the art. Also, although the compounds of the invention may be labeled in a variety of fashions with a variety of different radioisotopes, as those skilled in the art will recognize, such radiolabeling should be carried out in a manner such that the high binding affinity and specificity of the unlabeled cyclic platelet GPIIb/IIIa compounds of the invention to the GPIIb/IIIa receptor is not significantly affected. By not significantly affected, it is meant that the binding affinity and specificity is not affected more than about 3 log units, preferably not more than about 2 log units, more preferably not more than about 1 log unit, even more preferably not more than about 500%, and still even more preferably not more than about 250%, and most preferably the binding affinity and specificity is not affected at all.
For radiolabeled compounds, the label may appear at any position on Q. Preferred radiolabeled compounds of the invention are radiolabeled compounds wherein the radiolabel is located on the carbocyclic ring system of R31, the R5 substituent on J, and at R1 or R22. Even more preferred radiolabeled compounds of the invention are those of formula (II), wherein the radiolabel is located on the carbocyclic ring system of R31, or the R5 substituent on J. With regard to the preferred and more preferred direct radiolabeled compounds, the preferred radiolabel is a halogen label, especially an iodine radiolabel. For indirect radiolabeled compounds, the preferred metal nuclides are 99mTc and 111 ln. Preferred linking groups, Ln, and metal chelators, Ch, are
described below.
It has been discovered that the radiolabeled compounds of the invention are useful as
radiopharmaceuticals for non-invasive imaging to
diagnose present or potential thromboembolic disorders, such as arterial or venous thrombosis, including, for example, unstable angina, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, diabetes, thrombophlebitis, pulmonary emboli, or
platelet plugs, thrombi or emboli caused by prosthetic cardiac devices such as heart valves . The radiolabeled compounds of the invention are useful with both newly formed and older thrombi. The radiolabeled compounds of the invention may also be used to diagnose other present or potential conditions where there is overexpression of the GPIIb/IIIa receptors, such as with metastatic cancer cells. The subject compounds may be effectively employed in low doses, thereby minimizing any risk of toxicity. Also, the subject compounds are of a much smaller size than, for example, the radiolabeled 7E3 antibodies known in the art, allowing easier attainment of suitable target/background (T/B) ratio for detecting thrombi. The use of the radiolabeled compounds of the invention is further described in the utility section below.
In the present invention it has also been
discovered that the radiolabeled compounds above are useful as inhibitors of glycoprotein Ilb/IIIa
(GPIIb/IIIa), and thus the radiolabeled compounds of the invention may also be employed for therapeutic purposes, in addition to the diagnostic usage described above. As discussed above, GPIIb/IIIa mediates the process of platelet activation and aggregation. The radiolabeled compounds of the present invention inhibit the
activation and aggregation of platelets induced by all known endogenous platelet agonists.
The compounds herein described may have asymmetric centers. Unless otherwise indicated, all chiral, diastereomeric and racemic forms are included in the present invention. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. It will be appreciated that compounds of the present invention contain asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, from optically active starting materials. Two distinct isomers (cis and trans) of the peptide bond are known to occur; both can also be present in the compounds described herein, and all sύch stable isomers are contemplated,in the present invention. Unless otherwise specifically noted, the L- isomer of the amino acid is used at positions J, K, L, and M of the compounds of the present invention. Except as provided in the preceding sentence, all chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomer form is specifically
indicated. The D and L-isomers of a particular amino acid are designated herein using the conventional 3- letter abbreviation of the amino acid, as indicated by the following examples: D-Leu, D-Leu, L-Leu, or L-Leu.
When any variable (for example, R1 through R8, m, n, p, X, Y, etc.) occurs more than one time in any constituent or in any formula, its definition on each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 R11, then said group may optionally be substituted with up to two R11 and R11 at each occurrence is selected independently from the defined list of possible R11. Also, by way of example, for the group -N(R13)2, each of the two R13 substituents on N is independently selected from the defined list of possible R13.
When a bond to a substituent is shown to cross the bond connecting two atoms in a ring, then such
substituent may be bonded to any atom on the ring.
Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
By "stable compound" or "stable structure" is meant herein a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The term "substituted", as used herein, means that an one or more hydrogen on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound. When a substitent is keto (i.e., =O), then 2 hydrogens on the atom are replaced.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic
hydrocarbon groups having the specified number of carbon atoms; "haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen (for example -CVFW where v = 1 to 3 and w = 1 to (2v+1)); "alkoxy"
represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge; "cycloalkyl" is intended to include saturated ring groups, including mono-,bi- or poly-cyclic ring systems, such as
cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and adamantyl; and "biycloalkyl" is intended to include saturated bicyclic ring groups such as [3.3.0]bicyclooctane, [4.3.0]bicyclononane,
[4.4.0]bicyclddecane (decalin), [2.2.2]bicyclooctane, and so forth. "Alkenyl" is intended to include
hydrocarbon chains of either a straight or branched configuration and one or more unsaturated carbon-carbon bonds which may occur in any stable point along the chain, such as ethenyl, propenyl and the like; and "alkynyl" is intended to include hydrocarbon chains of either a straight or branched configuration and one or more triple carbon-carbon bonds which may occur in any stable, point along the chain, such as ethynyl, propynyl and the like. The phrase "boronic acid" as used herein means a group of the formula -B(R34) (R35), wherein R34 and R35 are independently selected from: -OH; -F; -NR13R14; or C1-C6-alkoxy; or R34 and R35 can alternatively be taken together to form: a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and,
optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and,
optionally, 1-4 heteroatoms independently selected from N, S, or O. Such cyclic boron esters, boron amides, or boron amide-esters may also be optionally substituted with 1-5 groups independently selected from R11.
Boron esters include boronic acid protecting groups, including moieties derived from diols, for example pinanediol and pinacol to form pinanediol boronic acid ester and the pinacol boronic acid, respectively. Other illustrations of diols useful for deriving boronic acid esters are perfluoropinacol, ethylene glycol, diethylene glycol, 1,2-ethanediol, 1,3-propanediol, 1,2-propanediol, 1,2-butanediol,
1,4-butanediol, 2,3-butanediol, 2,3-hexanediol,
1,2-hexanediol, catechol, 1,2-diisopropylethanediol, 5,6-decanediol, 1,2-dicyclohexylethanediol.
"Halo" or "halogen" as used herein refers to fluoro, chloro, bromo and iodo; and "counterion" is used to represent a small, negatively charged species such as chlori.de, bromide, hydroxide, acetate, sulfate and the like.
As used herein, "aryl" or "aromatic residue" is intended to mean phenyl or naphthyl. As used herein, "carbocycle" or "carbocyclic residue" is intended to mean any stable 3- to 7- membered monocyclic or bicyclic or 7- to 14-membered bicyclic or tricyclic or an up to 26-membered polycyclic carbon ring, any of which may be saturated, partially unsaturated, or aromatic. Examples of such carbocyles include, but are not limited to, cyclopropyl, cyclopentyl, cyclohexyl, phenyl, biphenyl, naphthyl, indanyl, adamantyl, or tetrahydronaphthyl (tetralin).
As used herein, the term "heterocycle" or
"heterocyclic ring system" is intended to mean a stable 5- to 7- membered monocyclic or bicyclic or 7- to 10- membered bicyclic heterocyclic ring which may be
saturated, partially unsaturated, or aromatic, and which consists of carbon atoms and from 1 to 4 heteroatoms selected independently from the group consisting of N, O and S and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen may
optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting
compound is stable. Examples of such heterocycles include, but are not limited to, benzopyranyl,
thiadiazine, tetrazolyl, benzofuranyl, benzothiophenyl, indolene, quinoline, isoquinolinyl or benzimidazolyl, piperidinyl, 4-piperidone, 2-pyrrolidone,
tetrahydrofuran, tetrahydroquinoline,
tetrahydroisoquinoline, decahydroquinoline,
octahydroisoquinoline, azocine, triazine "(including 1,2,3-, 1,2,4-, and 1,3,5-triazine), 6H-1,2,5- thiadiazine, 2H, 6H-1,5,2-dithiazine, thiophene, tetrahydrothiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, 2H-pyrrole, pyrrole, imidazole, pyrazole, thiazole, isothiazole, oxazole (including 1,2,4- and 1,3,4- oxazole), isoxazole, triazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, 3H- indole, indole, 1H-indazole, purine, 4H-quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine,
4aH-carbazole, carbazole, β-carboline, phenanthridine, acridine, perimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, isochroman, chroman, pyrrolidine, pyrroline,
imidazolidine, imidazoline, pyrazolidine, pyrazoline, piperazine, indoline, isoindoline, quinuclidine, or morpholine. Also included are fused ring and spiro compounds containing, for example, the above
heterocycles.
As used herein, the term "any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, aminό or sulfhydryl" means any group bonded to an O, N, or S atom, respectively, which is cleaved from the O, N, or S atom when the compound is administered to a mammalian subject to provide a compound having a remaining free hydroxyl, amino, or sulfhydryl group, respectively. Examples of groups that, when administered to a mammalian subject, are cleaved to form a free hydroxyl, amino or sulfhydryl, include but are not limited to, C1-C6 alkyl substituted with 0-3 R11, C3-C6 alkoxyalkyl substituted with 0-3 R11, C1-C6 alkylcarbonyl substituted with 0-3 R11, C1-C6 alkoxycarbonyl substituted with 0-3 R11, C1-C6
alkylaminocarbonyl substituted with 0-3 R11, benzoyi substituted with 0-3 R12, phenoxycarbonyl substituted with 0-3 R12, phenylaminocarbonyl substituted with 0-3 R12. Examples of groups that, when administered to a mammalian subject, are cleaved to form a free hydroxyl, amino or sulfhydryl, include hydroxy, amine or
sulfhydryl protecting groups, respectively.
As used herein, the term "amine protecting group" means any group known in the art of organic synthesis for the protection of amine groups. Such amine
protecting groups include those listed in Greene,
"Protective Groups in Organic Synthesis" John Wiley & Sons, New York (1981) and "The Peptides: Analysis, Sythesis, Biology, Vol. 3, Academic Press, New York (1981), the disclosure of which is hereby incorporated by reference. Any amine protecting group known in the art can be used. Examples of amine protecting groups include, but are not limited to, the following: 1) acyl types such as formyl, trifluoroacetyl, phthalyl, and p-toluenesulfonyl; 2) aromatic carbamate types such as benzyloxycarbonyl (Cbz or Z) and substituted
benzyloxycarbonyls, 1-(p-biphenyl)-1- methylethoxycarbonyl, and 9-fluorenylmethyloxycarbonyl (Fmoc); 3) aliphatic carbamate types such as tert- butyloxycarbonyl (Boc), ethoxycarbonyl,
diisopropylmethoxycarbonyl, and allyloxycarbonyl; 4) cyclic alkyl carbamate types such as cyclopentyloxycarbonyl and adamantyloxycarbonyl; 5) alkyl types such as triphenylmethyl and benzyl; 6) trialkylsilane such as trimethylsilane; and 7) thiol containing types such as phenylthiocarbonyl and
dithiasuccinoyl. Also included in the term "amine protecting group" are acyl groups such as azidobenzoyl, p-benzoylbenzoyl, o-benzylbenzoyl, p-acetylbenzoyl, dansyl, glycyl-p-benzoylbenzoyl, phenylbenzoyl,
m-benzoylbenzoyl, benzoylbenzoyl.
As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound of formula (I) is modified by making acid or base salts of the compound of formula (I).
Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
Pharmaceutically acceptable salts of the compounds of the invention can be prepared by reacting the free acid or base forms of these compounds with a
stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in
Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
The term "amino acid" as used herein means an organic compound containing both a basic amino group and an acidic carboxyl group. Included within this term are modified and unusual amino acids, such as those
disclosed in, for example, Roberts and Vellaccio (1983) The Peptides, 5: 342-429, the teaching of which is hereby incorporated by reference. Modified or unusual amino acids which can be used to practice the invention include, but are not limited to, D-amino acids,
hydroxylysine, 4-hydroxyproline, ornithine,
2,4-diaminobutyric acid, homoarginine, norleucine, N-methylaminobutyric acid, naphthylalanine,
phenylglycine, β-phenylproline, tert-leucine,
4-aminocyclohexylalanine, N-methyl-norleucine,
3,4-dehydroproline, 4-aminopiperidine-4-carboxylic acid, 6-aminocaproic acid, trans-4-(aminomethyl)- cyclohexanecarboxylic acid, 2-, 3-, and 4-(aminomethyl)- benzoic acid, 1-aminocyclopentanecarboxylic acid,
1-aminocyclopropanecarboxylic acid, and 2-benzyl-5- aminopentanoic acid.
The term "amino acid residue" as used herein means that portion of an amino acid (as defined herein) that is present in a peptide.
The term "peptide" as used herein means a linear compound that consists of two or more amino acids (as defined herein) that are linked by means of a peptide bond. The term "peptide" also includes compounds containing both peptide and non-peptide components, such as pseudopeptide or peptide mimetic residues or other non-amino acid components. Such a compound containing both peptide and non-peptide components may also be referred to as a "peptide analog".
A "pseudopeptide" or "peptide mimetic" is a compound which mimics the structure of an amino acid residue or a peptide, for example, by using linking groups other than amide linkages between the peptide mimetic and an amino acid residue (pseudopeptide bonds) and/or by using non-amino acid substituents and/or a modified amino acid residue.
A "pseudopeptide residue" means that portion of an pseudopeptide or peptide mimetic (as defined herein) that is present in a peptide.
The term "peptide bond" means a covalent amide linkage formed by loss of a molecule of water between the carboxyl group of one amino acid and the amino group of a second amino acid.
The term "pseudopeptide bonds" includes peptide bond isosteres which may be used in place of or as substitutes for the normal amide linkage. These substitute or amide "equivalent" linkages are formed from combinations of atoms not normally found in peptides or proteins which mimic the spatial
requirements of the amide bond and which should
stabilize the molecule to enzymatic degradation.
The terms "Ln", "linking group" and "linker", used interchangeably throughout, designate the group of atoms separating Q from the metal chelator, Ch.
The terms "activated Ln group", "activated Ln", "activated linking group" and "activated linker", used interchangeably throughout, refer to a linking group that bears one or more reactive group capable of reacting with, and forming a bond with, a chelator or a
Q.
The terms "Ch", "metal chelator", and "chelator" are used interchangeably throughout to designate a chemical moiety capable of binding to or complexing with a metal nuclide.
The term "cyclizing moiety" means the intermediate compound that serves as the precursor to the R31 group of Q. The term "ring substituted cyclizing moiety" is a cycliz'ing moiety bearin a substituent group one or more of its carbocyclic or heterocyclic rings.
The term "linker modified cyclizing moiety" refers to a cyclizing moiety that bears an activated Ln group.
The term "cyclic compound intermediate" means the intermediate compound that serves as the precursor to the Q group in the claimed compounds .
The term "linker modified cyclic compound
intermediate" means a cyclic compound intermediate that bears an activated Ln group.
The compounds of the present invention can be prepared in a number of ways well known to one skilled in the art of organic synthesis. Preferre-d methods include but are not limited to those methods described below.
The following abbreviations are used herein: Acm acetamidomethyl
D-Abu D-2-aminobutyric acid
5-Aca 5-aminocaproamide (5-aminohexanamide) b-Ala, b-Ala or
bAla 3-aminopropionic acid
Boc t-butyloxycarbonyl
Boc-iodo-Mamb t-butyloxycarbonyl-3-aminomethyl-4-iodo- benzoic acid
Boc-Mamb t-butyloxycarbonyl-3-aminomethylbenzoic acid
Boc-ON [2-(tert-butyloxycarbonyloxylimino)-2- phenylacetoni-trile
Cl2Bzl dichlorobenzyl
CBZ, Cbz or Z Carbobenzyloxy
DCC dicyclohexylcarbodiimide DIEA diisopropylethylamlne
di-NMeOrn N-aMe-N-gMe-ornithine
DMAP 4-dimethylaminopyridine
HBTU 2-(1H-Benzotriazol-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate
NMeArg or
MeArg a-N-methyl arginine
NMeAmf N-Methylaminomethylphenylalanine
NMeAsp a-N-methyl aspartic acid
NMeGly or
MeGly N-methyl glycine
NMe-Mamb N-methyl-3-aminomethylbenzoic acid
NMM N-methylmorpholine
OcHex O-cyclohexyl
OBzl O-benzyl
oSu O-succinimidyl
pNP p-nitrophenyl
TBTU 2-(1H-Benzotriazol-1-yl)-1,1,3,3- tetramethyluronium
tetrafluoroborate
Teoc 2-(Trimethylsilyl)ethyloxycarbonyl Tos tosyl
Tr trityl The following conventional three-letter amino acid abbreviations are used herein; the conventional one- letter amino acid abbreviations are not used herein:
Ala = alanine
Arg = arginine
Asn = asparagine
Asp = aspartic acid Cys = cysteine
Gin = glutamine Glu = glutamic acid
Gly = glycine
His = histidine
lie = isoleucine
Leu = leucine
Lys = lysine
Met = methionine
Nle = norleucine
Phe = phenylalanine
Phg = phenylglycine
Pro = proline
Ser = serine
Thr = threonine
Trp = tryptophan
Tyr = tyrosine
Val = valine
The compounds of the present invention can be synthesized using standard synthetic methods known to those skilled in the art. Preferred methods include but are not limited to those methods described below.
Generally, peptides are elongated by deprotecting the a-amine of the C-terminal residue and coupling the next suitably protected amino acid through a peptide linkage using the methods described. This deprotection and coupling procedure is repeated until the desired sequence is obtained. This coupling can be performed with the constituent amino acids in a stepwise fashion, or condensation of fragments (two to several amino acids), or combination of both processes, or by solid phase peptide synthesis according to the method
originally described by Merrifield, J. Am. Chem. Soc, 85, 2149-2154 (1963), the disclosure of which is hereby incorporated by reference.
The compounds of the invention may also be
synthesized using automated peptide synthesizing equipment. In addition to the foregoing, procedures for peptide synthesis are described in Stewart and Young, "Solid Phase Peptide Synthesis", 2nd ed. Pierce Chemical Co., Rockford, IL (1984); Gross, Meienhofer, Udenfriend, Eds., "The Peptides: Analysis, Synthesis, Biology, Vol. 1, 2, 3, 5, and 9, Academic Press, New York, (1980- 1987); Bodanszky, "Peptide Chemistry: A Practical
Textbook", Springer-Verlag, New York (1988); and
Bodanszky et al. "The Practice of Peptide Sythesis" Springer-Verlag, New York (1984), the disclosures of which are hereby incorporated by reference.
The coupling between two amino acid derivatives, an amino acid and a peptide, two peptide fragments, or the cyclization of a peptide can be carried out using standard coupling procedures such as the azide method, mixed carbonic acid anhydride (isobutyl chloroformate) method, carbodiimide (dicyclohexylcarbodiimide,
diisopropylcarbodiimide, or water-soluble carbodiimides) method, active ester (p-nitrophenyl ester, N- hydroxysuccinic imido ester) method, Woodward reagent K method, carbonyldiimidazole method, phosphorus reagents such as BOP-Cl, or oxidation-reduction method. Some of these methods (especially the carbodiimide) can be enhanced by the addition of 1-hydroxybenzotriazole.
These coupling reactions may be performed in either solution (liquid phase) or solid phase.
The functional groups of the constituent amino acids must be protected during the coupling reactions to avoid undesired bonds being formed. The protecting groups that can be used are listed in Greene, "Protective Groups in Organic Synthesis" John Wiley & Sons, New York (1981) and "The Peptides: Analysis, Sythesis, Biology, Vol. 3, Academic Press, New York (1981), the disclosure of which is hereby incorporated by reference.
The a-carboxyl group of the C-terminal residue is usually protected by an ester that can be cleaved to give the carboxylic acid. These protecting groups include: 1) alkyl esters such as methyl and t-butyl, 2) aryl esters such as benzyl and substituted benzyl, or 3) esters which can be cleaved by mild base treatment or mild reductive means such as trichloroethyl and phenacyl esters. In the solid phase case, the C-terminal amino acid is attached to an insoluble carrier (usually polystyrene). These insoluble carriers contain a group which will react with the carboxyl group to form a bond which is stable to the elongation conditions but readily cleaved later. Examples of which are: oxime resin
(DeGrado and Kaiser (1980) J. Org. Chem. 45, 1295-1300) chloro or bromomethyl resin, hydroxymethyl resin, and aminomethyl resin. Many of these resins are
commercially available with the desired C-terminal amino acid already incorporated.
The a-amino group of each amino acid must be protected. Any protecting group known in the art can be used. Examples of these are: 1) acyl types such as formyl, trifluoroacetyl, phthalyl, and p- toluenesulfonyl; 2) aromatic carbamate types such as benzyloxycarbonyl (Cbz) and substituted
benzyloxycarbonyls, 1-(p-biphenyl)-1- methylethoxycarbonyl, and 9-fluorenylmethyloxycarbonyl (Fmoc); 3) aliphatic carbamate types such as tert- butyloxycarbonyl (Boc), ethoxycarbonyl,
diisopropylmethoxycarbonyl, and allyloxycarbonyl; 4) cyclic alkyl carbamate types such as
cyclopentyloxycarbonyl and adamantyloxycarbonyl; 5) alkyl types such as triphenylmethyl and benzyl; 6) trialkylsilane such as trimethylsilane; and 7) thiol containing types such as phenylthiocarbonyl and
dithiasuccinoyl. The preferred a-amino protecting group is either Boc or Fmoc. Many amino acid derivatives suitably protected for peptide synthesis are
commercially available.
The a-amino protecting group is cleaved prior to the coupling of the next amino acid. When the Boc group is used, the methods of choice are trifluoroacetic acid, neat or in dichloromethane, or HCl in dioxane. The resulting ammonium salt is then neutralized either prior to the coupling or in situ with basic solutions such as aqueous buffers, or tertiary amines in dichloromethane or dimethylformamide. When the Fmoc group is used, the reagents of choice are piperidine or substituted
piperidines in dimethylformamide, but any secondary amine or aqueous basic solutions can be used. The deprotection is carried out at a temperature between 0 °C and room temperature.
Any of the amino acids bearing side chain
functionalities must be protected during the preparation of the peptide using any of the above-identified groups. Those skilled in the art will appreciate that the selection and use of appropriate protecting groups for these side chain functionalities will depend upon the amino acid and presence of other protecting groups in the peptide. The selection of such a protecting group is important in that it must not be removed during the deprotection and coupling of the a-amino group.
For example, when Boc is chosen for the a-amine protection the following protecting groups are acceptable: p-toluenesulfonyl (tosyl) moieties and nitro for arginine; benzyloxycarbonyl, substituted
benzyloxycarbonyls, tosyl or trifluoroacetyl for lysine; benzyl or alkyl esters such as cyclopentyl for glutamic and aspartic acids; benzyl ethers for serine and
threonine; benzyl ethers, substituted benzyl ethers or 2-bromobenzyloxycarbonyl for tyrosine; p-methylbenzyl, p-methoxybenzyl, acetamidomethyl, benzyl, or t- butylsulfonyl for cysteine; and the indole of tryptophan can either be left unprotected or protected with a formyl group.
When Fmoc is chosen for the a-amine protection usually tert-butyl based protecting groups are
acceptable. For instance, Boc can be used for lysine, tert-butyl ether for serine, threonine and tyrosine, and tert-butyl ester for glutamic and aspartic acids.
Once the elongation and cyclization of the peptide is completed all of the protecting groups are removed. For the liquid phase synthesis the protecting groups are removed in whatever manner as dictated by the choice of ' protecting groups. These procedures are well known to those skilled in the art.
When a solid phase synthesis is used, the peptide should be removed from the resin without simultaneously removing protecting groups from functional groups that might interfere with the cyclization process. Thus, if the peptide is to be cyclized in solution, the cleavage conditions need to be chosen such that a free a- carboxylate and a free a-amino group are generated without simultaneously removing other protecting groups. Alternatively, the peptide may be removed from the resin by hydrazinolysis, and then coupled by the azide method. Another very convenient method involves the synthesis of peptides on an oxime resin, followed by intramolecular nucleophilic displacement from the resin, which
generates a cyclic peptide (Osapay, Profit, and Taylor (1990) Tetrahedron Letters 43, 6121-6124) . When the oxime resin is employed, the Boc protection scheme is generally chosen. Then, the preferred method for removing side chain protecting groups generally involves treatment with anhydrous HF containing additives such as dimethyl sulfide, anisole, thioanisole, or p-cresol at 0 °C. The cleavage of the peptide can also be
accomplished by other acid reagents such as
trifluoromethanesulfonic acid/trifluoroacetic acid mixtures.
Unusual amino acids used in this invention can be synthesized by standard methods familiar to those skilled in the art ("The Peptides: Analysis, Sythesis, Biology, Vol. 5, pp. 342-449, Academic Press, New York (1981)). N-Alkyl amino acids can be prepared using procedures described in previously (Cheung et al.,
(1977) Can . J. Chem. 55, 906; Freidinger et al., (1982) J. Org. Chem. 48, 77 (1982)), which are incorporated here by reference.
The compounds of the present invention may be prepared using the procedures further detailed below.
Representative materials and methods that may be used in preparing the compounds of the invention are described further below.
Manual solid phase peptide synthesis was performed in 25 mL polypropylene filtration tubes purchased from BioRad Inc., or in 60 mL hour-glass reaction vessels purchased from Peptides International. Oxime resin (substitution level = 0.96 mmol/g) was prepared
according to published procedures (DeGrado and Kaiser (198C- J. Org. Chem . 45, 1295), or was purchased from Novabiochem (substitution level = 0.62 mmol/g). All chemicals and solvents (reagent grade) wexe used as supplied from the vendors cited without further
purification. t-Butyloxycarbonyl (Boc) amino acids and other starting amino acids may be obtained commercially from Bachem Inc., Bachem Biosciences Inc. (Philadelphia, PA), Advanced ChemTech (Louisville, KY), Peninsula Laboratories (Belmont, CA), or Sigma (St. Louis, MO). 2-(1H-Benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU) and TBTU were purchased from Advanced ChemTech. N-methylmorpholine (NMM), m-cresol, D-2-aminobutyric acid (Abu), trimethylacetylchloride, diisopropylethylamlne (DIEA), 3-cyanobenzoic acid and [2-(tert-butyloxycarbonyloxylimino)-phenylacetonitrile] (Boc-ON) were purchased from Aldrich Chemical Company. Dimethylformamide (DMF), ethyl acetate, chloroform
(CHCI3), methanol (MeOH), pyridine and hydrochloric acid
(HCl) were obtained from Baker. Acetonitrile,
dichloromethane (DCM), acetic acid (HOAc),
trifluoroacetic acid (TFA), ethyl ether, triethylamine, acetone, and magnesium sulfate were purchased from EM Science. Palladium on carbon catalyst (10% Pd) was purchased from Fluka Chemical Company. Absolute ethanol was obtained from Quantum Chemical Corporation. Thin layer chromatography (TLC) was performed on Silica Gel 60 F254 TLC plates (layer thickness 0.2 mm) which were purchased from EM Separations. TLC visualization was accomplished using UV light, iodine, ninhydrin spray and/or Sakaguchi spray. Melting points were determined using a Thomas Hoover or Electrothermal 9200 melting point apparatus and are uncorrected. HPLC analyses were performed on either a Hewlett Packard 1090, Waters Delta Prep 3000, Rainin, or DuPont 8800 system. NMR spectra were recorded on a 300 MHz General Electric QE-300, Varian 300, or Varian 400 spectrometer. Fast atom bombardment mass spectrometry (FAB-MS) was performed on a VG Zab-E double-focusing mass spectrometer using a Xenon FAB gun as the ion source or a Finnigan MAT 8230.
Boc-D-2-aminobutyric acid (Boc-D-Abu) was prepared by a modification of procedures previously reported in the literature (Itoh, Hagiwara, and Kamiya (1975) Tett. Lett . , 4393), as shown in the scheme below.
Figure imgf000138_0001
D-2-aminobutyric acid
D-2-aminobutyric acid (1.0 g, 9.70 mmol) was dissolved in 20 ml H2O and a solution of Boc-ON (2.62 g, 10.6 mmol) in 20 ml acetone was added. A white
precipitate formed which dissolved upon addition of triethylamine (3.37 ml, 24.2 mmol) to give a pale yellow solution (pH = 9, wet pH paper). The solution was stirred at room temperature overnight at which time the acetone was removed under reduced pressure. The
remaining aqueous layer was extracted with ether three times, acidified to pH 2 with concentrated HCl, and then extracted with ethyl acetate three times. The combined organic layers were dried over anhydrous magnesium sulfate and evaporated under reduced pressure to give t- butyloxycarbonyl-D-2-aminobutyric acid as an oil (2.05 g,greater than quantitative yield, contains solvent), which was used without further purification. 1H NMR (CDCI3) 0.98 (t, 3H), 1.45 (s, 9H), 1.73 (m, 1H), 1.90 (m, 1H), 4.29 (m, 1H), 5.05 (m, 1H). Synthesis of R31 Cyclizing Moistϊes
This section teaches the synthesis of certain cyclizing moieties that serve as intermediates to the R31 groups in Q. Later sections teach the synthesis of other cyclizing moieties.
Synthesis of Boc-aminnmethylbenzoic Acid, Boc- aminophenylacetic Acid and Boc-aminomethylphenylacetic
Acid Derivatives
Boc-aminomethylbenzoic acid derivatives useful as cyclizing moieties in the synthesis of the, compounds of the invention are prepared using standard procedures, for example, as described in Tett . Lett . , 4393 (1975); Modern Synthetic Reactions, H.O. House (1972); or
Harting et al. J. Am. Chem . Soc , 50: 3370 (1928), and as shown schematically below.
Figure imgf000139_0001
3-Aminomethylbenzoic acid•HCl 3-Cyanobenzoic acid (10.0 g, 68 mmol) was dissolved in 200 ml ethanol by heating in a 35-50°C water bath.
Concentrated HCl (6.12 ml, 73 mmol) was added and the solution was transferred to a 500 ml nitrogen-flushed round bottom flask containing palladium on carbon catalyst (1.05 g, 10% Pd/C). The suspension was stirred under an atmosphere of hydrogen for 38 hours, filtered through a scintered glass funnel, and washed thoroughly with H20. The ethanol was removed under reduced pressure and the remaining aqueous layer, which
contained a white solid, was diluted to 250 ml with additional H2O. Ethyl ether (250 ml) was added and the suspension was transferred to a separatory funnel. Upon vigorous shaking, all solids dissolved and the aqueous layer was then washed two times with ether, evaporated under reduced pressure to a volume of 150 ml, and lyophilized to give the title compound (3- aminomethylbenzoic acid-HCl) (8.10 g, 64%) as a beige solid. 1H NMR (D2O) 4.27 (s, 2H), 7.60 (t, 1H), 7.72 (d,1H), 8.06 (d, 2H). t-Butyloxycarbonyl-3-aminomethylbenzoic Acid (Boc-Mamb)
The title compound was prepared according to a modification of standard procedures previously reported in the literature (Itoh, Hagiwara, and Kamiya (1975) Tett. Lett . , 4393). 3-Aminomethylbenzoic acid
(hydrochloride salt) (3.0 g, 16.0 mmol) was dissolved in 60 ml H2O. To this was added a solution of Boc-ON (4.33 g, 17.6 mmol) in 60 ml acetone followed by
triethylamine (5.56 ml, 39.9 mmol). The solution turned yellow and the pH was adjusted to 9 (wet pH paper) by adding an additional 1.0 ml (7.2 mmol) triethylamine. The solution was stirred overnight at room temperature at which time the acetone was removed under reduced pressure and the remaining aqueous layer was washed three times with ether. The aqueous layer was then acidified to pH 2 with 2N HCl and then
extracted three times with ethyl acetate. The combined organic layers were washed three times with H2O, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. The material was recrystallized from ethyl acetate/ hexane, to give two crops of the title compound (2.58 g, 64%) as an off- white solid, mp 123-125°C ; 1H NMR (CDCI3) 1.47 (s, 9 H), 4.38 (br s, 2 H), 4.95 (br s, 1H), 7.45 (t, 1H), 7.55 (d, 1H), 8.02 (d, 2H).
Synthesis of t-Butyloxycarbonyl-3-aminophenylacetic Acid t-Butyloxycarbonyl-3-aminophenylacetic acids useful as intermediates in the synthesis of the compounds of the invention are prepared using standard procedures, for example, as described in Collman and Groh (1982) J. Am. Chem. Soc , 104: 1391, and as shown schematically below.
Figure imgf000141_0001
t-Butyloxycarbonyl-3-aminophenylacetic Acid
A solution of 3-aminophenylacetic acid (Aldrich, 10 g, 66 mmol), di-tert-butyl dicarbonate (15.8 g, 72 mmol), and DIEA (8.6 g, 66 mmol) in 50 ml of
dichloromethane was stirred overnight at room
temperature. The reaction mixture was concentrated, partitioned between dichloromethane-H2O, the water layer was separated, acidified to pH 3 with IN HCl, and extracted with dichloromethane. The extracts were washed with H2O, brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. This material was purified by recrystallization from heptane to provide the title compound (3.7 g, 22%) as a white solid, mp 105°C; 1H NMR (CDCI3) 7.35 (s, 1H), 7.25 (m, 3H), 6.95 (m, 1H), 6.60 (br s, 1H), 3.65 (s, 2H), 1.50 (s, 9H).
Synthesis of 2-Aminomethylbenzoic Acid•HCl and 2- Aminomethylphenylacetic Acid•HCl
2-Aminomethylbenzoic acid•HCl and 2- aminomethylphenylacetic acid•HCl useful as intermediates in the synthesis of the compounds of the invention are prepared using standard procedures, for example, as described in Naito et al J. Antibiotics, 30: 698 (1977); or Young and Sweet J. Am. Chem. Soc , 80: 800 (1958), and as shown schematically below.
Figure imgf000142_0001
2-Aminomethylphenylacetic Acid d-Lantam The title compound was prepared by modification of procedures previously reported in the literature (Naito et al. (1977) J. Antibiotics, 30: 698). To an ice-cooled suspension of 2-indanone (10.8 g, 82 mmol) and
azidotrimethylsilane (9.4 g, 82 mmol) in 115 ml of chloroform was added 25 ml of concentrated sulfuric acid at a rate to maintain the temperature bet-ween 30-40°C. After an additional 3 hours, the reaction mixture was poured onto ice, and the water layer was made basic with concentrated ammonium hydroxide. The chloroform layer was separated, washed with H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was purified by sublimination (145°C, <1 mm), followed by
recrystallization from benzene to give the title compound (5.4 g, 45%) as pale yellow crystals, mp 149- 150°C; 1H NMR (CDCI3) 7.20 (m, 5H), 4.50 (s, 2H), 3.60 (s, 2H). 2-Aminomethylphenylacetic Acid•HCl
The title compound was prepared by modification of procedures previously reported in the literature (Naito et al. (1977) J. Antibiotics, 30: 698). A mixture of 2- aminomethylphenylacetic acid d-lactam (6.4 g, 44 mmol) and 21 ml of 6N HCl was heated to reflux for 4 hours. The reaction mixture was treated with activated carbon (Norit A), filtered, evaporated to dryness, and the residual oil triturated with acetone. Filtration
provided the title compound (5.5 g, 62%) as colorless crystals, mp 168°C (dec); 1H NMR (D6-DMSO) 12.65 (br s, 1H) , 8.35 (br s, 3H), 7.50 (m, 1H), 7.35 (m, 3H), 4.05 (ABq, 2H), 3.80 (s, 2H).
2-Aminomethylbenzoic Acid g-Lactam The title compound was prepared by modification of procedures previously reported in the literature
(Danishefsky et al. (1975) J. Org. Chem. , 40: 796). A mixture of methyl o-toluate (45 g, 33 mol), N- bromosuccinimide (57 g, 32 mol), and dibenzoyl peroxide (0.64 g) in 175 ml of carbon tetrachloride was heated to reflux,, for 4 hours. The cooled reaction mixture was filtered, evaporated to dryness under reduced pressure, dissolved in 250 ml of methanol, and concentrated ammonium hydroxide (75 ml, 1.11 mol) was added. The reaction mixture was heated to reflux for 5 hours, concentrated, filtered, and the solid washed with H2O followed by ether. This material was purified by
recrystallization from H2O to give the title compound (11.0 g, 26%) as a white solid, mp 150°C; 1H NMR (CDCI3) 7.90 (d, 1H), 7.60 (t, 1H), 7.50 (t, 2H), 7.00 (br s, 1H), 4.50 (s, 2H).
2-Aminomethylbenzoic Acid•HCl
The title compound was prepared using the general procedure described above for 2-aminomethylphenylacetic acid•HCl. The lactam (3.5 g, 26 mmol) was converted to the title compound (2.4 g, 50%) as colorless crystals, mp 233°C (dec); 1H NMR (D6-DMSO) 13.40 (br s, 1H), 8.35 (br s, 3H), 8.05 (d, 1H), 7.60 (m, 3H), 4.35 (br s, 2H).
Synthesis of Cyclic Compound Intermediates This section teaches the synthesis of certain cyclic compound intermediates. These are the
intermediate compounds that serve as the precursor to the Q group in the claimed compounds, (QLn)dCh ; (Q)d'Ln-Ch. These compounds may be directly labeled with
radioisotopes, or may be modified by attaching linker group (s) and chelator (s). t-Butyloxycarbonyl-3-aminomethylbenzoic acid (Boc- Mamb) is coupled to oxime resin by a modification of the method described by DeGrado and Kaiser (1980) J. Org. Chem . 45, 1295 using 1 equivalent of the 3- aminom^thylbenzoic acid (with respect to the
substitution level of the resin), 1 equivalent of HBTU, and 3 equivalent of NMM. Alternatively, Boc-Mamb (1 equivalent) may be coupled to the oxime resin using 1 equivalent each of DCC and DMAP in methylene chloride. Coupling times range from 15 to 96 hours. The
substitution level is then determined using either the picric acid test (Sarin, Kent, Tam, and Merrifield, (1981) Anal. Biochem. 117, 145-157) or the quantitative ninhydrin assay (Gisin (1972) Anal . Chim . Acta 58, 248- 249). Unreacted oxime groups are blocked using 0.5 M trimethylacetylchloride / 0.5 M diisopropylethylamlne in DMF for 2 hours. Deprotection of the Boc protecting group is accomplished using 25% TFA in DCM for 30 minutes. The remaining amino acids or amino acid derivatives are coupled using between a two and ten fold excess (based on the loading of the first amino acid or amino acid derivative) of the appropriate amino acid or amino acid derivatives and HBTU in approximately 8 ml of DMF. The resin is then neutralized in situ using 3 eq. of NMM (based on the amount of amino acid used) and the coupling times range from 1 hour to several days. The completeness of coupling is monitored by qualitative ninhydrin assay, or picric acid assay in cases where the amino acid was coupled to a secondary amine. Amino acids are recoupled if necessary based on these results.
After the linear peptide had been assembled, the N- terminal Boc group is removed by treatment with 25% TFA in DCM for 30 minutes. The resin is then neutralized by treatment with 10% DIEA in DCM. Cyclization with concomitant cleavage of the peptide is accomplished using the method of Osapay and Taylor ((1990) J. Am.
Chem. Soc , 112, 6046) by suspending the resin in approximately 10 ml/g of DMF, adding one equivalent of HOAc (based on the loading of the first amino acid), and stirring at 50-60°C for 60 to 72 hours. Following filtration through a scintered glass funnel, the DMF filtrate is evaporated, redissolved in HOAc or 1:1 acetonitrile: H2O, and lyophilized to obtain protected, cyclized material. Alternatively, the material may be dissolved in methanol and precipitated with ether to obtain the protected, cyclized material. This is then treated using standard procedures with anhydrous hydrogen fluoride (Stewart and Young (1984) "Solid Phase Peptide Synthesis", 2nd. edition. Pierce Chemical Co., 85) containing 1 ml/g m-cresol or anisole as scavenger at 0°C for 20 to 60 minutes to remove side chain protecting groups. The crude product may be purified by reversed-phase HPLC using a 2.5 cm preparative Vydac C18 column with a linear acetonitrile gradient containing 0.1% TFA to produce pure cyclized material. The following N-a-Boc-protected amino acids may be used for the syntheses: Boc-Arg (Tos), Boc-N-a-MeArg (Tos), Boc- Gly, Boc-Asp (OcHex), Boc-3-aminomethyl-4-iodo-benzoic acid, Boc-D-Ile, Boc-NMeAsp (OcHex), Boc-NMe-Mamb, Boc-D- Phg, Boc-D-Asp(OBzl), Boc-L-Asp (OcHex), Boc-aMe- Asp (OcHex), Boc-bMe-Asp (OcHex), Boc-L-Ala, Boc-L-Pro, Boc-D-Nle, Boc-D-Leu, Boc-D-Val, Boc-D-2-aminobutyric acid (Boc-D-Abu), Boc-Phe, Boc-D-Ser (Bzl), Boc-D-Ala, Boc-3-aminomethylbenzoic acid (Boc-Mamb), Boc-D-Lys (2- ClZ), Boc-b-Ala, Boc-D-Pro, Boc-D-Phe, Boc-D- Tyr(Cl2Bzl), Boc-NMe-Amf (CBZ), Boc-aminotetralin- carboxylic acid, Boc-aminomethylnaphthoic acid, Boc-4- aminomethylbenzoic acid, or Boc-NMeGly.
Preferable N-a-Boc-protected amino acids useful in these syntheses are Boc-Arg (Tos), Boc-N-a-MeArg (Tos), Boc-Gly, Boc-Asp (OcHex), Boc-D-Leu, Boc-D-Val, Boc-D-2- aminobutyric acid (Boc-D-Abu), Boc-Phe, Boc-D-Ser (Bzl), Boc-D-Ala, Boc-3-aminomethylbenzoic acid (Boc-Mamb), Boc-D-Lys (2-ClZ), Boc-Ala,Boc-D-Pro, or Boc-NMeGly.
The synthesis of the compounds of the invention is further exemplified below. The Tables below set forth representative compounds of the present invention.
Cyclic Compound Intermediate 1 cyclo-(Gly-NMeArg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Gly, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described below for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.336 mmol scale to give the protected cyclic peptide (218 mg, 84%). The peptide (200 mg) and 200 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (158 mg, greater than quantitative yield; calculated as the acetate salt). Purification was accomplished by
reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 11% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (21% recovery, overall yield 16.3%).
Mass spectrum: M+H = 533.26. Cyclic Compound Intermediate 2
cyclo-(D-Ala-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Ala, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described below for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). Recoupling of the Boc-N-MeArg(Tos) residue was found to be necessary. The peptide was prepared on a 0.244 mmol scale to give the protected cyclic peptide (117 mg, 61%). The peptide (110 mg) and 110 mL of m- cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.25%/ min. gradient of 2 to 11% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid.
Mass spectrum: M+H = 547.23.
Cyclic Compound Intermediate 3 cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Abu, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described below for Cyclic Compound
Intermediate 4. The peptide was prepared on a 0.101 mmol scale to give the protected cyclic peptide (51 mg, 63%) . The peptide (43 mg) and 50 μL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (23 mg, 68.7%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative
Vydac C18 column (2.5 cm) using a 0.23%/min. gradient of 7 to 14% acetonitrile containing 0.1% trifluoroacetic acid and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (31% recovery; overall yield 12.4%).
Mass spectrum: M+H = 561.46.
Cyclic Compound Intermediate 3a cyclo-(Abu-NMeArg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Abu, K = NMeArg,
L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo- (D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. TBTU was used as the coupling reagent. The peptide was prepared on a 0.596 mmol scale to give the protected cyclic peptide (182 mg,38.4%). The peptide (176 mg) and 0.176 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (116 mg; 90.4%; calculated as the
fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.45%/ min. gradient of 9 to 27% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (1.92% recovery, overall yield 0.574%); FAB- MS: [M+H] = 561.39.
Cyclic Compound Intermediate 4 cyclo-(D-Val-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Val, K = NMeArg, L = Gly, M =
Asp, R1 = R2 = H
To a 25 ml polypropylene tube fitted with a frit was added Boc-Mamb (0.126 g, 0.5 mmol) and 6 ml of DMF. To this was added HBTU (0.194 g, 0.5 mmol), oxime resin (0.52 g, substitution level = 0.96 mmol/g), and N- methylmorpholine (0.165 ml, 1.50 mmol). The suspension was mixed at room temperature for 24 hours. The resin was then washed thoroughly (10-12 ml volumes) with DMF (3x), MeOH (1x), DCM (3x), MeOH (2x) and DCM (3x). The substitution level was determined to be 0.389 mmol/g by quantitative ninhydrin assay. Unreacted oxime groups were blocked by treatment with 0.5 M
trimethylacetylchloride/ 0.5M DIEA in DMF for 2 hours.
The following steps were then performed: (Step 1) The resin was washed with DMF (3x), MeOH (lx), DCM (3x), MeOH (2x), and DCM (3x). (Step 2) The t-Boc group was deprotected using 25% TFA in DCM for 30 minutes. (Step 3) The resin was washed with DCM (3x), MeOH (1x), DCM (2x), MeOH (3x) and DMF (3x) (Step 4) Boc-Asp (OcHex) (0.613 g, 1.94 mmol), HBTU (0.753 g, 1.99 mmol), 8 ml of DMF, and N-methylmorpholine (0.642 ml, 5.84 mmol) were added to the resin and the reaction allowed to proceed for 2.5 hours. (Step 5) The coupling reaction was found to be complete as assessed by the qualitative ninhydrin assay. Steps 1-5 were repeated until the desired sequence had been attained. The coupling of Boc-D-Val to NMeArg was monitored by the picric acid test.
After the linear peptide was assembled, the N- terminal t-Boc group was removed by treatment with 25% TFA in DCM (30 min.) The resin was washed thoroughly with DCM (3x), MeOH (2x) and DCM (3x), and then neutralized with 10% DIEA in DCM (2 x 1 min.) The resin was washed thoroughly with DCM (3x) and MeOH (3x) and then dried. Half of the resin (0.101 mmol) was cyclized by treating with 6 ml of DMF containing HOAc (5.8 mL,
0.101 mmol) and heating at 50°C for 72 hours. The resin was then filtered through a scintered glass funnel and washed thoroughly with DMF. The DMF filtrate was evaporated to an oil, redissolved in 1:1 acetonitrile: H2O, and lyophilized to give the protected cyclic peptide (49 mg, 60%). The peptide (42 mg) was treated with anhydrous hydrogen fluoride at 0°C, m the presence of 50 mL of m-cresol as scavenger, for 30 minutes to remove side chain protecting groups . The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (23 mg, 70%; calculated as the acetate salt). Purification was accomplished using reversed-phase HPLC with a preparative Vydac C18 column (2.5 cm) and a 0.23%/ minute gradient of 7 to 18% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound as a fluffy white solid (24% recovery; overall yield 9.4%); FAB-MS: [M+H] = 575.45.
Solution Phase Synthesis of Cyclic Compound Intermediate The following abbreviations are used below for TLC solvent systems: chloroform/methanol 95 : 5^ = CM;
chloroform/acetic acid 95:5 = CA;
chloroform/methanol/acetic acid 95:5 = CMA
BocNMeArg (Tos) -Gly-OBzl — 25 mmol BocNMeArg (Tos) (11.07 g, Bachem), 30 mmol Gly-OBzl tosylate (10.10 g, Bachem), 25 mmol HBTU (O-Benzotriazole-N,N,N',N',- tetramethyl-uronium-hexafluorophosphate; 9.48 g;
Advanced Chemtech), and 75 mmol DIEA
(diisopropylethylamlne; Aldrich) were dissolved in 25 ml CH2CI2. The reaction was allowed to proceed 1 hr, the solvent was evaporated under reduced pressure at 50° to a syrup, wich was dissolved in 400 ml ethyl acetate. This solution was extracted with (150 ml each) 2 x 5% citric acid, 1 x water, 2 x sat. NaHCO3, 1 x sat. NaCl. The organic layer was dried over MgSO4, and the solvent evaporated under reduced pressure. The resulting oil was triturated with petroleum ether and dried under high vacuum for a minimum of 1 hr. yield 14.7 g (99.5%); TLC Rf(CM) = 0.18 Rf(CA) = 0.10; NMR is consistent with structure; FABMS M+H+ = 590.43 (expected 590.26).
NMeArg (Tos) -Gly-OBzl — 14.5 g (BocNMeArg (Tos)-Gly-OBzl (24.5 mmol) was dissolved in 30 ml TFA, allowed to react for 5 min., and the solvent evaporated at 1 mm mecury pressure at r.t. The resulting syrup was dissolved in 400 ml ice cold ethyl acetate, and extracted with 100 ml ice cold sat. NaHCO3, the aqueous phase was extracted twice with 200 ml ethyl acetate, and the combined organic phases were extracted once with 25 ml sat. NaCl. The solvent was evaporated under reduced pressure giving a viscous oil that was triturated with 300 ml ether. The resulting solid was filtered and washed with ether, giving a hydroscopic compound that was dried in a vacuum desicςator: yield 10.33 g (86.2%); TLC Rf(CM) = 0.03; Rf(CMA) = 0.20; NMR is consistent with structure; FABMS M+H+ = 490.21 (expected 490.20).
Boc-D-Val-NMeArg (Tos) -Gly-OBzl — 9.80 mmol
NMeArg (Tos)-Gly-OBzl (4.80 g), 9.82 mmol Boc-D-Val (2.13 g, Bachem), and 10.0 mmol HBTU (3.79 g) were dissolved in 10 ml methylene chloride. The flask was placed on an ice bath, and 20 mmol DIEA (3.48 ml) was added. The reaction was allowed to proceed at 0° for 15 min and 2 days at r.t. The reaction mixture was diluted with 400 ml ethyl acetate, extracted (200 ml each) 2 x 5% citric acid, 1 x sat. NaCl, dried over MgSO4 and evaporated under reduced pressure. The resulting oil was
triturated with 50, then 30 ml ether for 30 min with efficient mixing: yield 4.58 g (69%); TLC Rf(CM) = 0.27 (also contains a spot near the origin, which is an aromatic impurity that is removed during trituration of the product in the next step); NMR is consistent with structure; FABMS M+H+ = 689.59 (expected 689.43).
Boc-D-Val-NMeArg (Tos) -Gly — 4.50 g Boc-D-Val- NMeArg (Tos)-Gly-OBzl (4.44 mmol) dissolved in 80 ml methanol was purged with N2 for 10 min. 1.30 g Pd/C catalyst (10% Fluka lot #273890) was then added, and then H2 was passed directly over the surface of the reaction. TLC showed the reaction to be complete within approximately 0.5 hr. After 1 hr. the catalyst was removed by filtering through a bed of Celite, and the solvent removed at 40° under reduced pressure. The resulting solid was triturated well with 50 ml refluxing ether, filtered, and washed with petroleum ether: yield 3.05 g (78%); TLC Rf(CM) = 0.03; Rf(CMA) = 0.37; NMR is consistent with structure; FABMS M+H+ = 599.45
(expected 599.29).
4-Nitrobenzophenone Oxime (Ox) — 50 g 4- nitrobenzophenone (220 mmol, Aldrich) and 30.6 g hydroxylamine hydrochloride (Aldrich, 440 mmol) were heated at reflux in 0.5 L methanol/pyridine (9:1) for 1 hr. The reaction mixture was evaporated under reduced pressure, dissolved in 500 ml ether, and extracted with 200 ml each of 5% citric acid (2 times) and sat. NaCl (1 time), dried over MgSO4, evaporated under reduced pressure and triturated with ether giving 44.35 g (83%) of the oxime as a mixture of the cis and trans isomers: TLC Rf(CM) = 0.50; Rf(CMA) = 0.82; NMR is consistent with structure; FABMS M+H+ = 242.07 (expected 242.07).
BocMamb-Ox — 22 mmol BocMamb (5.522 g), 20 mmol nitrobenzophenone oxime (4.84 g), and 20 mmol DMAP (4- dimethylaminopyridine; Aldrich) were dissolved in 40 ml CH2Cl2 . The flask was placed on an ice bath, and 21 mmol DCC (Dicyclohexylcarbodiimide; 4.33 g) was added. The reaction was allowed to proceed on ice for 30 min and at r.t. over night. The dicyclohexylurea formed was filtered, and washed with 40 ml methylene chloride. The filtrate was evaporated under reduced pressure at r.t. to a syrup, and dissolved in 400 ml ethyl acetate. This solution was extracted with (150 ml each) 2 x 5% citric acid, 1 x water, 2 x sat. NaHCO3, 1 x sat. NaCl. The organic layer was dried over MgSO4, and the solvent evaporated under reduced pressure. The resulting oil was triturated with petroleum ether and dried under high vacuum for a minimum of 1 hr. : yield 7.51 g (79%); TLC Rf(CM) = 0.41; Rf(CMA) = 0.66; NMR is consistent with structure; FABMS M+H+ = 476.30 (expected 476.18). TFA•MAMB-Ox — BocMamb-Ox , 7.4 g (15.5 mmol) was dissolved in 30 ml methylene chloride plus 10 ml TFA (25% TFA). The reaction was allowed to proceed at r.t. for 1 hr, and the solvent evaporated under reduced pressure at r.t. for 10 min, then at 40° for 15 min. The resulting syrup was triturated with ether (200 ml) at -5°, giving. The resulting crystals were filtered after 1 hr and washed well with ether: yield 7.22 g (95%); Rf(CMA) - 0.25; NMR is consistent with structure; FABMS M+H+ = 376.22 (expected 376.12).
Boc-Asp (OcHex) -Mamb-Ox — 20 mmol Boc-Asp (OcHex) (6.308 g, Bachem) and 44 mmol DIEA (7.66 ml) were dissolved in 20 ml DMF. 20 mmol HBTU (7.58 g. Advanced Chemtech) was added, and the reaction allowed to proceed for 2 minutes with vigorous stirring. TFA-Mamb-Ox (7.13 g, 15 mmol) was added, and the reaction allowed to proceed o.n. at r.t. The solvent was removed under reduced pressure giving an oil, which was dissolved in 500 ml ethyl acetate, and this solution was extracted with (150 ml each) 2 x 5% citric acid, 1 x water, 2 x sat. NaHCO3, 1 x sat. NaCl. The organic layer was dried over MgSO4, and the solvent evaporated under reduced pressure. The resulting oil was triturated with petroleum ether and dried under high vacuum: yield 9.76 g (97%); TLC Rf(CM) = 0.55; NMR is consistent with structure; FABMS M+H+ = 673.45 (expected 673.23). TFA Asp (OcHex) -MAMB-Ox — 15 mmol Boc-Asp (OcHex) -MAMB- Ox was dissolved in 50 ml 35% TFA in CH2Cl2, and allowed to react 90 min. The solvent was evaporated under reduced pressure at r.t. for 10 min, then at 40° for 15 min. To remove traces of TFA, 25 ml DMF was added and the solvent evaporated at 50°. The resulting syrup was triturated with ether (200 ml), then dried under high vacuum: yield 9.61 g (93%); Rf(CMA) = 0.45; NMR is consistent with structure; FABMS M+H+ = 573.56 (expected 573.23).
Boc-D-Val-NMeArg (Tos) -Gly-Asp (OcHex) -MAMB-Ox 10.0 mmol each TFA Asp (OcHex)-MAMB-Ox, Boc-D-Val-NMeArg (Tos)-Gly, and HBTU, plus 30 mmol DIEA were dissolved in 20 ml DMF. After 4 hr., the solvent was removed under reduced pressure, and the residue taken up in 600 ml ethyl acetate, which was extracted with 300 ml each of 5% citric acid, water and sat. NaCl. The organic layer was dried over MgSO4, evaporated under reduced pressure, triturated with ether and dried in vacuo: yield 9.90 g (86%); Rf(CM) = 0.10; NMR is consistent with structure; FABMS M+H+ = 1153.22 (expected 1153.47).
TFA-D-Val-NMeArg (Tos) -Gly-Asp (OcHex) -MAMB-Ox This compound was prepared from Boc-D-Val-NMeArg (Tos)-Gly- Asp (OcHex)-MAMB-Ox (9.8 g, 8.5 mmol) by treatment with TFA/CH2Cl2 (1:1) for 45 min. The solvent was evaporated and the product triturated with ether: yield 9.73 g (98%); Rf(CM) = 0.10; NMR is consistent with structure; FABMS M+H+ = 1053.22 (expected 1053.4). cyclo (·D-Val-NMeArg (Tos) -Gly-Asp (OcHex) -MAMB) TFA·D- Val-NMeArg (Tos)-Gly-Asp (OcHex)-MAMB-Ox (1.80 g, 1.54 mmol), and 2 mmol each of DIEA and acetic acid were dissolved in 200 ml DMF. The mixture was heated to 50° for 2 days, then evaporated under reduced pressure. The syrup was dissolved in 400 ml ethyl acetate/n-butanol (1:1), and extracted with 200 ml each of 5% citric acid (3x) and sat. NaCl (1x). The organic layer was dried over MgSO4 and triturated twice with 200 ml ether:
yield 1.07 g (86%); Rf(CM) = 0.10; NMR is consistent with structure; FABMS M+H+ = 811.25 (expected 811.38). cyclo (·D-Val-NMeArg-Gly-Asp-MAMB) 0.50 g cyclo (D-Val- NMeArg(Tos)-Gly-Asp (OcHex)-MAMB) was treated with 5 ml HF at 0°C, in the presence of 0.5 ml of anisole for 30 min. The HF was removed under reduced pressure and the crude peptide triturated with ether, ethyl acetate and ether. The resulting solid was dissolved in 10% acetic acid and lyophilized: yield 0.321 g (82% calculated as the acetate salt). The product was purified with a recovery of approximately 40% using the same method as described for the material synthesized by the solid phase procedure.
Crystallization Cyclic Compound Intermediate 4 Preparation of Salt Forms of the Compound of Cyclic
Compound Intermediate 4
It has been discovered that the compounds of the present invention may be isolated by crystallization of the compound from organic and aqueous solvents.
The zwitterion of Cyclic Compound Intermediate 4 was converted to the mesyl (methanesulfonate) salt of Cyclic Compound Intermediate 4 (Cyclic Compound
Intermediate 4 (methane-sulfonate)) by refluxing the zwitterion with stirring in isopropanol at 25 mg/ml and slowly adding a .solution of 1.0 molar equivalent
methanesulfonic acid (correcting for the water content of the zwitterion) dissolved in isopropanol. The heat was turned off and the solution cooled to 5°C in an ice bath. After stirring 1 hour, the solution was filtered and the solid rinsed three times with cold isopropanol and dried under vacuum to constant weight.
The following salts of the compound of Cyclic Compound Intermediate 4 were prepared using the same procedure, by adding 1.0 equivalent of the appropriate acid:
Cyclic Compound Intermediate 4 (biphenylsulfonate): zwitterion + 1.0 equivalent biphenylsulfonic acid.
Cyclic Compound Intermediate 4 (a- naphthalenesulfonate):
zwitterion + 1.0 equiv. a-naphthalenesulfonic acid.
Cyclic Compound Intermediate 4 (b- naphthalenesulfonate):
zwitterion + 1.0 equiv. b-naphthalenesulfonic acid. Cyclic Compound Intermediate 4 (benzenesulfonate): zwitterion + 1.0 equiv. benezene-sulfonic acid.
Cyclic Compound Intermediate 4 (p-toluenesulfonate): zwitterion + 1.0 equiv. p-toluene-sulfonic acid.
The following salts of the compound of Cyclic Compound Intermediate 4 were prepared by crystallization of the compound from aqueous systems. Cyclic Compound Intermediate 4 (sulfate):
10 mg amorphous Cyclic Compound Intermediate 4 (made by lyophilizing the zwitterion from a solution of 2 molar equivalents of acetic acid in water) dissolved per ml 1 N H2SO4, pH adjusted to 2.5. On standing at room temperature, a precipitate formed. This was filtered through a sintered glass funnel and dried under vacuum to constant weight. Cyclic Compound Intermediate 4 (methanesulfonate (mesyl)):
100 mg amorphous DMP728 dissolved per ml water + 1.2 molar equiv. methanesulfonic acid (this was obtained as a 4M aqueous solution). On standing at room
temperature, a large flat crystal was formed.
Cyclic Compound Intermediate 4 (benzenesulfonate): 100 mg zwitterion dissolved per ml water + 1.2 equiv. benzenesulfonic acid added. On standing at room temeprature, a precipitate formed. This was filtered through a sintered glass funnel, rinsed with a small volume of isopropanol, and dried under vacuum to constant weight. Cyclic Compound Intermediate 4 (p- toluenesulfonate):
100 mg zwitterion dissolved per ml water + 1.2 molar equiv. toluenesulfonic acid added. On standing at room temperature, a precipitate formed. This was filtered through a sintered glass funnel and dried under vacuum to constant weight.
Cyclic Compound Intermediate 4b cyclo-(D-Val-D-NMeArg-Gly-Asp-Mamb); J = D-Val, K = D- NMeArg, L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.596 mmol scale to give the protected cyclic peptide (186 mg, 38.6%). The peptide (183 mg) and 0.183 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and
lyophilized to generate the title compound (145 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (14.8% recovery, overall yield 5.3%); FABMS: [M+H] = 575.31.
Cyclic Compound Intermediate 5 cyclo-(D-Leu-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Leu, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.115 mmol scale to give the protected cyclic peptide (92.4 mg, 98%). The peptide (92.4 mg) and 93 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (45.7 mg, 63%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7 to 21% acetonitrile containing 0.1%
TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (29% recovery, overall yield 16.5%) ;FAB-MS: [M+H] = = 589.48.
Cyclic Compound Intermediate 7
cyclo-(D-Nle-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Nle, K = NMeArg,
L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val- NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was
prepared on a 0.586 mmol scale to give the
protected cyclic peptide (305 mg, 63.3%). The peptide (295 mg) and 0.295 mL of anisole were
treated with anhydrous hydrogen fluoride at 0 C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (207 mg, 95.4%; calculated as the fluoride salt).
Purification was accomplished by reversed-phase
HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 5.4 to 18%
acetonitrile containing 0.1% TFA and then
lyophilized to give the TFA salt of the title
compound as a fluffy white solid (44% recovery, overall yield
22.9%); FAB-MS: [M+H] = 589.26.
Cyclic Compound Intermediate 11 cyclo-(D-Phg-NMeArg-Gly-Asp-Mamb); the compound of
.formula (II) wherein J = D-Phg, K = NMeArg,
L - Gly, M = Asp, R1 = H, R2 = H The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the protected cyclic peptide (296 mg, 57.4%). The peptide (286 mg) and 0.286 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and
lyophilized to generate the title compound (210 mg,
98.9%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 5.4 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (24.2% recovery, overall yield 11.9%); FAB-MS: [M+H] = 609.27.
Cyclic Compound Intermediate 12 cyclo-(D-Phe-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Phe, K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the protected cyclic peptide (140 mg, 26.7%). The peptide (135 mg) and 0.135 mL of anisole were treated with anhydrous hydrogen fluoride at 0 C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and
lyophilized to generate the title compound (108 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column
(2.5 cm) using a 0.23%/ min. gradient of 7.2 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (35% recovery, overall yield 8.7%); FAB-MS :
[M+H] = 623.28. Solid Phase Synthesis of Cyclic Compound Intermediate
13f
cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Lys, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.586 mmol scale to give the protected cyclic peptide (349 mg, 58.9%). The peptide (334 mg) and 334 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound as a pale yellow solid (168 mg, 79.1%; calculated as the difluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 5.4 to 14.4% acetonitrile containing 0.1% TFA and then Lyophilized to give the TFA salt of the title compound as a fluffy white solid (33.6% recovery, overall yield 12.1%); FAB-MS: [M+H] = 604.32
Solution Phase Synthesis of Cyclic Compound Intermediate
13f
A Scheme depicting the synthesis described below appears immediately after the description.
Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb); the compound of formula (yy) wherein Part A - Boc-Asp (OBzl)
To a solution of Boc-Asp (OBzl) (45.80 g, 140 mmol) and HOSu (N-hydroxysuccinimide; 16.10 g, 140 mmol) in 300 ml p-dioxane at 5-10°C was added DCC (30.20 g, 140 mmol). The solution was stirred for 30 minutes at 5- 10°C then the solids were filtered and washed with dioxane (3 X 50 ml). The combined organics were
concentrated under reduced pressure to give a clear oil which crystallized to a colorless solid (42.98 g, 73%) when triturated with ethyl ether (3 x 100 ml). NMR is consistent with structure; MP = 98-99°C; DCI-MS : [M+NH4]
= 438.
Part B - Boc-Asp(OBzl)-Mamb
3-Aminomethylbenzoic acid-HCl (Mamb; 13.08 g, 70.0 mmol) was dissolved in 120 ml DMF and DIEA (24.32 ml,
140 mmol) was added, changing the pH from 4 to 7.5. The white suspension was stirred for 30 min at room
temperature before a solution of Boc-Asp (OBzl)-OSu
(29.40 g, 70.0 mmol) in DMF (50 ml) was added. The mixture was allowed to stir 24 hr, during which time it turned to a gold solution. The solution was added to 5% citric acid (2000 ml) and cooled to 5°C for 3 hr. The solids were then collected by filtration, washed with ice cold water (200 ml) and ice cold ethyl ether (100 ml), and dried under reduced pressure to give the title compound as a colorless solid (29.62 g, 92%); MP = 149- 151°C; DCI-MS: [M+NH4] = 474. Part C - HCl•B-Asp (OBzl)-Mamb
Boc-Asp (OBzl)-Mamb (7.92 g, 17.4 mmol) was
dissolved in 4N HCl in dioxane ( 50 ml), stirred for 2 hr, and the solution concentrated under reduced pressure to give the title compound as a colorless solid (6.80 g, 99%). DCI-MS: [M+NH4] = 374.
Part D - Boc-D-Lys(Tfa)-NMeArg(Tos)-Gly-OBzl
NMeArg (Tos)-Gly-OBzl (14.40 g, 29.4 mmol), Boc-D- Lys(Tfa) (10.00 g, 29.4 mmol), and HBTU (11.37 g, 62.0 mmol) were dissolved in methylene chloride (40 ml).
After cooling to 0°C, DIEA (10.44 g, 62.0 mmol) was added and the reaction was allowed to proceed 20 minutes at 0°C and 2 days at room temperature. The reaction mixture was diluted with ethyl acetate (800 ml),
extracted with 200 ml portions of 0.2 N HCl (1X), sat.
NaHCO3 (1X), and saturated NaCl (2X), dried (MgSO4), and evaporated under reduced pressure to a yellow solid. Purification by flash chromatography (silica gel; 5:1 EtOAc: acetonitrile) gave the title compound as a
colorless solid (20.34 g, 85%). MP 78-85°C; DCI-MS:
[M+NH4] = 831.
Part E - Boc-D-Lys(Tfa)-NMeArg(Tos)-Gly A solution of Boc-D-Lys (Tfa)-NMeArg (Tos)-Gly-OBzl (11.00 g, 13.5 mmol) in methanol (200 ml), was placed in a Parr shaker bottle, purged with N2 for 10 minutes, and treated with 10% palladium on carbon catalyst (10% Pd/C, 3.6 g). The shaker bottle was further purged with 7 pressurization-evacuation cycles, repressurized, and allowed to shake 90 minutes, during which time the calculated amount of hydrogen was consumed. The catalyst was removed by filtration through a bed of Celite and the filtrate was concentrated under reduced pressure yielding a solid. Trituration with refluxing ethyl ether (75 ml) gave pure product (9.18 g, 94%) as a colorless solid. DCI-MS: [M+H] = 724. Part F - Boc-D-Lys(Tfa)-NMeArg(Tos)-Gly-OSu
Boc-D-Lys (Tfa)-NMeArg (Tos)-Gly (8.00 g, 11.0 mmol), HOSu (1.25 g, 10.8 mmol) and DCC (2.22 g, 10.8 mmol) were dissolved in DMF (75 ml) and stirred at room temperature for 2 days. The solids were removed by filtration and washed with DMF (2 x 15 ml). The
filtrate was concentrated under reduced pressure and the resulting syrup dried under reduced pressure at 40°C to give a tan solid (6.50 g, 72%). MP = 66-69°C; FAB-MS : [M+H] = 821.
Part G - Boc-D-Lys(Tfa)-N-MeArg(Tos)-Gly-Asp(OBzl)-Mamb
A suspension of Boc-D-Lys (Tfa)-N-MeArg (Tos)-Gly-OSu (8.85 g, 10.8 mmol) and HCl•Asp (OBzl)-Mamb (4,24 g, 10.8 mmol) in 4:1 dioxane:DMF (100 ml) was treated with DIEA (1.39 g, 10.8 mmol) over 10 minutes. The resulting mixture was stirred 2 days at room temperature and concentrated under reduced pressure to a syrup. This syrup was dissolved in ethyl acetate (300 ml) and washed with 75 ml portions of 0.2N HCl (3X), sat. NaHCO3 (2X), H2O (1X), and saturated NaCl (1X). The organic layer was dried (MgSO4) and concentrated under reduced pressure at 40°C to a sticky amber solid (9.13 g, 78%) . MP = 90-93°C; FAB-MS: [M+H] = 1062.
Part H - HCl•D-Lys (Tfa)-N-MeArg (Tos)-Gly-Asp(OBzl)-Mamb
Boc-D-Lys (Tfa)-N-MeArg (Tos)-Gly-Asp (OBzl)-Mamb (8.30 g, 7.8 mmol) was partially dissolved in 4N HCl in dioxane (50 ml), stirred at room temperature for 30 min, and concentrated under reduced pressure to give a yellow solid. Trituration with warm EtOAc (60 ml) afforded the product (7.65 g, 98%) as a yellow solid. FAB-MS: [M+H] = 962. Part T - Cyclo-(D-Lys(Tfa)-N-MeArg (Tos)-Gly-Asp(OBzl)- Mamb)
HCl-D-Lys (Tfa)-N-MeArg (Tos)-Gly-Asp (OBzl)-Mamb (3.00 g, 3.0 mmol), DIEA (0.77 g, 6.0 mmol), and TBTU (0.98 g, 3.0 mmol) were dissolved in DMF (100 ml). The reaction was stirred at room temperature for 22 hours, and the pH was maintained at 7-8 by the addition of DIEA as necessary. The reaction was concentrated under reduced pressure and the resulting oil dissolved in 3.75:1 ethyl acetate : 1-butanol (110 ml). The organic solution was washed with 50 ml portions of 0.2 N HCl (2X), saturated NaHCO3 (1X), H2O (1X), and saturated NaCl (1X), dried (MgSO4), concentrated to a brown oil. Triturated with ethyl ether (100 ml) gave a brown solid which was purified by flash chromatography (silica gel; 5:1 EtOAc:EtOH) to give the title compound (1.62 g, 57%) as a colorless solid. MP = 128-130°C; FAB-MS : [M+H] = 944.
Part J - Cyclo-(D-Lys(Tfa)-N-MeArg-Gly-Asp-Mamb) Cyclo-(D-Lys (Tfa)-N-MeArg (Tos)-Gly-Asp (OBzl)-Mamb) (0.85 g, 0.9 mmol) was dissolved in TFA (10 ml) and cooled to -10°C. Triflie acid (trifluoromethanesulfonic acid; 10 ml) was slowly added to the stirred reaction while maintaining the temperature at -5°C. Anisole (2 ml) was added and stirring was continued for 3 hours at -5°C. The temperature of the reaction was decreased to -78°C, ethyl ether (200 ml) was added, and the reaction was stirred for 1 hour. The white sticky solids were removed by filtration and washed with ice cold ether (50 ml). The solids were dissolved in 1:1 acetone:H2O (10 ml) and lyophilized to give the product (0.63 g, 100%) as a fluffy colorless solid. FAB-MS : [M+H] = 700. Part K - Cyclo-(D-Lys-N-MeArg-Gly-Asp-Mamb)
Cyclo-(D-Lys (Tfa)-N-MeArg-Gly-Asp-Mamb) (0.63 g, 0.9 mmol) was dissolved in 1.0 M aqueous piperdine (10 ml) at 0°C and the reaction was allowed to slowly warm to room temperature over 3 hours. The solution was lyophilized to give a yellow solid. Purification was accomplished by preparative HPLC with a Vydac protein- peptide C-18 column (2.1 cm) using a 0.36%/min. gradient of 9 to 18% acetonitrile containing 0.1% TFA, and then lyophilized to give the title compound (0.20 g, 90%) as a colorless fluffy solid. MP = 138-142°C; FAB-MS: [M+H] = 604.
Solution Phase Synthesis of 13f
H-Gly-OBzl
Boc-N-MeAτg(Tos)-OH
Figure imgf000169_0002
Boc-N-MeArg(Tos)-Gly-OBzl
Figure imgf000169_0006
TFA
HBTU, DIEA
Boc-D-Lys(T fa)-OH
H-N-MeArg(Tos)-Gly-OBzl
Figure imgf000169_0003
Boc-D-Lys(Tfa)-N-MeArg(Tos)-Gly-OBzl
HBTU, DIEA
H
Figure imgf000169_0005
2. Pd/C
Figure imgf000169_0004
HOSu, DCC.
Boc-D-Lys(Tfa)-N-MeArg(Tos)-Gly-OSu
DMF
Figure imgf000169_0001
Cyclic Compound Intermediate 13r
cyclo-(D-Ile-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Ile,
K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the protected cyclic peptide (349 mg, 69.2%). The peptide (342 mg) and 0.342 mL of anisole were treated with anhydrous hydrogen fluoride at 0 C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and
lyophilized to generate the title compound (227 mg, 90%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 10.8 to 19.8% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (22.5% recovery, overall yield 12.1%); FAB-MS: [M+H] = 589.34. Cyclic Compound Intermediate 17 cyclo-(D-Met-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Met, K = NMeArg, L = Gly, M =
Asp, R1 = H, R2 = H The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used .for the attachment of Boc-Mamb to the resin. The peptide was prepared on a 0.179 mmol scale to give the protected cyclic peptide (105 mg, 69.7%). The peptide (105 mg) and 0.105 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (72 mg; 92.3% yield, calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 14.4 to 23.4% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (13.2% recovery, overall yield 7.4%); FAB-MS: [M+H] = 607.3. Cyclic Compound Intermediate 18 cyclo-(NMeGly-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = NMeGly, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H The title compound was prepared using the general procedure described above for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.43 mmol scale to give the protected cyclic peptide (205 mg, 60%). The peptide (200 mg) and 200 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate (18) as a pale yellow solid (148 mg, 97%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of- 7 to 22% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of (18) as a fluffy white solid (14.7% recovery, overall yield 7.9%); FAB-MS: [M+H] = 547.34.
Cyclic Compound Intermediate 24 cyclo-(Pro-NMeArg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Pro, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.43 mmol scale to give the protected cyclic peptide (170 mg, 48.8%). The peptide (164 mg) and 164 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate (24) as a pale yellow solid (101 mg, 79% ; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7 to 22% acetonitrile
containing 0.1% TFA and then lyophilized to give the TFA salt of (24) as a fluffy white solid (5.8% recovery, overall yield 2.1%);FAB-MS: [M+H] = 573.46.
Cyclic Compound Intermediate 25 cyclo-(D-Pro-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Pro, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was
prepared on a 0.43 mmol scale to give the protected cyclic peptide (211mg, 60.8%). The peptide (200 mg) and 200 mL of /n-cresol were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate (25) as a pale yellow solid
(145 mg, 93.3%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7 to 22% acetonitrile containing 0.1%
TFA and then lyophilized to give the TFA salt of (25) as a fluffy white solid (6.4% recovery, overall yield 3.3%); FAB-MS: [M+H] = = 573.35.
Cyclic Compound Intermediate 28c cyclo-(b-Ala-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = b-Ala, K = NMeArg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.586 mmol scale to give the protected cyclic peptide (264 mg, 57.5%). The peptide (258 mg) and 258 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound as a pale yellow solid (231 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 5.4 to 14.4% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (53.2% recovery, overall yield 32.5%); FAB-MS: [M+H] =
547.28:
Cyclic Compound Intermediate 28f
cyclo-(D-Tyr-NMeArg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Tyr,
K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the
general procedure described for cyclo-(D-Val-
NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was
prepared on a 0.313 mmol scale to give the
protected cyclic peptide (342 mg, greater than
quantitative yield). The peptide (331 mg) and
0.330 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to
generate the title compound (218 mg, greater than quantitative yield; calculated as the fluoride
salt). Purification was accomplished by reversed- phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18%
acetonitrile containing 0.1% TFA and then
lyophilized to give the TFA salt of the title
compound as a fluffy white solid (11.3% recovery, overall yield 10.8%); FAB-MS: [M+H] = 639.54.
Cyclic Compound Intermediate 29
cyclo-(Gly-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Gly, K = Arg,
L = Gly, M = Asp, R1 = R2 = H The title compound was prepared using the general procedure described above for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.283 mmol scale and half was cyclized to give the protected cyclic peptide (62 mg, 58%). The peptide (60 mg) and 60 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid
(48 mg, > quantitative yield; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.30%/ min. gradient of 0 to 9% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (36%
recovery, overall yield 19.9%); FAB-MS: [M+H] = 519.26.
Cyclic Compound Intermediate 30 cyclo-(D-Ala-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = D-Ala, K = Arg, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo- (D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.189 mmol scale to give the protected cyclic peptide (211 mg, Quantitative yield). The peptide (195 mg) and 195 mL of /n-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (125 mg, 83%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 11% acetoni"trile
containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (12.5% recovery, overall yield 13.8%); FAB-MS: [M+H] = 533.26.
Cyclic Compound Intermediate 31 cyclo-(Ala-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Ala, K = Arg,
L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.324 mmol scale to give the protected cyclic peptide (191 mg, 76.4%). The peptide (100 mg) and 100 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (75 mg, 97.4%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 11% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (15.5% recovery, overall yield 10.5%); FAB-MS: [M+H] = 533.25. Cyclic Compound Intermediate 32
cyclo-(D-Val-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = D-Val, K = Arg, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.193 mmol scale to give the protected cyclic peptide (199 mg, > quantitative yield). The peptide (193 mg) and 193 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (130 mg, 86%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 13% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (57% recovery, overall yield 58.1%); FAB-MS: [M+H] = 561.22.
Cyclic Compound Intermediate 33 cyclo-(D-Leu-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = D-Leu, K = Arg, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.202 mmol scale to give the protected cyclic peptide (152 mg, 93%). The peptide (150 mg) and 150 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (78 mg, 66%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 5 to 18% acetonitrile containing 0.1% trifluoroacetic acid and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (26% recovery, overall yield 14.8%); FAB-MS: [M+H] = 575.45.
Cyclic Compound Intermediate 34 cyclo-(D-Abu-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = D-Abu, K = Arg, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.193 mmol scale to give the protected cyclic peptide (210 mg, > quantitative yield). The peptide (206 mg) and 206 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (158 mg, 99%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 11% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (57% recovery, overall yield 72.2%); FAB-MS: [M+H] = 547.21.
Cyclic Compound Intermediate 35 cyclo-(D-Ser-Arg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Ser, K = Arg, L = Gly, M = Asp, R1 = R2 = H The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.193 mmol scale to give the protected cyclic peptide (224 mg, > quantitative yield). The peptide (210 mg) and 210 ml of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (145 mg, 89%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 2 to 13% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (22% recovery, overall yield 27%); FAB-MS : [M+H] = 549.31.
Cyclic Compound Intermediate 36 cyclo-(D-Phe-Arg-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Phe, K = Arg, L = Gly, M = Asp, R1 =
R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.266 mmol scale to give the protected cyclic peptide (202 mg,
90%). The peptide (157 mg) and 157 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (125 mg, > quantitative yield; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7 to 23% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (35% recovery, overall yield 29.3%); FAB-MS: [M+H] = 609.25 Cyclic Compound Intermediate 37 cyclo-(Phe-Arg-Gly-Asp-Mamb); the compound of formula
(II) wherein J = Phe, K = Arg, L = Gly,
M = Asp, R1 = R2 = H The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The peptide was prepared on a 0.335 mmol scale to give the protected cyclic peptide (306 mg, > quantitative yield). The peptide (275 mg) and 275 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 1 hour. The crude material was precipitated with ether, redissolved in aqueous HOAc, and lyophilized to generate the title compound as a pale yellow solid (214 mg, 98%; calculated as the acetate salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 23% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (32% recovery, overall yield 31.5%); FAB-MS: [M+H] = 609.26
Cyclic Compound Intermediate 40
cyclo-(D-Val-NMeAmf-Gly-Asp-Mamb); the compound of
formula (II) wherein J = D-Val, K = NMeAmf, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the
general procedure described for cyclo-(D-Val- NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4) . The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was
prepared on a 0.586 mmol scale to give the
protected cyclic peptide (189 mg, 39.9%). The
peptide (189 mg) and 0.189 mL of anisole were
treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (212 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of
10.8 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (8.1% recovery, overall yield 4.1%); FAB-MS : [M+H] = 595.23.
Cyclic Compound Intermediate 48a
The title compound may be synthesized using procedures described in Mosher et al. Tett. Lett. 29: 3183-3186, and as shown schematically below. This same procedure is a generally useful method for converting a primary amine into a guanidine functionality.
Figure imgf000182_0001
Cyclic Compound Intermediates 42-45
The synthesis of Cyclic Compound Intermediates 42- 45 is shown schematically below .
Figure imgf000183_0001
Cyelio Compound Intermediate 46 and 47
Cyclic Compound Intermediates 46 and 47 are prepared according to standard procedures, for example, as described in Garigipati, Tett . Lett . (1990) 31: 1969- 1972 and in Canadian Patent 2008311, as is shown schematically below. The aspartic acid group may be protected (e.g., with a phenacyl protection group) to avoid side reactions.
Figure imgf000184_0001
1) H2S/Pyr/Et3N
n = 0, 1 2) Mel/acetone
3) Amm. acetate
Cyclic Compound Intermediate 54 cyclo-(D-Val-NMeArg-b-Ala-Asp-Mamb); J = D-Val, K =
NMeArg,
L = b-Ala, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.586 mmol scale to give the protected cyclic peptide (227 mg, 46.9%). The peptide (219 mg) and 219 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate (54) as a pale yellow solid (150 mg, 93.2%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7.2 to 16.2% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of (54) as a fluffy white solid (43.6% recovery, overall yield 16.5%); FAB-MS: [M+H] = 589.32.
Cyclic Compound Intermediate 55-58
The synthesis of Cyclic Compound Intermediates 55- 58 is shown schematically below .
1) 25% TFA in DCM
2) 10% DIEA in DCM
BOC-Asp-Mamb-oxime
Figure imgf000185_0002
3) Br(CH2)nCOOH n = 1,2
DCC
Figure imgf000185_0001
Figure imgf000186_0001
Cyclic Compound Intermediate 58c cyclo-(D-Val-NMeArg-L-Ala-Asp-Mamb); the compound of formula (II) wherein J = D-Val,
K = NMeArg, L = L-Ala, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val- NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the
protected cyclic peptide (375 mg, 74.6%). The peptide (360 mg) and 0.360 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (220 mg, 83%; calculated as the fluoride salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18%
acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title
compound as a fluffy white solid (19.9% recovery, overall yield 10.6%); FAB-MS: [M+H] = 589.31. Cyclic Compound Intermediate 63 and 63a cyclo-(D-Val-NMeArg-Gly-a-MeAsp-Mamb); the compounds of formula (II) wherein J is D-Val; K is NMeArg; L is Gly;
M is a-MeAsp; R1 = R2 = H The title compound was prepared vising the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.794 mmol scale to give the protected cyclic peptide (237 mg, 36.1%). The peptide (237 mg) and 0.237 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous
acetonitrile, and lyophilized to generate the title compound (165 mg, 94.3%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid; isomer #1 (8.36% recovery, overall yield 2.5%); FAB-MS: [M+H] = 589.29; isomer #2 (9.16% recovery, overall yield 2.7%); FAB-MS: [M+H] = 589.27. Cyclic Compound Intermediates 64 and 64a
cyclo-(D-Val-NMeArg-Gly-B-MeAsp-Mamb); the
compounds of formula (II) wherein J = D-Val,
K = NMeArg, L = Gly, M = B-MeAsp, R1 = H, R2 = H The title compound was prepared using the general procedure described for cyclo-(D-Val- NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the
protected cyclic peptide (201 mg, 40.0%). The peptide (200 mg) and 0.200 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (162 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid; isomer #1 (12.7% recovery, overall yield 4.8%); FAB-MS: [M+H] =
589.43; isomer #2 (13.9% recovery, overall yield 5.3%); FAB-MS: [M+H] = 589.45.
Cyclic Compound Intermediate 64b cyclo-(D-Val-NMeArg-Gly-NMeAsp-Mamb); the compound of formula (II) wherein J = D-Val, K = NMeArg, L = Gly, M = NMeAsp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val-
NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the protected cyclic peptide (232 mg, 46.1%). The peptide (225 mg) and 0.225 mL of anisole were treated with anhydrous hydrogen fluoride at 0 C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (160 mg, 96.4%; calculated as the fluoride salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18%
acetonitrile containing 0.1% TFA and then
lyophilized to give the TFA salt of the title compound as a fluffy white solid (28.2% recovery, overall yield 10.9%); FAB-MS: [M+H] = 589.42.
Cyclic Compound Intermediate 64c cyclo-(D-Val-NMeArg-Gly-D-Asp-Mamb); the compound of formula (II) wherein J = D-Val,
K = NMeArg, L = Gly, M = D-Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described above for cyclo- (D-Val- NMeArg-Gly-Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.611 mmol scale to give the protected cyclic peptide (257 mg, 51.9%). The peptide (250 mg) and 0.250 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (192 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (44.4% recovery, overall yield 20.7%); FAB-MS: [M+H] = 575.42.
Cyclic Compound Intermediate 89e cyclo-(D-Abu-di-NMeOrn-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Abu, K = di-NMeOrn, L = Gly, M = Asp, R1 = R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val- NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. The peptide was prepared on a 0.498 mmol scale to give the
protected cyclic peptide (150 mg, 39.3%). The peptide (150 mg) and 0.150 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized. to generate the title compound (93 mg, 86%; calculated as the fluoride salt).
Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.45%/ min. gradient of 3.6 to 18%
acetonitrile containing 0.1% TFA and then
lyophilized to give the TFA salt of the title compound as a fluffy white solid (49.3% recovery, overall yield 14.2%); FAB-MS: [M+H] = 533.34.
Cyclic Compound Intermediate 89f cyclo-(D-Abu-NMeArg-Gly-D-Asp-Mamb); compound of formula (II) wherein J = D-Abu, K = NMeArg, L = Gly, M =
D-Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. TBTU was used as the coupling reagent. The peptide was prepared on a 0.596 mmol scale, to give the protected cyclic peptide (273 mg, 57.6%). The peptide (263 mg) and 0.263 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (218 mg; greater than quantitative yield; calculated as the fluoride salt) . Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 10.8 to 19.8% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (40.4% recovery, overall yield 21.9%); FAB-MS: [M+H] = 561.37.
Cyclic Compound Intermediate 89g cyclo-(D-Abu-D-NMeArg-Gly-Asp-Mamb); the compound of formula (II) J = D-Abu, K = D-NMeArg, L = Gly, M = Asp,
R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-Mamb to the oxime resin. TBTU was used as the coupling reagent. The peptide was prepared on a 0.596 mmol scale to give the protected cyclic peptide (241 mg, 50.8%). The peptide (235 mg) and 0.235 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (168 mg; 98.3%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 12.6 to 21.6% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (2.3% recovery, overall yield 0.99%); FABMS: [M+H] = 561.-36.
Cyclic Compound Intermediate 89h Cyclo-(D-Ala-p-guanidinyl-Phe-Gly-Asp-Mamb);
the compound of formula (II) wherein J = D-Ala, K = p- guanidinyl-Phe, L = Gly, M = Asp R1 = H, R2 = H
Figure imgf000192_0001
Dissolved 25 mg (38.3 mmoles) of cyclo-(D-Ala-p- amino-Phe-Gly-Asp-Mamb) (TFA salt), 14.3 mg (114.9 umoles) formamidine sulfonic acid, and 18.7 mg (153.2 umoles) of 4-dimethyl-aminopyridine in 5 ml of ethanol in a 10 ml round bottom flask. Refluxed the mixture for 3 hours, then added an additional 14.3 mg of formamidine sulfonic acid and 18.7 mg of 4-dimethyl-aminopyridine. After refluxing for an additional 3 hours, the reaction was found to be -75% complete by reversed-phase HPLC. The ethanol was evaporated under reduced pressure, and the residue was purified on a preparative Vydac C18 column (2.5 cm) using a 0.45%/min. gradient of 0 to 18% acetonitrile containing 0.1% TFA.
Lyophilization afforded the TFA salt of the title compound as a white solid (28% recovery), overall yield 26.4%); FAB-MS: [M+H] = 581.30.
Cyclic Compound Intermediate 89i cyclo-(D-Abu-(DiNMe,guanidinyl-Orn)-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Abu, K = diNMe, guanidinyl-Orn , L = Gly, D = Asp, R1 = H, R2 = H
Figure imgf000193_0001
Dissolved 10.53 mg (16.3 mmoles) of cyclo-(D-Abu- diNMeOrn-Gly-Asp-Mamb) (TFA salt), 6.08 mg (48.99 umoles) formamidine sulfonic acid, and 8.00 mg (65.57 umoles) of 4-dimethyl-aminopyridine in 2.5 ml of ethanol in a 10 ml round bottom flask. Refluxed the mixture for 2 hours and then stirred at room temperature overnight. Refluxed for one hour, added an additional 6.08 mg of formamidine sulfonic acid and 8.00 mg of 4- dimethylaminopyridine and then refluxed for an
additional 2 hours. Evaporated the ethanol under reduced pressure and purified the residue on a
preparative Vydac C18 column (2.5 cm) using a 0.45%/min. gradient of 3.6 to 18% acetonitrile containing 0.1% TFA. Lyophilization afforded the TFA salt of the title compound as a white solid (57.2% recovery), overall yield 53.5%); FAB-MS: [M+H] = 575.34. Cyclic Compound Intermediates 89j cyclo-(D-Abu-Di-NMeLys-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Abu, K = Di-NMeLys, L = Gly,
M = Asp, R1 = H, R2 = H cyclo-(D-Abu-NMeLys-Gly-Asp-Mamb); the compound of formula (II) wherein J = D-Abu, K = NMeLys, L = Gly, M =
Asp, R1 = H, R2 = H Di-N-methyl amino acid derivatives may be prepared using methods which have been described previously
(Olsen, J. Org. Chem. (1970) 35: 1912) or,
alternatively, through the use of NaH/CH3l. The mono-
NMe-Lysine amino acid was obtained as a side product during the synthesis of the corresponding di-NMe-lysine derivative. The title compounds were prepared using conventional solution phase peptide chemistry techniques described previously. Cyclo-(D-Abu-diNMeLys-Gly-Asp- Mamb) was obtained in 0.31% overall yield, FAB-MS: [M+H] = 547.3. Cyclo-(D-Abu-NMeLys-Gly-Asp-Mamb) was obtained in 0.25% overall yield, FAB-MS: [M+H] = 533.3. Cyclic Compound Intermediate 90
cyclo-(D-Val-NMeArg-Gly-Asp-2-aminomethylphenylacetic acid)
The title compound was prepared by a modification of the general solution-phase chemistry route. This approach employed an amino acid succinimide ester coupling to the aromatic cyclizing moiety, and the dinitrobenzophenone oxime as shown schematically below in the Scheme below (n = 1).
Figure imgf000195_0001
Boc-Asp (OcHex)-2-aminomethylphenylacetic Acid
To a suspension of 2-aminomethylphenylacetic acid•HCl (4.0 g, 20 mmol) in H2O (20 ml) was added NaHCO3 (5.0 g, 60 mmol), followed by a solution of Boc- Asp (OcHex)-OSu (7.5 g, 18 mmol) in THF (20 ml). The reaction mixture was stirred at room temperature for 3 hours, filtered, diluted with H2O, acidified with IN HCl, and extracted with ethyl acetate. The extracts were washed with H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated with ether to provide the title compound (7.0 g, 83%) as a white powder. 1H NMR (D6-DMSO) 12.40 (br s, 1H), 8.30 (br t, 1H), 7.20 (m, 5H), 4.65 (m, 1H), 4.35 (q, 1H), 4.25 (m, 2H), 3.65 (s, 2H), 2.70 (dd, 1H), 2.55 (dd, 1H), 1.70 (m, 4H), 1.40 (s, 9H), 1.35 (m, 6H). 4 ,4'-Dinitrobenzophenone Oxime
The title compound was prepared by modification of procedures previously reported in the literature
(Chapman and Fidler (1936) J. Chem . Soc, 448; Kulin and Leffek (1973) Can . J. Chem . , 51: 687). A solution of chromic anhydride (20 g, 200 mmol) in 125 ml of H2O was added dropwise over 4 hours, to a suspension of bis (4- nitrophenyl) methane (25 g, 97 mmol) in 300 ml of acetic acid heated to reflux. The reaction mixture was heated at reflux for 1 hour, cooled to room temperature, and poured into water. The solid was collected by
filtration, washed with H2O, 5% sodium bicarbonate, H2O, and air-dryed to provide a 1:1 mixture of bis (4- nitrophenyl) methane/4, 4'-dinitrobenzophenone via 1H NMR. This material was oxidized with a second portion of chromic anhydride (20 g, 200 mmol), followed by an identical work-up procedure to provide the crude
product. Trituration with 200 ml of benzene heated to reflux for 16 hours provided 4,4'-dinitrobenzophenone (20.8 g, 79%) as a yellow powder. A solution of hydroxylamine hydrochloride (10.2 g, 147 mmol) was added to a suspension of 4,4'- dinitrobenzophenone (19 g, 70 mmol) in 100 ml of ethanol. The reaction mixture was heated to reflux for 2 hours, cooled to room temperature, and the solid collected by filtration. Recrystallization from ethanol provided the title compound (14.0 g, 70%) as pale yellow crystals, mp 194°C; 1H NMR (D6-DMSO) 12.25 (s, 1H), 8.35 (d, 2H), 8.20 (d, 2H), 7.60 (d, 4H).
4,4'-Dinitrobenzophenone Oxime Boc-Asp (OcHex) -2- aminomethylphenylacetate
To an ice-cooled solution of Boc-Asp (OcHex)-2- ammomethylphenylacetic acid (3.5 g, 7.6 mmol) and 4,4'- dinitrobenzophenone oxime (2.2 g, 7.5 mmol) in 50 ml of ethyl acetate and 5 ml of DMF was added DCC (1.6 g, 7.8 mmol). The reaction mixture was stirred at room
temperature for 8 hours, filtered, diluted with ethyl acetate, washed with saturated sodium bicarbonate solution, H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was purified by column
chromatography on silica gel (EM Science, 230-400 mesh) using 10:1 dichloromethane/ethyl acetate to give the title compound (4.3 g, 78%) as pale yellow crystals. 1H NMR (D6-DMSO) 8.30 (dd, 5H), 7.80 (d, 2H), 7.65 (d, 2H), 7.15 (m, 5H), 4.65 (m, 1H), 4.35 (q, 1H), 4.15 (m, 2H), 3.90 (s, 2H), 2.70 (dd, 1H), 2.50 (dd, 1H), 1.70 (m, 4H), 1.40 (s, 9H), 1.35 (m, 6H).
4,4'-Dinitrobenzophenone Oxime Boc-D-Val-NMeArg (Tos)- Gly-Asp (OcHex)-2-aminomethylphenylacetate To a solution of 4,4'-dinitrobenzophenone oxime Boc-Asp (OcHex)-2-aminomethylphenylacetate (1.5 g, 2 mmol) in 4 ml of dichloromethane was added 2 ml of trifluoroacetic acid. The reaction mixture was stirred at room temperature for 1 hour, diluted with
dichloromethane, and evaporated to dryness under reduced pressure. The oily residue was concentrated under high vacuum to remove traces of excess trifluoroacetic acid. To a solution of the crude TFA salt and Boc-D-Val- NMeArg (Tos)-Gly (1.2 g, 2 mmol) in 5 ml of DMF was added TBTU (640 mg, 2 mmol) and DIEA (780 mg, 6 mmol). The reaction mixture was stirred at room temperature for 16 hours, concentrated under high vacuum, diluted with ethyl acetate, washed with 5% citric acid, H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated with ether to provide the title compound (2.3 g, 95%) as a yellow powder. This material was used without further purification. cyclo-(D-Val-NMeArg(Tos)-Gly-Asp(OcHex)-2- aminomethylphenylacetic acid)
To a solution of 4,4'-dinitrobenzophenone oxime Boc-D-Val-NMeArg (Tos)-Gly-Asp (OcHex)-2- aminomethylphenylacetate (1.2 g, 1 mmol) in 4 ml of dichloromethane was added 2 ml of trifluoroacetic acid. The reaction mixture was stirred at room temperature for
3 hours, diluted with dichloromethane, and evaporated to dryness under reduced pressure. The oily residue was concentrated under high vacuum to remove traces of excess trifluoroacetic acid. To a solution of the crude TFA salt in 100 ml of DMF was added acetic acid (0.50 ml, 8.7 mmol) and DIEA (1.52 ml, 8.7 mmol). The reaction mixture was stirred at 60°C for 3 days, concentrated under high vacuum, diluted with ethyl acetate, and the solution allowed to
crystallize overnight. Filtration provided the title compound (563 mg, 68%) as a yellow powder. 1H NMR (D6- DMSO) 8.70 (d, 1H), 8.40 (br s, 1H), 8.30 (br s, 1H), 8.05 (t, 1H), 7.65 (d, 2H), 7.25 (d, 2H), 7.20 (m, 4H), 7.10 (br d, 1H), 6.80 (br s, 1H), 6.60 (br s, 1H), 5.10 (dd, 1H), 4.65 (m, 1H), 4.55 (m, 1H), 4.40 (m, 2H), 3.85 (m, 2H), 3.65 (d, 1H), 3.45 (m, 2H), 3.05 (m, 2H), 2.80 (s, 3H), 2.80 (m, 1H), 2.60 (dd, 1H), 2.30 (s, 3H), 1.70 (m, 6H), 1.30 (m, 9H), 0.95 (d, 3H), 0.80 (d, 3H);
DCI(NH3)-MS: [M+H] = 825. cvclo-(D-Val-NMeArg-Gly-Asp-2-aminomethylphenylacetic acid)
A mixture of 352 mg (0.43 mmol) of cyclo-(D-Val- NMeArg (Tos)-Gly-Asp (OcHex)-2-aminomethylphenylacetic acid) and 352 μl of anisole was treated at 0°C with 5 ml of HF for 20 minutes. The excess HF was removed under reduced pressure, the residue triturated with ether, dissolved in 50% acetonitrile/H2O, and lyophilized to provide the crude cyclic peptide•HF salt as an off-white powder. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile
containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (225 mg, 75%) as a fluffy white solid; 1H NMR (D6-DMSO) 8.70 (d, 1H), 8.35 (d, 1H), 8.20 (t, 1H), 8.00 (t, 1H), 7.45 (t, 1H), 7.20 (m, 3H), 7.10 (m, 1H), 7.00 (br s, 4H), 5.10 (dd, 1H), 4.50 (dt, 1H), 4.40 (m, 2H), 3.85 (dt, 2H), 3.65 (d, 1H), 3.50 (dd, 1H), 3.45 (d, 1H), 3.10 (m, 2H), 2.90 (s, 3H), 2.75 (dd, 1H), 2.55 (dd, 1H), 2.00 (m, 1H), 1.85 (m, 1H), 1.65 (m, 1H), 1.30 (m, 2H), 0.95 (d, 3H), 0.85 (d, 3H); FAB-MS: [M+H] = 589.
Cyclic Compound Intermediate 91 cyclo-(D-Val-NMeArg-Gly-Asp-2-aminomethylbenzoic acid)
The title compound was prepared by, the general solution-phc.se procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-2-aminomethylphenylacetic acid), and as shown schematically above in the Cyclic Compound Intermediate 90 Scheme (n = 0). The cyclic peptide (192 mg, 0.24 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (20 mg, 12%) as a fluffy white solid; 1H NMR
(D6-DMSO) 8.75 (d, 1H), 8.50 (d, 1H), 7.65 (t, 1H), 7.60 (t, 1H), 7.50 (m, 2H), 7.40 (m, 3H), 7.00 (br s, 4H), 5.05 (dd, 1H), 4.50 (t, 1H), 4.30 (m, 2H), 4.10 (dd, 1H), 3.70 (m, 2H), 3.15 (q, 2H), 3.05 (s, 3H), 2.80 (dd, 1H), 2.55 (dd, 1H), 2.10 (m, 1H), 1.95 (m, 1H), 1.60 (m, 1H), 1.40 (m, 2H), 1.05 (d, 3H), 0.95 (d, 3H); FAB-MS: [M+H] = 575.
Cyclic Compound Intermediate 92 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminophenylacetic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), and as shown schematically in the Scheme below. The cyclic peptide (360 mg, 0.44 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative LiChrospher RP-18 column (5 cm) using a 2.3% / minute gradient of 22 to
90% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (150 mg, 50%) as a fluffy white solid; 1H NMR (D6-DMSO) 12.40 (br s, 1H), 8.95 (s, 1H), 8.55 (m, 2H), 8.45 (t, 1H), 7.90 (d, 1H), 7.50 (m, 1H), 7.20 (t, 1H), 7.00 (br s, 4H), 6.90 (m,
2H), 5.15 (dd, 1H), 4.65 (q, 1H), 4.55 (t, 1H), 3.65 (m, 2H), 3.60 (dd, 1H), 3.10 (m, 2H), 2.85 (s, 3H), 2.85 (d, 1H), 2.70 (dd, 2H), 2.00 (m, 2H), 1.75 (m, 1H), 1.35 (m, 2H), 0.90 (d, 3H), 0.85 (d, 3H); FAB-MS : [M+H] = 575.
Cyclic Compound Intermediate 87, 88 cyclo-(D-Val-NMeArg-Gly-Asp-4-aminomethylbenzoic acid); the compound of formula (III) wherein J = D-Val, K =
NMeArg, L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb). The DCC/DMAP method was used for attachment of Boc-4-aminomethylbenzoic acid to the oxime resin. The peptide was prepared on a 0.43 mmol scale to give the protected cyclic peptide (212mg, 60.8%). The peptide (200 mg) and 200 mL of m-cresol were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether,
redissolved in aqueous HOAc, and lyophilized to generate the crude peptide as a pale yellow solid (152 mg, 97% ; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 7 to 22% acetonitrile containing 0.1% TFA. Two peaks were isolated to give isomer #1 (87) (17.1% recovery, overall yield 9.3%) and isomer #2 (88) (13.4% recovery, overall yield 7.3%); FAB-MS: [M+H] = 575.41 (isomer #1; 87); 575.44 (isomer #2; 88).
R1 or R2 Substituted Intermediates Cyclic compound intermediates which incorporate substituents at R1 or R2 are synthesized from the corresponding substituted cyclizing moieties. The following Schemes, discussions, and examples teach the preparation of this class of cyclizing moiety and the corresponding cyclic compound intermediates. t-Butyloxycarbonyl-N-methyl-3-aminomethylbenzoic Acid
(Boc-NMeMamb) The title compound can be prepared according to standard procedures, for examples, as disclosed in Olsen, J. Org. Chem . (1970) 35: 1912), and as shown schematically below.
Figure imgf000202_0001
Synthesis of Aminomethyibenzoic Acid Analogs Cyclizing moieties of the formula below may be prepared using standard synthetic procedures, for example, as shown in the indicated reaction schemes shown below.
Figure imgf000203_0001
For R = CH3, CH2CH3, CH2CH2CH3, CH2CH2CH2CH3, CH(CH3)2, C(CH3)3, CH (CH3) CH2CH3, benzyl, cyclopentyl, cyclohexyl; see Scheme 1.
For R = CH3, CH2CH2CH2CH3, phenyl; see Scheme 2. For R = CH3, phenyl; see Scheme 3 and 4.
Figure imgf000204_0001
Figure imgf000205_0001
O 3-[1'-(t-butyloxycarbonyl)amino]ethylbenzoic acid
(BOC-MMMAMB)
The title compound for the purpose of this
invention was prepared according to the Scheme 4
(above).
3-Acetylbenzoic acid (0.50 g, 3 mmol),
hydroxylamine hydrochloride (0.70 g, 10 mmol) and pyridine (0.70 ml, 9 mmol) were refluxed in 10 ml ethanol, for 2 h. Reaction mixture was concentrated, residue triturated with water, filtered and dried. Oxime was isolated as a white solid (0.51 g ; 94.4% yield). 1HNMR (CD3OD) 7.45-8.30(m, 4H), 2.30(s, 3H) . MS (CH4-CI)
[M+H-O] = 164.
A solution of the oxime (0.51 g, 3 mmol) in ethanol, containing 10% Pd on carbon (1.5 g) and cone. HCl (0.25 ml, 3 mmol) was hydrogenated at 30 psi H2 pressure in a Parr hydrogenator for 5 h. Catalyst was filtered and the filtrate concentrated. Residue was triturated with ether. Amine hydrochloride was isolated as a white solid (0.48 g ; 85.7% yield). IHNMR (CD3OD) 7.6-8.15(m, 4H), 4.55(q, 1H), 1.70(s, 3H). MS [M+H] = 166.
Amine hydrochloride (0.40 g, 2 mmol) was dissolved in 15 ml water. A solution of BOC-ON (0.52 g, 2.1 mmol) in 15 ml acetone was added, followed by the addition of triethylamine (0.8 ml, 6 mmol). Reaction was allowed to proceed for 20 h. Reaction mixture was concentrated, partitioned between ethyl acetate and water. Aqueous layer was acidified to pH 2 using 10% HCl solution.
Product was extracted in ethyl acetate, which after the usual work up and recrystallization from ethyl
acetate/hexane, gave the title compound as a white solid (0.30 g ; 57% yield), m.p. 116-118° C.
1HNMR (CDCI3) 7.35-8.2(m, 4H), 4.6(bs, 1.5H), 1.50(d, 3H), 1.40(s, 9H). MS (NH3-CI) [M+NH4] = 283.
3-[l'-(t-butyloxycarbonyl)amino]benzylbenzoic acid
(BOC-phMAMB) The title compound for the purpose of this
invention was prepared according to the Scheme 4
(above), by the procedure similar to that for the methyl derivative. A solution of 3-benzoylbenzoic acid (2.00 g, 9 mmol), hydroxylamine hydrochloride (2.00 g, 29 mmol) and pyridine (2.00 ml, 25 mmol) in ethanol was refluxed for 12 h. After the usual extractive work up, white solid was obtained (2.41 g). The product still contained traces of pyridine, but was used in the next step without further purification.
The crude product (2.00 g, ~8 mmol) was dissolved in 200 ml ethanol. 10% Pd-C (2.00 g) and con. HCl (1.3 ml, 16 mmol) were added. Reaction mixture was
hydrogenated at 30 psi for 1 h. The catalyst was filtered and the reaction mixture concentrated. Upon trituration of the residue with ether and drying under vacuum, amine hydrochloride was obtained as a white solid (2.12 g ; 97% yield). 1HNMR (CD3OD) 7.4-8.15 (m, 10H), 5.75 (s, 1H). MS (CH4-CI) [M+H-OH] = 211.
Amine hydrochloride (1.00 g, 4 mmol) was converted to its BOC-derivative by a procedure similar to the methyl case. 0.60 g (48% yield) of the recrystallized (from ethanol/hexane) title compound was obtained as a white solid, m.p. 190-192° C. 1HNMR (CD3OD) 7.2-8.0(m, 10H), 5.90 (2s, 1H, 2 isomers), 1.40(s, 9H). MS (NH3-CI) [M+NH4-C4H8] = 289
Cyclic Compound Intermediates 68 and 68a cyclo-(D-Val-NMeArg-Gly-Asp-MeMamb); the compound of formula (II) wherein J = D-Val,
K = NMeArg, L = Gly, M = Asp, R1 = CH3, R2 = H
MeMAMB cyclizing moiety was prepared via Scheme 4 (described earlier). The title compound was made by following the solution phase synthetic route to attach MeMAMB to the tripeptide. Cyclization gave the protected cyclic peptide. Deprotection was achieved by treatment of the peptide (390 mg) and anisol (0.390 ml) with anhydrous HF at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in 10% aqueous acetic acid, and lyophilized to give a mixture of the two isomers (330 mg; greater than quantitative yield; calculated as the acetate salt). Purification and the separation of the isomers was accomplished by Reverse- Phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.48%/min gradient of 7 to 23% acetonitrile containing 0.1% TFA. Fractions collected at Rf 24.1 min and 26.8 min were lyophilized to give the TFA salts of the isomers 1 and 2 respectively. FAB-MS (Isomer 1): [M+H] = 589.31; FAB-MS (isomer 2): [M+H] = 589.31. Cyclic Compound Intermediates 76 and 76a cyclo-(D-Val-NMeArg-Gly-Asp-PhMamb); the compound of formula (II) wherein J = D-Val,
K = NMeArg, L = Gly, M = Asp, R1 = Ph, R2 = H PhMAMB cyclizing moiety was prepared via Scheme 4 (described earlier). The title compound was made by following the solution phase synthetic route to attach PhMAMB to the tripeptide. Cyclization gave the protected cyclic peptide. Deprotection was achieved by treatment of the peptide (470 mg) and anisol (0.470 ml) with anhydrous HF at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in 10% aqueous acetic acid, and lyophilized to give a mixture of the two isomers (310 mg; 82.4% overall recovery).
Purification and the separation of the isomers was accomplished by Reverse-Phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.55%/min gradient of 18 to 36% acetonitrile containing 0.1% TFA. Fractions collected at Rf 22 min and 24.6 min were lyophilized to give the TFA salts of the isomers 1 and 2 respectively. FAB-MS (Isomer 1): [M+H] = 651.33; FAB-MS (isomer 2): [M+H] = 651.33. Cyclic Compound Intermediate 79
cyclo-(D-Val-NMeArg-Gly-Asp-NMeMamb); the compound
of formula (II) wherein J = D-Val,
K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = CH3 The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-NMeMamb to the oxime resin. The peptide was prepared on a 0.456 mmol scale to give the protected cyclic peptide (406 mg, greater than quantitative yield). The peptide (364 mg) and 0.364 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (251 mg, 93.5%; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 9 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (34.2% recovery, overall yield 29.9%); FABMS: [M+H] = 589.33.
Ring-Substituted R31-Cyclizing Moieties Cyclizing moieties possessing an aromatic ring that bears a substituent group may be prepared using the methods taught in the following examples and Schemes. Synthesis of 4, 5, and 6-Substituted 3-
Aminomethylbenzoic Acid•HCl, and 4, 5, and 6-Substituted t-Butyloxycarbonyl-3-aminomethylbenzoic Acid Derivatives
4, 5, and 6-Substituted 3-aminomethylbenzoic acid'HCl, and 4, 5, and 6-substituted t- butyloxycarbonyl-3-aminomethylbenzoic acid derivatives useful as intermediates in the synthesis of the
compounds of the invention are prepared using standard procedures, for example, as described in Felder et al Hel v . Chim . Acta , 48: 259 (1965); de Diesbach Hel v.
Chim . Acta , 23: 1232 (1949); Truitt and Creagn J. Org. Chem . , 27: 1066 (1962); or Sekiya et al Chem . Pharm .
Bull . , 11: 551 (1963), and as shown schematically below.
Figure imgf000210_0001
Synthesis of 4-Chloro-3-aminomethylbenzoic Acid•HCI
The title compound was prepared by modification of procedures previously reported in the literature (Felder et al (1965) Hel v. Chim . Acta , 48: 259). To a solution of 4-chlorobenzoic acid (15.7 g, 100 mmol) in 150 ml of concentrated sulfuric acid was added N-hydroxymethyl dichloroacetamide (23.7 g, 150 mmol) in portions. The reaction mixture was stirred at room temperature for 2 days, poured onto 375 g of ice, stirred for 1 hour, the solid was collected by filtration, and washed with H2O. The moist solid was dissolved in 5% sodium bicarbonate solution, filtered, and acidified to pH 1 with
concentrated HCl. The solid was collected by filtration, washed with H2O, and air-dryed overnight to give 4- chloro-3-dichloroacetylaminomethylbenzoic acid (26.2 g, 89%) as a white powder. A suspension of 4-chloro-3- dichloroacetylaminomethylbenzoic acid (26.2 g, 88 mmol) in 45 ml of acetic acid, 150 ml of concentrated HCl, and 150 ml of H2O was heated to reflux for 3 hours, filtered while hot, and allowed to cool to room temperature. The solid was collected by filtration, washed with ether, washed with acetone-ether, and air-dryed overnight to give the title compound (7.6 g, 39%) as off-white crystals, mp 278-9°C; 1H NMR (D6-DMSO) 13.40 (br s, 1H), 8.75 (br s, 3H), 8.20 (s, 1H), 7.95 (dd, 1H), 7.70 (d, 1H), 4.20 (br s, 2H). t-Butyloxycarbonyl-4-chloro-3-aminomethylbenzoic Acid
A suspension of 4-chloro-3-aminomethylbenzoic acid•HCl (6.7 g, 30 mmol) and triethylamine (9.3 g, 92 mmol) in 50 ml of H2O, was added to a solution of Boc-ON (9.2 g, 38 mmol) in 50 ml of tetrahydrofuran cooled to 0°C. The reaction mixture was stirred at room
temperature overnight, and the volatile compounds were removed by concentration under reduced pressure. The residue was diluted with H2O, washed with ether, acidified to pH 3 with IN HCl, and extracted with ethyl acetate. The extracts were washed with H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated with ether-hexane to provide the title compound (7.4 g, 87%) as a white powder, mp 159°C (dec); 1H NMR (D6-DMSO) 13.20 (br s, 1H), 7.90 (s, 1H), 7.80 (dd, 1H), 7.60 (br s, 1H), 7.55 (d, 1H), 4.20 (br d, 2H), 1.40 (s, 9H).
Synthesis of 3-Aminomethyl-6-iodobenzoic Acid•HCl The title compound was prepared by modification of procedures previously reported in the literature (Felder et al. (1965) Helv. Chim. Acta, 48: 259). To a
solution of 6-iodobenzoic acid (24.8 g, 100 mmol) in 150 ml of concentrated sulfuric acid was added N- hydroxymethyl dichloroacetamide (23.7 g, 150 mmol) in portions. The reaction mixture was stirred at room temperature for 7 days, poured onto 375 g of ice, and stirred for 1 hour. The solid was then collected by filtration, and washed with H2O. The moist solid was dissolved in 5% sodium bicarbonate solution, filtered, and acidified to pH 1 with concentrated HCl. The solid was collected by filtration, washed with H2O, and air- dried overnight to give 3-dichloroacetyl-aminomethyl-6- iodobenzoic acid (32.0 g, 82%) as a white powder.
A suspension of 3-dichloroacetylaminomethyl-6- iodobenzoic acid (32.0 g, 82 mmol) in 51 ml of acetic acid, 170 ml of concentrated HCl, and 125 ml ofH2Owas heated to reflux for 3 hours, and filtered while hot, and allowed to cool to room temperature. The solid was collected by filtration, washed with ether, washed with acetone-ether, and air-dried overnight to give the title compound (13.2 g, 51%) as a beige powder; 1H NMR (D6- DMSO) 13.50 (br s, 1H), 8.50 (br s, 3H), 8.05 (d, 1H), 7.85 (s, 1H), 7.40 (d, 1H), 4.05 (br s, 2H). t-Butyloxycarbonyl-3-Aminomethyl-6-Iodobenzoic Acid
A suspension of 3-aminomethyl-6-iodobenzoic acid•HCl (8.0 g, 26 mmol) and triethylamine (8.7 g, 86 mmol) in 32 ml of H2O, was added to a solution of Boc-ON (8.0 g, 32 mmol) in 23 ml of tetrahydrofuran cooled to 0°C. The reaction mixture was stirred at room
temperature for overnight, and the volatile compounds were removed by concentration under reduced pressure. The residue was diluted with H2O, washed with ether, acidified to pH 3 with 1N HCl, and extracted with ethyl acetate. The extracts were washed with H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated from ether to provide the title compound (5.7 g, 59%) as a white powder; mp 182°C (dec); 1H NMR (D6-
DMSO) 13.35 (br s, 1H), 7.95 (d, 1H), 7.60 (s, 1H), 7.50 (br t, 1H), 7.10 (d, 1H), 4.10 (d, 2H), 1.40 (s, 9H).
Other examples of ring-substituted R31 cyclizing moieties prepared using the general procedure described above for t-butyloxycarbonyl-3-aminomethyl-6-iodobenzoic acid are tabulated below.
Figure imgf000214_0001
4-Bromo and 6-Bromo derivatives useful as
intermediates in the synthesis of the compounds of the invention may be prepared as described above for t- butyloxycarbonyl-3-aminomethyl-6-iodobenzoic acid. 4- Hydroxy and 6-Hydroxy derivatives useful as
intermediates in the synthesis of the compounds of the invention may be prepared as described in Sekiya et al Chem . Pharm . Bull . , 11: 551 (1963). 5-Nitro and 5-Amino derivatives useful as intermediates in the synthesis of the compounds of the invention may be prepared as described in Felder et al Helv. Chim . Acta, 48: 259 (1965). The 5-amino derivative may be converted to the 5-iodo, 5-bromo, 5-chloro, or 5-fluoro derivatives via the diazonium salt as described in Org. Syn . Coll . Vol . , 2: 130 (1943); 2: 299 (1943); 2: 351 (1943); and 3: 185 (1955).
Figure imgf000214_0002
Synthesis of Cyclic Compound Intermediates Using Ring Substituted R31 Cyclizing Moieties.
Cyclic compound intermediates in which the
cyclizing moiety contains an aromatic ring bearing a substituent group may be prepared as taught in the following examples.
Cyclic Compound Intermediate 93 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-4- chlorobenzoic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb). The cyclic peptide (240 mg, 0.28 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative LiChrospher RP-18 column (5 cm) using a 1.4% / minute gradient of 22 to 90% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (80 mg, 39%) as a fluffy white solid; 1H NMR (D6-DMSO) 9.00 (d, 1H), 8.50 (d, 1H), 8.45 (t, 1H), 7.60 (d, 2H), 7.45 (s, 1H), 7.45 (d, 2H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.45 (m, 2H), 4.20 (m, 2H), 4.10 (d, 1H), 3.55 (d, 1H), 3.10 (m, 2H), 2.90 (s, 3H), 2.65 (dd, 1H), 2.50 (m, 1H), 2.05 (m, 2H), 1.50 (m, 1H), 1.30 (m, 2H), 1.05 (d, 3H), 0.85 (d, 3H); FAB-MS: [M+H] = 609. Cyclic Compound Intermediate 94 cyclo-(D-Val-NMeArg-Gly-Asp-iodo-Mamb);
the compound of formula (VII) wherein J = D-Val, K = NMeArg, L = Gly, M = Asp, R1 = R2 = H, R10 = H,
R10a = l
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-iodo-Mamb to the oxime resin. The peptide was prepared on a 1.05 mmol scale to give the protected cyclic peptide (460 mg, 46.8%). The peptide (438 mg) and 0.5 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetic acid, and
lyophilized to generate the title compound (340 mg, 95.6%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 12.6 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (39.7% recovery, overall yield 16.6%); 1H NMR (D6-DMSO) d 9.05 (d, 1H), 8.55 (d, 1H), 8.55 (t, 1H), 7.90
(d, 1H), 7.65 (d, 1H), 7.55 (t, 1H), 7.20 (d, 1H), 7.15 (s, 1H),7.00 (br s, 4H), 5.15 (dd, 1H), 4.50
(g, 1H), 4.30 (m, 3H), 3.95 (dd, 1H), 3.60 (d, 1H), 3.10 (m, 2H), 3.00 (s, 3H), 2.75 (dd, 1H), 2.55
(dd, 1H), 2.10 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 1.10 (d, 3H), 0.90 (d, 3H); FAB-MS: [M+H] = 701.37. Cyclic Compound Intermediate 95
cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-4- methoxybenzoic acid) The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb). The cyclic peptide (600 mg, 0.71 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.33% / minute gradient of 7 to 18% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (104 mg, 32%) as a fluffy white solid; 1H NMR (D6-DMSO) 12.40 (br s, 1H), 8.25 (d, 1H), 8.20 (br s, 1H), 8.00 (br s, 2H), 7.85 (d, 1H), 7.75 (s, 1H), 7.65 (br s, 1H), 7.05 (d, 1H), 7.05 (br s, 4H), 5.00 (dd, 1H), 4.60 (q, 1H), 4.30 (d, 1H), 4.25 (d, 2H), 3.85 (s, 3H), 3.85 (dd, 1H), 3.70 (dd, 1H), 3.10 (q, 2H), 3.00 (s, 3H), 2.70 (m, 1H), 2.50 (m, 1H), 2.10 (m, 1H), 1.90 (m, 1H), 1.65 (m, 1H), 1.35 (m, 2H), 1.00 (d, 3H), 0.90 (d, 3H); FAB-MS: [M+H2O+H] = 623.
Cyclic Compound Intermediate 96 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-4- methylbenzoic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb). The cyclic peptide (210 mg, 0.25 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative LiChrospher RP-18 column (5 cm) using a 2.3% / minute gradient of 22 to 90% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (75 mg, 42%) as a fluffy white solid; 1H NMR (D6-DMSO) 12.30 (br s, 1H), 8.85 (d, 1H), 8.55 (d, 1H), 8.30 (t, 1H), 7.75 (d, 1H), 7.55 (m, 2H), 7.40 (s, 1H), 7.20 (s, 1H), 7.00 (br s, 4H), 5.20 (dd, 1H), 4.55 (q, 1H), 4.45 (dd, 1H), 4.30 (m, 2H), 4.05 (dd, 1H), 3.60 (d, 1H), 3.10 (q, 2H), 3.00 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 2.25 (s, 3H), 2.10 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 1.10 (d, 3H), 0.90 (d, 3H); FAB-MS: [M+H] = 589.
Cyclic Compound Intermediate 97 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyI-6- chlorobenzoic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (550 mg, 0.65 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (254 mg, 54%) as a fluffy white solid; 1H NMR (D6-DMSO) 12.30 (br s, 1H), 9.05 (d, 1H), 8.45 (m, 2H), 7.50 (t, 1H), 7.35 (d, 1H), 7.30 (m, 2H), 7.10 (s, 1H), 7.05 (br s, 4H), 5.15 (dd, 1H), 4.45 (dd, 1H), 4.40 (q, 2H), 4.05 (dt, 2H), 3.55 (dd, 1H), 3.15 (q, 2H), 3.10 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 2.05 (m, 2H), 1.65 (m, 1H), 1.35 (m, 2H), 1.10 (d, 3H), 0.90 (d, 3H); FABMS: [M+H] = 609. Cyclic Compound Intermediate 99
cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-6- methoxybenzoic acid) The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (256 mg, 0.30 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (137 mg, 63%) as a fluffy white solid; 1H NMR
(D6-DMSO) 8.45 (d, 1H), 8.40 (d, 1H), 8.30 (t, 1H), 7.65 (d, 1H), 7.50 (t, 1H), 7.40 (s, 1H), 7.35 (d, 1H), 7.05 (d, 1H), 7.00 (br s, 4H), 5.20 (dd, 1H), 4.55 (dd, 1H), 4.50 (q, 1H), 4.35 (dd, 1H), 4.25 (dd, 1H), 3.95 (dd, 1H), 3.90 (s, 3H), 3.55 (d, 1H), 3.10 (q, 2H), 3.00 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 2.05 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 1.10 (d, 3H), 0.95 (d, 3H); FAB-MS: [M+H] = 605. Cyclic Compound Intermediate 100 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl- 6- methylbenzoic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (230 mg, 0.28 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (54 mg, 27%) as a fluffy white solid; 1H NMR (D6-DMSO) 12.30 (br s, 1H), 8.80 (d, 1H), 8.40 (d, 1H), 8.30 (t, 1H), 7.45 (m, 2H), 7.15 (q, 2H), 7.00 (s, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.45 (m, 3H), 4.05 (m, 2H), 3.55 (dd, 1H), 3.10 (q, 2H), 3.05 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 2.30 (s, 3H), 2.05 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 1.05 (d, 3H), 0.90 (d, 3H); FABMS: [M+H] = 589.
Cyclic Compound Intermediate 100a cyclo-(D-Abu-NMeArg-Gly-Asp-3-aminomethyl-6- chlorobenzoic acid)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (330 mg, 0.40 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 1.0% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (114 mg, 41%) as a fluffy white solid; 1H NMR (D6-DMSO) 9.00 (d. 1H), 8.40 (m, 2H), 7.50 (m, 1H), 7.40 (d, 1H), 7.30 (m, 2H), 7.15 (s, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.65 (q, 1H), 4.50 (dd, 1H), 4.40 (q, 1H), 4.05 (dd, 1H), 3.95 (dd, 1H), 3.65 (dd, 1H), 3.10 (q, 2H), 3.05 (s, 3H), 2.75 (dd, 1H), 2.50 (m, 1H), 1.95 (m, 1H), 1.75 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 0.95 (t, 3H); FAB-MS: [M+H] = 595.4.
Cyclic Compound Intermediate 89d cyclo-(D-Abu-NMeArg-Gly-Asp-iodo-Mamb); the
compound of formula (VII) wherein J = D-Abu,
K = NMeArg, L = Gly, M = Asp, R1 = R2 = H,
R10 = H, R10a = I
The title compound was prepared using the general procedure described above for cyclo-(D-Val-NMeArg-Gly- Asp-Mamb) (Cyclic Compound Intermediate 4). The
DCC/DMAP method was used for attachment of Boc-iodo-Mamb to the oxime resin. The peptide was prepared on a 3.53 mmol scale to give the protected cyclic peptide (4.07 g, greater than quantitative yield). The peptide (4.07 g) and 4.0 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetic acid, and lyophilized to generate the title compound (2.97 g, greater than quantitative yield; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.16%/ min. gradient of 16.2 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (28.7% recovery, overall yield 30.2%); FAB-MS: [M+H] = 687.33.
Cyclic Compound Intermediate 100b cyclo-(D-Abu-NMeArg-Gly-Asp-3-aminomethyl-6-iodobenzoic acid) The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (350 mg, 0.38 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 1.0% / minute gradient of 10 to 38% acetonitrile containing 0.1%
trifluoroacetic acid to give the TFA salt of the title compound (150 mg, 49%) as a fluffy white solid; 1H NMR (D6-DMSO) 8.90 (d, 1H), 8.40 (m, 2H), 7.70 (d, 1H), 7.50 (m, 1H), 7.30 (m, 1H), 7.05 (s, 1H), 7.00 (d, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.65 (q, 1H), 4.45 (dd, 1H), 4.40 (q, 1H), 4.00 (q, 1H), 3.90 (q, 1H), 3.65 (dd, 1H), 3.10 (q, 2H), 3.05 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 1.95 (m, 1H), 1.75 (m, 2H), 1.60 (m, 1H), 1.40 (m, 2H), 0.95 (t, 3H); FAB-MS: [M+H] = 687.3. Cyclic Compound Intermediate 100c cyclo-(D-Abu-NMeArσ-Gly-Asp-3-aminomethyl-6- methylbenzoic acid)
(the compound of formula (VII) wherein J = D-Abu, K =
NMeArg, L = Gly, M = Asp, R10 = Me)
The title compound was prepared by the general solution-phase procedure described above for cyclo-(D- Val-NMeArg-Gly-Asp-Mamb), except that 4,4'- dinitrobenzophenone oxime was employed. The cyclic peptide (130 mg, 0.16 mmol) was deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 1.0% / minute gradient of 10 to 38% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (31 mg, 28%) as a fluffy white solid; 1H NMR (D6-DMSO) 8.70 (d, 1H), 8.40 (d, 1H), 8.30 (t, 1H), 7.50 (m, 1H), 7.45 (m, 1H), 7.15 (q, 2H), 7.05 (s, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.65 (q, 1H), 4.45 (m, 2H), 4.00 (m, 2H), 3.65 (dd, 1H), 3.10 (q, 2H), 3.05 (s, 3H), 2.75 (dd, 1H), 2.50 (m, 1H), 2.30 (s, 3H), 2.00 (m, 1H), 1.75 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 0.95 (t, 3H); FAB-MS: [M+H] = 575.4.
Figure imgf000223_0001
Scheme 5: procedure for synthesis of cyclic compound intermediate. Solid-Phase Synthesis of Cyclic Compound Intermediate
101 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-4-iodobenzoic
Acid)
The title compound was prepared using the general procedure previously described for cyclo-(D-Val-NMeArg- Gly-Asp-Mamb). The DCC/DMAP method was used for
attachment of Boc-iodo-Mamb to the oxime resin. The peptide was prepared on a 1.05 mmol scale to give the protected cyclic peptide (460 mg, 46.8%). The peptide (438 mg) and 0.5 mL of anisole were treated with
anhydrous hydrogen fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetic acid, and lyophilized to generate the title compound (340 mg, 95.6%; calculated as the acetate salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.23% / minute gradient of 12.6 to 22.5% acetonitrile containing 0.1% trifluoroacetic acid and then
lyophilized to give the TFA salt of the title compound as a fluffy white solid (39.7% recovery, overall yield 16.6%; 1H NMR (D6-DMSO) d 9.05 (d, 1H), 8.55 (d, 1H), 8.55 (t, 1H), 7.90 (d, 1H), 7.65 (d, 1H), 7.55 (t, 1H), 7.20 (d, 1H), 7.15 (s, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.50 (q, 1H), 4.30 (m, 3H), 3.95 (dd, 1H), 3.60 (d, 1H), 3.10 (m, 2H), 3.00 (s, 3H), 2.75 (dd, 1H), 2.55 (dd, 1H), 2.10 (m, 2H), 1.60 (m, 1H), 1.35 (m, 2H), 1.10 (d, 3H), 0.90 (d, 3H); FAB-MS: [M+H] = 701.37.
Solution-Phase Synthesis of Cyclic Compound Intermediate
102 cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-6-iodobenzoic
Acid)
The title compound was prepared according to the method of Scheme 6, shown below.
c
Figure imgf000225_0001
Scheme 6
1. Boc-Asp (OcHex)-3-aminomethyl-6-iodobenzoic Acid
To a suspension of 3-aminomethyl-6-iodobenzoic acid•HCl (4.9 g, 16 mmol) in H2O (16 ml) was added
NaHCO3 (3.9 g, 47 mmol), followed by a solution of Boc- Asp (OcHex)-OSu (5.9 g, 14 mmol) in THF (16 ml). The reaction mixture was stirred at room temperature overnight, filtered, diluted with H2O, acidified with IN HCl, and extracted with ethyl acetate. The extracts were washed with H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated with ether to provide the title compound (6.7 g, 82%) as a white powder. 1H NMR d (D6-DMSO) 8.45 (br t, 1H), 7.90 (d, 1H), 7.60 (s, 1H), 7.15 (m, 2H), 4.65 (m, 1H), 4.35 (m, 1H), 4.25 (d, 2H), 2.70 (m, 1H), 2.55 (m, 1H), 1.70 (m, 4H), 1.40 (s, 9H), 1.35 (m, 6H).
2. 4,4'-Dinitrobenzophenone Oxime
The title compound was prepared by modification of procedures previously reported in the literature
(Chapman and Fidler (1936) J. Chem . Soc, 448; Kulin and Leffek (1973) Can . J. Chem . , 51: 687). A solution of chromic anhydride (20 g, 200 mmol) in 125 ml of H2O was added dropwise over 4 hours, to a suspension of bis (4- nitrophenyl) methane (25 g, 97 mmol) in 300 ml of acetic acid heated to reflux. The reaction mixture was heated at reflux for 1 hour, cooled to room temperature, and poured into water. The solid was collected by
filtration, washed with H2O, 5% sodium bicarbonate, H2O, and air-dryed to provide a 1:1 mixture of bis (4- nitrophenyl) methane/4,4'-dinitrobenzophenone via 1H NMR. This material was oxidized with a second portion of chromic anhydride (20 g, 200 mmol), followed by an identical work-up procedure to provide the crude
product. Trituration with 200 ml of benzene heated to reflux for 16 hours provided 4,4'-dinitrobenzophenone (20.8 g, 79%) as a yellow powder.
A solution of hydroxylamine hydrochloride (10.2 g, 147 mmol) was added to a suspension of 4,4'- dinitrobenzophenone (19 g, 70 mmol) in 100 ml of
ethanol. The reaction mixture was heated to reflux for 2 hours, cooled to room temperature, and the solid
collected by filtration. Recrystallization from ethanol provided the title compound (14.0 g, 70%) as pale yellow crystals, mp 194°C; 1H NMR (D6-DMSO) d 12.25 (s, 1H), 8.35 (d, 2H), 8.20 (d, 2H), 7.60 (d, 4H). 3. 4,4'-Dinitrobenzophenone Oxime Boc-Asp (OcHex)-3- aminomethyl-6-iodobenzoate
To an ice-cooled solution of Boc-Asp (OcHex)-3- aminomethyl-6-iodobenzoic acid (3.3 g, 5.7 mmol) and 4,4'-dinitrobenzophenone oxime (1.7 g, 5.9 mmol) in 32 ml of ethyl acetate was added DCC (1.2 g, 5.8 mmol). The reaction mixture was stirred at room temperature for 3 hours, filtered, diluted with ethyl acetate, washed with saturated sodium bicarbonate solution, H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was purified by column chromatography on silica gel (EM Science, 230-400 mesh) using 10:1 dichloromethane/ethyl acetate to give the title compound (1.8 g, 36%) as pale yellow crystals. 1H NMR (D6-DMSO) d 8.40 (dd, 5H), 7.90
(m, 5H), 7.45 (s, 1H), 7.20 (m, 2H), 4.65 (m, 1H), 4.35 (m, 1H), 4.20 (m, 2H), 2.75 (dd, 1H), 2.50 (dd, 1H), 1.70 (m, 4H), 1.40 (s, 9H), 1.35 (m, 6H).
4. Boc-D-Val-NMeArg (Tos)-Gly
To a mixture of Boc-NMeArg (Tos) (11.07 g, 25 mmol), and Gly-OBzl tosylate (10.10 g, 30 mmol) in 25 ml of dichloromethane was added HBTU (9.48 g, 25 mmol) and DIEA (9.69 g, 75 mmol). The reaction mixture was stirred at room temperature for 1 hour, concentrated under high vacuum, diluted with ethyl acetate, washed with 5% citric acid, H2O, saturated sodium bicarbonate solution, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. The resulting oil was triturated with petroleum ether to provide Boc-NMeArg (Tos)-Gly-OBzl (14.7 g, 100%); FAB-MS: [M+H] = 590.43. This material was used without further purification.
A solution of Boc-NMeArg (Tos) -Gly-OBzl (14.5 g, 24.6 mmol) in 30 ml of trifluoroacetic acid was stirred at room temperature for 5 minutes, and evaporated to dryness under reduced pressure. The oily residue was diluted with cold ethyl acetate, washed with cold saturated sodium bicarbonate solution, the aqueous phase was extracted with ethyl acetate. The combined organics were washed with brine, evaporated to dryness under reduced pressure, and the resulting oil triturated with ether. The resulting solid was filtered, washed with ether, and dried in a vacuum desiccator to provide NMeArg (Tos)-Gly-OBzl (10.3 g, 86%); FAB-MS: [M+H] = 490.21. This material was used without further
purification.
To a solution of NMeArg (Tos)-Gly-OBzl (4.80 g, 9.8 mmol), and Boc-D-Val (2.13 g, 9.8 mmol) in 10 ml of dichloromethane, cooled in an ice-bath, was added HBTU (3.79 g, 10.0 mmol) and DIEA (2.58 g, 20.0 mmol). The reaction mixture was stirred at room temperature for 48 hours, diluted with ethyl acetate, washed with 5% citric acid, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. The resulting oil was triturated with ether to provide Boc- D-Val-NMeArg (Tos)-Gly-OBzl (4.58 g, 68%); FAB-MS : [M+H] = 689.59. This material was used without further
purification.
A solution of Boc-D-Val-NMeArg (Tos)-Gly-OBzl (4.50 g, 6.53 mmol) in 80 ml of methanol was purged with nitrogen gas, 1.30 g of 10% Pd/C was added, and hydrogen gas was passed over the reaction. After 1 hour the catalyst was removed by filtration through a bed of celite, and the solvent removed under reduced pressure. The resulting solid was triturated with ether, filtered, and washed with petroleum ether to provide Boc-D-Val- NMeArg (Tos)-Gly (3.05 g, 78%); 1H NMR (D6-DMSO) d 7.90 (br t, 1H), 7.65 (d, 2H), 7.30 (d, 2H), 7.00 (d, 1H), 6.85 (br d, 1H), 6.60 (br s, 1H), 5.00 (dd, 1H), 4.15 (t, 1H), 3.70 (m, 2H), 3.05 (m, 2H), 2.90 (s, 3H), 2.35 (s, 3H), 1.90 (m, 2H), 1.55 (m, 1H), 1.35 (s, 9H), 1.25 (m, 2H), 0.80 (br t, 6H); FAB-MS: [M+H] = 599.45.
5. 4,4'-Dinitrobenzophenone Oxime Boc-D-Val-NMeArg (Tos)- Glv-Asp (OcHex)-3-aminomethyl-6-iodobenzoate
To a solution of 4,4'-dinitrobenzophenone oxime Boc-Asp (OcHex)-3-aminomethyl-6-iodobenzoate (0.5 g, 0.59 mmol) in 1 ml of dichloromethane was added 0.5 ml of trifluoroacetic acid. The reaction mixture was stirred at room temperature for 90 minutes, diluted with
dichloromethane, and evaporated to dryness under reduced pressure. The oily residue was concentrated under high vacuum to remove traces of excess trifluoroacetic acid.
To a solution of the crude TFA salt and Boc-D-Val- NMeArg (Tos)-Gly (0.52 g, 0.87 mmol) in 3.8 ml of DMF was added TBTU (0.28 g, 0.87 mmol) and DIEA (0.33 g, 2.58 mmol). The reaction mixture was stirred at room
temperature overnight, concentrated under high vacuum, diluted with ethyl acetate, washed with 5% citric acid, H2O, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was triturated with ether to provide the title compound (0.48 g, 61%) as a powder. This material was used without further purification.
6. cyclo-(D-Val-NMeArg(Tos)-Gly-Asp(OcHex)-3- aminomethyl-6-iodobenzoic Acid)
To a solution of 4,4'-dinitrobenzophenone oxime Boc-D-Val-NMeArg (Tos)-Gly-Asp(OcHex)-3-aminomethyl-6- iodobenzoate (0.48 g, 0.36 mmol) in 1 ml of
dichloromethane was added 0.5 ml of trifluoroacetic acid. The reaction mixture was stirred at room
temperature for 45 minutes, diluted with
dichloromethane, and evaporated to dryness under reduced pressure. The oily residue was concentrated under high vacuum to remove traces of excess trifluoroacetic acid.
To a solution of the crude TFA salt in 38 ml of DMF was added acetic acid (0.09 ml, 1.57 mmol) and DIEA (0.26 ml, 1.49 mmol). The reaction mixture was stirred at 60°C for 3 days, concentrated under high vacuum, diluted with ethyl acetate, washed with 5% citric acid, brine, dried over anhydrous magnesium sulfate, and evaporated to dryness under reduced pressure. This material was purified by column chromatography on silica gel (EM Science, 230-400 mesh) using 10:1
chloroform/isopropanol to give the title compound (0.13 g, 38%) as a powder; 1H NMR (D6-DMSO) d 8.95 (d, 1H), 8.50 (t, 1H), 8.45 (d, 1H), 7.70 (d, 1H), 7.60 (d, 2H), 7.30 (d, 3H), 7.05 (d, 1H), 7.00 (s, 1H), 6.80 (br s, 1H), 6.60 (br s, 1H), 5.10 (dd, 1H), 4.65 (m, 1H), 4.45 (m, 1H), 4.35 (m, 1H), 4.00 (m, 1H), 3.55 (dd, 1H), 3.05 (m, 2H), 3.00 (s, 3H), 2.70 (dd, 1H), 2.55 (dd, 1H), 2.35 (s, 3H), 2.05 (m, 1H), 1.90 (m, 1H), 1.75 (m, 1H), 1.65 (m, 1H), 1.35 (m, 13H), 1.15 (d, 3H), 0.85 (d, 3H); FAB(GLYC)-MS: [M+H] = 937.
7. cyclo-(D-Val-NMeArg-Gly-Asp-3-aminomethyl-6- iodobenzoic Acid)
The cyclic peptide (490 mg, 0.52 mmol) was
deprotected with excess HF in the presence of anisole as scavenger. Purification was accomplished by reversed- phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.8% / minute gradient of 10 to 38% acetonitrile containing 0.1% trifluoroacetic acid to give the TFA salt of the title compound (194 mg, 46%) as a fluffy white solid; 1H NMR (D6-DMSO) d 12.30 (br s, 1H), 9.00 (d, 1H), 8.40 (m, 2H), 7.70 (d, 1H), 7.50 (m, 1H), 7.30 (m, 1H), 7.05 (d, 1H), 7.00 (s, 1H), 7.00 (br s, 4H), 5.15 (dd, 1H), 4.40 (d, 1H), 4.40 (q, 2H), 4.0 (m, 2H), 3.55 (dd, 1H), 3.15 (q, 2H), 3.10 (s, 3H), 2.70 (dd, 1H), 2.50 (m, 1H), 2.05 (m, 2H), 1.65 (m, 1H), 1.35 (m, 2H), 1.15 (d, 3H), 0.90 (d, 3H); FAB -MS : [M+H] = 701.
Table A shows the FAB-MS obtained for certain cyclic compound intermediates.
Figure imgf000231_0001
Figure imgf000232_0002
Other ring substituted cyclizing moieties can be synthesized as taught in the following schemes and discussion. The moiety of the formula above where Z = NH2 can be synthesized by at least two different routes For example, starting with 4-acetamidobenzoic acid (Aldrich Chemical Co.), a Friedel-Crafts alkylation with N-hydroxymethyldichloroacetamide would give the dichloroacetyl derivative of 3-aminomethyl-4- acetamidobenzoic acid (Felder, Pitre, and Fumagalli (1964), Helv. Chim . Acta, 48, 259-274). Hydrolysis of the two amides would give 3-aminomethyl-4-aminobenzoic acid.
C
Figure imgf000232_0001
Alternatively, starting with 3-cyano-4-nitrotoluene, oxidation with chromium trioxide followed by reduction will give 3-aminomethyl-4-aminobenzoic acid.
Figure imgf000233_0001
a] CrO3 b] H2-catalyst
The moiety of the formula above where Y = CH2NH2 can be synthesized from 3,5-dicyanotoluene by oxidation of the methyl group with chromium trioxide followed by reduction.
Figure imgf000233_0002
a] CrO3 b] H2-catalyst The moiety of the formula above where Z = CH2NH2 can be synthesized from 3-cyano-4-methylbenzoic acid (K & K Rare and Fine Chemicals). Bromination using N- bromosuccinimide would give 4-bromomethyl-3-cyanobenzoic acid. A nucleophilic substitution reaction at the bromomethyl position using an amide anion would produce the protected amine. Amide anions which could be used in this reaction include potassium phthalimide (Gabriel synthesis), and the anion of trifluoroacetamide (Usui (1991), Nippon Kagaku Kaishi , 206-212) used in this example. Reduction of the nitrile would produce the second aminomethyl group, which would be protected by reaction with di-t-butyl dicarbonate. Removal of the trifluoroacetamide protecting group using aqueous piperidine would give the moiety. γ
Figure imgf000234_0001
Alternatively, the moiety can be prepared from 4- bromobenzoic acid as shown in the scheme .
Figure imgf000234_0002
a] H2SO4, HOCH2NHCOCHCI2 b]H+, boc-ON
c] CuCN, DMF d] H2-catalyst
These ring substituted cyclizing moieties can be used to synthesize cyclic compound intermediates.
Cyclic Compound Intermediate 113
Cyclo (D-Val-NMeArg-Gly-Asp-Mamb (4-NH2)
Figure imgf000235_0001
This compound can be prepared using the procedure described above for Cyclo (D-Val-NMeArg-Gly-Asp-Mamb substituting the ring substituted cyclizing moiety where Z = NH2.
Cyclic Compound Intermediates 114, 115 and 116
Figure imgf000235_0002
X1 = 2-propyl, ethyl, or p-hydroxyphenylmethyl X2 = H.
Compounds cyclo (D-Val-NMeArg-Gly-Asp-Mamb ( 5-CH2NHX2), cyclo (D-Abu-NMeArg-Gly-Asp-Mamb (5-CH2NHX2), and cyclo (D- Tyr-NMeArg-Gly-Asp-Mamb (5-CH2NHX2) can be prepared via the methods described above using the ring substituted cyclizing moiety where Y = CH2NH2.
Cyclic Compound Intermediates 117, 118 and 119.
Figure imgf000236_0001
X1 = 2-propyl, ethyl, or p-hydroxyphenylmethyl
X2 = H
Compounds cyclo (D-Val-NMeArg-Gly-Asp-Mamb (4-CH2NHX2 ), cyclo (D-Abu-NMeArg-Gly-Asp-Mamb(4-CH2NHX2), and cyclo(D- Tyr-NMeArg-Gly-Asp-Mamb (4-CH2NHX2) can be prepared via the procedures described above using the ring
substituted cyclizing moiety where Z = CH2NH2.
Other R31 Cyclizing Moieties
Alternatives to Mamb useful as cyclizing moieties R31 in the cyclic peptides of the invention include aminoalkyl-naphthoic acid and aminoalkyl- tetrahydronaphthoic acid residues. Representative aminoalkyl-naphthoic acid and aminoalkyl- tetrahydronaphthoic acid intermediates useful in the synthesis of cyclic peptides of the present invention are described below. The synthesis of these intermediates is outlined below in Scheme 7.
Figure imgf000237_0001
8-Amino-5,6,7,8-tetrahydro-2-naphthoic Acid Hydrochloride (8)
The title compound was prepared according to a modification of standard procedures previously reported in the literature (Earnest, I., Kalvoda, J., Rihs, G., and Mutter, M., Tett. Lett., Vol. 31, No. 28, pp 4011- 4014, 1990).
As shown above in Scheme 7, 4-phenylbutyric acid (1) was converted to the ethyl ester (2) which was acylated via aluminum chloride and acetylchloride to give 4-acetylphenylbutyric acid ethyl ester (3). This ester was subjected to saponification to give 4- acetylphenylbutyric acid (4). Subsequently, the acetyl group was oxidized to give 4-carboxyphenylbutyric acid (5) which was converted to the l-tetralin-7-carboxylic acid (6) using aluminum chloride in a Friedel-Crafts cyclization with resonably high yield. At that point, the tetralone was split into two portions and some was converted to the oxime (7) using sodium acetate and hydroxylamine hydrochloride. The oxime was subjected to hydrogenolysis to give the racemic mixture of 8-amino- 5,6,7,8-tetrahydro-2-naphthoic acid as the hydrochloride (8) for use as an intermediate for incorporation into the cyclic peptide.
Part A - A solution of 4-phenylbutyric acid (50.0 g, 0.3 mol) in ethanol (140 mL) with concentrated sulfuric acid (0.53 mL) was stirred at reflux over 5 hours. The cooled solution was poured into ice water and extracted with ethyl acetate. The combined organic layers were
backwashed with brine, dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure to give 4-phenylbutyric acid ethyl ester (56.07 g, 0.29 mol, 97%) as a yellow liquid. 1H NMR (CDCI3) d 7.3-7.1 (m, 5H), 4.1 (q, 2H, J=7.1 Hz), 2.7 (t, 2H, J=7.7 Hz), 2.3 (t, 2H, J=7.5 Hz), 1.95 (quintet, 2H, J=7.5 Hz), 1.25 (t, 3H, J=7.1 Hz).
Part B - To a solution of aluminum chloride (153 g, 1.15 mol), and acetyl chloride (38.5 mL, 42.5 g, 0.54 mol) in dichloromethane (1500 mL) was added, dropwise, a solution of 4-phenylbutyric acid ethyl ester (50.0 g, 0.26 mol) in dichloromethane (500 mL). All was stirred at ambient temperature for 15 minutes. The solution was poured into cold concentrated hydrochloric acid (2000 mL) and then extracted with dichloromethane. The combined organic layers were backwashed with brine, dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure to give 4- acetylphenylbutyric acid ethyl ester (53.23 g, 0.23 mol, 88%) as a dark yellow liquid. 1H NMR (CDCI3) d 7.9 (d, 2H, J=8.1 Hz), 7.25 (d, 2H, J=8.4 Hz), 4.1 (q, 2H, J=7.1 Hz), 2.75 (t, 2H, J=7.6 Hz), 2.6 (s, 3H), 2.35 (t, 2H, J=7.6 Hz), 2.0 (quintet, 2H, J=7.5 Hz), 1.25 (t, 3H, J=7.1 Hz).
Part C -To a solution of 4-acetylphenylbutyric acid ethyl ester (50.0 g, 0.21 mol) in ethanol (1250 mL) was added, dropwise, a solution of sodium hydroxide (50.0 g) in water (1250 mL). All was stirred at reflux over 4 hours. The solution was concentrated to half volume and then acidified to a pH equal to 1.0 using hydrochloric acid (1N). The resulting precipitate was collected and washed with water to give 4-acetylphenylbutyric acid (53.76 g, 0.26 mol, 99%) as a white solid, mp = 50-52°C; 1H NMR (CDCI3) d 7.9 (d, 2H, J=8.1 Hz), 7.25 (d, 2H, J=9 . 1 Hz ) , 2 . 75 ( t , 2H, J=7 . 7 Hz ) , 2 . 6 ( s , 3H ) , 2 . 4 (t , 2H, J=7 . 3 Hz ) , 2 . 0 ( quintet , 2H, J=7 . 4 Hz ) .
Part D -To a solution of sodium hypochlorite (330 mL, 17.32 g, 0.234 mol) in a solution of sodium hydroxide
(50%, 172 mL), warmed to 55°C, was added, portionwise as a solid, 4-acetylphenylbutyric acid (16.0 g, 0.078 mol) while keeping the temperature between 60-70°C. All was stirred at 55°C over 20 hours. The cooled solution was quenched by the dropwise addition of a solution of sodium bisulfite (25%, 330 mL). The mixture was then transferred to a beaker and acidified by the careful addition of concentrated hydrochloric acid. The
resulting solid was collected, washed with water and dried, then triturated sequentially with chlorobutane and hexane to give 4-carboxyphenylbutyric acid (15.31 g, 0.074 mol, 95%) as a white solid, mp = 190-195°C; 1H NMR (DMSO) d 12.55 (bs, 1H), 8.1 (s, 1H), 7.85 (d, 2H, J=8.1 Hz), 7.3 (d, 2H, J-8.1 Hz), 2.7 (t, 2H, J=7.5 Hz), 2.2 (t, 2H, J=7.4 Hz), 1.8 (quintet, 2H, J=7.5 Hz).
Part E - A mixture of 4-carboxyphenylbutyric acid (10.40 g, 0.05 mol), aluminum chloride (33.34 g, 0.25 mol) and sodium chloride (2.90 g, 0.05 mol) was heated with continual stirring to 190°C over 30 minutes. As the mixture cooled to 60°C, cold hydrochloric acid (1N, 250 mL) was carefully added. The mixture was extracted with dichloromethane. The combined organic layers were backwashed with dilute hydrochloric acid and water, dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure. The resulting solid was triturated with chlorobutane to give 1-tetralon-7- carboxylic acid (9.59 g, 0.05 mol, 100%) as a brown solid, mp = 210-215°C; 1H NMR (DMSO) d 8.4 (s, 1H), 8.1 (d, 2H, J=8.0 Hz), 7.5 (d, 1H, J=7.9 Hz), 3.0 (t, 2H, J=6.0 Hz), 2.65 (t, 2H, J=6.6 Hz), 2.1 (quintet, 2H, J=6.3 Hz) . Part F - A solution of l-tetralon-7-carboxylic acid (1.0 g, 0.0053 mol) and sodium acetate (1.93 g, 0.024 mol) and hydroxylamine hydrochloride (1.11 g, 0.016 mol) in a mixture of methanol and water (1:1, 15 mL) was stirred at reflux over 4 hours. The mixture was cooled and then added was more water (50 mL). The solid was collected, washed with water and dried, then triturated with hexane to give 1-tetralonoxime-7-carboxylic acid (0.78 g, 0.0038 mol, 72%) as a white solid, mp = 205-2l5°C; 1H NMR (DMSO) d 11.3 (s, 2H) , 8.4 (s, 1H), 7.8 (d, IH, J=7.7 Hz), 7.3 (d, 1H, J=7.7 Hz), 2.8 (t, 2H, J=5.9 Hz), 2.7 (d, 2H, J=6.6 Hz), 1.9-1.7 (m, 2H).
Part G - A mixture of 1-tetralonoxime-7-carboxylic acid (0.75 g, 0.0037 mol) in methanol (25 mL) with
concentrated hydrochloric acid (0.54 mL, 0.20 g, 0.0056 mol) and palladium on carbon catalyst (0.10 g, 5% Pd/C) was shaken for 20 hours at ambient temperature under an atmosphere of hydrogen (60 psi). The reaction mixture was filtered over Celite@ and washed with methanol. The filtrate was evaporated to dryness under reduced
pressure and the residue was purified by flash
chromatography using hexane:ethyl acetate :: 1 : 1 to give the racemic mixture of 8-amino-5,6,7,8-tetrahydro-2- naphthoic acid hydrochloride (0.225 g, 0.001 mol. 27%) as a white solid, mp = 289-291°C; 1H NMR (DMSO) d 8.55 (bs, 3H), 8.2-8.1 (m, 1H), 7.85-7.8 (m, 1H), 7.35-7.25 (m, 1H), 4.5 (m, 1H), 2.9-2.8 (m, 2H), 2.1-1.9 (m, 3H), 1.85-1.7 (m, 1H). N- (BOC) -8-Aminomethyl-5 , 6 , 7 , 8-tetrahydro-2-naphthoic
Acid ( 12 ) As shown above in Scheme 7, the remaining tetralone was then converted to -the methyl ester (9). Using a procedure from Gregory, G.B. and Johnson, A.L, JOC, 1990, 55, 1479, the tetralone methyl ester (9) was converted, first, to the cyanohydrin by treatment with trimethylsilylcyanide and zinc iodide and then, via the in situ dehydration with phosphorous oxychloride in pyridine, to the methyl 8-cyano-5,6-dihydro-2-naphthoate (11). This naphthoate was divided into two portions and some was subjected to hydrogenolysis, N-BOC-protection and saponification to give N-(BOC)-8-aminomethyl- 5,6,7,8-tetrahydro-2-naphthoic acid (12) as an
intermediate for incorporation into the cyclic peptide.
Part A - A mixture of 1-tetralon-7-carboxylic acid (7.0 g, 0.037 mol) in methanol (13.6 mL, 10.8 g, 0.30 mol) with a catalytic amount of hydrochloriic acid (0.07 mL, 0.12 g, 0.0012 mol) was stirred at reflux over 5 hours. The cooled reaction mixture was poured into ice water and extracted with ethyl acetate. The combined organic layers were backwashed with water and brine, dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure. The resulting solid was purified by flash chromatography using hexane: ethyl
acetate :: 75 : 25. The resulting solid was triturated with hexane to give l-tetralon-7-carboxylic acid methyl ester (3.61 g, 0.018 mol, 49%) as a yellow solid, mp = 170- 172°C; 1H NMR (CDCI3) d 8.7 (s, 1H), 8.15 (d, 1H, J=8.1 Hz), 7.35 (d, 1H, J=8.1 Hz), 3.95 (s, 3H), 3.05 (d, 2H, J=6 . 1 Hz ) , 2 . 7 (t , 2H, J=6 . 4 Hz ) , 2 . 15 ( quintet , 2H , J=6 . 2 Hz ) .
Part B - A solution of l-tetralon-7-carboxylic acid methyl ester (3.50 g, 0.017 mol), trimethylsilylcyanide (1.98 g, 0.02 mol) and zinc iodide (0.10 g) in benzene (20 mL) was stirred at ambient temperature over 15 hours. Then added, sequentially and dropwise, was pyridine (20 mL) and phosphorous oxychloride (4.0 mL, 6.55 g, 0.0425 mol). The reaction mixture was stirred at reflux over 1 hour then evaporated to dryness under reduced pressure. The residue was taken up in
chloroform, backwashed with water, dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure to give methyl 8-cyano-5, 6-dihydro-2- naphthoate (1.70 g, 0.008 mol, 47%) as a yellow solid, mp = 73-75°C; 1H NMR (CDCl3) d 8.0-7.9 (m, 1H), 7.3-7.2 (m, 1H), 6.95 (t, 1H, J=4.8 Hz), 3.95 (s, 3H), 2.9 (t, 2H, J=8.3 Hz), 2.6-2.4 (m, 3H)
Part C - A mixture of methyl 8-cyano-5, 6-dihydro-2- naphthoate (0.80 g, 0.0038 mol) in methanol (25 mL) with concentrated hydrochloric acid (0.56 mL) and palladium on carbon catalyst (0.40 g, 5% Pd/C) was shaken for 20 hours at ambient temperature under an atmosphere of hydrogen (50 psi). The reaction mixture was filtered over Celite and washed with methanol. The filtrate was evaporated to dryness under reduced pressure and the residue was triturated with hexane to give the racemic mixture of methyl 8-aminomethyl-5,6,7,8-tetrahydro-2- naphthoate (0.80 g, 0.0037 mol, 97%) as a white solid, mp = 172-179°C; 1H NMR (DMSO) d 8.2-8.0 (m, 4H), 7.9-7.7 (m, 6H), 7.5-7.2 (m, 4H), 3.9-3.8 (m, 7H), 3.3-2.7 (m, 10H), 2.0-1.6 (m, 8H). Part D - A solution of methyl 8-aminomethyl-5,6,7,8- tetrahydro-2-naphthoate (0.78 g, 0.0036 mol) and triethylamine (0.55 mL, 0.40 g, 0.004 mol) in aqueous tetrahydrofuran (50%, 75 mL) was added, portionwise as a solid, 2-(tert-butoxycarbonyloxyimino)-2- phenylacetonitrile (0.99 g, 0.004 mol). All was stirred at ambient temperature over 3 hours. The solution was concentrated to half volume and extracted with
diethylether. The aqueous layer was then acidified to a pH of 1.0 using hydrochloric acid (1N) and then extraced with ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure. The residue was purified by flash chromatography using hexane: ethyl acetate::8:2 to give methyl N-(BOC)-8-aminomethyl-5,6,7,8-tetrahydro- 2-naphthoate (0.54 g, 0.0017 mol, 47%) as a white solid, mp = 72-80°C; 1H NMR (DMSO) d 13.8 (s, 1H), 7.8-7.65 (m, 3H), 7.6-7.5 (m, 3H) , 7.25-7.20 (m, 1H), 7.15-7.05 (m, 1H), 3.9-3.8 (m, 1H), 3.2-2.8 (m, 4H), 1.8-1.6 (m, 3H), 1.4 (s, 6H).
Part E - To a solution of methyl N-(BOC)-8-aminomethyl- 5,6,7,8-tetrahydro-2-naphthoate (0.50 g, 0.0016 mol) in ethanol (12.5 mL) was added, dropwise, a solution of sodium hydroxide (0.50 g) in water (12.5 mL). All was stirred a reflux over 4 hours. The reaction mixture was concentrated to half volume and then acidified to a pH equal to 1.0 using hydrochloric acid (IN). The residue was puified by flash chromatography using a gradient of hexane :ethyl acetate :: 1 : 1 to ethyl acetate to ethyl acetate: methanol :: 9 : 1 to give the racemic mixture of the title compound, N-(BOC)-2-aminomethyl-5,6,7,8- tetrahydro-2-naphthoic acid (0.19 g, 0.00062 mol, 39%) as a white solid, mp = 172-176°C; 1H NMR (DMSO) d 7.8 (s, 1H), 7.65 (d, 1H, J=8.1 Hz), 7.15 (d, 1H, J=8.1 Hz), 7.1-7.0 (m, 1H), 3.2-3.1 (m, 2H), 3.0-2.7 (m, 4H), 1.8- 1.6 (m, 4H), 1.4 (s, 9H).
N-(BOC)-8-aminomethyl-2-naphthoic acid (14)
The remaining naphthoate (11) was treated with 2,3- dichloro-5, 6-dicyano-1,4-benzoquinone (DDQ) in dioxane to aromatize the adjacent ring to give the methyl 8- cyano-2-naphthoate (13). Then, the nitrile was reduced via hydrogentation and the methyl ester saponified to the carboxylic acid. This acid was then N-BOC-protected to give N-(BOC)-8-aminomethyl-2-naphthoic acid (14) as an intermediate for incorporation into the cyclic peptide.
Part A - A solution of methyl 8-cyano-5,6-dihydro-2- naphthoate (1.0 g, 0.0047 mol) and 2,3-dichloro-5,6- dicyano-1,4-benzoquinone (1.07 g, 0.0047 mol) in dioxane (50 mL) was stirred at 120°C over 16 hours. The reaction mixture was poured into ice water and extracted with ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure. The residue was purified by flash chromatography using ethyl acetate to give methyl 8-cyano-2-naphthoate (0.72 g, 0.0034 mol, 73%) as a tan solid, mp = 178-182°C; 1H NMR (CDCl3) d 8.95 (s, 1H), 8.3-8.2 (m, 1H), 8.15-8.10 (m, 1H), 8.0-7.95 (m, 2H), 7.7-7.6 (m, 1H), 4.05 (s, 1H). Part B - A mixture of methyl 8-cyano-2-naphthoate (1.0 g, 0.0047 mol) in methanol (35 mL) with concentrated hydrochloric acid (0.69 mL) andpalladium on carbon catalyst (0.20 g, 5% Pd/C) was shaken for 6 hours at ambient temperature under anatmosphere of hydrogen (50 psi). The reaction mixture was filtered over Celite@ and washed with methanol. The filtrate was evaporated to dryness under reduced pressure and the residue was triturated with hexane to give methyl 8-aminomethyl-2- naphthoate (0.76 g, 0.0035 mol, 75%) as an oil. 1H NMR (DMSO) d 8.75 (s, 1H), 8.5 (bs, 2H) , 8.2-8.05 (m, 3H), 7.75-7.70 (m, 2H), 4.6 (s, 2H), 3.95 (m, 3H).
Part C - To a solution of methyl 8-aminomethyl-2- naphthoate (0.75 g, 0.0035 mol) in dry tetrahydrofuran (50 mL), cooled to 0°C, was added a solution of lithium hydroxide (0.5 M, 5.83 mL). All was stirred at ambient temperature over 20 hours. Another aliquot of lithium hydroxide was added and all was stirred for an
additional 20 hours. The solid was collected and the filtrate was evaporated to dryness under reduced pressure. The solids were triturated with diethyl ether to give 8-aminomethyl-2-naphthoic acid (0.67 g, 0.0033 mol, 95%) as a white solid, mp = 223-225°C; 1H NMR
(DMSO) d 8.6 (s, 1H), 8.1-7.9 (m, 1H), 7.8-7.7 (m, 4H), 7.55-7.5 (m, 1H), 7,45-7.35 (m, 2H), 4.2 (s, 2H).
Part D - A solution of 8-aminomethyl-2-naphthoic acid (0.50 g, 0.00025 mol) and triethylamine (0.038 mL, 0.028 g, 0.000275 mol) in aqueous tetrahydrofuran (50%, 5 mL) was added, portionwise as a solid, 2- ( tert- butoxycarbonyloxyimino)-2-phenylacetonitrile (0.068 g, 0.000275 mol). All was stirred at ambient temperature over 5 hours. The solution was concentrated to half volume and extracted with diethylether. The aqueous layer was then acidified to a pH of 1.0 using
hydrochloric acid (1N) and then extraced with ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate and evaporated to dryness under reduced pressure to give the title compound, N- (BOC)-8-aminomethyl-2-naphthoic acid (0.050 g, 0.00017 mol) as a white solid, mp = 190-191°C; 1H NMR (DMSO) d 13.1 (bs, 1H), 8.8 (s, 1H), 8.0 (q, 2H, J=7.9 Hz), 7.9 (d, 1H, J=8.1 Hz), 7.6 (t, 1H, J=7.5 Hz), 7.65-7.55 (m, 2H), 4.6 (d, 2H, J=5.5 Hz), 1.4 (s, 9H).
Cyclic Compound Intermediates 89a and 89b
cyclo-(D-Val-NMeArg-Gly-Asp-aminotetralincarboxylic acid); the compound of formula (VIII) wherein J =
D-Val, K = NMeArg, L = Gly, M = Asp,
R1 = R2 = H
The title compound was prepared using the
general procedure described for cyclo-(D-Val-
NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-aminotetralin-carboxylic acid to the oxime
resin. The peptide was prepared on a 0.164 mmol scale to give the protected cyclic peptide (69 mg, 49.3%). The peptide (69 mg) and 0.069 mL of
anisole were treated with anhydrous hydrogen
fluoride at 0°C for 30 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (59.7 mg, greater than quantitative yield;
calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a
preparative Vydac C18 column (2.5 cm) using a 0.23%/ min. gradient of 16.2 to 27% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid. Two isomers were obtained; isomer #1 (12.5% recovery, overall yield 6.2%, FAB-MS: [M+H] = 615.34; isomer #2 (18.6% recovery, overall yield 9.3%, FAB-MS: [M+H] = 615.35.
Cyclic Compound Intermediate 89c cyclo-(D-Val-NMeArg-Gly-Asp-aminomethylnaphthoic acid); the compound of formula (IX) wherein J = D- Val, K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H
The title compound was prepared using the general procedure described for cyclo- (D-Val-
NMeArg-Gly-Asp-Mamb) (Cyclic Compound Intermediate 4). The DCC/DMAP method was used for attachment of Boc-aminomethyl-naphthoic acid to the oxime resin. The peptide was prepared on a 0.737 mmol scale to give the protected cyclic peptide (463 mg, 73.1%). The peptide (463 mg) and 0.463 mL of anisole were treated with anhydrous hydrogen fluoride at 0°C for 20 minutes. The crude material was precipitated with ether, redissolved in aqueous acetonitrile, and lyophilized to generate the title compound (349 mg, greater than quantitative yield; calculated as the fluoride salt). Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.5 cm) using a 0.45%/ min. gradient of 4.5 to 22.5% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy white solid (12.1% recovery, overall yield 7.8%); FAB-MS: [M+H] = 625.32. Synthesis of Linker Modified Cyclic Compound
Intermediates Linker modified cyclic compound intermediates can be synthesized either by incorporating an appropriately protected linker into a cyclizing moiety and then synthesizing the linker modified cyclic compound intermediate or by attaching the linker to a cyclic compound intermediate.
Linker Modified Cyclizing Moieties
Linker modified cyclizing moieties can be
synthesized either by attaching the linker to a ring substituted cyclizing moiety synthesized as described above or by incorporating an appropriately protected linker into the synthesis of the cyclizing moiety. For example, the ring substituted cyclizing moiety described above where X = NH2 can be reacted with the succinimidyl linker, RCOOSu (R = -(CH2)5-NH2 or CH2- C6H5-p-NH2), to give a linker attached at position X via an amide group.
Figure imgf000249_0001
a] Boc-ON b] RCOOSu The ring substituted cyclizing moiety with X = OH can be reacted with a linker derived from tetraethylene glycol. This linker consists of four ethylene units separated by ether groups, and bearing a Z-protected amine group at one end of the tether, and a leaving group such as tosylate at the other end of the tether. This will give a linker attached at position X via an ether group.
The ring substituted cyclizing moiety with Z = NH2 can be reacted with (Z-NH(CH2)5CO)2O to give a linker attached at position Z via an amide group.
Figure imgf000250_0001
Linkers can be attached to the ring substituted cyclizing moiety with Z = OH. Attachment of the linkers to the ring will require the linker having a leaving group suitable for reaction with a phenolate ion. Such leaving groups include halides, aryl sulfonates (e.g., tosylate) and alkyl tosylates (e.g., mesylate). For example, an alkyl chain bearing a tosyl group at one end of the chain and a protected amine at the other end is used. The literature provides several examples of alkylation at a phenolic group in the presence of a carboxylic acid group (See, for example Brockmann, Kluge, and Muxfeldt (1957), Ber . Deutsch . Chem . Ges . , 90, 2302.
Figure imgf000250_0002
The ring substituted cyclizing moiety with Z = CH2NH2 can be reacted with Z-NH(CH2)n-COOSu to give linkers attached at position Z via an amidomethyl group.
Figure imgf000251_0001
The previous examples have demonstrated the use of linkers which terminate in a protected amine. Linkers that terminate in a carboxylic acid or ester groups may also be desirable. Several such linkers can be attached to the cyclizing moieties described above. For example, in the following scheme, t-Boc protected 3- aminomethyl-4-hydroxybenzoic acid is treated with benzyl chloroacetate and base to introduce a short linker terminating in an ester.
Figure imgf000251_0002
A linker can be attached to the ring substituted cyclizing moiety where Y = NH2. As shown in Scheme 8, hydrolysis of the methyl ester of t-Boc protected methyl 3-aminomethyl-5-aminobenzoate under mild base
conditions, followed by treatment with benzyl acrylate (Lancaster Synthesis, Inc.) and acetic acid catalyst would produce the Michael addition product. Even though this linker modifed cyclizing moiety contains an
unprotected secondary amine, it could be used directly in a solid phase synthesis. However, amine protection, if desired, could be accomplished by treatment with benzyl chloroformate and a mild base.
Figure imgf000252_0001
Scheme 8
The linker can also be incorporated into the synthesis of the cyclizing moieties . One example is the synthesis of linker modified cyclizing moiety 5-Aca- Mamb .
Synthesis of Boc-Mamb (Z-5-Aca)
This synthesis is depicted in Scheme 9, below.
Part A - Methyl 3-Nitro-5-hydroxymethylbenzoate
To a solution of monomethyl 3-nitroisophthalate
(396.0 g, 1.76 mol) in anhydrous THF (1000 ml) was added 2.0 M BMS (borane methylsulfide complex) in THF (880 ml, 1.76 mol) dropwise over 1 hour. The resulting solution was heated to reflux for 12 hours, and MeOH (750 ml) was slowly added to quench the reaction. The solution was concentrated to give a yellow solid which was
recrystalized from toluene (297.5 g, 80%). 1H NMR (CDCI3) : 8.71-8.70 (m, 1H), 8.41-8.40 (m, 1H), 8.31-8.30 (m, 1H), 4.86 (s, 2H), 3.96 (s, 3H), 2.47 (s, 1H) ; MP = 76.5-77.5°C; DCI-MS: [M+H] = 212. Part B - 3-Carbomethoxy-5-nitrobenzyl Methanesulfonate Methyl 3-nitro-5-hydroxymethylbenzoate (296.0 g, 1.40 mol) and proton sponge (360.8 g, 1.68 mol) were dissolved in ethylene dichloride (150 ml). Triflic anhydride (292.3 g, 1.68 mol) dissolved in ethylene dichloride (800 ml) was added dropwise to the suspension over 90 minutes and the mixture allowed to stir 18 hour under nitrogen. The reaction was quenched with H2O
(2000 ml), the two layers were separated, and the organic layer was washed with 1000 ml portions of 1 N HCl, H2O, saturated NaHCO3, H2O, and saturated NaCl. The organic layer was dried (MgSO4) and concentrated under reduced pressure. The resulting yellow solid was recrystalized from toluene to give the title compound as a tan solid (366.8 g, 91%). 1H NMR (CDCI3) : 8.84-8.85 (m, 1H), 8.45-8.46 (m, 1H), 8.40-8.39 (m, 1H), 5.35 (s, 2H), 3.98 (s, 3H), 3.10 (s, 3H); MP = 96-97°C; DCI-MS: [M+NH4] = 307.
Part C - Methyl 3-Azidomethyl-5-nitrobenzoate
3-Carbomethoxy-5-nitrobenzyl methanesulfonate
(300.0 g, 1.04 mol) and sodium azide (81.0 g, 1.25 mol) were suspended in DMF (1700 ml) and stirred at room temperature for 5 hours. The reaction was diluted with ethyl acetate (2000 ml), washed with 1000 ml portions of H2O (2X) and saturated NaCl (1X), dried (MgSO4), and concentrated under reduced pressure. The resulting amber syrup was dried under vacuum at 40°C to yield the title compound as a tan solid (226.5 g, 92%). 1H NMR ( CDCI3 ) : 8 . 60 ( s , 1H ) , 8 . 26 ( s , 1H ) , 8 . 20 ( s , 1H ) , 4 . 52 ( s , 2H ) , 3 . 88 ( s , 3H) ; MP = 44-46°C .
Part D - Methyl 3-Amino-5-aminomethylbenzoate
A solution of Methyl 3-Azidomethyl-5-nitrobenzoate (15.50 g, 65.7 mmol) and benzene sulfonic acid (22.14 g, 140 mmol) in warm methanol (320 ml) was placed in a Parr shaker bottle and purged with nitrogen for 15 minutes. Palladium on carbon catalyst (10% Pd/C, 4.0 g) was added and the shaker bottle was further purged with 7
pressurization-evacuation cycles, repressurized, and allowed to shake 18 hours, during which time the
required amount of hydrogen was consumed. The catalyst was removed by filtration through a bed of Celite and the filtrate was concentrated under reduced pressure yielding a tan oil. Trituration with refluxing EtOAc (2 X 150 ml) followed by cooling 12 hours at -5°C gave a tan solid which was collected by filtration, washed with EtOAc (2 X 50 ml) and dried under vacuum (25.82 g, 80%). 1H NMR (CD3OD) : 8.25-8.23 (m, 1H), 8.07-8.06 (m, 1H),
7.86-7.80 (m, 5H), 7.49-7.42 (m, 6H), 4.29 (s, 2H), 3.97 (s, 3H).
Part E - Methyl 3-Amino-5-(t-butoxycarbonylamino)- methylbenzoate
A solution of methyl 3-amino-5-aminomethylbenzoate (19.32 g, 39.0 mmol), TEA (7.89 g, 78.0 mmol), and di-t- butyl dicarbonate (8.51 g, 39.0 mmol) in MeOH (350 ml) was allowed to react 24 hours at room temperature and concentrated to yield a colorless solid. Purification by flash chromatography (silica gel; 1:1 hexane :EtOAc) gave the product (9.21 g, 84%) as a colorless solid. 1H NMR (CD3OD) : 7.26-7.25 (m, 2H), 6.86-6.85 (m, 1H), 4.16 ( s , 2H) , 3 . 88 ( s , 3H) , 1 . 48 ( s , 9H) ; MP = 57- 65°C . ESI- MS : [M+H ] = 281 .
Part F - Boc-Mamb (Z-5-Aca)-OMe
N-CBZ-e-aminocaproic acid (7.77 g, 29.3 mmol) and TEA (2.97 g, 29.3 mmol) were dissolved in anhydrous THF (250 ml) and cooled to -20°C. Isobutylchloroformate (4.00 g, 29.3 mmol) was added dropwise and the mixture allowed to react for 5 minutes at -20°C. Methyl 3- Amino-5-(t-butoxycarbonylamino)methylbenzoate (8.20 g,
29.3 mmol) dissolved in anhydrous THF (50 ml) was cooled to -20°C and added to the reaction. The reaction mixture was allowed to slowly warm to room temperatures and was stirred for an additional 2 days. The solids were removed by filtration and the filtrate was
concentrated under reduced pressure. The resulting residue was dissolved in EtOAc (125 ml) and washed with two 50 ml portions each of 0.2 N HCl, saturated NaHCO3, and saturated NaCl. The organic layer was dried (MgSO4) and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel; 1:2 hexane : EtOAc), and recrystallization from CCl4 to give the title compound (10.09 g, 65%) as a colorless solid. 1H NMR (CDCI3) : 8.03-7.63 (m, 3H), 7.32-7.28 (m, 5H), 5.12-4.92 (m, 4H), 4.27-4.25 (m, 2H), 3.85 (s, 3H), 3.17-3.12 (m, 2H), 2.34-2.28 (m, 2H), 1.72-1.66 (m, 2H), 1.48-1.53 (m, 2H), 1.43 (s, 9H), 1.36-1.34 (m, 2H); MP = 52-54°C. ESI-MS: [M+H] = 528. Part G - Boc-Mamb (Z-5-Aca)
Boc-Mamb (Z-5-Aca) -OMe (22.58 g, 43.0 mmol) was dissolved in 1 : 1 1 N NaOH:MeOH (500 ml) and allowed to stir 18 hours at room temperature. The reaction was partitioned between EtOAc (300 ml) and H2O (200 ml) and the two layers were separated. The pH of the aqueous layer was lowered to 4.5, and the resulting oily precipitate was extracted into EtOAc (2 X 300 ml). The organic extract was dried (MgSO4) and concentrated to a yellow solid. The solid was triturated with refluxing CCI4 (3 X 100 ml) to give the product (14.17 g, 64%) as a colorless solid. 1H NMR (CD3OD) : 8.04 (s, 1H), 7.71- 7.66 (m, 2H), 7.30-7.23 (m, 5H), 5.02 (s, 2H), 4.24 (s, 2H), 3.32 (s, 3H), 3.11 (t, J = 6.8 Hz., 2H), 2.34 (t, J = 6.8 Hz, 2H), 1.74-1.35 (m, 15H); MP = 168-169°C. DCI- MS: [M+NH4] = 531.
Figure imgf000256_0001
Scheme 10 teaches how a linker attached to the cyclizing moiety via a reverse amide functional group can also be synthesized. Reduction of the nitro group of monomethyl 3-nitroisophthalate (Fluka) using palladium on carbon would give monomethyl 3- aminoisophthalate, which can be converted to the corresponding nitrile by the Sandmeyer procedure.
Treatment of this ester with a mono-protected diamine would yield the corresponding amide. The protecting group on the diamine must be stable to hydrogenation conditions. The Scheme demonstrates the used of the Teoc (2-trimethylsilylethyloxycarbonyl) group, but others familiar to those skilled in the art can also be used. Reduction of the nitrile using palladium on carbon would give the linker modified cyclizing moiety.
Figure imgf000258_0001
Linkers attached at position Y of the ring
substituted cyclizing moieties via an ether linkage can be synthesized, starting from 3-hydroxy-5-aminobenzoic acid. A Sandmeyer reaction can be used to convert the amine to a 3-hydroxy-5-cyanobenzoic acid. Alkyklation as above introduces the linker. Reduction of the nitrile using palladium on carbon catalyst would provide the aminomethyl group. Protection of the amine with the t-Boc group using di-t-butyl dicarbonate would provide linker modified cyclizing moieties ready for use in a solid phase synthesis. This is shown in Scheme 11.
Figure imgf000259_0001
h Linkers terminating in a carboxylic acid group can be synthesized using cyclic anhydrides. Scheme 12 illustrates such a synthesis using succinic anhydride. Reaction of t-Boc protected methyl 3-aminomethyl-5- aminobenzoate with succinic anhydride would give the carboxylic acid linker. Activation of the carboxylic acid and condensation with benzyl carbazate (Lancaster Synthesis, Inc.) would give the protected hydrazide. This hydrazide serves to protect the carboxylic acid during the remainder of the synthesis. Hydrolysis of the methyl ester provides the linker modified cyclizing moiety in a form ready to be used in the solid phase synthesis. After synthesis is complete, removal of the Cbz protecting group from the hydrazide opens the way for the preparation of an azide and azide coupling to the chelator (Hofmann, Magee, and Lindenmann (1950) J. Amer. Chem . Soc , 72, 2814). This is shown in Scheme 12.
Figure imgf000260_0001
Linkers can also be incorporated into the syntheses of alternate cyclizing moieties. For example, a linker modified heterocyclic cyclizing moiety can be
synthesized from 4-amino-6-carbethoxy-l- hydroxymethylpyrimidine (Boger (1994), J. Amer. Chem . Soc , 116, 82-92). The alcohol would be converted to the amine in three steps. First, treatment with toluenesulfonyl chloride and base would give the tosylate, which on treatment with sodium azide would give the azide. Reduction of the azide over palladium on carbon catalyst would yield the diamine. The large difference in nucleophilicity of the two amines will allow the selective protection of the aminomethyl group using di-t-butyl dicarbonate. Attachment of a protected linker, such as Z-5-Aca, to the remaining amine would be accomplished using mixed anhydride or symmetrical anhydride chemistry. Finally, hydrolysis of the ethyl ester would give the linker modified heterocyclic cyclizing moiety ready to be coupled to solid phase synthesis resin. This is shown in Scheme 13.
H ch
Figure imgf000261_0001
The preparation of the tetraethylene glycol tether discussed above is shown in Scheme 14. The synthesis begins with 1-amino-11-azido-3,6,9-trioxaundecane
(Bertozzi and Bednarski (1990), J. Org . Chem . , 56, 4326- 4329). Reduction of the azide with palladium on carbon catalyst gives the amine, which is protected with the Cbz group (designated as "Z" in Scheme 14, and
thereafter). The alcohol is now converted to the tosylate using toluenesulsonyl chloride and base.
1) H2, Pd/C
N3-(CH2CH2O)4-H
Figure imgf000262_0001
Z-NH-(CH2CH2O)4-H
2) Z-Cl, Et3N
Ts-Cl, Et3N
Figure imgf000262_0002
Z-NH-(CH2CH2O)4-Ts
Scheme 14 A second type of linker composed of ethylene glycol units is shown in the next Scheme. This linker bears a carboxylic acid group on one end, allowing it to be attached to cyclizing moieties containing amine
functional groups. The synthesis begins with the Cbzprotected amino alcohol described above. Treatment of the alcohol with ethyl diazoacetate and rhodium (II) acetate dimer would give the e glycolic acid ester having the tetraethylene glycol tail. Hydrolysis of the ethyl ester would provide the linker ready to be coupled to the cyclizing moiety. This is shown in Scheme 15.
N2CH2CO2Et
Z-NH-(CH2CH2O)4-H
Figure imgf000262_0004
Z-NH-(CH2CH2O)4-CH2CO2Et
Rh2(OAc)4
NaOH, H2O
Figure imgf000262_0003
Z-NH-(CH2CH2O)4-CH2CO2H
Scheme 15 As taught below, these linker modified cyclizing moieties can be used to synthesize linker modified cyclic compound intermediates. Linker Modified Cyclic Compound 1
Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb (5-Aca))
The synthesis of the title compound is depicted in Scheme 16, shown below.
To a 60 ml peptide reaction vessel was added oxime resin (1.61 g, substitution level = 0.62 mmol/g). The resin was swelled by washing once with DMF (30 ml). To the reaction vessel was added Boc-Mamb (Z-5-Aca) (513 mg, 1.0 mmol), HBTU (379 mg, 1.0 mmol), and DIEA (0.52 ml, 3 mmol). The suspension was mixed at room temperature for 96 hr. The resin was washed thoroughly with 30 ml portions of DMF (3X), MeOH (IX), DCM (3X), MeOH (2X), and DCM (3X). The substitution level was determined to be 0.381 mmol/g by the picric acid test. Unreacted oxime groups were blocked by treatment with 30 ml of 0:5 M trimethylacetylchloride/0.5 M DIEA in DMF for 2 hours.
The following steps were then performed: (Step 1) The resin was washed with 30 ml portions of DMF (3X), MeOH (1X), DCM (3X), MeOH (2X), and DCM (3X). (Step 2) The resin was washed with 30 ml of 50% TFA in DCM, and the t-Boc group was deprotected using 30 ml of 50% TFA in DCM for 30 minutes. (Step 3) The resin was washed thoroughly with DCM (3X), MeOH (1X), DCM (2X), MeOH (3X), and DMF (3X). (Step 4) Boc-Asp (OBzl) (0.982 g, 3.04 mmol), HBTU (1.153 g, 3.04 mmol), DIEA (1.59 ml,
9.14 mmol), and DMF (14 ml) were added to the resin and the reaction was allowed to proceed for 22 hours. (Step 5) The completeness of the coupling reaction was monitored by the picric acid test . Steps 1-5 were repeated until the. desired sequence had been attained.
After the linear peptide was assembled, the N- terminal t-Boc group was removed first washing with 50% TFA in DCM, followed by treatment with 30 ml of 50% TFA in DCM for 30 minutes. The resin was washed thoroughly with DCM (3X), MeOH (2X), DCM (3X), and then neutralized with 30 ml portions of 10 DIEA in DCM (2 X 1 min.) The resin was washed with DCM (3X) and MeOH (3X), and dried under vacuum to give 1.965 g of brown resin. The resin was cyclized by suspending in DMF (20 ml) containing HOAc (35 μl, 0.609 mmol) and heating at 50°C for 72 hours. The resin was filtered in a scintered glass funnel and washed thoroughly with 10 ml of DMF (3X). The DMF filtrate was evaporated, and the resulting oil was redissolved in 1:1 acetonitrile :H2O (20 ml), and lyophilized to give the protected cyclic peptide (342 mg). Purification was accomplished using reversed-phase HPLC with a preparative Vydac C18 column (2.1 cm) and an isocratic mobile phase of 1:1 acetonitrile :H2O
containing 0.1% TFA. Lyophilization of the product fraction gave purified protected peptide (127 mg).
The peptide (120 mg, 0.11 mmol) was deprotected by treating with TFA (1 ml) and triflic acid (1 ml) containing anisole (0.2 ml) for three hours at -10°C. The peptide was precipitated by the addition of ether and cooling to -35°C for 1.5 hours. The peptide was collected by filtration, washed with ether, and dried. The resulting solid was dissolved in 1:1 acetone :H2O (12 ml) and the pH is adjusted to 4-6 by treatment with Bio- Rad AGl-8X acetate ion exchange resin. The resin was filtered and washed with water. The filtrate was lyophilized to give HPLC pure peptide (75 mg, overall yield 13.5%); FAB-MS: [M+H] = 703.3951.
Figure imgf000265_0001
Linker Modified Cyclic Compound 2
Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb (5-Aca))
The title compound was prepared using the general procedure described for cyclo-(D-Val-NMeArg-Gly-Asp- Mamb(5-Aca)). The peptide was prepared on a 1.35 mmol scale to give the crude cyclic protected peptide (1.05 g, 73%). The peptide (500 mg) was deprotected by treating with TFA (4 ml) and triflic acid (4 ml) containing anisole (0.8 ml) for three hours at -10°C. The peptide was precipitated by the addition of ether and cooling to -35°C for 1.5 hours. The peptide was collected by filtration, washed with ether, and dried. The resulting solid was dissolved in 1:1 acetone :H2O (50 ml) and lyophilized. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C18 column (2.1 cm) using a 0.36%/min. gradient of 9 to 18% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy colorless solid (218 mg, 69% recovery, overall yield 37%); FAB-MS: [M+H] = 689.3735.
Linker Modified Cyclic Compounds 3-8
Figure imgf000266_0001
R = -(CH2)5-NH2 or CH2-C6H5-P-NH2 X1 = 2-propyl, ethyl, or p-hydroxyphenylmethyl
Compounds cyclo (D-Val-NMeArg-Gly-Asp-Mamb (4-NHCOR), cyclo (D-Abu-NMeArg-Gly-Asp-Mamb(4-NHCOR), and cyclo (D- Tyr-NMeArg-Gly-Asp-Mamb (4-NHCOR) can be prepared via the procedure described above.
Linkers can be incorporated into the synthesis of cyclic compound intermediates.
Linker Modified Cyclic Compounds 9,10 and 11
Figure imgf000267_0001
X = CH2CH2, CH2CH2CH2, CH2CH2CH2CH2
Cyclo (O-2-aminoethyl-D-Tyr)-NMeArg-Gly-Asp-Mamb),
Cyclo (O-3-aminopropyl-D-Tyr)-NMeArg-Gly-Asp-Mamb), Cyclo (O-4-amino-butyl-D-Tyr)-NMeArg-Gly-Asp-Mamb):
These compounds can be prepared using the procedure described above for Cyclo (D-Tyr-NMeArg-Gly-Asp-Mamb) using linker modified D-Tyr. The O-derivatized D-Tyr can be prepared' via the alkylation of boc-D-Tyr with the aminoprotected 2-bromoethylamine (or 3-bromopropylamine, 4-bromobutylamine) in the presence of a base.
Linkers can also be attached to cyclic compound intermediates. Linker Modified Cyclic Compound 12
Cyclo-(D-Lys(5-Aca)-NMeArg-Gly-Asp-Mamb) The preparation of the title compound is depicted in Scheme 17, shown below.
A solution of cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) (100 mg, 0.12 mmol), Boc-5-aminocaproic acid
hydroxysuccinimide ester (47 mg, 0.144 mmol), and Et3N (50 μl, 0.36 mmol) in DMF (1.50 ml) was allowed to react at room temperature for 60 minutes. The progress of the reaction was monitored by normal phase TLC (90:8:2 CHCl3:MeOH:HOAc) using the ninhydrin and Sakaguchi tests. The DMF was removed under reduced pressure. The crude conjugate was treated with TFA (3 ml) at room temperature for 45 minutes to remove the t-Boc
protecting group. The TFA was removed under reduced pressure and the conjugate was purified using reversed- phase HPLC with a preparative Vydac C18 column (2.1 cm) using 6% acetonitrile containing 0.1% TFA for 20 minutes, followed by a 3.0%/min. gradient of 6 to 36% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy colorless solid (80 mg, 70%); FAB-MS: [M+H] =
Figure imgf000269_0001
Linker Modified Cyclic Compound 13
Cyclo-([3-(4-hydroxyphenyl)propyl-D-Lys]-NMeArg-Gly-Asp- Mamb)
A solution of N-succinimidyl-3-(4-hydroxyphenyl)- propionate (Bolton-Hunter reagent; 0.022 g, 0.08 mmol) and DIEA (0.02 ml, 0.10 mmol) in dioxane (5 ml) was added to a solution of cyclo [D-Lys-N-MeArg-Gly-Asp-MAMB] (0.026 g, 0.04 mmol) in pH 9 phosphate buffer (5 ml) and the reaction was allowed to stir for 2 days at room temperature. The solution was lyophilized and the resulting white solid was purified by reversed-phase preparative HPLC on a Vydac C-18 column (2.1 cm) using a 0.36%/min. gradient of 9 to 18% acetonitrile containing 0.1% TFA to give the product (0.018 g, 60%) as a colorless solid. MP = 146-155°C; ESI-MS: [M] = 751
Linker Modified Cyclic Compound 14 Cyclo ((N-E-Tyr-D-Lys)-NMeArg-Gly-Asp-Mamb)
Figure imgf000270_0001
The desired compound can be prepared from the reaction of Cyclo (D-Lys-NMeArg-Gly-Asp-Mamb) with boc- Tyr-OSu in a solvent such as DMF in the presence of a base such as triethylamine, followed by deprotection.
Linker Modified Cyclic Compound 15 Cyclo ((N-E-(4-aminophenylacetyl)-D-Lys)-NMeArg-Gly-Asp- Mamb)
Figure imgf000271_0001
The desired compound can be prepared from the reaction of Cyclo (D-Lys-NMeArg-Gly-Asp-Mamb) with succinimidyl fmoc-4-aminophenylacetate in a solvent such as DMF in the presence of a base such as triethylamine, followed by deprotection.
Linker Modified Cyclic Compound 16 Cyclo ( (N-E- ( 4-amino-2-hydroxybenzoyl ) -D-Lys ) -NMeArg-Gly- Asp-Mamb)
Figure imgf000271_0002
The desired compound can be prepared from the reaction of Cyclo (D-Lys-NMeArg-Gly-Asp-Mamb) with succimidyl 4-amino-2-hydroxybenzoate in a solvent such as DMF or THF in the presence of a base such as
triethylamine.
A variety of linker modifed cyclic compounds can be synthesized using bifunctional cross-linking reagents developed for the derivatization of proteins. These reagents consist of two electrophilic groups, such as active esters or isocyanates, separated by a spacer. The reagents can be homobifunctional, meaning that the two reactive groups are identical, or
heterobifunctional. The spacer can be aliphatic or aromatic and may contain additional functionality to modify the lipophilicity of the conjugates, or to allow cleavage of the chain. The following examples will illustrate the use of several commercially available cross-linking reagents using as a starting point a cyclic compound intermediate synthesized with the 4- aminomethyl Mamb unit. In the first example, the cyclic compound is treated with an excess of DSS (disuccinimidyl suberate, Pierce Chemical Co.) in either aqueous or organic solvent at a pH of between 7 and 9. These are typical reaction conditions for these cross-linking reagents. The excess of cross-linker minimizes the amount of dimeric species formed. The pH of 7-9 allows the amine to react at a reasonable rate but does not produce any appreciable hydrolysis of the second reactive group and prevents reaction with the guanidino group on arginine. The active ester at the end of the linker is stable enough to allow purification by HPLC or flash
chromatography. Once purified, the linker modified cyclic compound can be conjugated to a chelator containing a nucleophilic group, such as an amine or thiol. This is depicted in Scheme 18.
Figure imgf000273_0001
Heterobifunctional reagents are typically used to achieve very selective activatation of peptides and proteins. In the following example SMPB (succinimidyl 4-(p-maleimidophenyl) butyrate, Pierce Chemical Co.) is used to modify an amine-containing cyclic compound and prepare it for coupling to a thiol-containing chelator. Treatment of the cyclic compound with SMPB under slightly basic conditions gives the linker modified cyclic compound in which the linker terminates in a maleimido group. Selectivity is achieved because the maleimido group shows low reactivity towards amine groups, and dimerization is minimized. After
purification, the maleimido group can be coupled to a thiol-containing chelator. This is depicted in Scheme 19.
Figure imgf000274_0001
Linkers containing interior functional groups can be prepared with the reagents shown in Scheme 20. EGS (ethylene glycolbis (succinimidylsuccinimidate), Sigma Chemical Co.) is a bis-succinimidyl ester which reacts preferentially with amines. Dimethyl 3,3'- dithiobispropionimidate (DTBP, also called the Wang and Richards reagent, Pierce Chemical Co.) also reacts preferentially with amines. The disulfide is cleaved by thiols. Meares and coworkers have shown ( Int . J.
Cancer : Supplement 2, 1988, 99-102) that 111In labeled antibody-chelate conjugates joined by a disulfide- containing linker show more rapid clearance of
radioactivity from mice than conjugates which did not contain a cleavable linker. The third example of Scheme 20 demonstrates the use of BSOCOES (bis [2- (succinimidooxycarbonyloxy) ethyl] sulfone, Pierce
Chemical Co.), a homobifunctional cross-linker which contains an interior sulfone group. This reagent produces a carbamate group on conjugation with an amine,
Figure imgf000275_0001
Scheme 21 illustrates the use of bisisocyanates and bisisothiocyanates in the preparation of linker modified cyclic compounds. These reagents react with amines to for urea and thiourea groups, respectively. The reagents would be used in excess to minimize the formation of dimers. The isocyanate and isothiocyanate groups at the end of the linkers are sufficiently stable to allow purification of the products.
Figure imgf000276_0001
Chelators
The present invention also provides novel reagents useful for the preparation of radiopharmaceuticals. These reagents consist of a chelator, Ch, attached via a linking group, Ln, to a cyclic compound intermediate, Q. These reagents can be synthesized in several ways, either by attaching a chelator to a linker modified cyclic compound intermediate or by attaching a chelator bearing a linking group to the cyclic compound
intermediate. Preferably, the chelator is attached to linker modified cyclic compound intermediate. Any chelator can be used in this invention provided it forms a stable complex to a radioactive isotope.
Typically the radioactive isotope is a metal or
transition metal and the complex with the chelator is a metal chelate complex. Examples of metal chelate complexes can be found in a recent review (S. Jurisson et. al., Chem Rev., 1993, 93, 1137-1156) herein
incorporated by reference.
The chelators can be attached to the linkers by a variety of means known to those skilled in the art. In general, a reactive group on the linker can react with the chelator or alternatively a reactive group on the chelator can react with the linker. Suitable reactive groups include active esters, isothiocyanates, alkyl and aryl halides, amines, thiols, hydrazines, maleimides, and the like. Several linker modified cyclic compounds bearing reactive groups are described in the examples below . Representative chelators include:
diethylenetriamine- pentaacetic acid (DTPA),
ethylenediamine-tetraacetic acid (EDTA), 1,4,7,10- tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA), 1,4,7,10-tetraaza-cyclododecane-N,N',N''- triacetic acid, hydroxybenzyl-ethylene-diamine diacetic acid, N,N'-bis (pyridoxyl- 5-phosphate) ethylene diamine, N,N'-diacetate, 3,6,9-triaza-12- oxa-3,6,9- tricarboxymethylene-10-carboxy-13-phenyl-tridecanoic acid, 1,4,7-triazacyclononane-N,N',N"'-triacetic acid, 1,4,8,11- tetraazacyclo-tetradecane-N,N'N'',N'''- tetraacetic acid, 2,3-bis(S- benzoyl) mercaptoacetamido- propanoic acid and the chelators described below. Other chelators may include metal binding regions derived from metal binding proteins such as, for example,
metallothionmes which are sulfhydryl-rich cytoplasmic proteins present in vertebrates, invertebrates and fungi. Synthesis of Chelators
Synthesis of 4,5 bis((S- benzoyl)mercaptoacetamido)pentanoic acid (mapt). The chelator was synthesized as described in
Fritzberg et. al., Appl. Radiat. Isot. 1991, 42, 525- 530.
Synthesis of (S- benzoyl)mercaptoacetylglycylglycylglycine (MAG3)
The chelator was synthesized as described in
Brandau, W. et al., Appl. Radiat . Isot . 1988, 39, 121- 129.
Synthesis of Succinimidyl 6-Boc-hydrazinopyridine-3- carboxylate (SHNH) The chelator was synthesized as described in
Schwartz et. al., 1990, European Patent Application 90301949.5. Synthesis of N-[4-(Carboxy)benzyl]-N,N'-bis[(2- triphenylmethylthio)ethyl]glycinamide N- hydroxysuccinimide ester
The synthesis of the title compound is depicted below in Scheme 22.
Part A - S-Triphenylmethyl-2-aminoethanethiol
A solution of cysteamine hydrochloride (79.5 g, 0.7 mol) in TFA (500 ml) was treated with triphenylmethanol (182 g, 0.7 mol), and stirred at room temperature for one hour. TFA was removed under reduced pressure at a temperature of 45°C and the resulting dark orange oil was dissolved in EtOAc (700 ml). The EtOAc solution was washed with cold 2N NaOH (3 X 350 ml), H2O (2 X 350 ml), saturated NaHCO3 (350 ml), and saturated NaCl (350 ml). The combined aqueous washings were back extracted with EtOAc (350 ml). The combined organic layers were dried (MgSO4) and concentrated to a yellow solid. Trituration with ether (500 ml) gave product (97.2 g, 43%) as a colorless solid, MP 90-92°C (D. Brenner et al., J.
Inorg. Chem. 1984, 23, 3793-3797, MP 93-94°C).
Concentration of the ether triturant to a volume of 100 ml and cooling produced an additional 40.9 g of product, MP 89-91°C, for a combined yield of 62%.
Part B - N-2-Bromoacetyl-S-triphenylmethyl-2- aminoethanethiol
A solution S-triphenylmethyl-2-aminoethanethiol (48 g, 0.15 mol) and Et3N (20.9 ml, 0.15 mol) in DCM (180 ml) was slowly added to a stirred solution of
bromoacetyl bromide (13.9 ml, 0.15 mol) in DCM (100 ml) at a temperature of -20°C. The reaction was allowed to warm to room temperature over a one hour period. The reaction was washed with 500 ml portions of H2O, 0.2 N HCl, saturated NaHCO3, and saturated NaCl. The organic solution was dried (MgSO4) and concentrated to an oil. This oil was crystallized from DCM-hexane to give product (54.9 g, 83%) as a colorless solid, MP 137- 139.5°C (J.A. Wolff, Ph.D. Thesis, Massachusetts
Institute of Technology, February 1992, MP 130-135°C.
Part C - N,N'-Bis [(2- triphenylmethylthio)ethyl]glycinamide
A solution of N-2-Bromoacetyl-S-triphenylmethyl-2- aminoethanethiol (35.2 g, 0.08 mol), S-triphenylmethyl- 2-aminoethanethiol (25.5 g, 0.08 mol), and Et3N (16.7 ml, 0.12 mol) in DCM (375 ml) was kept at room
temperature for 24 hours. The solution was washed with 200 ml portions of H2O (1X), saturated NaHCO3 (2X), H2O (1X), and saturated NaCl (1X), dried (MgSO4), and concentrated to give a viscous oil. The oil was dissolved in 70:30 DCM:EtOAc (150 ml) and cooled in an ice bath. The solid which formed was removed by filtration. The filtrate was concentrated to a viscous oil. This oil was purified by flash chromatography over 200-400 mesh, 60A silica gel using 70:30 DCM:EtOAc mobile phase to give product (34.4 g, 63%) as a
colorless, amorphous foamy solid. 1H NMR (CDCI3) 7.42- 7.18 (m, 30H), 3.12-3.01 (m, 4H), 2.48-2.27 (m, 6H).
Part D - Methvl 4-(Methanesulfonylmethyl) benzoate
A solution of methyl 4-(hydroxymethyl) benzoate (10.8 g, 0.065 mol) and proton sponge (19.5 g, 0.091 mol) in DCM (200 ml) was treated with methanesulfonic anhydride (13.94 g, 0.08 mol) and stirred at room temperature for 20 hours. The reaction mixture was washed with 100 ml portions of H2O (1X), 1N HCl (2X), H2O (1X), saturated NaHCO3 (1X), and H2O (1X). The organic phase was dried (MgSO4) and concentrated to give 15.5 g of pale yellow solid. Recrystallization from CCI4 (150 ml) using decolorizing carbon gave product (14.2 g, 90%) as colorless needles, MP 91-94°C.
Part E - N-[4-(Carbomethoxy)benzyn-N,N'-bisr(2- triphenylmethylthio)ethyl]glycinamide
A solution of N,N'-Bis[(2-triphenyl- methylthio)ethyl]glycinamide (16.27 g, 0.024 mol) and methyl 4- (methanesulfonylmethyl) benzoate (4.88 g, 0.02 mol) in ethylene dichloride (200 ml) was heated to reflux for 28 hours. The reaction was washed with 200 ml portions of saturated NaHCO3 and H2O, dried (MgSO4), and concentrated to a light brown oil (30 g). This oil was purified by flash chromatography over 200-400 mesh, 60A silica gel using DCM:EtOAc mobile phase to give product (9.9 g, 60%) as a colorless, amorphous foamy solid. 1H NMR (CDCI3) 7.90 (d, 2H, J = 6.5 Hz), 7.49- 7.18 (m, 32H), 3.91 (s, 3H), 3.47 (s, 2H), 3.01 (q, 2H, J = 6.2 Hz), 2.88 (s, 2H), 2.43 (t, 2H, J = 6.2 Hz), 2.39-2.27 (m, 4'H).
Part F - N-[4-(Carboxy)benzyl]-N,N'-bisr (2-triphenyl- methylthio)ethyl]glycinamide
A mixture of N-[4-(carbomethoxy)benzyl]-N,N'- bis[(2-triphenylmethylthio)ethyl]glycinamide (6.00 g, 7.26 mmol) in dioxane (65 ml) and IN NaOH (65 ml) was stirred at room temperature for 24 hours. The mixture was acidified with 2.5 M citric acid (100 ml) and the gummy precipitate which formed was extracted into EtOAc (400 ml). The EtOAc solution was washed with H2O (3 X 200 ml) and saturated NaCl (100 ml), dried (MgSO4), and concentrated to give product (5.90 g, 100%) as a colorless, amorphous foamy solid. 1H NMR (CDCI3) 7.96 (d, 2H, J = 8.1 Hz), 7.40-7.16 (m, 32H), 3.71 (s, 3H), 3.49 (s, 2H), 3.00 (q, 2H, J = 5.4 Hz), 2.91 (s, 2H), 2.44 (t, 2H, J = 5.4 Hz), 2.38-2.30 (m, 4H). Part G - N-[4-(Carboxy)benzyl]-N,N'-bisr(2- triphenylmethylthio)ethyl]glycinamide N- hydroxysuccinimide ester
A solution of N-[4-(carboxy)benzyl]-N,N'-bis[(2- triphenylmethylthio)ethyl]glycinamide (450 mg, 0.55 mmol) and N-hydroxysuccinimide (76 mg, 0.66 mmol) in DCM (10 ml) was treated with a solution of WSCD•HCl (122 mg, 0.66 mmol) in DCM (7 ml) and stirred at room temperature for 22 hours. The reaction mixture was concentrated and the solids redissolved in EtOAc (60 ml). The EtOAc solution was washed with H2O (2 X 25 ml), 0.1 N NaOH (35 ml), H2O (2 X 25 ml), and saturated NaCl (35 ml), dried (Na2SO4), and concentrated to give product (469 mg, 93%) as a colorless solid.
Figure imgf000283_0001
Synthesis of N-[ 2-(Benzoylthio)propionyl]glycylglycyl-g- Amino-butyric Acid (Bz-Me-MΑG2-gaba). The title compound was prepared according to Scheme 23 from N-(2-mercaptopropionyl)-glycine (1), which is commercially available from Aldrich. The protection of the thiol group in compound 1 is achieved by reacting with benzoyi chloride under basic conditions to give compund 2. The carboxylic group can be activated by forming its succinimide ester (3), which reacts with glycyl-g-aminobutyric acid in 90% methanol solution to give the benzoyl-protected Me-MAG2-gaba (4). The spectral (IR, 1H NMR and FAB-MS) data are completely consistent with the proposed formulation.
Figure imgf000284_0001
Step 1: N-[2-(benzoylthiol)propionyl]glycine
(2). Sodium hydroxide (4.5 g, 0.109 mol) and N-(2- mercaptopropionyl) glycine (8.20 g, 0.05 mol) were dissolved in a mixture of water (40 mL) and toluene (30 mL). The temperature was lowered to 5-15 °C using an ice bath. Benzoyl chloride (4.6 mL, 0.051 mol) in toluene (10 mL) was added dropwise with vigorously stirring. After addition, the mixture was stirred at 5- 15 °C for another 30 min., and then at room temperature for 2 hr. The organic layer was separated, washed with H2O (2x20 mL), and discarded. Aqueous fractions were combined and acidified to pH ~ 1.5 using concentrated HCl while white solid formed. The precipitate was collected by filtration, washed with H2O and small amount of ethanol, and dried under vacuum. The yield was 13.0 g (97%). Anal. Calcd (found) for C12H13NO4S : C, 53.90 (53.89); H, 4.90 (4.81); N, 5.24 (5.22). IR (KBr disk, in cm-1): 3375 (s, nN_H) 3200-2500 (br, nO-H); 1745 (vs, thioester nC=O); 1663, 1625 (vs, amide and
carboxylic nC=O). 1H NMR (DMSO-d6, d in ppm): 1.47 (d, 3H, CH3, J = 7.0 Hz); 3.79 (d, 2H, CH2, J = 5.9 Hz);
4.40 (q, 1H, CH, J = 7.0 Hz); 7.53 (m, 2H, =CH); 7.69 (m, 1H, =CH); 7.90 (dd, 2H, =CH, J = 7.0 Hz); 8.59 (t, 1H, NH, J = 5.8 Hz); 12.6 (bs, 1H, COOH ). DCI-MS: m/z = 268 ( [M+H]+) .
Step 2: N-[2-(Benzoylthio)propionyl]glycine Sυccinimide Ester (3). To a suspension of N- hydroxysuccinimide (5.80 g, 0.05 mol) and N-[2- (benzoylthiol)propionyl]glycine (13.35 g, 0.05 mol) in dry THF (400 mL) was added DCC (12.0 g, 0.052 mol) in the same solvent (100 mL THF) at 5-10 °C. The mixture was stirred at 5 - 10 °C for 2hr, and then at room temperature for 2 days. To the reaction mixture was added 2-3 mL of acetic acid and then stirred for another 2 hr. The solid was filtered off, washed with 2x150 mL of THF. The organic fractions were combined and the solvent was removed under reduced pressure to give a white solid, which was collected, washed with diethyl ether, and dried in air. The yield was 14.5 g (80%). Anal. Calcd (found) for C16H16N2O6S: C, 52.72 (52.70); H 4.43 (4.21); N, 7.69 (7.69). IR (KBr disk, in cm-1): 3290 (s, nN-H); 1820 (m, succinimide nC=O); 1785 (m, ester nC=O); 1735 (vs, thioester nC=O); 1600 (vs, amide nC=O). 1H NMR (CDCl3, d in ppm): 1.57 (d, 3H, CH3, J = 7.0 Hz); 2.79 (s, 4H, CH2); 4.33 (q, 1H, CH, J = 7.0 Hz); 4.39 (m, 2H, CH2); 7.00 (t, 1H, NH, J = 5.8 Hz);
7.44 (m, 2H, =CH); 7.59 (m, 1H, =CH); 7.93 (dd, 2H, =CH, J = 7.0 Hz). DCI-MS: m/z = 365 ([M+H]+).
Step 3: N-[2- (Benzoylthio)propionyl]glycylglycyl-g-Axnino- butyric Acid (Bz-Me-MAG2-gaba, 4). N-[2-
(Benzoylthio)-propionyl]glycine succinimide ester (1.82 g, 5 mmol) and glycyl-g-aminobutyric acid (0.80 g, 5 mmol) were suspended in a mixture of methanol (150 mL) and water (30 mL). The mixture was heated to reflux for 5 hr, during which time the cloudy mixture became a clear solution. The solution was then cooled to room temperature and was kept stirring overnight.
Evaporation of solvents under reduced pressure give a white solid, which was purified by washing with water, and dried under vacuum. The yield was 1.85 g (93%). Anal. Calcd (found) for C18H23N3O6S: C, 52.78 (52.69); H, 5.66 (5.70); N, 10.27 (10.17). IR (KBr disk, in cm-1): 3380, 3320 (s, nN_H); 3100-2500 (br, nO-H); 1725 (vs, thioester nC=O); 1680, 1640, 1624 (vs, amide nC=O). 1H NMR (DMSO-d6, d in ppm): 1.49 (d, 3H, CH3, J = 7.0 Hz); 1.62 (qin, 2H, CH2, J = 7.1 Hz); 2.21 (t, 2H, CH2COOH, J = 7.5 Hz); 3.05 (qart, 2H, NH-CH2, J = 7.0 Hz); 3.67 (d, 2H, NH-CH2, J = 5.7 Hz); 3.75 (d, 2H, NH-CH2, J = 7.0 Hz); 4.42 (q, 1H, CH,
J = 7.0 Hz); 7.57 (m, 2H, =CH); 7.70 (m, 1H, -CH); 7..80 (t, 1H, NH, J = 3.0 Hz); 7.90 (dd, 2H, =CH, J = 7.0 Hz); 8.14 (t, 1H, NH, J = 5.70 Hz); 8.57 (t, 1H, NH, J = 5.90 Hz), 12.0 (bs, 1H, COOH). DCI-MS: m/z = 410
([M+H]+).
Synthesis of N-[2- (Benzoylthio)prooionyl]glycylglycylglycine (Bz-Me-MAG3)
The title compound was synthesized as described for Bz-Me-MAG2-gaba by substituting glycylglycine for glycyl-g-aminobutyric acid. The yield was 83%. Anal. Calcd (found) for C16H19N3O6S: C, 50.39 (50.59); H,
5.02(5.78); N, 11.02 (10.70). IR (KBr disk, in cm-1): 3380, 3300 (s, nN_H); 3100-2500 (br, nO-H); 1738 (vs, thioester nC=O); 1680, 1660 (vs, amide nC=O). 1H NMR (DMSO-d6, d in ppm): 1.48 (d, 3H, CH3, J = 7.05 Hz); 3.78 (m, 4H, CH2); 3.85 (d, 2H, CH2, J = 6.00 Hz); 4.41 (m, 1H, CH); 7.52 (m, 2H, =CH); 7.70 (m, 1H, =CH), 7.90 (m, 2H, =CH); 8.15 (t, 1H, NH, J = 3.00 Hz); 8.51 (t, 1H, NH, J = 3.00 Hz); 8.80 (t, 1H, NH, J = 3.00 Hz). FAB-MS: m/z = 382 ([M+H]+). ESI-MS: m/z = 381.9
([M+H]+).
Synthesis of N-[2-(Benzoylthio)propiony]glycylglycyl-4- Amino-methylcyclohexane Carboxylic Acid (Bz-Me-MAG2- ACA).
Synthesis of Bz-Me-MAG2-ACA involves several steps (Scheme 24). Compound 1 could be easily converted to its chloride 2, which reacted with 4-trans-amino- methylcyclohexane carboxylic acid to give compound 3. Deprotection of 3 using hydrazine in ethanol, followed by addition of HCl produces 4. Reaction of 4 with Bz- Me-MAG-Succ in methanol in presence of Et3N afforded Bz- Me-MAG2-ACA 5.
Figure imgf000288_0001
Step 1: Phthaloylglycyl Chloride. Phthaloylglycine (40 g) was suspended in chloroform (400 mL), followed by addition of thionyl chloride (60 mL). The mixture was heated to reflux for 2 hr, during which time the mixture became a homogeneous clear solution. The solvent and excess of thionyl chloride was removed under reduced pressure to give an off-white solid, which was dried under vacuum and used without further purification. 1H NMR was consistent with the proposed structure.
Step 2: 4-trans- [(Phthaloylglycyl)aminomethyl]cyclohexane Carboxylic Acid. Suspended were 4-trans-aminomethylcyclohexane carboxylic acid (7.85 g, 50 mmol) and K2CO3 (5 g, 50 mmol) in DMF (150 mL). To the suspension was added phthaloylglycyl chloride (11.85 g, 50 mmol) in
acetonitrile (150 mL). The reaction mixture was refluxed for 3 hr and then filtered while hot. Solvents were removed under reduced pressure to give an oil.
Upon addition of diethyl ether (50 mL), a white solide formed. The solid was collected by filtration, washed with diethyl ether, and dried in air. The yield was
10.32 g (60%). 1H NMR (in DMSO-d6, d in ppm relative to TMS) : 0.87-2.00 (m, 9H, CH2 and CH from cyclohexane ring); 2.10 (m, 1H, CHCOOH); 2.92 (t, 2H, CH2, J = 4.6 Hz); 4.19 (s, 2H, CH2); 7.85 (m, 4H, -CH=); 8.21 (t, 1H, NH, J = 4.1 Hz).
Step 3: Glycyl-4-trans-(Aminomethyl)cyclohexane Carboxylic Acid Hydrochloride (Gly-ACA-HCl). To a suspension of 4-trans- [(Phthaloylglycyl)aminomethyl]cyclohexane carboxylic acid
(10.32 g, 30 mmol) in ethanol (300 mL) was added 85% hydrazine hydrate (100 mL) . The mixture was heated to reflux for 12 hr, during which time a white precipitate formed. After solvent was removed, 2 N HCl (200 mL) was added to the residue. The mixture was warmed up to 60- 70 °C for 20 min and the solid was filtered off and discarded. The filtrate was concentrated to 1/3 of its original volume. The mixture was cooled in an ice bath for 2 hr. The precipitate was collected by filtration, washed with a small amount of water and ethanol, and dried under vacuum. The yield was 3.45 g (45%). 1H NMR (in D2O, d in ppm relative to TMS): 1.04 (m, 2H, CH2); 1.45 (m, 2H, CH2); 1.57 (m, 1H, CH), 1.81-2.05 (m, 4H, CH2); 2.35 (m, 1H, CHCOOH); 3.15 (d, 2H, CH2, J = 4.9 Hz); 3.84 (s, 2H, CH2).
Step 4: N-[2-(Benzoylthio)propiony]glycylglycyl-4- Amino-methylcyclohexane Carboxylic Acid (Bz-Me-MAG2- ACA). Gly-ACA-HCl (1.25 g, 5 mmol), Et3N (1.0 g, 10 mmol) and Bz-Me-MAG-Succ (1.82 g, 5 mmol) were suspended in a mixture of methanol (200 mL) and acetonitrile (100 mL). The mixture was refluxed overnight. Solvents were removed under reduced pressure to give a white solid residue, to which was added 6 N HCl (10 mL). The solid was separated by filtration, washed with water and small amount of ethanol, and dried under vacuum. The yield was 1.35 g (58%). Anal. Calcd (found) for C22H29N3O6S: C, 57.00 (58.41);
H, 6.31 (6.70); N, 9.06 (9.72). IR (KBr disk, in cm-1):
3600-2000 (br, OH---N); 3270 (s, nN-H); 1720, 1655, 1625, and 1565 (vs, nC=O). FAB-MS: m/z = 464 (M+1). 1H NMR (in DMSO-d6, d in ppm relative to TMS): 0.81-1.90 (m, 9H, CH2 and CH from cyclohexane ring); 1.48 (d, 3H, CH3, J = 5.2 Hz); 2.10 (t, 1H, CHCOOH, J = 9.0 Hz); 2.91 (t, 2H, CH2, J = 4.6 Hz); 3.68 (d, 2H, CH2, 4.2 Hz); 3.75 (d, 2H, CH2, J = 4.1 Hz); 4.42 (q, 1H, CH, J = 5.2 Hz); 7.50 (t, 2H, -CH=, J = 5.8 Hz); 7.71 (t, 2H, -CH=, J = 5.4 Hz); 7.91 (d, 1H, -CH=, J = 6.4 Hz); 8.14 (t, 1H, NH, J = 4.2 Hz); 8.60 (t, 1H, NH, J = 4.1 Hz), 12.00 (bs, 1H, COOH).
Synthesis of 3,4-Bis[3-(Benzoylthioacetyl)amido]benzoic Acid (Bz-MABA).
To a solution of S-benzoylthioacetyl chloride
(8.69g, 40 mmol), freshly prepared from the reaction of S-benzoylthioacetic acid with excess of thionyl chloride in chloroform, in dry THF (300 mL) was added 3,4- diaminobenzoic acid (3.04 g, 20 mmol) while the solution became brown. The solution was refluxed over night, during which time a precipitate formed. The mixture was cooled, and the solid was separated by filtration, washed with THF, ethanol and diethyl ether, and dried under vacuum to give a pale gray solid. The yield was 5.8 g (54%). Anal. Calcd (found) for C25H20N2O6S2: C, 59.04 (58.82); H, 3.96 (4.04); N, 5.51 (5.46). IR (KBr disk, in cm-1): 3600-2000 (br, OH----N); 3340 (s, nN_H);
1690, 1670, 1655, 1610 and 1595 (s or m, nC=O). FAB-MS: m/z = 509 (M+1). 1H NMR (in CDCI3, d in ppm relative to TMS): 4.12 and 4.14 (s, 4H, CH2); 7.50-8.30 (m, 13H, aromatic H's); 9.85 and 9.89 (s, 2H, NH); 12.99 (bs, 1H, COOH).
Synthesis of 2-(S- Triphenylmethylmercapto)ethylaminoacetyl-S- triphenylmethyl-L-cysteine ethyl ester (Tr2-MA-MAMA).
Figure imgf000291_0001
a: Triphenylmethanol, TFA; b: bromoacetyl bromide, TEA, THF; c: S-triphenylmethyl- 2-aminoethanethiol, TEA, methylene
chloride
Scheme 25 S-Triphenylmethyl-L-cysteine ethyl ester (2): To a solution of L-cysteine ethyl ester hydrochloride (18.6 g, 0.1 mole) in 200 mL TFA was added triphenylmethanol (52 g, 0.2 mole). The resulting dark brown solution was allowed to stir for 2 h at room temperature under nitrogen. The solvent was removed in vacuo and ethanol (100 mL) added to the residue. A I M solution of sodium ethoxide (50 mL) was added to the ethanolic solution and stirred for 90 min. during which time the solution turned cloudy. The mixture was filtered, the filtrated was concentrated in vacuo to give an oily residue.
Flash column chromatography using ethyl acetate:hexane (1:3) and ethyl acetate gave the desired product
(containing some ethyl acetate which is difficult to remove) which was stored under vacuum.
N-Bromoacetyl-S-triphenylmethyl-L-cysteine ethyl ester (3) : A solution of S-triphenylmethyl-L-cysteine ethyl ester (18 g, 46 mmol.) and triethylamine (6.4 mL, 46 mmol.) in dry THF (250 mL) under nitrogen was cooled to 0 °C. A solution of bromoacetyl bromide (9.28 g, 46 mmol.) in dry THF (60 mL) was added dropwise during which time the solution timed cloudy. The reaction mixture was stirred at 0 °C for 1 h and then at room temperature for 1 h. The reaction mixture was filtered and the filtrate was concentrated in vacuo to give an oil. The oil was partitioned between methylene chloride and water (60 mL each), the organic layer washed with 5% HCl, NaHCO3, dried (magnesium sulfate), filtered, and the volatiles removed to give the desired product (69%).
2-(S-Triphenylmethylmercapto)ethylaminoacetyl-S- triphenylmethyl-L-cysteine ethyl ester (4) : To a solution of N-bromoacetyl-S-Triphenylmethyl-L-cysteine ethyl ester (1.0 g, 1.98 mmol.) and triethylamine (0.4 mL, 2.9 mmol.) in methylene chloride (10 mL) was added S-triphenylmethyl-2-aminoethanethiol (0.64 g, 2.0 mmol.). The reaction mixture allowed to stir at room temperature for seven days. Water (10 mL) was added. The organic layer was washed with NaHCO3 (2x10 mL), water (2x10 mL), and brine (10 mL), dried (magnesium sulfate), and concentrated in vacuo to give a foamy product. Flash chromatography using ethyl
acetate :hexane (3:1) gave the product in 22% yield. MS (M+H) = 751, calculated 751.3
The synthesis of a chelator having a single carboxylic acid group availible for attaching the linker is shown in Scheme 26. The synthesis begins with the N- alkylation of Cys(Acm)OMe with bromoacetaldehyde dimethylacetal. The secondary amine of the alkylation product is now protected from further reaction with the Teoc group. Other protecting groups which are stable to both mild acid and mild base, and can be removed in the presence of sulfur may also be used. The Teoc group is introduced by the use of 2-(trimethylsilyl)ethyl p- nitrophenyl carbonate. The acetal is now hydrolyzed with mild aqueous acid and the aldehyde is reductively aminated with S-triphenylmethyl-2-aminoethanethiol. The one free amine of the chelator is protected with the Teoc group and the methyl ester is hydrolyzed with aqueous base to give the carboxylic acid ready for reaction with the reactive group of a linker modified cyclic compound.
Figure imgf000294_0001
A chelator having one additional amine available for conjugation to the linker modified cyclic compound can be synthesized according to the procedure of Scheme 27. Acm protected thioglycolic acid would be coupled to N-t-butoxycarbonylethylenediamine using any of the standard coupling methods of peptide synthesis. The Boc protecting group would be removed by the use of TFA, and the resulting amine would be coupled to Boc-Cys (Acm)-OH. Removal of the Boc protecting group provides the S- protected chelator in a form appropriate for reaction with the reactive group of a linker modified cyclic compound. u
V j
Figure imgf000295_0001
Also subject to this invention are reagents of the formula (QLn)dCh for radiolabeling which comprise more than one linker modified cyclic compound intermediate attached to a chelator as well as reagents of the formula (Q)d'Ln-Ch, having two or more cyclic compound intermediates attached to a common linker that also bears a chelator.
An example of a reagent comprising two linker modified cyclic compound intermediates attached to a chelator is shown below (Schemes 28 and 29). Other representative examples are shown in the following schemes. In this scheme, amine groups on two linker intermediate compounds react with the shown two
activated ester groups to afford a compound of this invention of formula (QLn)2Ch.
Figure imgf000296_0001
The sulfur protecting group, Pg, shown above, as well as all Pg groups claimed herein, may be any sulfur protecting group capable of being displaced upon reaction with the metal nuclide. Such protecting groups are well known by those skilled in the art. Examples of suitable protecting are taught in U.S. Patents Nos.
4,897,255, 4,965,392, and 4,980,147, each of which is herebu incorporated herein by reference.
Figure imgf000297_0001
Chelators useful in the synthesis of these reagents are described in Chervu et. al., U.S. Patent 4,883,862 and Bergstein et. al., U.S. Patent 5, 279,811. The synthesis of other useful chelators is described in the following schemes.
The following examples illustrate how three such chelators could be prepared. Scheme 30 outlines the synthesis of a N2S2 ligand having two carboxylic acid group to which the targeting cyclic compound can be conjugated. The synthesis begins with an alkylation reaction on the two amines of DL-2,3-diaminosuccinic acid (Sigma Chemical Co.), using S-triphenylmethyl-2- bromoethanethiol. The secondary amines must now be protected to avoid self-condensation when the carboxylic acids are activated. This can be accomplished with any of the standard amine protecting groups. The Z group would be a good choice because it can be removed under acidic conditions (HBr/HOAc or
TFA/trifluoromethanesulfonic acid) at the same time as the trityl protection on sulfur.
Figure imgf000298_0001
The synthesis of a second N2S2 having two
carboxylic acid groups is shown in Scheme 31.
Alkylation of ethylenediamine-N,N '-dipropionic acid (American Tokyo Kasei) with S-triphenylmethyl-2- bromoethanethiol would give the N2S2 ready for
conjugation. The amines are tertiary and no additional protection is required.
Figure imgf000299_0001
Scheme 32 outlines the synthesis of an N2S2 ligand having two additional amine groups for conjugation to targeting cyclic compounds bearing reactive
electrophilic groups (e.g., active esters) . A reductive amination reaction between benzyl amine and glyoxal would give N,N'-dibenzylethylenediamine. Alkylation of the two amines with N-(3-bromopropyl)phthalimide would give the fully protected tetraamine. The benzyl protection on the two secondary amines would be removed by catalytic reduction, and the free amines would then be alkylated with S-triphenylmethyl-2-bromoethanethiol to give the fully protected ligand. Selective
deprotection of the primary amines would be accomplished with hydrazine.
Figure imgf000300_0001
Reagents having twp targeting groups and one chelator bound to a common linker can be synthesized according to the route shown in Scheme 33. Reaction of benzylamine with N-(3-bromopropyl)phthalimide will yield N,N-bis(3-phthalimidopropyl)benzylamine (Niitsu and Samejima (1986), Chem . Pharm . Bui . , 34, 1032-1038).
Treatment with hydrazine will remove the phthalimido protecting groups. N,N-Bis (3-aminopropyl) benzylamine would then be reacted with succinic anhydride to give the diacid, which would be converted to the bis active ester with DCC and N-hydroxysuccinimide. This bis active ester would then be conjugated to a linker modified cyclic compound. Hydrogenation to remove the benzyl protecting group and conjugation with an activated chelator would yield the final product.
S
Figure imgf000301_0001
More than two compounds Q and more than one chelator can be joined together by using starburst or cascade dendrimers as linkers. Dendrimers are
constructed by adding branched segments onto a functionalized core, producing a product having twice the number of functional groups as the original core. This addition of branched units can be carried through several generations to product large polyfunctional molecules. One example is the PAMAM (polyamidoamine) dendrimers (Aldrich Chemical Co.), which use
ethylenediamine as the initiator core. Scheme 34 shows the generalized preparation of a radiopharmaceutical based on PAMAM dendrimer containing targeting cyclic compounds and chelators in a 2:1 ratio. For this structure a generation = 0 (n = 1) dendrimer would have two targeting cyclic compounds and one chelator. A generation = 1 (n = 2) dendrimer would have four targeting cyclic compounds and two dendrimers. The ratio and absolute number of targeting cyclic compounds and chelators would be controlled by the stoichiometry of the conjugation reactions.
Figure imgf000303_0001
A similar system, called the multiple antigen peptide (MAP) system was developed by Posnett, McGrath, and Tarn (J. Biol . Chem. , 263, (1988), 1719) to
facilitate the generation of antibodies. This system constructs a branching network on a solid support using the two amino groups of lysine. Because the two different amino groups on lysine can be orthogonally protected, this system allows a higher level of control of the conjugation reactions. In Scheme 35 a MAP system terminating in four lysine groups is conjugated first to four targeting cyclic compounds at the alpha amino groups, and them to four chelators at the epsilon amino groups.
Figure imgf000304_0001
Synthesis of Radiolabeled Compounds
The radiolabeled cyclic platelet glycoprotein
Ilb/IIIa compounds of the present invention can be synthesized using standard synthetic methods known to those skilled in the art, using radioisotopes of halogens (such as chlorine, fluorine, bromine and iodine), technetium and indium, as well as others.
Preferable radioisotopes include 123 l , 125l, 131l, 99rnTc, and 111In.
The cyclic platelet glycoprotein Ilb/IIIa
compounds of the invention may be labeled either directly (that is, by incorporating the radiolabel directly into the compounds) or indirectly (that is, by incorporating the radiolabel into the compounds through a chelator which has been incorporated into the compounds. For direct labeling, as those skilled in the art will recognize, the labeling may be isotopic or nonisotopic. With isotopic labeling, one group already present in the cyclic compound is substituted with (exchanged for) the radioisotope. With nonisotopic labeling, the radioisotope is added to the cyclic compounds without substituting with (exchanging for) an already existing group.
Generally, labeled compounds are prepared by procedures which introduce the labeled atom at a late stage of the synthesis. This allows for maximum radiochemical yields, and reduces the handling time of radioactive materials. When dealing with short half- life isotopes, a major consideration is the time required to conduct synthetic procedures, and
purification methods. Protocols for the synthesis of radiopharmaceuticals are described in Tubis and Wolf, Eds., "Radiopharmacy", Wiley- Interscience, New York (1976); Wolf, Christman, Fowler, Lambrecht, "Synthesis of Radiopharmaceuticals and Labeled Compounds Using Short-Lived Isotopes", in Radiopharmaceuticals and Labeled Compounds, Vol 1, p. 345-381 (1973), the disclosures of each of which are hereby incorporated herein by reference, in their entirety. Various procedures may be employed in preparing the radiolabeled compounds of the invention where the radiolabel is a halogen. Some common synthetic
methodologies for isotopic halogen labeling of aromatic compounds such as the type present here are
iododediazonization, iododeborobation,
iododestannylation, iododesilation, iododethallation, and halogen exchange reactions. The most common synthetic methodology for nonisotopic halogen labeling of aromatic compounds such as the type present here is iododeprotonation or electrophilic aromatic substitution reactions. These methods and additional procedures are described in Merkushev, Synthesis, 923 (1988), and
Seevers et al, Chem. Rev., 82: 575 (1982), the
disclosures of each of which are hereby incorporated herein by reference, in their entirety.
By way of example, isotopically radiolabeled 4, 5 and 6-halo t-butyloxycarbonyl-3-aminomethylbenzoic acid derivatives may be prepared using the general procedures described above for the synthesis of the unlabeled compounds. In carrying out such radiolabeling, it is important that the half-life of the isotope chosen be much longer than the handling time of the reaction sequences. Known starting materials include the 2, 3, and 4-iodo (123l, 125l, and 131l) benzoic acids.
The iodo-radiolabeled Mamb derivatives may also be isotopically prepared from the anilines by the Sandmeyer reaction as described in Ellis et at Aust. J. Chem., 26: 907 (1973).
Alternatively, such compounds may prepared by way of isotopic labeling from the unlabeled bromo or iodo derivatives by various two step reaction sequences, such as through the use of trialkylsilyl synthons as
described in Wilson et at J. Org. Chem., 51: 483 (1986) and Wilbur et al J. Label. Compound. Radiopharm., 19: 1171 (1982), the use of trialkylsilyl synthons as described in Chumpradit et al J. Med. Chem., 34: 877 (1991) and Chumpradit et al J. Med. Chem., 32: 1431 (1989), and the use of boronic acid synthons as described in Kabalka et al J. Label. Compound.
Radiopharm., 19: 795 (1982) and Koch et al Chem. Ber., 124:2091 (1991). These synthetic transformations are outlined in the Scheme 36 below.
Figure imgf000307_0001
Although the foregoing protocol may be employed in preparing radiolabeled compounds of the present
invention, to maximize radiochemical yields, to reduce the handling time of radioactive materials, and to prepare short half-life halogen labeled compounds, it is preferable to perform the isotopic halogen labeling as one of the final steps in the cyclic compound synthesis. The following provides exemplary proceudres for such late stage labeling.
The unlabeled iodo compounds are versatile
precursors which can be converted to the labeled
derivatives by any of the two step reaction sequences described above. Useful functionality to incorporate into the Mamb portion of the cyclic compound includes the bromo, the nitro, the trialkylsilyl, the
trialkyltin, and the boronic acid groups. The synthesis and application of each of these precursors is described above.
The least complex means of radioiodination of the cyclic compounds of the present invention via isotopic labeling during the final stages of their preparation is the substitution of radioactive iodide for a stable iodine atom already present in the molecule. This can often be done by heating the compound with radioactive iodide in an appropriate solvent as described in Ellis et al., Aust. J. Chem., 26: 907 (1973). When applied to aromatic iodides, the extremely small quantities and low concentration of radioactive iodide employed leads to the incorporation of only modest specific activity.
This reaction sequence is outlined in the Scheme e 37.
Figure imgf000309_0001
The cyclic compounds may also be isotopically iodo- labeled during the final stages of their preparation from the anilines by the Sandmeyer reaction as described in Ellis et al., Aust. J. Chem., 26: 907 (1973). This approach leads to a labeled cyclic compound with high specific activity. To avoid complications in the synthesis of the cyclic compound, the nitro group provides an ideal synthon for the aniline.
Alternatively, the cyclic compounds may be
isotopically labeled late in the reaction scheme from the unlabeled bromo or iodo derivatives by various two step reaction sequences, as described above, such as through the use of trialkylsilyl synthons as described in Wilson et al., J. Org. Chem., 51: 4833 (1986) and Wilbur et al., J. Label. Compound. Radiopharm., 19: 1171 (1982), through the use of trialkylsilyl synthons as described in Chumpradit et al., J. Med. Chem., 34: 877 (1991) and Chumpradit et al., J. Med. Chem., 32: 1431 (1989), and through the use of boronic acid synthons as described in Kabalka et al., J. Label. Compound.
Radiopharm., 19: 795 (1982) and Koch et al., Chem. Ber., 124:2091 (1991).
A related approach where the isotopic halogen radiolabeling may be carried out late in the synthesis scheme involves converting the substituted Mamb derivatives to cyclic compounds that already incorporate the trialkylsilyl, trialkyltin, or boronic acid groups. The synthesis of each Mamb derivative has been described in an earlier section.
The forgoing synthetic transformations on the cyclic compounds are outlined in the Scheme 38.
Figure imgf000311_0001
Labeled iodo derivatives may also be readily prepared nonisotopically from the amino, hydroxy, or methoxy substituted cyclic compounds as described in Arora et al J. Med. Chem., 30:918 (1987). Electrophilic aromatic substitution reactions are enhanced by the presence of such electron-donating substituents. This synthetic sequence is outlined in Schemes 39 and 40.
Figure imgf000313_0001
Figure imgf000314_0001
As an alternate approach to the incorporation of a radiolabeled halogen, the methyl substituted cyclic compounds may be converted to the a-halotoluene derivative with NBS or NCS under free-radical
halogenation conditions. The benzylic halides may be smoothly replaced by radiolabeled iodide through a nucleophilic substitution reaction. This synthetic sequence is outlined in Scheme 41.
Figure imgf000314_0002
Scheme 4 1
Although primarily illustrated for the radiolabeled iodo compounds, the above described process chemistry can be used to prepare any radioactive halogen isotope.
18F derivatives of these cyclic compounds can be prepared by conjugation of 18F functionalized phenyl intermediates. 18F-functionalized cyclic compounds can be prepared as shown in Scheme 42 (R.H. Mach et al., J. Med. Chem., 1993, 36,3707-3720). Reaction of p- trimethylammonium-benzaldehyde with [18F] CsF/aqueous DMF at 120 °C for 10 min. (aqueous [18F] KF/kryptofix/ACN can also be used to generate the 18F-phenyl compounds from the corresponding trimethylammonium or nitro groups), followed by LAH/THF/pentane and 57% aqueous HI gives the p-18F-benzyl iodide.
Figure imgf000315_0001
Scheme 42
Reaction with the amine funtionality of the cyclic compound intermediate cyclo (D-Lys-NMeArg-Gly-Asp-Mamb) or the linker modifed cyclic compound Cyclo (D-Val- NMeArg-Gly-Asp-Mamb (5-Aca)) can give the 18F labeled products suitable for use in positron emission
tomography (PET):
Figure imgf000316_0001
Various procedures may also be employed in preparing the radiolabeled compounds of the invention where the radiolabel is a metal, such as where the radiolabel is technetium or indium. These procedures are utilized for labeling compounds of this invention of formulae:
(QLn)dCh and (Q)d'Ln-Ch. Exemplary procedures for such technetium or indium labeling are disclosed, for example, in Cerqueira et al., Circulation, Vol. 85, No. 1, pp. 298-304 (1992), Pak et al., J. Nucl. Med., Vol. 30, No. 5, p. 793, 36th Ann. Meet. Soc. Nucl. Med.
(1989), Epps et al., J. Nucl. Med., Vol. 30, No. 5, p. 794, 36th Ann. Meet. Soc. Nucl. Med. (1989), Pak et al., J. Nucl. Med., Vol. 30, No. 5, p. 794, 36th Ann. Meet. Soc. Nucl. Med. (1989), and Dean et al., J. Nucl. Med., Vol. 30, No. 5, p. 794, 36th Ann. Meet. Soc. Nucl. Med. (1989), the disclosures of each of which are hereby incorporated herein by reference, in their entirety. In additon, specific procedures are provided in the
examples below.
Another useful method for labeling the cyclic compounds of the present invention involves preparing a 99mTc chelator (at the tracer level) and conjugating it to either a cyclic compound intermediate or a linker modified cyclic compound. This method is termed the prechelate approach. As shown, for example, in the scheme below, 4,5-bis(S- benzoyl)mercaptoacetamidopentanoic acid (1) is complexed with 99mTcO4 under reducing conditions to form (2).
Then (2) is converted to the active ester (3) containing the tetrafluorophenyl group. Complex (3) then may be reacted with an appropriate cyclic compound intermediate such as (5) or (6), to yield radiolabeled compounds (4). Another appropriate technetium chelator is 2,3-bis(S- benzoyl)mercaptoacetamido-propanoic acid (7). HPLC purification of the 99mTc complex may be performed at each step. This approach is depicted in Scheme 43.
Figure imgf000318_0001
Examples
Section A. Reagents for Radiolabeling
Example 1
Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb(5-Aca)) - N-[4- (carboxy)benzyl]-N,N'-bis[(2-triphenylmethylthio)ethyl]- glycinamide Conjugate
A solution of N-[4-(carboxy)benzyl]-N,N'-bis[(2- triphenylmethylthio)ethyl]glycinamide N- hydroxysuccinimide ester (0.017 mmol), cyclo-(D-Val- NMeArg-Gly-Asp-Mamb(5-Aca)) (13.9 mg, 0.015 mmol), and Et3N (6.25 μl, 0.045 mmol) in DMF (350 μl) was allowed to stir at room temperature for 14 hours. The progress of the reaction was monitored by normal phase TLC
(90:8:2 CHCI3 :MeOH: HOAc) using the ninhydrin and
Sakaguchi tests. The DMF was removed under reduced pressure. The conjugate was purified using reversed- phase HPLC with a preparative Vydac C18 column (2.1 cm) using a 1.0%/min. gradient of 18 to 36% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy colorless solid (11 mg, 53%); FAB-MS: [M+H] =
Example 2
Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) - N-[4- (carboxy)benzyl]-N,N'-bis[(2-triphenylmethylthio)ethyl]- glycinamide Conjugate
A solution of N-[4-(carboxy)benzyl]-N,N'-bis[(2- triphenylmethylthio)ethyl]glycinamide N- hydroxysuccinimide ester (30 mg, 0.033 mmol), cyclo-(D- Lys-NMeArg-Gly-Asp-Mamb) (23.8 mg, 0.029 mmol), and Et3N (12 μl, 0.087 mmol) in DMF (0.60 ml) was allowed to stir at room temperature for 63 hours. The progress of the reaction was monitored by normal phase TLC (90:8:2 CHCl3:MeOH:HOAc) using the ninhydrin and Sakaguchi tests. The DMF was removed under reduced pressure. The conjugate was purified using reversed-phase HPLC with a preparative Vydac C18 column (2.1 cm) using a 0.9%/min. gradient of 18 to 36% acetonitrile containing 0.1% TFA and then lyophilized to give the TFA salt of the title compound as a fluffy colorless solid (24 mg, 60%); ESI- MS: [M] = 1397.3. Example 3
Cyclo(D-Val-NMeArg-Gly-Asp-Mamb(N-hydrazino- nicotinyl-5-Aca)) TFA salt Part A. Synthesis of Cyclo (D-Val-NMeArg-Gly-Asp-Mamb (N- boc-hydrazino-nicotinyl-5-Aca)) TFA salt
To a solution of cyclo (D-Val-NMeArg-Gly-Asp-Mamb (5- Aca) (10 mg, 0.011 mmol), succinimidyl boc- hydrazinonicotinate (4.6 mg, 0.0132 mmol) in DMF (0.3 mL) was added triethylamine (0.0061 mL, 0.044 mmol) and the reaction stirred at room temperature under nitrogen for 24 hours. The solvent was removed in vacuo and the residue dissolved in a solution of acetonitrile-water and lyophilized overnight to give an off-white solid.
Purification of part of the product was accomplished by reversed-phase HPLC on a preparative Vydac C-18 column using a 2.0%/min. gradient of 6.3-72% aqueous
acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H = 938.4849, calc. 938.4848).
Part B. Deprotection to Cyclo (D-Val-NMeArg-Gly-Asp- Mamb (N-hydrazinonicotinyl-5-Aca)) TFA salt
Cyclo (D-Val-NMeArg-Gly-Asp-Mamb (N-boc- hydrazinonicotinyl-5-Aca) TFA salt was dissolved in a mixture of 98:2 TFA:anisole (2 mL) and the reaction mixture stirred for 15 min. The solvent was removed in vacuo and the residue disolved in a solution of
acetonitrile-water and lyophilized to give a white solid. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C-18 column using a
2.0%/min. gradient of 6.3-72% aqueous acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H =
838.4324, calc. 838.4324).
Example 4
Cyclo (D-Abu-NMeArg-Gly-Asp-Mamb (N-hydrazino- nicotinyl-5-Aca)) TFA salt
Part A. Synthesis of Cyclo (D-Abu-NMeArg-Gly-Asp-Mamb (N- boc-hydrazino-nicotinyl-5-Aca)) TFA salt
To a solution of cyclo (D-Abu-NMeArg-Gly-Asp-Mamb (5- Aca) TFA salt (10 mg, 0.0109 mmol), succinimidyl boc- hydrazinonicotinate (4.55 mg, 0.0131 mmol) in DMF (0.4 mL) was added triethylamine (0.0061 mL, 0.044 mmol) and the reaction stirred at room temperature under nitrogen for 24 hours. The solvent was removed in vacuo and the residue dissolved in a solution of acetonitrile-water and lyophilized overnight to give an off-white solid. Purification of part of the product was accomplished by reversed-phase HPLC on a preparative Vydac C-18 column using a 2.0%/min. gradient of 6.3-72% aqueous
acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H = 924.4699, calc. 924.4692). Part B. Deprotection to Cyclo (D-Abu-NMeArg-Gly-Asp- Mamb(N-hydrazino-nicotinyl-5-Aca)) TFA salt
Cyclo (D-Abu-NMeArg-Gly-Asp-Mamb (N- hydrazinonicotinyl-5-Aca)) TFA salt: Cyclo (D-Abu- NMeArg-Gly-Asp-Mamb (N-boc-hydrazinonicotinyl-5-Aca)) TFA salt was dissolved in a mixture of 98:2 TFA:anisole (2 mL) and the reaction mixture stirred for 15 min. The solvent was removed in vacuo and the residue disolved in a solution of acetonitrile-water and lyophilized to give a white solid. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C-18 column using a 2.07%/min. gradient of 6.3-85.5% aqueous acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H = xx, calc. xx).
Example 5 Cyclo ((N-E-hydrazinonicotinyl-D-Lys)-NMeArg-Gly- Asp-Mamb) TFA salt
Part A. Synthesis of Cyclo ( (N-E-boc-hydrazinonicotinyl- D-Lys)-NMeArg-Gly-Asp-Mamb) TFA salt To a solution of cyclo (D-Lys-NMeArg-Gly-Asp- Mamb) .2TFA (4.2 mg, 0.005 mmol), succinimidyl boc- hydrazinonicotinate (2.1 mg, 0.006 mmol) in DMF (0.15 mL) was added triethylamine (0.003 mL, 0.02 mmol) and the reaction stirred at room temperature under nitrogen for 48 hours. The solvent was removed in vacuo and the residue dissolved in a solution of acetonitrile-water and lyophilized overnight to give an off-white solid. Purification was accomplished by reversed-phase HPLC on a preparative Vydac C-18 column using a 1.7%/min.
gradient of 6.3-85.5% aqueous acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H = 839.4157, calc. 839.4164).
Part B. Deprotection to Cyclo ((N-E-hydrazinonicotinyl-D- Lys)-NMeArg-Gly-Asp-Mamb) TFA salt Cyclo ((N-E-hydrazinonicotinyl-D-Lys)-NMeArg-Gly- Asp-Mamb) TFA salt: Cyclo ( (N-E-boc-hydrazinonicotinyl- D-Lys)-NMeArg-Gly-Asp-Mamb) TFA salt (3 mg) was
dissolved in a mixture of 98:2 TFA:anisole (2 mL) and the reaction mixture stirred for 15 min. The solvent was removed in vacuo and the residue disolved in a solution of acetonitrile-water and lyophilized to give a white solid. Purification was accomplished by reversed- phase HPLC on a preparative Vydac C-18 column using a 2.0%/min. gradient of 6.3-72% aqueous acetonitrile containing 0.1% TFA and lyophilized to give the TFA salt of the title compound as a fluffy solid. MS (M+H = 739.3629, calc. 739.3640). Example 6.
Cyclo-([DTPA-D-Lys]-NMeArg-Gly-Asp-Mamb) Conjugate
To a solution of 250 mg (2 mmol.) of cyclo (D-Lys- NMeArg-Gly-Asp-Mamb) in 208 mL of 0.1 M Borate (pH 9.88) at room temperature was added DTPA anhydride (743 mg, 10 mmol.) with constant stirring. The reaction was allowed to stir for 2 h. The crude mixture of products obtained after removal of the solvent was purified by preparative HPLC (Vydac Ciβ column, gradient of 0-50% ACN containing 0.1% TFA over 60 min., flow rate 20 mL/min). Two major components were isolated. Component A is Cyclo-([DTPA-D- Lys]-NMeArg-Gly-Asp-Mamb). MS: 979.1 (M+H+)
Example 7.
[Cyclo-(D-Lys-NMeArg-Gly-Asp-Marnb)]2 - DTPA Conjugate
Component B from the synthesis described in Example 6 is [Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb)]2 - DTPA. MS: 1565.4 (M+)
Section B. Radiolabeled Compounds Direct Labeling
Example 8. Cyclo-((125I)D-Tyr-NMeArg-Gly-Asp-Mamb)
To a 5 mL vial was added 22 mCi (45 μL) aqueous Na125I, 100 μL 0.5 M phosphate buffer pH 7.5, 4.5 μL 1 N HCl, 75 μg of the cyclic compound intermediate Cyclo-(D- Tyr-NMeArg-Gly-Asp-Mamb) dissolved in 75 μL 0.1% aqueous TFA, and 50 μg Chloramine-T dissolved in 50 μL H2O. The reaction was allowed to proceed for 1 minute then 50 μg of sodium metabisulfite dissolved in H2O was added. The product was purified by preparative HPLC. (Zorbax-Rx C18 column, flow = 1 mL/min, gradient from 100% A to 100% B over 30 minutes; Solvent A = 0.1% TFA in H2O, Solvent B = 40% ethanol in A. The product had a retention time of 30 min.
Example 9.
[(125I)N-3-(4-hydroxyphenyl)propionyl]-Cyclo-(D-Lys- NMeArg-Gly-Asp-Mamb)
To a 5 mL vial was added 11.4 mCi (25 μL) aqueous Na125I, 100 μL 0.5 M phosphate buffer pH 7.5, 4.5 μL 1 N HCl, 50 μg of the linker modified cyclic compound [N-3- (4-hydroxyphenyl)propionyl]-Cyclo-(D-Tyr-NMeArg-Gly-Asp- Mamb) dissolved in 50 μL 0.1% aqueous TFA, and 50 μg Chloramine-T dissolved in 50 μL H2O. The reaction was allowed to proceed for 1 minute then 50 μg of sodium metabisulfite dissolved in H2O was added. The product was purified by preparative HPLC, using the condition described in Example 10. The product had a retention time of 32 min.
Indirect Labeling
Example 10.
99mTcO (MAMA)-Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb (5-Aca) Part A. Deprotection The trityl protecting groups on the reagent described in Example 1 are removed: To a separate, clean 10 cc vial was added the reagent and 0.1 mL
trifluoroacetic acid (TFA). The solid dissolved to give a yellow solution.
Part B. Synthesis of 99mTc-glucoheptonate
A Glucoscan® vial was reconstituted with 1.0 mL Milli-Q H2O. 0.2 mL of the solution was removed and added to a clean 10 cc vial followed by -200 mCi
99mTcO4-. The reaction proceeded at room temperature for 20 minutes.
Part C. Synthesis of 99mTcO (MAMA)-Cyclo-(D-Val-NMeArg- Gly-Asp-Mamb (5-Aca))
To the deprotected reagent solution from Part A was added 0.2 mL 5 N NaOH, and 0.4 mL 0.2 M phosphate buffer pH 6. The pH was measured and adjusted as needed to 6.
This solution was immediately added to the 99mTc- glucoheptonate solution vial, crimped and heated at 100
°C for 15 minutes. After cooling -2 minutes, 20 μL of the solution was analyzed by HPLC using Method 1. (See
Table 1)
Example 11.
99mTcO (MAMA)-Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) Part A. Deprotection
The trityl protecting groups on the reagent
described in Example 2 are removed: To a separate, clean 10 cc vial was added the reagent and 0.1 mL
trifluoroacetic acid (TFA). The solid dissolved to give a yellow solution. Part B. Synthesis of 99mTcO (MAMA)-Cyclo-(D-Lys-NMeArg- Gly-Asp-Mamb)
To the deprotected reagent solution from Part A was added 0.2 mL 5 N NaOH, and 0.4 mL 0.2 M phosphate buffer pH 6. The pH was measured and adjusted as needed to 6. This solution was immediately added to the 99mTc- glucoheptonate solution vial, generated as described in Example 11, Part B, crimped and heated at 100 °C for 15 minutes. After cooling ~2 minutes, 20 μL of the
solution was analyzed by HPLC using Method 1. (See Table 1)
Example 12.
99mTc (tricine)2-Cyclo(D-Val-NMeArg-Gly-Asp- Mamb (hydrazino-nicotinyl-5-Aca)) To a solution of 70 mg tricine in 1.0 mL of water was added 0.05 mL 1.0 N NaOH to raise the pH to 7. 0.1 - 1.0 mL of 99mTcO4- in saline (10 - 100 mCi) was added followed by 10 μg of the reagent described in Example 3 dissolved in 100 μL of 0.1 N HCl and 100 μg of SnCl2 . 2H2O dissolved in 0.1 N HCl. The reaction proceeded at room temperature for 45 minutes. The product was analyzed by HPLC using the method 1 and by TLC using method 2. (see Table 1)
Example 13. 99mTc (EDDA)-Cyclo(D-Val-NMeArg-Gly-Asp-Mamb(hydrazino- nicotinyl-5-Aca))
To a solution of 10 mg ethylenediamine-N,N'- diacetic acid (EDDA) in 1.0 mL of water was added 0.05 mL 1.0 N NaOH to raise the pH to 7. 0.1 - 1.0 mL of 99mTcO4- in saline (10 - 100 mCi) was added followed by 50 μg of the reagent described in Example 3 dissolved in 100 μL of 0.1 N HCl and 100 μg of SnCl2 · 2H2O dissolved in 0.1 N HCl. The reaction proceeded at room
temperature for 45 minutes. The product was analyzed by HPLC using the method 1 and by TLC using method 2. (see Table 1) Example 14.
99mTc (tricine)2-Cyclo(D-Abu-NMeArg-Gly-Asp- Mamb(hydrazino-nicotinyl-5-Aca)) To a solution of 70 mg tricine in 1.0 mL of water was added 0.05 mL 1.0 N NaOH to raise the pH to 7. 0.1 - 1.0 mL of 99mTcO4- in saline (10 - 100 mCi) was added followed by 10 μg of the reagent described in Example 4 dissolved in 100 μL of 0.1 N HCl and 100 μg of SnCl2 · 2H2O dissolved in 0.1 N HCl. The reaction proceeded at room temperature for 45 minutes. The product was analyzed by HPLC using the method 1 and by TLC using method 2. (see Table 1) Example 15.
99mTc (tricine)2-Cyclo(D-Lys-NMeArg-Gly-Asp- Mamb (hydrazino-nicotinyl-5-Aca)) To a solution of 70 mg tricine in 1.0 mL of water was added 0.05 mL 1.0 N NaOH to raise the pH to 7. 0.1 - 1.0 mL of 99mTcO4- in saline (10 - 100 mCi) was added followed by 10 μg of the reagent described in Example 5 dissolved in 100 μL of 0.1 N HCl and 100 μg of SnCl2 · 2H2O dissolved in 0.1 N HCl. The reaction proceeded at room temperature for 45 minutes. The product was analyzed by HPLC using the method 1 and by TLC using method 2. (see Table 1)
Table 1. Analytical and Yield Data for 99mTc Labeled Reagents
Figure imgf000329_0001
Example 16.
Cyclo-([111In-DTPA-D-Lys]-NMeArg-Gly-Asp-Mamb)
50 μL of 111InCl3 (-100 mCi/mL in 0.05 M HCl) obtained from DuPont-NEN Products, Billerica, MA, was combined with an equal volume of freshly prepared 1.0 M ammonium acetate. After about five minutes, 0.1 - 1 mg of the reagent described in Example 6 dissolved in 0.25 mL water was added. The reaction proceeded at room temperature for 30 minutes. The product was analyzed by HPLC using method 3. Example 17.
111In-DTPA-[Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb)]2
To 0.5 mL of a solution of the reagent described in Example 7 in water (0.9 mg/1 mL) was added 111InCl3 (~3 mCi) in 0.5 mL of 1 N NH4OAc solution. The mixture was allowed to stand at room temperature for 30 minutes then analyzed by HPLC using method 3. (See Table 2)
Table 2. Analytical and Yield Data for 111In-labeked Reagents
Figure imgf000330_0001
Section C. 99mTc Labeled Reagents Via the Prechelate Approach.
The 99mTc-labeled reagents described in these examples were synthesized using the prechelate approach, The prechelate approach involves the steps: (1)
chelation of 99πιTc by the chelator; (2) activation of a non-coordinated carboxylic group on the resulting complex by forming its tetrafluorophenyl (TFP) ester; and (3) conjugation of the TFP-ester complex by forming an amide bond with a cyclic compound intermediate or linker modified cyclic compound.
Example 18 .
Cyclo- ( [ [ 99mTcO (mapt ) ] --D-Lys ] -NMeArg-Gly-Asp-Mamb) Part A. Chelation of 99mTc
To a clean 10 cc vial was added 0.35 mL Bz-mapt ( 3.0 mg/mL in 1 N NaOH), 0.10 mL SnCl2·2H2O (10 mg/mL in 1 N HCl), and 200 mCi 99mTcO4- in saline. The vial was crimped and placed in a 100 °C water bath for 25
minutes. After cooling ~2 minutes, 10 μL of the
solution was analyzed by HPLC using Method 1. Part B. Activation
To the solution from Part A was added 0.3 mL 0.5 M sodium phosphate pH 6, 0.3 mL 2, 3, 5, 6-tetrafluorophenol (100 mg/mL in 90% acetonitrile), 0.3 mL 1-(3- dimethylamino-propyl)-3-ethylcarbodiimide (100 mg/mL in 90% acetonitrile), and -0.1 mL 1 N HCl. The pH was adjusted as needed to pH 6. The vial was crimped and heated at 40 °C for 25 minutes. After cooling - 2 minutes, 20 μL of the solution was analyzed by HPLC using Method 1.
Part C. Conjugation
1.0 - 2.5 mg of the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40° C for 30 minutes. After cooling -2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3) Example 19.
Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb([99mTcO(mapt)]--5-Aca)) 1.0 - 2.5 mg of the linker modified cyclic compound Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb (5-Aca)) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Example 18, Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling -2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3) Example 20.
Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb([99mTcO(mapt)]--5-Aca))
1.0 - 2.5 mg of the linker modified cyclic compound Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb(5-Aca)) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Example 18, Part B. Using 1N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling -2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3)
Example 21. Cyclo-([(r99mTcO(mapt)]--5-Aca)D-Lys]-NMeArg-Gly-Asp- Mamb)
1.0 - 2.5 mg of the linker modified cyclic compound Cyclo- ( (5-Aca) D-Lys-NMeArg-Gly-Asp-Mamb) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Example 18, Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling ~2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3)
Example 22.
Cyclo-([[99mTcO(MeMAG2gaba)]"-D-Lys]-NMeArg-Gly-Asp-Mamb)
Part A. Chelation
To a 10 mL vial was added 100-250 mCi 99mTcO4- in 1.0 mL of saline, 1.0 mL of Bz-MeMAG2gaba solution (1 mg/1 mL in 0.5M pH 12 phosphate buffer), followed by of 0.15-0.20 mL of SnCl2·2H2O solution (15 mg/3 mL in IN HCl). The pH was adjusted to -11 and the mixture was heated for 30 min at 100 °C. The solution was analyzed by HPLC using Method 1.
Part B. Activation
To the solution from Part A was added 0.2 mL of IN HCl, 0.5 mL of tetrafluorophenol solution (100 mg/mL in 90% CH3CN) , and 0.5 mL of (1-[3-(dimehtylamino)propyl]- 3-ethylcarbodiimide chloride) solution (100 mg/mL in 90% CH3CN). The pH was adjusted to 6.0 and the mixture was heated at 50 °C for 30 min. Part C. Conjugation
1.0 - 2.5 mg of the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling -2 minutes, 25 μL of the
solution was analyzed by HPLC using Method l.(See Table 3) Example 23 .
Cyclo- (D-Val-NMeArg-Gly-Asp-Mamb ( [ 99mTcO (MeMAG2gaba ) ] --5-
Aca) )
1.0 - 2.5 mg of the linker modified cyclic compound Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb (5-Aca)) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Example 22, Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling -2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3) Example 24.
Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb([99mTcO(MeMAG2gaba)]--5- Aca)) 1.0 - 2.5 mg of the linker modified cyclic
compound Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb (5-Aca)) was dissolved in 0.3 mL 0.5 M pH 9 phosphate buffer and added to the solution from Example 22, Part B. Using 1 N NaOH, the pH was adjusted to 9. The reaction was heated at 40 °C for 30 minutes. After cooling -2 minutes, 25 μL of the solution was analyzed by HPLC using Method 1. (See Table 3)
Example 25.
Cyclo-([[99mTcO(MAG3)]--D-Lys]-NMeArg-Gly-Asp-Mamb)
This example was synthesized following the
procedure described in Example 22, substituting Bz-MAG3 as the chelator. (See Table 3) Example 26.
Cyclo-([[99mTcO(Me-MAG3)]--D-Lys]-NMeArg-Gly-Asp-Mamb) This example was synthesized following the
procedure described in Example 22, substituting Bz-Me- MAG3 as the chelator. (See Table 3)
Example 27.
Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb([99mTcO(MeMAG2ACA)]--5-
Aca))
The title compund was prepared according to the procedure procedure described in Example 22,
substituting Bz-Me-MAG2-ACA as the chelator in Part A and using Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb (5-Aca)) as the linker modifed cyclic compound in Part C. (See Table 3) Example 28.
Cyclo- ( [ [ 99mTcO (MABA) ] --D-Lys ] -NMeArg-Gly-Asp-Mamb)
Part A. Chelation
To a 10 mL vial was added 50-300 mCi 99mTcO4- in 0.5mL of saline, followed by 0.5 mL of Bz-MABA solution (1 mg/l mL in 0.5 M pH 12 phosphate buffer) and 0.15 mL of Na2S2O4 solution (5mg/mL in 0.5 M in pH 11.5 phosphate buffer) The pH was adjusted to 10-12 using 1 N NaOH and the mixture was heated for 30 min. at 100 °C then analyzed by HPLC using method 1.
Part B. Activation
To the solution from Part A was added 0.2 mL of 1 N HCl, 0.5 mL of TFP solution (50 mg/0.5 mL in 90% CH3CN), and 0.5 mL of DCI solution (50 mg in 0.5 mL in 90% CH3CN). The pH was adjusted to 6 if necessary and the mixture was heated at 45-50 °C for 30 min then analyzed by HPLC using method 1.
Part C. Conjugation
To the solution from Part B was added 2-3 mg of the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp- Mamb) dissolved in 0.5 mL 0.5 M phosphate buffer pH 9 and pH was then adjusted to 9.5-10. The solution was heated at 50 °C for 30 min, then analyzed by HPLC using method 1.
(See Table 3)
Example 29.
Cyclo-(D-Val-NMeArg-Gly-Asp-Mamb([99mTcO(MABA)]--5-Aca))
The title compound was synthesized following the procedure described in Example 28, substituting the linker modified cyclic compound Cyclo-(D-Val-NMeArg-Gly- Asp-Mamb (5-Aca)) for the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) in Part C.
Example 30.
Cyclo-(D-Abu-NMeArg-Gly-Asp-Mamb([99mTcO(MABA)]--5-Aca))
The title compound was synthesized following the procedure described in Example 28, substituting the linker modified cyclic compound Cyclo-(D-Abu-NMeArg-Gly- Asp-Mamb (5-Aca)) for the cyclic compound intermediate
Cyclo- (D-Lys-NMeArg-Gly-Asp-Mamb) in Part C.
Example 31.
Cyclo-([[99mTCO(MA-MAMA)]-D-Lys]-NMeArg-Gly-Asp-Mamb) Part A. Deprotection.
The trityl groups on the chelator MA-MAMA were removed by dissolving 6 mg in 1 mL of anhydrous trifluoroacetic acid (TFA). The resulting yellow solution was allowed to stand at room temperature for 5 minutes. Triethylsilane (0.5 mL) was added to the yellow solution to give a clear two-layered mixture. Volatiles were removed under reduced pressure to give a white residue. Part B. Hydrolysis of the Ethyl Ester.
To the white residue from Part A was added 0.5 mL of 5 N NaOH and 1 mL of THF. The mixture was heated in a water bath (100 °C) for 5 minutes, by which time most of THF was evaporated. To the reaction mixture was added 3 mL of 0.5 M phosphate buffer pH 11.5. The pH was adjusted to 10-12 and sodium dithionite (15-30 mg) was added. The mixture was filtered and the total volume was adjusted to 6 mL using 0.5 M pH 11.5 phosphate buffer. Part C. Chelation.
To a 10 mL vial was added 50-150 mCi 99rnTcO4- in
0.5 mL of saline, followed by 0.5 mL of ligand
solution from Part B. The pH was adjusted to 10-12
using 1 N NaOH and the mixture was heated for 30 min at 100 °C then analyzed by HPLC using method 1.
Part D. Activation.
To the solution from Part C was added 0.2 mL of 1 N HCl, 0.5 mL of TFP solution (50 mg/0.5 mL 90% CH3CN), and 0.5 mL of DCI solution (50 mg in 0.5 mL 90% CH3CN).
The pH was adjusted to 6 if necessary and the mixture was heated at 45-50 °C for 30 min.then analyzed by
HPLC using method 1. Part E. Conjugation.
To the solution from Part D was added 2.5 mg of the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp- Mamb) dissolved in 0.5 mL 0.5 M phosphate buffer pH 9 and the pH was then adjusted to 9.5-10. After heating at 50 °C for 30 min, the solution was analyzed by HPLC using method 1.
Example 32 .
Cyclo- (D-Val-NMeArg-Gly-Asp-Mamb ( [ 99mTcO (MA-MAMA) ] -5-Aca ) )
The title compound was synthesized following the procedure described in Example 31, substituting the linker modified cyclic compound Cyclo-(D-Val-NMeArg-Gly- Asp-Mamb (5-Aca)) for the cyclic compound intermediate Cyclo-(D-Lys-NMeArg-Gly-Asp-Mamb) in Part E.
Table 3. Analytical and Yield Data for 99mTc-labeled
Reagents
Figure imgf000338_0001
Figure imgf000339_0001
Analytical Methods
HPLC Method 1
Column: Vydac C18, 250 mm x 4.6 mm, 300 A pore size
Solvent A: 10 mM sodium phosphate, pH 6.0
Solvent B: 10C % acetonitrile
Gradient :
0%B 30 %B 75%B
0' 15 ' 25'
Flow rate: 1 .0 mL/min
Detection by Nal probe TLC Method 2
ITLC-SG strip, 1 cm x 7.5 cm, developed in 1:1 acetone : water.
HPLC Method 3
Column: Vydac C18, 250 mm x 4.6 mm, 300 A pore size Solvent A: 10 mM sodium phosphate, pH 6.0
Solvent B: 75% acetonitrile in Solvent A
Gradient :
5% B 5%B 100%B
0' 5' 40'
Flow rate: 1.0 mL/min
Detection by Nal probe
Utility The radiolabeled compounds of the invention are useful as radiopharmaceuticals for imaging a thrombus such as may be present in a patient with unstable angina, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, diabetes,
thrombophlebitis, pulmonary emboli, or prosthetic cardiac devices such as heart valves, and thus may be used to diagnose such present or potential disorders. The patient may be any type of a mammal, but is
preferably a human. The radiolabeled compounds may be used alone, or may be employed as a composition with a radiopharmaceutically acceptable carrier, and/or in combination with other diagnostic or therapeutic agents. Suitable radiopharmaceuticals carriers and suitable amounts thereof are well known in the art, and can be found in, for example, Remington's Pharmaceutical
Sciences, Gennaro, A.R., ed., Mack Publishing Company, Easton, PA (1985), and The United States Pharmacopia - The National Formulary, 22nd Revision, Mack Printing Company, Easton, PA (1990), standard reference texts in the pharmaceutical field. Other materials may be added, as convenient, to stabilize the composition, as those skilled in the art will recognize, including
antioxidizing agents such as sodium bisulfite, sodium sulfite, ascorbic acid, gentisic acid or citric acid (or their salts) or sodium ethylenediamine tetraacetic acid (sodium EDTA), as is well known in the art. Such other materials, as well as suitable amounts thereof, are also described in Remington's Pharmaceutical Sciences and The United States Pharmacopia - The National Formulary, cited above.
The present invention also includes
radiopharmaceutical kits containing the labeled
compounds of the invention. Such kits may contain the labeled compounds in sterile lyophilized form, and may include a sterile container of a radiopharma-ceutically acceptable reconstitution liquid. Suitable
reconstitution liquids are disclosed in Remington's Pharmaceutical Sciences and The United States
Pharmacopia - The National Formulary, cited above. Such kits may alternatively contain a sterile container of a composition of the radiolabeled compounds of the invention. Such kits may also include, if desired, other conventional kit components, such as, for example, one or more carriers, one or more additional vials for mixing. Instructions, either as inserts or labels, indicating quantities of the labeled compounds of the invention and carrier, guidelines for mixing these components, and protocols for administration may also be included in the kit. Sterilization of the containers and any materials included in the kit and lyophilization (also referred to as freeze-drying) of the labeled compounds of the invention may be carried out using conventional sterilization and lyophilization
methodologies known to those skilled in the art.
To carry out the method of the invention, the radiolabeled compounds are generally administered intravenously, by bolus injection, although they may be administered by any means that produces contact of the compounds with platelets. Suitable amounts for
administration will be readily ascertainable to those skilled in the art, once armed with the present
disclosure. The dosage administered will, of course, vary depending up such known factors as the particular compound administered, the age, health and weight or the nature and extent of any symptoms experienced by the patient, the amount of radiolabeling, the particular radionuclide used as the label, the rate of clearance of the radiolabeled compounds from the blood. Acceptable ranges for administration of radiolabeled materials are tabulated, for example, in the Physicians Desk Reference (PDR) for Nuclear Medicine, published by Medical Exonomics Company, a well-known reference text. A discussion of some of the aforementioned
considerations is provided in Eckelman et al., J. Nucl. Med., Vol. 209, pp. 350-357 (1979). By way of general guidance, a dosage range of the radiolabeled compounds of the invention may be between about 1 and about 40 mCi.
Once the radiolabeled compounds of the invention are administered, the presence of thrombi may be visualized using a standard radioscintographic imaging system, such as, for example, a gamma camera or a computed tomographic device, and thromboembolic
disorders detected. Such imaging systems are well known in the art, and are discussed, for example, in Macovski, A., Medical Imaging Systems, Information and Systems Science Series, Kailath, T., ed., Prentice-Hall, Inc., Englewood Cliffs, NJ (1983). Particularly preferred are single-photon emission computed tomography (SPECT) and positron emission tomography (PET). Specifically, imaging is carried out by scanning the entire patient, or a particular region of the patient suspected of having a thrombus formation, using the
radioscintographic system, and detecting the
radioisotope signal. The detected signal is then converted into an image of the thrombus by the system. The resultant images should be read by an experienced observer, such as, for example, a nuclear medicine physician. The foregoing process is referred to herein as "imaging" the patient. Generally, imaging is carried out about 1 minute to about 48 hours following administration of the radiolabeled compound of the invention. The precise timing of the imaging
will be dependant upon such factors as the half-life of the radioisotope employed, and the clearance rate of the compound administered, as will be readily apparent to those skilled in the art. Preferably, imaging is carried out between about 1 minute and about 4 hours following administration.
The advantage of employing the radiolabeled compounds oi the invention, which have the ability to localize specifically and with high affinity in thrombi, to detect the presence of thrombi and/or to diagnose thromboembolic disorders in a patient, will be readily apparent to those skilled in the art, once armed with the present disclosure.
Arteriovenous Shunt Model: Adult mongrel dogs of either sex (9-13kg) were anesthetized with
pentobarbital sodium (35 mg/kg,i.v.) and ventilated with room air via an endotracheal tube (12 strokes/min, 25 ml/kg) . For arterial pressure determination, the left carotid artery was cannulated with a saline-filled polyethylene catheter (PE-240) and connected to a
Statham pressure transducer (P23ID; Oxnard,CA). Mean arterial blood pressure was determined via damping the pulsatile pressure signal. Heart rate was monitored using a cardiotachometer (Biotach, Grass Quincy, MA) triggered from a lead II electrocardiogram generated by limb leads. A jugular vein was cannulated (PE-240) for drug administration. The both femoral arteries and femoral veins were cannulated with silicon treated
(Sigmacote, Sigma Chemical Co. St Louis, MO), saline filled polyethylene tubing (PE-200) and connected with a 5 cm section of silicon treated tubing (PE-240) to form an extracorporeal arterio-venous shunts (A-V). Shunt patency was monitored using a doppler flow system (model VF-1, Crystal Biotech Inc, Hopkinton, MA) and flow probe (2-2.3 mm, Titronics Med. Inst., Iowa City, IA) placed proximal to the locus of the shunt. All parameters were monitored continuously on a polygraph recorder (model 7D Grass) at a paper speed of 10 mm/min or 25 mm/sec.
On completion of a 15 min post surgical
stabilization period, an occlusive thrombus was formed by the introduction of a thrombogenic surface ( 4-0 braided silk thread, 5 cm in length, Ethicon Inc., Somerville, NJ) into the shunt one shunt with the other serving as a control. Two consecutive Ihr shunt periods were employed with the test agent administered as an infusion over 5 min beginning 5 min before insertion of the thrombogenic surface. At the end of each 1 hr shunt period the silk was carefully removed and weighed and the % incorporation determined via well counting.
Thrombus weight was calculated by subtracting the weight of the silk prior to placement from the total weight of the silk on removal from the shunt. The results are shown in Table 4. Arterial blood was withdrawn prior to the first shunt and every 30 min thereafter for
determination of blood clearance, whole blood collagen- induced platelet aggregation, thrombin-induced platelet degranulation (platelet ATP release), prothrombin time and platelet count. Template bleeding time was also performed at. 30 min intervals.
Canine Deep Vein Thrombosis Model: This model incorporates the triad of events (hypercoagulatible state, period of stasis, low shear environment)
essential for the formation of a venous fibrin-rich actively growing thrombus. The procedure was as follows: Adult mongrel dogs of either sex (9-13 kg) were anesthetized with pentobarbital sodium (35 mg/kg,i.v.) and ventilated with room air via an endotracheal tube (12 strokes/min, 25 ml/kg). For arterial pressure determination, the right femoral artery was cannulated with a saline-filled polyethylene catheter (PE-240) and connected to a Statham pressure transducer (P23ID; Oxnard,CA). Mean arterial blood pressure was determined via damping the pulsatile pressure signal. Heart rate was monitored using a cardiotachometer (Biotach, Grass Quincy, MA) triggered from a lead II electrocardiogram generated by limb leads. The right femoral vein was cannulated (PE-240) for drug administration. A 5 cm segment of both jugular veins was isolated, freed from fascia and circumscribed with silk suture. A microthermister probe was placed on the vessel which serves as an .indirect measure of venous flow. A balloon embolectomy catheter was utilized to induce the 15 min period of stasis during which time a hypercoagulatible state was then induced using 5 U thrombin (American Diagnosticia, Greenwich CT)
administered into the occluded segment. Fifteen minutes later, flow was reestablished by deflating the balloon.The agent was infused during the first 5 min of reflow and the rate of incorporation monitored using gamma scintigraphy. The results for Examples 12 and 19 are shown in Figure 1. Example 33
Table 4. Experimental Data from the Arteriovenous Shunt Model
(mean± SEM, T/B = thrombus/background)
Figure imgf000346_0001
Platelet Aggregation Assay: Canine blood was collected into 10 ml citrated Vacutainer tubes. The blood was centrifuged for 15 minutes at 150 x g at room temperature, and platelet-rich plasma (PRP) was removed. The remaining blood was centrifuged for 15 minutes at 1500 x g at room temperature, and platelet-poor plasma (PPP) was removed. Samples were assayed on a
aggregometer (PAP-4 Platelet Aggregation Profiler), using PPP as the blank (100% transmittance). 200 μl of PRP was added to each micro test tube, and transmittance was set to 0%. 20 μl of various agonists (ADP,
collagen, arachidonate, epinephrine, thrombin) were added to each tube, and the aggregation profiles were plotted (% transmittance versus time). The results were expressed as % inhibition of agonist-induced platelet aggregation. For the IC50 evaluation, the test compounds were added at various concentrations prior to the activation of the platelets.
Platelet-Fibrinogen Binding Assay: Binding of 125l-fibrinogen to platelets was performed as described by Bennett et al. (1983) Proc. Natl. Acad. Sci. USA 80: 2417-2422, with some modifications as described below. Human PRP (h-PRP) was applied to a Sepharose column for the purification of platelet fractions. Aliquots of platelets (5 X 108 cells) along with 1 mM calcium chloride were added to removable 96 well plates prior to the activation of the human gel purified platelets (h- GPP). Activation of the human gel purified platelets was achieved using ADP, collagen, arachidonate,
epinephrine, and/or thrombin in the presence of the ligand, 125l-fibrinogen. The 125I-fibrinogen bound to the activated, platelets was separated from the free form by centrifugation and then counted on a gamma counter. For an IC50 evaluation, the test compounds were added at various concentrations prior to the activation of the platelets.
The novel cyclic glycoprotein Ilb/IIIa compounds of the invention may also possess thrombolytic efficacy, that is, they are capable of lysing (breaking up) already formed platelet-rich fibrin blood clots, and thus may useful in treating a thrombus formation, as evidenced by their activity in the tests described below. Preferred cyclic compounds of the present invention for use in thrombolysis would include those compounds having an IC50 value (that is, the molar concentration of the cyclic compound capable of
achieving 50% clot lysis) of less than about 1 mM, more preferably an IC50 value of less than about 0.1 mM, even more preferably an IC50 value of less than about 0.01 mM, still more preferably an IC50 value of less than about 0.001 mM, and most preferably an IC50 value of about 0.0005 mM.
IC50 determinations may be made using a standard thrombolysis assay, as described below. Another class of preferred thrombolytic compounds of the invention would include those compounds which have a Kd of < 100 nM, preferably < 10 nM, most preferably 0.1 to 1.0 nM.
Thrombolytic Assay: Venous blood was obtained from the arm of a healthy human donor who was drug-free and aspirin free for at least two weeks prior to blood collection, and placed into 10 ml citrated Vacutainer tubes. The blood was centrifuged for 15 minutes at 1500 x g at room temperature, and platelet rich plasma (PRP) was removed. To the PRP was then added 1 x 10-3 M of the agonist ADP, epinephrine, collagen, arachidonate, serotonin or thrombin, or a mixture thereof, and the PRP incubated for 30 minutes. The PRP was centrifuged for 12 minutes at 2500 x g at room temperature. The
supernatant was then poured off, and the platelets remaining in the test tube were resuspended in platelet poor plasma (PPP), which served as a plasminogen source. The suspension was then assayed on a Coulter Counter
(Coulter Electronics, Inc., Hialeah, FL), to determine the platelet count at the zero time point. After obtaining the zero time point, test compounds were added at various concentrations. Test samples were taken at various time points and the platelets were counted using the Coulter Counter. To determine the percent of lysis, the platelet count at a time point subsequent to the addition of the test compound was subtracted from the platelet count at the zero time point, and the resulting number divided by the platelet count at the zero time point. Multiplying this result by 100 yielded the percentage of clot lysis achieved by the test compound. For the IC50 evaluation, the test compounds were added at various concentrations, and the percentage of lysis caused by the test compounds was calculated.
The disclosures of each patent and publication cited in this document are hereby incorporated herein by reference, in their entirety.
Various modifications in the invention, in addition to those shown and described herein will be readily apparent to those skilled in the art from the foregoing description. Such modifications are intended to be within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A reagent for preparing a radiopharmaceutical of formulae:
(QLn)dCh ; (Q)d'Ln-Ch, wherein, d is 1-3, d' is 2-20, Ln is a linking group, Ch is a metal chelator, and Q is a compound of formula (I):
Figure imgf000350_0001
or a pharmaceutically acceptable salt or
prodrug form thereof, wherein: R31 is a C6-C14 saturated, partially saturated, or aromatic carbocyclic ring system, substituted with 0-4 R10 or R10a, and optionally bearing a bond to Ln; a heterocyclic ring system, optionally substituted with 0-4 R10 or R10a, and optionally bearing a bond to Ln;
R32 is selected from:
-C(=O)-; -C (=S ) - -S (=O) 2- ;
-S (=O) - ;
-P (=Z ) ( ZR13 ) -;
Z is S or O; n" and n' are independently 0-2; R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11; a bond to Ln; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13,
-C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N (R13) 2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C(=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy;
R1 and R21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R12; when n' is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R21 and R23 are independently selected from: hydrogen;
C1-C4 alkyl, optionally substituted with
1-6 halogen;
benzyl; R22 and R23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2 , R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms; R1 and R2, where R21 is H, can
alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R12;
R11 is selected from one or more of the
following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13,
-C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N(R13) 2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H,
-SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C(=NH)NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy,
C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C3-C6 cycloalkoxy, C1-C4 alkyl (alkyl being substituted with 1-5 groups selected independently from:
-NR13R14, -CF3, NO2, -SO2R13a, or
-S(=O)R13a), aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said heterocyclic ring being substituted with 0-2 R12;
R12 is selected from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl,
C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B(R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl) -OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13,
-NR13C (=O) OR13a, -NR13C (=O) N (R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a);
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
R2 is H or C1-C6 alkyl;
R10 and R10a are selected independently from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C3-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O) N (R13) 2, -C(=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B(R34) (R35) , C3-C6 cycloalkoxy,
-OC (=O) R13, -C (=O) R13, -OC (=O) OR13a,
-OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13,
-NR13C (=O)OR13a, -NR13C (=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H,
-SO2R13a, -S (=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl (including -CVFW where v = 1 to 3 and w = 1 to
(2v+1)), C1-C4 haloalkoxy, C1-C4
alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with -N(R13)2, -CF3, NO2, or -S(=O)R13a);
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein:
R3 is H or C1-C8 alkyl; R4 is H or C1-C3 alkyl;
R5 is selected from:
hydrogen;
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11; a bond to Ln; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13,
-C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, =NOR13, -B (R34) (R35), -OCH2CO2H, 2-(1-morpholino) ethoxy,
-SC(=NH)NHR13, N3, -Si(CH3)3, (C1-C5 alkyl)NHR16;
(C0-C6 alkyl) X;
Figure imgf000357_0001
independently 0,1;
Figure imgf000357_0002
- (CH2)mS(O)p, (CH2)2X, where m = 1,2 and p' = 0-2; wherein X is defined below; and
R3 and R4 may also be taken together to form (CH2)nX
Figure imgf000357_0003
-CH2CHCH2-, where
n = 0,1 and X is
Figure imgf000357_0004
R3 and R5 can alternatively be taken together to form -(CH2)t- of -CH2S(O)p'C(CH3)2-, where t = 2-4 and p' = 0-2; or R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is a D-isomer or L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is selected from:
-(C1-C7 alkyl) X;
Figure imgf000358_0001
(CH2)q-X, wherein each q is independently 0-2 and
substitution on the phenyl is at the 3 or 4 position; -(CH2)q-
Figure imgf000359_0001
(CH2)q-X, wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
(C1-C6 alkyl)
Figure imgf000359_0002
-(CH2)mO-(C1-C4 alkyl)-X, where m = 1 or 2;
-(CH2)mS(O)p'-(C1-C4 alkyl)-X, where m = 1 or 2 and p' = 0-2; and
X is selected from:
Figure imgf000359_0003
R1:. -N(R13)R13;
-C(=NH) (NH2); -SC (=NH)-NH2; -NH- C(=NH) (NHCN); -NH-C(=NCN) (NH2);
-NH-C(=N-OR13) (NH2);
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000359_0004
-(CH2)qCH(CH2)q-, wherein each q is independently 1 or 2 and wherein n = 0 or 1 and X is -NH2 or -NH-
Figure imgf000360_0001
R1:;
L is -Y (CH2)vC(=O)-, wherein: Y is NH, N(C1-C3 alkyl), O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
-NR17-CH-C(=O)-
Figure imgf000360_0002
(CH(R4))q.
Figure imgf000360_0003
R8
wherein: q' is 0-2; R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35 ) , -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having
1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl
(said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a, -NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R35 are independently selected from:
-OH,
-F,
-N(R13)2, or
C1-C8-alkoxy;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O. 2. A reagent of Claim 1, wherein:
R31 is bonded to (C(R23)R22)n" and (C(R21)R1)n' at 2 different atoms on said carbocyclic ring.
3. A reagent of Claim 1, wherein: n" is 0 and n' is 0;
n" is 0 and n' is 1;
n" is 0 and n' is 2;
n" is 1 and n' is 0;
n" is 1 and n' is 1;
n" is 1 and n' is 2;
n" is 2 and n' is 0;
n" is 2 and n' is 1; or
n" is 2 and n' is 2.
4. A reagent of Claim 1 wherein R6 is methyl, ethyl, or propyl.
5. A reagent of Claim 1 wherein:
R32 is selected from:
-C(=O)-;
-C(=S)- -S(=O)2-;
R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11, C2-C8 alkenyl substituted with 0-2 R11, C2-C8 alkynyl substituted with 0-2 R11,
C3-C8 cycloalkyl substituted with 0-2
R11, C6-C10 bicycloalkyl substituted with 0-2 R11; a bond to Ln; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2,
-CH2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C(=NH)NHR13, NO2;
R1 and R21 can alternatively join to form a 5-7 membered carbocyclic ring
substituted with 0-2 R12; when n1 is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms; R22 and R23 can alternatively join to form a
3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2, R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R1 and R2, where R21 is H, can alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2 R12; R11 is selected from one or more of the
following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -CH2N(R13)2, -N(R13)2, -NHC (=NH) NHR13,
-C(=NH)NHR13, =NOR13, NO2;
C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C1-C4 alkyl (substituted with -NR13R14, -CF3, NO2, -SO2R13, or -S(=O)R13a) aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with
0-2 R12;
R3 is H or CH3; R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SNHR16, where s
= 3-5; a bond to Ln; R3 and R5 can alternatively be taken together to form -(CH2)t- ( t = 2-4) or
-CH2SC(CH3)2-; or
R7 is selected from: - (C1-C7 alkyl) X;
Figure imgf000365_0001
wherein each q is
independently 0-2 and substitution on the phenyl is at the 3 or 4 position; wherein each q
Figure imgf000366_0001
is
independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
-
Figure imgf000366_0002
- (CH2 ) mO- (C1 -C4 alkyl ) -X, where m = 1 or
2 ;
-(CH2)mS-(C1-C4 alkyl)-X, where m = 1 or 2; and
X is selected from:
-NH-C(=NH) (NH2), -NHR13, -C (=NH) (NH2), -SC(NH)-NH2;
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000366_0003
-CH2CHCH2-, wnere
n = 0 or 1 and X is -NH2 or -NH- C(=NH) (NH2);
L is -Y (CH2)vC (=O) -, wherein: Y is NH, N (C1-C3 alkyl ) , O, or S ; and v = 1 or 2 ;
M is a D-isomer or L-isomer amino acid of
structure
-
Figure imgf000367_0001
wherein: q' is 0-2;
R17 is H, C1-C3 alkyl ;
R8 is selected from :
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R35 are independently selected from:
-OH,
-F,
-NR13R14, or C1-C8-alkoxy;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from
N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from
N, S, or O.
6. A reagent of Claim 1, wherein:
R31 is selected from the group consisting of: (a) a 6 membered saturated, partially saturated or aromatic carbocyclic ring substituted with 0-3 R10 or R10a, and optionally bearing a bond to Ln; (b) a 8-11 membered saturated, partially saturated, or aromatic fused bicyclic carbocyclic ring substituted with 0-3 R10 or R10a ' and optionally bearing a bond to Ln; or (c) a 14 membered saturated, partially saturated, or aromatic fused tricyclic carbocyclic ring substituted with 0-3 R10 or R10a' and optionally bearing a bond to Ln .
7. A reagent of Claim 1, wherein:
R31 is selected from the group consisting of:
(a) a 6 membered saturated, partially saturated, or aromatic carbocyclic ring of formulae:
Figure imgf000369_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted with 0-3 R10, and optionally bears a bond to Ln;
(b) a 10 membered saturated, partially saturated, or aromatic bicyclic carbocyclic ring of formula:
Figure imgf000369_0002
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R10, and optionally bears a bond to Ln;
(c) a 9 membered saturated, partially saturated, or aromatic bicyclic carbocyclic ring of formula:
Figure imgf000370_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, wherein said carbocyclic ring is substituted independently with 0- 4 R10, and optionally bears a bond to Ln.
8. A reagent of Claim 1, wherein:
R31 is selected from (the dashed bond may be a single or double bond):
Figure imgf000370_0002
; or
Figure imgf000371_0001
wherein R31 may be independently substituted with 0-3 R10 or R10a, and optionally bears a bond to Ln; n" is 0 or 1; and n' is 0-2.
9. A reagent of Claim 1, wherein:
R1 and R22 are independently selected from: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C (=O)OR13a, -NR13C (=O)N(R13)2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino) ethoxy, C1-C4 alkyl (alkyl being substituted with -N(R13)2, -CF3, NO2, or -S(=O)R13a).
10. A reagent of Claim 1, wherein:
R31 is selected from:
Figure imgf000372_0001
wherein R31 may be independently substituted with 0-3 R10 or R10a, and may optionally bear a bond to Ln;
R32 is -C(=O) -; n" is 0 or 1; n' is 0-2; R1 and R22 are independently selected from H, C1-C4 alkyl, phenyl, benzyl,
phenyl-(C2-C4) alkyl, C1-C4 alkoxy; and a bond to Ln;
R21 and R23 are independently H or C1-C4 alkyl;
R2 is H or C1-C8 alkyl; R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl; R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
R10 and R10a are selected independently from:
H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy; J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4) (R5)C(=O)- , wherein:
R3 is H or CH3; R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)sNHR16, where s = 3-5; and a bond to Ln; or
R3 and R5 can alternatively be taken together to form -(CH2)t- (t = 2-4) or
-CH2SC(CH3)2-; or R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is
Figure imgf000374_0001
Figure imgf000375_0001
0 or 1;
-(CH2) rX, where r = 3-6;
Figure imgf000375_0002
-(CH2)mS(CH2)2X, where m = 1 or 2;
-(C3-C7 alkyl)-NH-(C1-C6 alkyl);
Figure imgf000375_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2); or
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000375_0004
CH2CHCH2-, where n = 0 or 1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y (CH2)vC(=O)-, wherein: Y is NH, O, or S ; and v = 1 or 2 ;
M is a D-isomer or L-isomer amino acid of
structure
-NR17-CH-C(=O)-
Figure imgf000376_0002
(CH(R4))q,
Figure imgf000376_0001
R8
wherein: q' is 0-2;
R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B (R34 ) (R35),
-NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl
(said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13.
11. The reagent of Claim 1 that is a 1,3- disubstituted phenyl compound of the formula (Il) :
Figure imgf000377_0001
wherein: the shown phenyl ring in formula (II) may be substituted with 0-3 R10, and may optionally bear a bond to Ln;
R10 is selected independently from: H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl,
phenyl- (C1-C4) alkyl, or a bond to Ln; R2 is H or methyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4) (R5) C (=O) -, wherein: R3 is H or CH3;
R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), -(CH2)SNHR16, where s = 3-5, or a bond to Ln;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein: R6 is H or C1-C8 alkyl;
R7 is:
Figure imgf000379_0001
0 or 1;
-(CH2)rX, where r = 3-6;
-
Figure imgf000379_0002
-(CH2)mS (CH2)2X, where m = 1 or 2;
-(C3-C7 alkyl)-NH-(C1-C6 alkyl)
Figure imgf000379_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; and X is -NH2 or -NHC (=NH) (NH2), provided that X is not -NH2 when r = 4; or
R6 and R7 are alternatively be taken together to form
(CH2)nX
Figure imgf000380_0001
-CH2CHCH2-, where n = 0,1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y(CH2)vC(=O)-, wherein:
Y is NH, O, or S; and v = 1,2;
M is a D-isomer or L-isomer amino acid of
structure
wherein:
Figure imgf000380_0002
q' is 0-2; R17 is H, C1-C3 alkyl; R8 is selected from: -CO2R13,-SO3R13, -SO2NHR14, -B (R34)(R35),
-NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S , or O) ,
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13 .
12. The reagent of Claim 1 that is a 1,3-disubstituted phenyl compound of the formula (II):
Figure imgf000381_0001
wherein : the phenyl ring in formula ( II ) may be substituted with 0-3 R10 or R10a;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl, or phenyl- (C2- C4)alkyl;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10
alkyl)aryl, or C3-C10 alkoxyalkyl;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein:
R3 is H or CH3; R4 is H;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3-C6
cycloalkylmethyl, C1-C6 cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SR16, where s = 3-5; or a bond to Ln;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-;
R16 is selected from:
an amine protecting group;
1-2 amino acids; 1-2 amino acids substituted with an amine protecting group; K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein: R6 is H or C3-C8 alkyl; R7 is
Figure imgf000383_0001
1;
-(CH2)rX, where r = 3-6;
Figure imgf000383_0002
-(CH2)mS(CH2)2X, where m = 1 or 2; -(C4-C7 alkyl)-NH-(C1-C6 alkyl) k
Figure imgf000383_0003
-(CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; -(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and X is -NH2 or -NHC(=NH) (NH2), provided that X is not -NH2 when r = 4; or
L is -YCH2C (=O) -, wherein: Y is NH or O;
M is a D-isomer or L-isomer amino acid of structure
-NR17-CH-C(=O)-
Figure imgf000384_0001
(CH(R4))q,
Figure imgf000384_0002
R8 , wherein: q1 is 1;
R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2H or -SO3R13,
13. The reagent of Claim 1 that that is a compound of formula (II) above, wherein: the phenyl ring in formula (II) bears a bond to Ln, and may be further substituted with 0-2 R10 or R10a ;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy; R1 is H;
R2 is H;
R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of formula -N (R3)CH(R5)C(=O)-, wherein: R3 is H and R5 is H, CH3, CH2CH3, CH(CH3)2,
CH(CH3)CH2CH3, CH2CH2CH3, CH2CH2CH2CH3,
CH2CH2SCH3, CH2CH(CH3)2, (CH2)4NH2, (C3-C5 alkyl) NHR16;
or
R3 is CH3 and R5 is H; or
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of formula
-N(CH3)CH(R7)C(=O)-, wherein:
R7 is -(CH2)3NHC(=NH)(NH2); L is -NHCH2C(=O)-; and M is a D-isomer or L-isomer amino acid of structure
wherein:
Figure imgf000386_0001
q' is 1;
R4 is H or CH3;
R17 is H;
R8 i s
-CO2H;
-SO3H .
14. The reagent of Claim 1 that that is a compound of formula (II) above, wherein: the phenyl ring in formula (II) bears a bond to Ln;
R1 and R2 are independently selected from H,
methyl;
J is selected from D-Val, D-2-aminobutyric acid, D-
Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, βAla,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala, Nε-p-azidobenzoyl-D-Lys, Nε-p- benzoylbenzoyl-D-Lys, Nε-tryptophanyl-D-Lys,
Nε-o-benzylbenzoyl-D-Lys, Nε-p-acetylbenzoyl- D-Lys, Nε-dansyl-D-Lys, Nε-glycyl-D-Lys, Nε- glycyl-p-benzoylbenzoyl-D-Lys, Nε-p- phenylbenzoyl-D-Lys, Nε-m-benzoylbenzoyl-D- Lys, Nε-o-benzoylbenzoyl-D-Lys;
K is selected from NMeArg, Arg;
L is selected from Gly, β-Ala, Ala;
M is selected from Asp; CMeAsp; βMeAsp; NMeAsp; D- Asp.
15. The reagent of Claim 1, wherein: R31 bears a bond to Ln;
R1 and R2 are independently selected from H,
methyl;
J is selected from: D-Val, D-2-aminobutyric acid, D-Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, βAla,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala; K is selected from NMeArg;
L is Gly;
M is selected from Asp; OdeAsp; βMeAsp; NMeAsp;
D-Asp.
16. A reagent as in one of claims 1-15, wherein Ch is selected from the group:
r
Figure imgf000388_0001
Figure imgf000389_0001
Figure imgf000390_0001
A1, A2, A3, A4, A5, A6, and A7 are
independently selected at each occurrence from the group: NR40R41, S, SH, S(Pg), O, OH, PR42R43, P(O)R42R43, P(S)R42R43,
P(NR44)R42R43;
W is a bond, CH, or a spacer group selected from the group: C1-C10 alkyl substituted with 0-3 R52, aryl substituted with 0-3 R52, cycloaklyl substituted with 0-3 R52, heterocycloalkyl substituted with 0-3 R52, aralkyl substituted with 0-3 R52 and alkaryl substituted with 0-3 R52; Wa is a C1-C10 alkyl group or a C3-C14
carbocycle;
R40, R41, R42, R43, and R44 are each
independently selected from the group: a bond to Ln, hydrogen, C1-C10 alkyl substituted with 0-3 R52, aryl
substituted with 0-3 R52, cycloaklyl substituted with 0-3 R52,
heterocycloalkyl substituted with 0-3 R52, aralkyl substituted with 0-3 R52, alkaryl substituted with 0-3
R52substituted with 0-3 R52 and an electron, provided that when one of R40 or R41 is an electron, then the other is also an electron, and provided that when one of R42 or R43 is an electron, then the other is also an electron; additionally, R40 and R41 may combine to form =C(C1-C3 alkyl) (C1-C3 alkyl);
R52 is independently selected at each
occurrence from the group: a bond to Ln, =O, F, Cl, Br, I, -CF3, -CN, -CO2R53, -C(=O)R53, -C(=O)N(R53)2, -CHO, -CH2OR53, -OC(=O)R53, -OC(=O)OR53a, -OR53,
-OC(=O)N(R53)2, -NR53C(=O)R53,
-NR54C(=O)OR53a, -NR53C(=O)N(R53)2,
-NR54SO2N(R53)2, -NR54SO2R53a, -SO3H, -SO2R53a, -SR53, -S(=O)R53a, -SO2N(R53)2, -N(R53)2, -NHC(=NH)NHR53, -C (=NH) NHR53, =NOR53, NO2, -C(=O)NHOR53,
-C(=O)NHNR53R53a, -OCH2CO2H,
2-(1-morpholino) ethoxy, C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, aryl substituted with 0-2 R53, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O;
R53, R53a, and R54 are independently selected at each occurrence from the group: a bond to Ln, C1-C6 alkyl, phenyl, benzyl, C1-C6 alkoxy, halide, nitro, cyano, and
trifluoromethyl; and Pg is a thiol protecting group capable of being displaced upon reaction with a radionuclide.
17. A reagent as in one of Claims 1-15, wherein Ch is selected from the group:
'
Figure imgf000393_0001
wherein : A1, A2, A3, A4, A5, A6, and A7 are
independently selected at each occurrence from the group: NR40R41, S, SH, S (Pg), OH;
W is a bond, CH, or a spacer group selected from the group: C1-C3 alkyl substituted with 0-3 R52;
Wa is a methylene group or a C3-C6 carbocycle;
R40, R41, R42, R43, and R44 are each
independently selected from the 'group: a bond to Ln, hydrogen, C1-C10 alkyl substituted with 0-3 R52, and an
electron, provided that when one of R40 or R41 is an electron, then the other is also an electron, and provided that when one of R42 or R43 is an electron, then the other is also an electron; additionally, R40 and R41 may combine to form, =C(C1-C3 alkyl) (C1-C3 alkyl);
R52 is independently selected at each
occurrence from the group: a bond to Ln, =O, F, Cl, Br, I, -CF3, -CN, -CO2R53, -C(=O)R53, -C(=O)N(R53)2, -CHO, -CH2OR53,
-OC(=O)R53, -OC(=O)OR53a, -OR53,
-OC(=O)N(R53)2, -NR53C(=O)R53,
-NR54C (=O) OR53a, -NR53C (=O) N (R53)2,
-NR54SO2N(R53)2, -NR54SO2R53a, -SO3H, -SO2R53a, -SR53, -S(=O)R53a, -SO2N(R53)2, -N(R53)2, -NHC(=NH)NHR53, -C (=NH) NHR53, =NOR53, NO2, -C(=O)NHOR53,
-C(=O)NHNR53R53a, -OCH2CO2H,
2-(1-morpholino) ethoxy; and
R53, R53a, and R54 are independently selected at each occurrence from the group: a bond to Ln, C1-C6 alkyl.
18. A reagent as in one of Claims 1-15, of formula:
(QLn)dCh, wherein d is 1; and Ch is selected from:
Figure imgf000395_0001
wherein: A1 and A4 are SH or SPg;
A2 and A3 are NR41;
W is independently selected from the
group:
CHR52, CH2CHR52, CH2CH2CHR52 and CHR52C=O; and
R41 and R52 are independently selected from hydrogen and a bond to Ln, and,
Figure imgf000396_0001
wherein:
A1 is NH2 or N=C (C1-C3 alkyl) (C1-C3
alkyl);
W is a bond;
A2 is NHR40, wherein R40 is heterocycle substituted with R52, wherein the heterocycle is selected from the group: pyridine, pyrazine, proline, furan, thiofuran, thiazole, and diazine, and R52 is a bond to Ln.
19. A reagent as in one of Claims 1-15, of formula:
(QLn)dCh, wherein d is 1; and wherein Ch is:
Figure imgf000396_0002
wherein:
A1 is NH2 or N=C(C1-C3 alkyl) (C1-C3 alkyl); W is a bond;
A2 is NHR40, wherein R40 is heterocycle
substituted with R52, wherein the heterocycle is selected from pyridine and thiazole, and R52 is a bond to Ln.
20. A reagent as in one of Claims 1-15, wherein Ln
is: a bond between Q and Ch; or,
a compound of formula: M1-[Y1(CR55R56)h(Z1)h"Y2)h'-M2 wherein:
Ml is -[(CH2)gZ1]g'-(CR55R56)g"-;
M2 is -(CR55R56)g"-[Zl(CH2)g]g--;
g is independently 0-10;
g' is independently 0-1;
g" is 0-10;
h is 0-10;
h" is 0-10;
h" is 0-1
Y1 and Y2, at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C (=O) O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3,
NHC(=O), (NH)2C(=O), (NH)2C=S; Z1 is independently selected at each occurrence from a C6-C14 saturated, partially saturated, or aromatic
carbocyclic ring system, substituted with 0-4 R57; a heterocyclic ring system, optionally substituted with 0-4 R57;
R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5
R57;
(C1-C10 alkyl) aryl wherein the aryl
is substituted with 0-5 R57;
R57 is independently selected at each
occurrence from the group: hydrogen, OH, NHR58, C(=O)R58, OC(=O)R58,
OC(=O)OR58, C(=O)OR58, C(=O)NR58-, C≡N, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively, when attached to an additional
molecule Q, R57 is independently selected at each occurrence from the group: O, NR58, C=O, C(=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO,
SO2, SO3, NHC(=O), (NH)2C(=O),
(NH)2C=S; and,
R58 is independently selected at each
occurrence from the group: hydrogen; C1- C6 alkyl; benzyl, and phenyl.
21. A reagent as in Claim 16, wherein Ln is: a compound of formula: M1-[Y1(CR55R56)h(Z1)h"Y2]h'-M2 wherein:
M1 is -[(CH2)gZ1]g'-(CR55R56)g"-;
M2 is -(CR55R56)g"-[Z1(CH2)g]g'-;
g is independently 0-10;
g' is independently 0-1;
g" is 0-10;
h is 0-10;
h' is 0-10;
h" is 0-1
Y1 and Y2, at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C (=O)O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3, NHC(=O), (NH)2C(=O), (NH)2C=S;
Z2 is independently selected at each
occurrence from a C6-C14 saturated, partially saturated, or aromatic carbocyclic ring system, .substituted with 0-4 R57; a heterocyclic ring system, optionally substituted with 0-4 R57; R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5 R57; (C1-C10 alkyl) aryl wherein the aryl is substituted with 0-5 R57;
R57 is independently selected at each occurrence from the group: hydrogen,
OH, NHR58, C(=O)R58, OC(=O)R58,
OC(=O)OR58, C(=O)OR58, C(=O)NR58-,
CΞN, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively,
when attached to an additional
molecule Q, R57 is independently
selected at each occurrence from the group: O, NR58, C=O, C (=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO,
SO2, SO3, NHC(=O), (NH)2C (=O),
(NH)2C=S, and R57 is attached to an additional molecule Q; and, R58 is independently selected at each occurrence from the group: hydrogen; C1-C6 alkyl; benzyl, and phenyl.
22. A reagent as in Claim 17, wherein Ln is:
-(CR55R56)g"- [Y1(CR55R56)hY2]h,-(CR55R56)g"-, wherein: g" is 1-10;
h is 0-10;
h" is 1-10;
Y1 and Y2, at each occurrence, are
independently selected from: a bond, O, NR56, C=O, C(=O)O,
OC(=O)O,
C(=O)NH-, C=NR56, S, SO, SO2, SO3,
NHC(=O), (NH)2C(=O), (NH)2C=S;
R55 and R56 are independently selected at
each occurrence from: hydrogen;
C1-C10 alkyl substituted with 0-5
R57;
(C1-C10 alkyl) aryl wherein the aryl
is substituted with 0-5 R57;
R57 is independently selected at each
occurrence from the group: hydrogen, OH, NHR58, C(=O)R58, OC(=O)R58, OC(=O)OR58, C(=O)OR58, C(=O)NR58-,
C≡N, SR58, SOR58, SO2R58,
NHC(=O)R58, NHC (=O) NHR58,
NHC (=S) NHR58; or, alternatively, when attached to an additional molecule Q, R57 is independently selected at each occurrence from the group: O, NR58, C=O, C (=O)O,
OC(=O)O, C(=O)N-, C=NR58, S, SO, SO2, SO3, NHC(=O), (NH)2C(=O), (NH)2C=S, and R57 is attached to an additional molecule Q; and,
R58 is independently selected at each occurrence from the group: hydrogen; C1-C6 alkyl; benzyl, and phenyl.
23. A reagent as in Claim 18, wherein Ln is:
-(CR55R56)g"-[Y1(CR55R56)hY2]h'-(CR55R56)g"-, wherein: g" is 1-5;
h is 0-5;
h' is 1-5;
Y1 and Y2, at each occurrence, are
independently selected from:
O, NR56, C=O, C(=O)O, OC(=O)O, C(=O)NH-, C=NR56, S, SO, SO2, SO3,
NHC(=O), (NH)2C(=O), (NH)2C=S;
R55 and R56 are independently selected at each occurrence from: hydrogen;
C1-C10 alkyl;
(C1-C10 alkyl) aryl.
24. A reagent as in Claim 19, wherein Ln is:
-(CR55R56)g"-[Y1(CR55R56)hY2]h'- (CR55R56)g"-, wherein: g" is 1-5;
h is 0-5;
h' is 1-5;
Y1 and Y2, at each occurrence, are
independently selected from: O, NR56, C=O, C(=O)O, OC(=O)O,
C(=O)NH-, C=NR56, S,
NHC(=O), (NH)2C(=O), (NH)2C=S;
R55 and R56 are independently selected at each occurrence from: hydrogen.
25. The reagents of Claim 1, which are:
Figure imgf000403_0001
Figure imgf000403_0002
Figure imgf000404_0001
Figure imgf000405_0001
26. A kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of Claim 23.
27. A kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of Claim 24.
28. A kit for preparing a radiopharmaceutical comprising a predetermined quantity of a sterile,
pharmaceutically acceptable reagent of Claim 25.
29. A radiopharmaceutical comprising a complex of a
reagent of Claims 1-15 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu,
43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
30. A radiopharmaceutical comprising a complex of a reagent of Claim 16 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111ln, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
31. A radiopharmaceutical comprising a complex of a
reagent of Claim 17 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
32. A radiopharmaceutical comprising a complex of a
reagent of Claim 18 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
33. A radiopharmaceutical comprising a complex of a
reagent of Claim 19 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Tl.
34. A radiopharmaceutical comprising a complex of a
reagent of Claim 20 and a radionuclide selected from the group 99mTc, 94mTc, 95Tc, 111In, 62Cu, 43Sc, 45Ti, 67Ga, 68Ga, 97Ru, 72As, 82Rb, and 201Ti.
35. A radiopharmaceutical comprising a complex of a
reagent of Claim 21 and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
36. A radiopharmaceutical comprising a complex of a
reagent of Claim 22 and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
37. A radiopharmaceutical comprising a complex of a reagent of Claim 23 and a radionuclide selected from the group 99mTc, 111In, and 62Cu.
38. A radiopharmaceutical comprising a complex of a
reagent of Claim 24 and a radionuclide selected from the group 99mTc, and 111In.
39. The radiopharmaceuticals of Claim 29, which are:
Figure imgf000407_0001
Figure imgf000408_0001
Figure imgf000409_0001
Figure imgf000410_0001
Figure imgf000411_0001
Figure imgf000412_0001
Figure imgf000413_0001
40. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 29, and (ii) scanning the mammal using a radioimaging devise.
41. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising
(i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 30, and (ii) scanning the mammal using a radioimaging devise.
42. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 31, and (ii) scanning the mammal using a radioimaging devise.
43. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 32, and (ii) scanning the mammal using a radioimaging devise.
44. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 33, and (ii) scanning the mammal using a radioimaging devise.
45. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 34, and (ii) scanning the mammal using a radioimaging devise.
46. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising
(i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 35, and (ii) scanning the mammal using a radioimaging devise.
47. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 36, and (ii) scanning t'he mammal using a radioimaging devise.
48. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 37, and (ii) scanning the mammal using a radioimaging devise.
49. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising
(i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 38, and (ii) scanning the mammal using a radioimaging devise.
50. A method for visualizing sites of platelet
deposition in a mammal by radioimaging, comprising (i) administering to said mammal an effective amount of a radiopharmaceutical of Claim 39, and (ii) scanning the mammal using a radioimaging devise.
51. A direct radiolabeled compound of formula (I):
Figure imgf000415_0001
or a pharmaceutically acceptable salt or
prodrug form thereof wherein: R31 is a C6-C14 saturated, partially saturated,
or aromatic carbocyclic ring system
substituted with 0-4 R10 or R10a;
R32 is selected from: -C (=O) - ;
-C (=S ) - -S (=O) 2- ;
-S (=O) -;
-P (=Z ) (ZR13) - ;
Z is S or O; n" and n' are independently 0-2;
R1 and R22 are independently selected from the following groups: hydrogen,
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11; C3-C10 cycloalkyl substituted with 0-2 R11; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12; =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C (=O) OR13a, -NR13C (=O) N (R13) 2, -NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH)NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy;
R1 and R21 can alternatively join to form a 3- 7 membered carbocyclic ring substituted with 0-2 R12; when n' is 2, R1 or R21 can alternatively be taken together with R1 or R21 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between said carbon atoms;
R22 and R23 can alternatively join to form a 3-7 membered carbocyclic ring substituted with 0-2 R12; when n" is 2 , R22 or R23 can
alternatively be taken together with R22 or R23 on an adjacent carbon atom to form a direct bond, thereby to form a double or triple bond between the adjacent carbon atoms;
R1 and R2, where R21 is H, can
alternatively join to form a 5-8 membered carbocyclic ring substituted with 0-2
R12;
R11 is selected from one or more of the
following: =O, F, Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13,
-OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13,
-C(=O)NHNR13R13a, -OCH2CO2H,
2-(1-morpholino) ethoxy,
C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, C3-C6 cycloalkoxy, C1-C4 alkyl (alkyl being substituted with 1-5 groups selected independently from:
-NR13R14, -CF3, NO2, -SO2R13a, or
-S(=O)R13a), aryl substituted with 0-2 R12, a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, said
heterocyclic ring being substituted with 0-2 R12; R12 is selected from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3- C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl) -OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C (=O) OR13a, -NR13C (=O) N (R13) 2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2,
C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino) ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a);
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form
-(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl; R21 and R23 are independently selected from: hydrogen;
C1-C4 alkyl, optionally substituted with 1-6 halogen;
benzyl;
R2 is H or C1-C8 alkyl; R10 and R10a are selected independently from one or more of the following: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3-
C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)N(R13)2, -C(=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B(R34) (R35), C3-C6 cycloalkoxy,
-OC(=O)R13, -C(=O)R13,-OC(=O)OR13a,
-OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13,
-NR13C (=O) OR13a, -NR13C (=O) N (R13) 2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H, -SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2,
C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl (including -CVFW where v = 1 to 3 and w = 1 to
(2v+1)), C1-C4 haloalkoxy, C1-C4
alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino, -OCH2CO2H, 2-(1-morpholino) ethoxy, C1-C4 alkyl
(alkyl being substituted with -N(R13)2, -CF3, NO2, or -S(=O)R13a); J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or C1-C8 alkyl;
R4 is H or C1-C3 alkyl; R5 is selected from:
hydrogen;
C1-C8 alkyl substituted with 0-2 R11; C2-C8 alkenyl substituted with 0-2 R11; C2-C8 alkynyl substituted with 0-2 R11 ; C3-C10 cycloalkyl substituted with 0-2
R11; aryl substituted with 0-2 R12; a 5-10-membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, or O, said
heterocyclic ring being substituted with 0-2 R12; =O, F,. Cl, Br, I, -CF3, -CN, -CO2R13, -C(=O)R13, -C(=O)N(R13)2, -CHO, -CH2OR13, -OC(=O)R13, -OC(=O)OR13a, -OR13,
-OC(=O)N(R13)2, -NR13C(=O)R13,
-NR14C(=O)OR13a, -NR13C(=O)N(R13)2,
-NR14SO2N(R13)2, -NR14SO2R13a, -SO3H, -SO2R13a, -SR13, -S(=O)R13a, -SO2N(R13)2, -N(R13)2, -NHC(=NH)NHR13, -C (=NH) NHR13, =NOR13, NO2, -C(=O)NHOR13, -C(=O)NHNR13R13a, =NOR13, -B (R34) (R35), -OCH2CO2H, 2-(1-morpholino) ethoxy, -SC(=NH)NHR13, N3, -Si(CH3)3, (C1-C5 alkyDNHR16;
-(C0-C6 alkyl) X;
Figure imgf000422_0001
independently 0,1;
Figure imgf000422_0002
-(CH2)mS(O)p' (CH2)2X, where m = 1,2 and p' = 0-2; wherein X is defined below; and
R3 and R4 may also be taken together to form (CH2)nX
Figure imgf000422_0003
I
-CH2CHCH2-, where
n 0, 1 and X is
Figure imgf000422_0004
R3 and R5 can alternatively be taken together to form -(CH2)t- or -CH2S(O)p'C(CH3)2-, where t = 2-4 and p' = 0-2; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5; R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is a D-isomer or L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is selected from:
- (C1-C7 alkyl) X;
Figure imgf000423_0001
wherein each q is independently 0-2 and
substitution on the phenyl is at the 3 or
4 position;
Figure imgf000423_0002
wherein each q is independently 0-2 and substitution on the cyclohexyl is at the 3 or 4 position;
Figure imgf000423_0003
-(CH2)mO- (C1-C4 alkyl)-X, where m = 1 or 2 ; -(CH2)mS(O)p'-(C1-C4 alkyl)-X, where m =
1 or 2 and p' = 0-2; and
X is selected from:
Figure imgf000424_0001
-C(=NH) (NH2); -SC (=NH)-NH2; -NH- C(=NH) (NHCN); -NH-C(=NCN) (NH2);
-NH-C(=N-OR13) (NH2); R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000424_0003
-(CH2)qCH (CH2)q-, w.herei.n eac.h q is independently 1 or 2 and wherein n = 0 or 1 and X is -NH2 or
Figure imgf000424_0002
L is -Y(CH2)vC(=O)-, wherein: Y is NH, N(C1-C3 alkyl), O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
-
Figure imgf000425_0001
wherein: q' is 0-2;
R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B (R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O),
-SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13;
R34 and R35 are independently selected from:
-OH,
-F,
-N(R13)2, or C1-C8-alkoxy;
R34 and R35 can alternatively be taken together form:
a cyclic boron ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms
independently selected from N, S, or O; a divalent cyclic boron amide where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from
N, S, or O;
a cyclic boron amide-ester where said chain or ring contains from 2 to 20 carbon atoms and, optionally, 1-4 heteroatoms independently selected from
N, S, or O; and wherein the radiolabel is selected from the group: 123I, 125I, 131I, 18F, 11C, 13N, 15 O, 75Br.
52. A radiolabeled compound of Claim 51, wherein:
R31 is bonded to (C(R23)R22)n" and
(C(R21)R1)n' at 2 different atoms on said carbocyclic ring.
53. A radiolabeled compound of Claim 51, wherein: n" is 0 and n' is 0;
n" is 0 and n' is 1; n" is 0 and n' is 2;
n" is 1 and n' is 0;
n" is 1 and n' is 1;
n" is 1 and n' is 2;
n" is 2 and n' is 0;
n" is 2 and n' is 1; or
n" is 2 and n' is 2.
54. A radiolabeled compound of Claim 51 wherein
R6 is methyl, ethyl, or propyl.
55. A radiolabeled compound of Claim 51, wherein: R31 is selected from the group consisting of:
(a) a 6 membered saturated, partially saturated or aromatic carbocyclic ring substituted with 0-3 R10 or R10a;
(b) a 8-11 membered saturated, partially saturated, or aromatic fused bicyclic carbocyclic ring substituted with 0-4 R10 or R10a; or
(c) a 14 membered saturated, partially saturated, or aromatic fused tricyclic carbocyclic ring substituted with 0-4 R10 or R10a.
56. A radiolabeled compound of Claim 51, wherein: R31 is selected from the group consisting of: (a) a 6 membered saturated, partially saturated, or aromatic carbocyclic ring of formula:
Figure imgf000428_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10;
(b) a 10 membered saturated, partially saturated, or aromatic bicyclic
carbocyclic ring of formula:
Figure imgf000428_0002
, wherein any of the bonds forming the carbocyclic ring may be a single or doubie bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10 or R10a;
(c) a 9 membered saturated, partially saturated, or aromatic bicyclic
carbocyclic ring of formula:
Figure imgf000429_0001
wherein any of the bonds forming the carbocyclic ring may be a single or double bond, and wherein said carbocyclic ring is substituted independently with 0-4 R10 or RlOa.
57. A radiolabeled compound of Claim 51, wherein:
R31 is selected from (the dashed bond may be a single or double bond) :
Figure imgf000429_0002
n" is 0 or 1 ; and n ' is 0-2 .
58. A radiolabeled compound of Claim 51, wherein:
R1 and R22 are independently selected from: phenyl, benzyl, phenethyl, phenoxy, benzyloxy, halogen, hydroxy, nitro, cyano, C1-C5 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C7-C10 arylalkyl, C1-C5 alkoxy, -CO2R13, -C (=O)NHOR13a, -C(=O)NHN(R13)2, =NOR13, -B (R34) (R35), C3-
C6 cycloalkoxy, -OC(=O)R13, -C(=O)R13,- OC(=O)OR13a, -OR13, -(C1-C4 alkyl)-OR13, -N(R13)2, -OC(=O)N(R13)2, -NR13C(=O)R13, -NR13C (=O)OR13a, -NR13C (=O) N (R13) 2,
-NR13SO2N(R13)2, -NR13SO2R13a, -SO3H,
-SO2R13a, -S(=O)R13a, -SR13, -SO2N(R13)2, C2-C6 alkoxyalkyl, methylenedioxy, ethylenedioxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 alkylcarbonyloxy, C1-C4 alkylcarbonyl, C1-C4 alkylcarbonylamino,
-OCH2CO2H, 2-(1-morpholino)ethoxy, C1-C4 alkyl (alkyl being substituted with
-N(R13)2, -CF3, NO2, or -S(=O)R13a).
59. A radiolabeled compound of Claim 51, wherein: R31 is selected from:
Figure imgf000431_0001
wherein R31 may be substituted
independently with 0-3 R10 or R10a;
R32 is -C(=O) n" is 0 or 1; n' is 0-2;
R1 and R22 are independently selected from H, C1-C4 alkyl, phenyl, benzyl,
phenyl-(C2-C4) alkyl, C1-C4 alkoxy;
R21 and R23 are independently H or C1-C4 alkyl; R2 is H or C1-C8 alkyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form
-(CH2)2-5- or -(CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl; R10 and R10a are selected independently from:
H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or CH3;
R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)SNHR16, where s = 3-5; or
R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
R3 and R5 can alternatively be taken together to form -(CH2)t- (t = 2-4) or
-CH2SC(CH3)2-; or R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl;
R7 is
Figure imgf000433_0001
0 or 1;
-(CH2)rX, where r = 3-6;
Figure imgf000434_0001
-(CH2)mS(CH2)2X, where m = 1 or 2;
-(C3-C7 alkyl) -NH-(C1-C6 alkyl)
Figure imgf000434_0002
-(CH2)m-O-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2;
- (CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2); or
R6 and R7 can alternatively be taken together to form
(CH2)nX
Figure imgf000434_0003
-CH2CHCH2-, where n = 0 or 1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y (CH2)vC (=O)-, wherein:
Y is NH, O, or S; and v = 1 or 2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000435_0001
wherein: q' is 0-2;
R17 is H, C1-C3 alkyl;
R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2,
-PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a, -NHCONHSO2R13a, -SO2NHCONHR13.
60. A radiolabeled compound of Claim 51 that is a radiolabeled 1,3-disubstituted phenyl the formula (II):
Figure imgf000436_0001
wherein: the shown phenyl ring in formula (II) may be further substituted with 0-3 R10;
R10 is selected independently from: H, C1-C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl, or
phenyl-(C1- C4) alkyl;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may
alternatively be taken together to form -(CH2)2-5- or -(CH2)O(CH2)-; R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure
-N(R3)C(R4) (R5)C(=O)-, wherein:
R3 is H or CH3; R4 is H or C1-C3 alkyl;
R5 is H, C1-C8 alkyl, C3-C6 cycloalkyl, C3- C6 cycloalkylmethyl, C1-C6
cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3,
CH2CH2SCH3, (CH2)SNH2,
-(CH2)SNHC(=NH) (NH2), - (CH2)SNHR16, where s = 3-5; or R16 is selected from:
an amine protecting group;
1-2 amino acids; or
1-2 amino acids substituted with an amine protecting group;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-; or
R4 and R5 can alternatively be taken together to form -(CH2)u-, where u = 2-5;
K is an L-isomer amino acid of structure
-N(R6)CH(R7)C(=O)-, wherein:
R6 is H or C1-C8 alkyl; R7 is:
Figure imgf000438_0001
0 or 1;
-(CH2)rX, where r = 3-6;
Figure imgf000438_0002
- (CH2)mS(CH2)2X, where m = 1 or 2,
-(C3-C7 alkyl)-NH-(C1-C6 alkyl)
Figure imgf000438_0003
-(CH2)m-O-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2;
-(CH2)m-S-(C1-C4 alkyl) -NH-(C1-C6 alkyl), where m = 1 or 2; and
X is -NH2 or -NHC(=NH) (NH2), provided that X is not -NH2 when r = 4; or R6 and R7 are alternatively be taken together to form
(CH2)nX
Figure imgf000439_0002
-CH2CHCH2-, where n = 0,1 and X is -NH2 or -NHC(=NH) (NH2);
L is -Y (CH2)VC(=O)-, wherein:
Y is NH, O, or S; and v = 1,2;
M is a D-isomer or L-isomer amino acid of
structure
Figure imgf000439_0001
wherein: q' is 0-2; R17 is H, C1-C3 alkyl; R8 is selected from:
-CO2R13,-SO3R13, -SO2NHR14, -B(R34) (R35), -NHSO2CF3, -CONHNHSO2CF3, -PO(OR13)2, -PO(OR13)R13, -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected independently from N, S, or O) , -SO2NH-heteroaryl (said heteroaryl being 5-10-membered and having 1-4 heteroatoms selected
independently from N, S, or O), -SO2NHCOR13, -CONHSO2R13a,
-CH2CONHSO2R13a, -NHSO2NHCOR13a,
-NHCONHSO2R13a, -SO2NHCONHR13.
61. A radiolabeled compound of Claim 51 that is a
radiolabeled 1, 3-disubstituted phenyl of the formula (II):
Figure imgf000440_0001
wherein: the phenyl ring in formula (II) may be further substituted with 0-3 R10 or R10a;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy;
R1 is H, C1-C4 alkyl, phenyl, benzyl, or phenyl- (C2- C4) alkyl;
R2 is H or methyl;
R13 is selected independently from: H, C1-C10
alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl,
C4-C12 alkylcycloalkyl, aryl, -(C1-C10
alkyl) aryl, or C3-C10 alkoxyalkyl;
R14 is OH, H, C1-C4 alkyl, or benzyl;
J is β-Ala or an L-isomer or D-isomer amino acid of structure -N(R3)C(R4)(R5)C(=O)-, wherein:
R3 is H or CH3; R4 is H;
R5 is H, C1-C6 alkyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C1-C6 cycloalkylethyl, phenyl, phenylmethyl, CH2OH, CH2SH, CH2OCH3, CH2SCH3, CH2CH2SCH3, (CH2)SNH2,
(CH2)SNHC(=NH) (NH2), (CH2)SR16, where s = 3-5;
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-;
R16 is selected from:
an amine protecting group;
1-2 amino acids;
1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of structure -N(R6)CH(R7)C(=O)-, wherein
R6 is H or C3-C8 alkyl;
R7 is
1;
Figure imgf000442_0001
-(CH2)rX, where r = 3-6;
Figure imgf000442_0002
~(CH2)mS(CH2)2X, where m = 1 or 2;
-(C4-C7 alkyl) -NH-(C1-C6 alkyl)
Figure imgf000442_0003
- (CH2)m-O-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2;
- (CH2)m-S-(C1-C4 alkyl)-NH-(C1-C6 alkyl), where m = 1 or 2; and X is -NH2 or -NHC (=NH) (NH2), provided that X is not -NH2 when r = 4; or
L is -YCH2C(=O)-, wherein:
Y is NH or O;
M is a D-isomer or L-isomer amino acid of structure
Figure imgf000443_0001
, wherein: q ' is 1 ;
R17 is H, C1-C3 alkyl ;
R8 is selected from:
-CO2H or -SO3R13,
62. A radiolabeled compound of Claim 51 that is a
radiolabeled compound of formula (II) above, wherein: the phenyl ring in formula (II) may be further
substituted with 0-2 R10 or R10a;
R10 or R10a are selected independently from: H, C1- C8 alkyl, phenyl, halogen, or C1-C4 alkoxy; R1 is H; R2 is H; R13 is selected independently from: H, C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl;
R13a is C1-C10 alkyl, C3-C10 cycloalkyl, C4-C12
alkylcycloalkyl, aryl, -(C1-C10 alkyl) aryl, or C3-C10 alkoxyalkyl; when two R13 groups are bonded to a single N, said R13 groups may alternatively be taken together to form -(CH2)2-5- or - (CH2)O(CH2)-;
R14 is OH, H, C1-C4 alkyl, or benzyl;
is β-Ala or an L-isomer or D-isomer amino acid of formula -N(R3)CH(R5)C(=O)-, wherein:
R3 is H and R5 is H, CH3, CH2CH3, CH(CH3)2,
CH(CH3)CH2CH3, CH2CH2CH3, CH2CH2CH2CH3,
CH2CH2SCH3, CH2CH(CH3)2, (CH2)4NH2, (C3-C5 alkyl) NHR16;
or
R3 is CH3 and R5 is H; or
R3 and R5 can alternatively be taken together to form -CH2CH2CH2-;
R16 is selected from:
an amine protecting group;
1-2 amino acids; 1-2 amino acids substituted with an amine protecting group;
K is an L-isomer amino acid of formula
-N(CH3)CH(R7)C(=O)-, wherein:
R7 is - (CH2)3NHC(=NH)(NH2);
L is -NHCH2C(=O)-; and
M is a D-isomer or L-isomer amino acid of structure
wherein:
Figure imgf000445_0001
q' is 1;
R4 is H or CH3;
R17 is H;
R8 is
-CO2H;
-SO3H .
63. A radiolabeled compound of Claim 51 that is a
radiolabeled compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein:
R1 and R2 are independently selected from H,
methyl; J is selected from D-Val, D-2-aminobutyric acid, D- Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, βAla,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala, Nε-p-azidobenzoyl-D-Lys, Nε-p- benzoylbenzoyl-D-Lys, Nε-tryptophanyl-D-Lys, Nε-o-benzylbenzoyl-D-Lys, Nε-p-acetylbenzoyl- D-Lys, Nε-dansyl-D-Lys, Nε-glycyl-D-Lys, Nε- glycyl-p-benzoylbenzoyl-D-Lys, Nε-p- phenylbenzoyl-D-Lys, Nε-m-benzoylbenzoyl-D-
Lys, Nε-o-benzoylbenzoyl-D-Lys;
K is selected from NMeArg, Arg; L is selected from Gly, β-Ala, Ala;
M is selected from Asp; MeAsp; βMeAsp; NMeAsp; D- Asp.
64. A radiolabeled compound of Claim 51 that is a
radiolabeled compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein:
R1 and R2 are independently selected from H,
methyl;
J is selected from: D-Val, D-2-aminobutyric acid, D-Leu, D-Ala, Gly, D-Pro, D-Ser, D-Lys, -βAla,
Pro, Phe, NMeGly, D-Nle, D-Phg, D-Ile, D-Phe, D-Tyr, Ala;
K is selected from NMeArg; L is Gly;
M is selected from Asp; βdeAsp; βMeAsp; NMeAsp;
D-Asp.
65. The radiolabeled compounds of Claim 51 that are: the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-2-aminobutyric acid; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Leu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Ala; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Gly; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Pro; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Lys; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is β-Ala; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is NMeGly; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is methyl (isomer 1); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is methyl (isomer 2); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 is phenyl (isomer 1); R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein J = D-Met, K = NMeArg, L = Gly, M = Asp, R1 = H, R2 = H; the radiolabeled compound of formula (II) wherein J = D-Abu, K = diNMe-guanidinyl-Orn ,
L = Gly, M = Asp, R1 = H, R2 = H; the radiolabeled compound of formula (II) wherein J = D-Abu, K = diNMe-Lys, L = Gly, M = Asp, R1 = H, R2 = H; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- azidobenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-tryptophanyl-
D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-o- benzylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp.
The radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- acetylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-dansyl-D-
Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-glycyl-D-
Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-glycyl-p- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-p- phenylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-m- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is Nε-o- benzoylbenzoyl-D-Lysine; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (III) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000451_0001
the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is D- NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Nle; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Phg; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Phe; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (V) wherein R1 and R2 are H; J is D-Ile; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000452_0001
the radiolabeled compound of formula (V) wherein n"=1; R1, R2, and R22 are H; J is D- Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (V) wherein n"=0; R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000452_0002
the radiolabeled compound of formula (VI) wherein R2 and R22 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000453_0001
the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is Cl; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R1 0 a is I; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is I; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is Me; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10a are H; R10 is Cl; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10a are H; R10 is MeO; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10a are H; R10 is Me; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1,R2, and R10 are H; R10a is Cl; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (VII) wherein R1, R2, and R10 are H; R10a is I; J is
D-Abu; K is NMeArg; L is Gly; and M is Asp.
The radiolabeled compound of formula (VII) wherein R1, R2, and R10 are H; R10a is Me; J is D-Abu; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Tyr; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is NMeAmf; L is Gly; and M is Asp; the radiolabeled compound of formula (II) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is βMeAsp; the radiolabeled compound of formula (II) wherein R1 is H; R2 is CH3; J is D-Val; K is
NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula (III) wherein R1 and R2 are H; J is D-Val; K is NMeArg; L is Gly; and M is Asp; the radiolabeled compound of formula
(VIII) wherein J is D-Val; K is NMeArg; L is Gly; and M is Asp;
Figure imgf000455_0001
66. A radiolabeled compound as in one of Claims
51-65 wherein the radiolabel is selected from the group: 18F, 11C, 123I, and 125I.
67. A radiolabeled compound of Claim 66 wherein
the radiolabel is 123I.
68. A radiopharmaceutical composition comprising a radiopharmaceutically acceptable carrier and a radiolabeled compound of any of Claims 51-67.
69. A method of determining platelet deposition in a mammal comprising administering to said mammal a radiopharmaceutical composition comprising a compound of any of Claims 51-67, and imaging said mammal.
70. A method of diagnosing a disorder associated with platelet deposition in a mammal
comprising administering to said mammal a radiopharmaceutical composition comprising a compound of any of Claims 51-67, and imaging said mammal.
PCT/US1994/003256 1993-03-30 1994-03-29 RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS WO1994022494A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
AU65248/94A AU6524894A (en) 1993-03-30 1994-03-29 Radiolabeled platelet gpiib/iiia receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders
UA95094341A UA32577C2 (en) 1993-03-30 1994-03-29 Radiolabeled compound as imaging agents for the diagnosis of arterial and venous thrombi, radiopharmaceuticals and radiopharmaceutical composition and kits for its preparation
BR9406055A BR9406055A (en) 1993-03-30 1994-03-29 Reagent for the preparation of radiopharmaceutical product sets radiopharmaceutical products methods for visualizing the deposition sites of the pallets radio-labeled compounds radiopharmaceutical composition method for determining platelet deposition and method for diagnosing a disorder
DE69425138T DE69425138T2 (en) 1993-03-30 1994-03-29 RADIOACTIVELY MARKED BLOOD PLATE GPIIb / IIIa RECEPTOR ANTAGONISTS AS IMAGING ACTIVE SUBSTANCES FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISEASES
RO95-01701A RO114895B1 (en) 1993-03-30 1994-03-29 Radiopharmaceutical product and reagent for preparing such product, method for viewing mammal platelet deposit position and radiolabeled compound
JP6522205A JP3042887B2 (en) 1993-03-30 1994-03-29 Radiolabeled platelet GP IIb / IIIa receptor antagonist as imaging agent for diagnosing thromboembolic disease
AT94912870T ATE194293T1 (en) 1993-03-30 1994-03-29 RADIOACTIVELY LABELED PLATEET GPIIB/IIIA RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR DIAGNOSING THROMBOEMBOLIC DISEASES
RU95118183A RU2145608C1 (en) 1993-03-30 1994-03-29 Cyclic peptides, radiopharmaceutical preparation, and radioactively labeled cyclic peptides
EP94912870A EP0692982B1 (en) 1993-03-30 1994-03-29 RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS
DK94912870T DK0692982T3 (en) 1993-03-30 1994-03-29 Radiolabeled platelet GPIIb / IIIa receptor antagonists as imaging agents for the diagnosis of thromboembolic disorder
BR9406820A BR9406820A (en) 1993-03-30 1994-03-29 Reagent for the preparation of radiopharmaceutical product sets radiopharmaceutical products methods for visualizing the deposition sites of the pallets radio-labeled compounds radiopharmaceutical composition method for determining platelet deposition and method for diagnosing a disorder
BG100036A BG100036A (en) 1993-03-30 1995-09-29 RADIOLABELED PLATELED GPIIb/IIIa RECEPTOR ANTAGONISTA AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS
NO953886A NO307568B1 (en) 1993-03-30 1995-09-29 Compound suitable for radiolabeling, kit for the manufacture of a radiopharmaceutical and this pharmaceutical, direct radiolabelled compound and radiopharmaceutical preparation
FI954655A FI954655A (en) 1993-03-30 1995-09-29 Radiolabeled GPIIb / IIIa platelet receptor antagonists as imaging agents in the diagnosis of thromboembolic disorders
KR1019950704228A KR960701666A (en) 1993-03-30 1995-09-29 RADIOLABELED PLATELET GP Ⅱb / IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS
LVP-95-296A LV11106B (en) 1993-03-30 1995-10-27 Radiolabeled platelet gpiib/iiia receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders
GR20000401959T GR3034272T3 (en) 1993-03-30 2000-08-30 RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US4033693A 1993-03-30 1993-03-30
US08/040,336 1993-03-30
US08/218,861 1994-03-28
US08/218,861 US5879657A (en) 1993-03-30 1994-03-28 Radiolabeled platelet GPIIb/IIIa receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders

Publications (1)

Publication Number Publication Date
WO1994022494A1 true WO1994022494A1 (en) 1994-10-13

Family

ID=21910453

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/003256 WO1994022494A1 (en) 1993-03-30 1994-03-29 RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS

Country Status (26)

Country Link
US (2) US5879657A (en)
EP (2) EP0692982B1 (en)
JP (1) JP3042887B2 (en)
KR (1) KR960701666A (en)
CN (1) CN1122577A (en)
AT (1) ATE194293T1 (en)
AU (2) AU6524894A (en)
BG (1) BG100036A (en)
BR (2) BR9406055A (en)
CA (1) CA2159445A1 (en)
DE (1) DE69425138T2 (en)
DK (1) DK0692982T3 (en)
ES (1) ES2149266T3 (en)
FI (1) FI954655A (en)
GR (1) GR3034272T3 (en)
HU (1) HUT72889A (en)
LV (1) LV11106B (en)
NO (1) NO307568B1 (en)
NZ (1) NZ263970A (en)
PL (1) PL178252B1 (en)
PT (1) PT692982E (en)
RU (1) RU2145608C1 (en)
TW (1) TW445267B (en)
UA (1) UA32577C2 (en)
WO (1) WO1994022494A1 (en)
ZA (1) ZA942262B (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996031243A1 (en) * 1995-04-03 1996-10-10 The Du Pont Merck Pharmaceutical Company Ternary radiopharmaceutical complexes
WO1996040637A1 (en) * 1995-06-07 1996-12-19 The Du Pont Merck Pharmaceutical Company Stable reagents for the preparation of radiopharmaceuticals
WO1997033627A2 (en) * 1996-03-13 1997-09-18 Du Pont Pharmaceuticals Company New ternary radiopharmaceutical complexes
US5736122A (en) * 1991-02-08 1998-04-07 Diatide, Inc. Technetium-99m labeled peptides for thrombus imaging
WO1998014220A2 (en) * 1996-10-02 1998-04-09 Du Pont Pharmaceuticals Company TECHNETIUM-99m LABELED CHELATOR INCORPORATED CYCLIC PEPTIDES
US5849260A (en) * 1991-02-08 1998-12-15 Diatide, Inc. Technetium-99M labeled peptides for thrombus imaging
US5879659A (en) * 1996-03-13 1999-03-09 Dupont Pharmaceuticals Company Ternary radiopharmaceutical complexes
WO1999040882A2 (en) * 1998-02-06 1999-08-19 Trustees Of The University Of Pennsylvania STEREOSELECTIVE Tc-99m LIGANDS
US5968476A (en) * 1992-05-21 1999-10-19 Diatide, Inc. Technetium-99m labeled peptides for thrombus imaging
US5968979A (en) * 1995-02-07 1999-10-19 Brusilow Enterprises Llc Triglycerides and ethyl esters of phenylalkanoic acid and phenylalkenoic acid useful in treatment of various disorders
US5997845A (en) * 1991-02-08 1999-12-07 Diatide, Inc. Technetium-99M labeled peptides for imaging inflammation
WO2000021980A1 (en) * 1998-10-13 2000-04-20 Du Pont Pharmaceuticals Company Chelator incorporated arg-gly-asp (rgd) mimetic synthetic disintegrins as imaging agents
US6056940A (en) * 1993-04-08 2000-05-02 Diatide, Inc. Radiolabeled compounds for thrombus imaging
WO2000057787A2 (en) * 1999-03-26 2000-10-05 Du Pont Pharmaceuticals Company Method for localization of blood clots
US6132697A (en) * 1996-06-10 2000-10-17 G. D. Searle & Co. Radiopharmaceutical compositions capable of localizing at sites of thrombus
US6254852B1 (en) 1999-07-16 2001-07-03 Dupont Pharmaceuticals Company Porous inorganic targeted ultrasound contrast agents
EP1293214A2 (en) * 1996-10-07 2003-03-19 Bristol-Myers Squibb Pharma Company LTB4 antagonists and radiopharmaceuticals for imaging infection and inflammation
US6808698B1 (en) 1999-03-26 2004-10-26 Bristol-Myers Squibb Pharma Company Method for localization of blood clots
WO2015016616A1 (en) 2013-07-30 2015-02-05 연세대학교 산학협력단 Saxatilin-fc fusion protein and use thereof
US9095559B2 (en) 2011-09-30 2015-08-04 Horizon Therapeutics, Inc. Methods of therapeutic monitoring of nitrogen scavenging drugs
US9289406B2 (en) 2012-11-21 2016-03-22 Horizon Therapeutics, Inc. Methods of administering and evaluating nitrogen scavenging drugs for the treatment of hepatic encephalopathy
US9561197B2 (en) 2012-04-20 2017-02-07 Horizon Therapeutics, Llc Methods of therapeutic monitoring of phenylacetic acid prodrugs
US9914692B2 (en) 2016-05-25 2018-03-13 Horizon Therapeutics, Llc Procedure for the preparation of 4-phenyl butyrate and uses thereof
US10668040B2 (en) 2017-09-11 2020-06-02 Horizon Therapeutics, Llc Treatment of urea cycle disorders in neonates and infants

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6083481A (en) * 1992-05-21 2000-07-04 Diatide, Inc. Thrombus imaging agents
US5879657A (en) * 1993-03-30 1999-03-09 The Dupont Merck Pharmaceutical Company Radiolabeled platelet GPIIb/IIIa receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders
US6171578B1 (en) * 1999-04-14 2001-01-09 Diatide, Inc. Benzodiazepine derivatives for imaging thrombi
US6685914B1 (en) * 1999-09-13 2004-02-03 Bristol-Myers Squibb Pharma Company Macrocyclic chelants for metallopharmaceuticals
US6518244B2 (en) 2000-03-09 2003-02-11 Intimax Corporation Combinations of heparin cofactor II agonist and platelet IIb/IIIa antagonist, and uses thereof
US7045286B2 (en) 2000-07-25 2006-05-16 The Trustees Of The University Of Pennsylvania Methods of detecting molecules expressing selected epitopes via fluorescent dyes
US6743592B1 (en) 2000-07-25 2004-06-01 The Trustees Of The University Of Pennsylvania Methods, systems and kits for immuno-detection of epitopes expressed on molecules
US7524628B2 (en) * 2000-07-25 2009-04-28 The Trustees Of The University Of Pennsylvania Method for detecting molecules expressing a selected epitope via fluorescent dyes
US7341831B2 (en) * 2001-07-18 2008-03-11 The Trustees Of The University Of Pennsylvania Method for immuno-detection of epitopes
AU2003233662B2 (en) 2002-05-23 2010-04-01 Trustees Of The University Of Pennsylvania Fas peptide mimetics and uses thereof
US7319149B2 (en) 2003-06-13 2008-01-15 Bristol-Myers Squibb Pharma Company Chelants and macrocyclic metal complex radiopharmaceuticals thereof
US7317104B2 (en) 2003-06-13 2008-01-08 Bristol-Myers Squibb Pharma Company Chelants and macrocyclic metal complex radiopharmaceuticals thereof
CN101913973B (en) * 2003-07-24 2013-11-20 皇后医学中心 Preparation and use of alkylating agents
CA2539384C (en) * 2003-09-17 2012-08-28 Board Of Regents, The University Of Texas System Mechanism-based targeted pancreatic beta cell imaging and therapy
WO2005081908A2 (en) * 2004-02-20 2005-09-09 The Trustees Of The University Of Pennsyvania Reagents, kits and methods for immunodetection of epitopes on molecules
WO2005081898A2 (en) 2004-02-20 2005-09-09 The Trustees Of The University Of Pennsylvania Binding peptidomimetics and uses of the same
EP1719529A4 (en) * 2004-02-25 2008-05-21 Astellas Pharma Inc Contrast medium for thrombus formation
KR101699142B1 (en) 2004-06-18 2017-01-23 암브룩스, 인코포레이티드 Novel antigen-binding polypeptides and their uses
US7292198B2 (en) * 2004-08-18 2007-11-06 Ruckus Wireless, Inc. System and method for an omnidirectional planar antenna apparatus with selectable elements
FI20050753A (en) 2004-09-03 2006-03-04 Licentia Oy New peptides
US8691761B2 (en) * 2006-10-16 2014-04-08 Jean E. F. Rivier Somatostatin receptor 2 antagonists
EP2433963B1 (en) 2006-10-16 2014-06-04 The Salk Institute for Biological Studies Receptor (SSTR2)-selective somatostatin antagonists
DK2474525T3 (en) 2006-12-26 2020-07-13 Lantheus Medical Imaging Inc Ligands for imaging cardiac innervation
EP2150248A4 (en) 2007-01-16 2011-06-29 Univ Johns Hopkins Glutamate receptor antagonists and methods of use
CA2699394C (en) 2007-09-17 2020-03-24 The Regents Of The University Of California Internalizing human monoclonal antibodies targeting prostate cancer cells in situ
GB2453589A (en) 2007-10-12 2009-04-15 King S College London Protease inhibition
AU2012229156A1 (en) 2011-03-11 2013-10-31 Board Of Regents Of The University Of Nebraska Biomarker for coronary artery disease
WO2017095823A1 (en) 2015-11-30 2017-06-08 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
CN116419747A (en) 2020-08-07 2023-07-11 福蒂斯治疗公司 CD46 targeting immunoconjugates and methods of use thereof

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0341915A2 (en) * 1988-05-09 1989-11-15 Smithkline Beecham Corporation Anti-aggregatory peptides
WO1991002750A1 (en) * 1989-08-18 1991-03-07 Biogen, Inc. Novel inhibitors of thrombin
EP0425212A2 (en) * 1989-10-23 1991-05-02 Smithkline Beecham Corporation Cyclic anti-aggregatory peptides
US5023233A (en) * 1989-07-28 1991-06-11 Merck & Co., Inc. Fibrinogen receptor antagonists
US5041380A (en) * 1982-08-04 1991-08-20 La Jolla Cancer Research Foundation Tetrapeptide
US5192380A (en) * 1989-12-19 1993-03-09 The Yokohama Rubber Co., Ltd. Pneumatic radial tire for passenger vehicle including a particular carcass line
US5192746A (en) * 1990-07-09 1993-03-09 Tanabe Seiyaku Co., Ltd. Cyclic cell adhesion modulation compounds
US5192745A (en) * 1987-05-21 1993-03-09 Merrell Dow Pharmaceuticals Inc. Cyclic anticoagulant peptides
WO1993007170A1 (en) * 1991-09-30 1993-04-15 The Du Pont Merck Pharmaceutical Company CYCLIC COMPOUNDS USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa
US5236898A (en) * 1987-05-21 1993-08-17 Merrell Dow Pharmaceuticals Inc. Cyclic anticoagulant peptides
WO1993015770A1 (en) * 1992-02-05 1993-08-19 Mallinckrodt Medical, Inc. Radiolabelled peptide compounds
US5279812A (en) * 1989-10-03 1994-01-18 Merrell Dow Pharmaceuticals Inc. Radiolabeled anticoagulant peptides

Family Cites Families (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444690A (en) * 1982-02-25 1984-04-24 University Patents, Inc. Technetium chelates
US4614517A (en) * 1982-08-04 1986-09-30 La Jolla Cancer Research Foundation Tetrapeptide
US4517686A (en) * 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4589881A (en) * 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4661111A (en) * 1982-08-04 1987-04-28 La Jolla Cancer Research Foundation Polypeptide
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
US4673562A (en) * 1983-08-19 1987-06-16 The Children's Medical Center Corporation Bisamide bisthiol compounds useful for making technetium radiodiagnostic renal agents
US4638051A (en) * 1984-04-30 1987-01-20 The Johns Hopkins University Brain imaging radiopharmaceuticals
US4670545A (en) * 1984-05-11 1987-06-02 University Patents, Inc. Chelating agents for technetium-99M
US4963688A (en) * 1984-07-19 1990-10-16 University Of Florida Compounds for site-enhanced delivery of radionuclides and uses thereof
US4897255A (en) * 1985-01-14 1990-01-30 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
US5175343A (en) * 1985-01-14 1992-12-29 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
CA1336076C (en) * 1985-01-14 1995-06-27 Alan R. Fritzberg Metal radionuclide labeled proteins for diagnosis and therapy
US4746505A (en) * 1985-04-26 1988-05-24 President And Fellows Of Harvard College Technetium radiodiagnostic fatty acids derived from bisamide bisthiol ligands
US4988621A (en) * 1985-05-24 1991-01-29 La Jolla Cancer Research Foundation Peptides in cell detachment and aggregation
US4879237A (en) * 1985-05-24 1989-11-07 La Jolla Cancer Research Foundation Use of peptides in control of cell attachment and detachment
US5082930A (en) * 1986-05-29 1992-01-21 Mallinckrodt Medical, Inc. Coupling agents for joining radionuclide metal ions with biologically useful proteins
US4861869A (en) * 1986-05-29 1989-08-29 Mallinckrodt, Inc. Coupling agents for joining radionuclide metal ions with biologically useful proteins
US5279811A (en) * 1987-02-18 1994-01-18 The Du Pont Merck Pharmaceutical Company Ester-substituted diaminedithiols and radiolabeled complexes thereof
US4965392A (en) * 1987-03-26 1990-10-23 Neorx Corporation Chelating compounds for metal-radionuclide labeled proteins
EP0284071B1 (en) * 1987-03-26 1994-06-08 Neorx Corporation Metal-radionuclide-labeled proteins and glycoproteins for diagnosis and therapy
WO1989000051A1 (en) * 1987-07-07 1989-01-12 Cytrx Biopool Ltd. Fibrin-binding compound and method
DE3728600A1 (en) * 1987-08-27 1989-03-09 Hoechst Ag METHOD FOR MARKING SUBSTANCES WITH TECHNETIUM OR RHENIUM
CA1335725C (en) * 1987-09-25 1995-05-30 Stephen W. Hadley Method of diagnosing blood clots using fibrin-binding proteins
EP0394326B1 (en) * 1987-12-10 1996-07-31 La Jolla Cancer Research Foundation Methods for the production of conformationally stabilized cell adhesion peptides
ATE124269T1 (en) * 1988-02-09 1995-07-15 Mallinckrodt Inc METHOD FOR PRODUCING A METAL RADIONUCLIDE LABELED PROTEIN.
US5202451A (en) * 1988-02-17 1993-04-13 Neorx Corporation Anchimeric radiometal chelating compounds
CA1324954C (en) * 1988-03-10 1993-12-07 Erkki I. Ruoslahti Inhibition of cell migration with synthetic peptides
US4883862A (en) * 1988-04-13 1989-11-28 Albert Einstein College Of Medicine - Of Yeshiva University Mercaptosuccinyl glycyl-glycyl-glycine a complex thereof with Tc-99m, and methods of making the same
WO1989010759A1 (en) * 1988-04-29 1989-11-16 Mallinckrodt, Inc. Diaminedithiol chelating agents for radiopharmaceuticals
KR940009084B1 (en) * 1988-05-18 1994-09-29 체크 포인트 시스템스, 인코오퍼레이티드 Antenna system for magnetic and resonant circuit detection
US4988496A (en) * 1988-05-31 1991-01-29 Neorx Corporation Metal radionuclide chelating compounds for improved chelation kinetics
US5218128A (en) * 1988-06-15 1993-06-08 Centocor, Inc. Bifunctional coupling agents and radionuclide labeled compositions prepared therefrom
US5144043A (en) * 1988-06-15 1992-09-01 Centocor Cleavable bifunctional coupling agents
AU3876789A (en) * 1988-06-20 1990-01-12 Washington University Use of modified tpa to detect blood clots
US5180816A (en) * 1988-08-24 1993-01-19 Centocor One vial method for labeling protein/linker conjugates with technetium-99M
US4925650A (en) * 1988-11-16 1990-05-15 Mallinckrodt, Inc. Technetium -99m complex for examining the renal function
MY106120A (en) * 1988-12-05 1995-03-31 Novartis Ag Peptide derivatives.
KR910700063A (en) * 1988-12-20 1991-03-13 알. 더글라스 암스트롱 Polypeptide-polymer conjugates having wound healing activity
US5270030A (en) * 1988-12-29 1993-12-14 Bio-Technology General Corp. Fibrin binding domain polypeptide and method of producing
GB8902362D0 (en) * 1989-02-03 1989-03-22 Amersham Int Plc Cationic complexes of technetium-99m
US5053503A (en) * 1989-02-17 1991-10-01 Centocor Chelating agents
US5095111A (en) * 1989-03-17 1992-03-10 The John Hopkins University Thiolactone bifunctional chelating agents for diagnostic and therapeutic products
US5120829A (en) * 1989-03-20 1992-06-09 La Jolla Cancer Research Foundation Hydrophobic attachment site for adhesion peptides
FR2655339B2 (en) * 1989-04-19 1992-04-10 Medgenix Group Sa COMPOUNDS AND COMPLEXES USEFUL IN PARTICULAR IN MEDICAL IMAGING.
CA2031528C (en) * 1989-06-16 1997-03-18 Linda M. Gustavson Radionuclide metal chelates for the radiolabeling of proteins
US5250666A (en) * 1989-06-16 1993-10-05 Neorx Corporation Radionuclide metal chelates for the radiolabeling of proteins
GB8914020D0 (en) * 1989-06-19 1989-08-09 Antisoma Ltd Synthetic peptides for use in thrombus detection
FR2650672A1 (en) * 1989-08-02 1991-02-08 Medgenix Group Sa PROCESS FOR MARKING PROTEINS OR POLYPEPTIDES WITH TECHNETIUM-99M, CONJUGATE OBTAINED, USE AS IMAGING AGENT, KIT FOR RECONSTITUTION OF THESE CONJUGATES
EP0489061A4 (en) * 1989-08-24 1992-10-14 Australian Nuclear Science And Technology Organisation Radio-labelled antibodies for imaging
GB8919488D0 (en) * 1989-08-29 1989-10-11 Amersham Int Plc New cores for technetium radiopharmaceuticals
US4952562A (en) * 1989-09-29 1990-08-28 Rorer Pharmaceutical Corporation Anti-thrombotic peptides and pseudopeptides
EP0593452A1 (en) * 1989-11-30 1994-04-27 Mallinckrodt Medical, Inc. Method for preparing a metal-radionuclide-labelled protein
US5080884A (en) * 1989-12-12 1992-01-14 Medi-Physics, Inc. Hydrocarbylphenyl diaminodithiol radionuclide complexes and their use in imaging
EP0432988B1 (en) * 1989-12-12 1994-02-16 AMERSHAM INTERNATIONAL plc Hydrocarbylphenyl diaminodithiol derivatives
US5026913A (en) * 1989-12-12 1991-06-25 Medi-Physics, Inc. Hydrocarbylphenyl diaminodithiol derivatives
CA2079606A1 (en) * 1990-04-06 1991-10-07 Michael D. Pierschbacher Method and composition for treating thrombosis
CS136091A3 (en) * 1990-05-10 1992-04-15 Zymo Genetics Agents for determining thrombi and their application
WO1992005154A1 (en) * 1990-09-14 1992-04-02 Mallinckrodt Medical, Inc. Novel nitrogen-sulfur ligands useful in radiographic imaging agents
US5175256A (en) * 1990-09-28 1992-12-29 Neorx Corporation Protein labeling reagents
US5116598A (en) * 1990-10-29 1992-05-26 Mallinckrodt Medical, Inc. N4 technetium-99 m complexes for use as radiopharmaceuticals
US5037631A (en) * 1990-10-29 1991-08-06 Mallinckrodt Medical, Inc. Technetium-99M complex for examinating the renal function
WO1992007870A1 (en) * 1990-11-02 1992-05-14 Genentech, Inc. Platelet aggregation inhibitors
US5965107A (en) * 1992-03-13 1999-10-12 Diatide, Inc. Technetium-99m labeled peptides for imaging
US5443815A (en) * 1991-11-27 1995-08-22 Diatech, Inc. Technetium-99m labeled peptides for imaging
DK0570493T3 (en) * 1991-02-08 2000-06-26 Diatide Inc Technetium-99m-labeled polypeptides for imaging.
WO1992016520A1 (en) * 1991-03-20 1992-10-01 British Technology Group Ltd Macrocyclic thioether ligands and their use as intermediates for binding ions to substrates
WO1992019274A1 (en) * 1991-05-08 1992-11-12 Mallinckrodt Medical, Inc. Technetium chelates to be used for determining the renal function
US5225180A (en) * 1991-09-10 1993-07-06 Diatech, Inc. Technetium-99m labeled somatostatin-derived peptides for imaging
US5635477A (en) * 1991-09-30 1997-06-03 The Dupont Merck Pharmaceutical Company Cyclic compounds useful as inhibitors of platelet glycoprotein IIB/IIIA
EP0629133B1 (en) * 1992-01-03 2000-11-29 Rhomed, Incorporated Peptide-metal ion pharmaceutical applications
EP0630264B1 (en) * 1992-02-06 2002-11-06 BioSynthema Inc. Ligands for improving metal chelate formation kinetics
AU683015B2 (en) * 1992-03-13 1997-10-30 Diatech, Inc. Technetium-99m labeled peptides for imaging inflammation
US5326856A (en) * 1992-04-09 1994-07-05 Cytogen Corporation Bifunctional isothiocyanate derived thiocarbonyls as ligands for metal binding
GB9209641D0 (en) * 1992-05-02 1992-06-17 Johnson Matthey Plc Improvements in radiolabelling
EP0641222B1 (en) * 1992-05-21 2000-09-06 Diatide, Inc. TECHNETIUM-99m LABELED PEPTIDES FOR THROMBUS IMAGING
EP0672059A1 (en) * 1992-11-18 1995-09-20 The Du Pont Merck Pharmaceutical Company CYCLIC COMPOUNDS LINKED BY A HETEROCYCLIC RING USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa
AU6415894A (en) * 1993-03-29 1994-10-24 Du Pont Merck Pharmaceutical Company, The Cyclic compounds useful as inhibitors of platelet glycoprotein iib/iiia
US5879657A (en) * 1993-03-30 1999-03-09 The Dupont Merck Pharmaceutical Company Radiolabeled platelet GPIIb/IIIa receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders
JPH09501910A (en) * 1993-06-18 1997-02-25 ラ ホヤ キャンサー リサーチ ファウンデイション Methods and compositions for the treatment of thrombosis

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041380A (en) * 1982-08-04 1991-08-20 La Jolla Cancer Research Foundation Tetrapeptide
US5192745A (en) * 1987-05-21 1993-03-09 Merrell Dow Pharmaceuticals Inc. Cyclic anticoagulant peptides
US5236898A (en) * 1987-05-21 1993-08-17 Merrell Dow Pharmaceuticals Inc. Cyclic anticoagulant peptides
EP0341915A2 (en) * 1988-05-09 1989-11-15 Smithkline Beecham Corporation Anti-aggregatory peptides
US5023233A (en) * 1989-07-28 1991-06-11 Merck & Co., Inc. Fibrinogen receptor antagonists
WO1991002750A1 (en) * 1989-08-18 1991-03-07 Biogen, Inc. Novel inhibitors of thrombin
US5279812A (en) * 1989-10-03 1994-01-18 Merrell Dow Pharmaceuticals Inc. Radiolabeled anticoagulant peptides
EP0425212A2 (en) * 1989-10-23 1991-05-02 Smithkline Beecham Corporation Cyclic anti-aggregatory peptides
US5192380A (en) * 1989-12-19 1993-03-09 The Yokohama Rubber Co., Ltd. Pneumatic radial tire for passenger vehicle including a particular carcass line
US5192746A (en) * 1990-07-09 1993-03-09 Tanabe Seiyaku Co., Ltd. Cyclic cell adhesion modulation compounds
WO1993007170A1 (en) * 1991-09-30 1993-04-15 The Du Pont Merck Pharmaceutical Company CYCLIC COMPOUNDS USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa
WO1993015770A1 (en) * 1992-02-05 1993-08-19 Mallinckrodt Medical, Inc. Radiolabelled peptide compounds

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736122A (en) * 1991-02-08 1998-04-07 Diatide, Inc. Technetium-99m labeled peptides for thrombus imaging
US6248304B1 (en) 1991-02-08 2001-06-19 Berlex Laboratories, Inc. Scintigraphic imaging agents
US5997845A (en) * 1991-02-08 1999-12-07 Diatide, Inc. Technetium-99M labeled peptides for imaging inflammation
US5879658A (en) * 1991-02-08 1999-03-09 Diatide, Inc. Technetium-99m labeled peptides for thrombus imaging
US5849260A (en) * 1991-02-08 1998-12-15 Diatide, Inc. Technetium-99M labeled peptides for thrombus imaging
US5968476A (en) * 1992-05-21 1999-10-19 Diatide, Inc. Technetium-99m labeled peptides for thrombus imaging
US5744120A (en) * 1993-03-30 1998-04-28 The Dupont Merick Pharmaceutical Company Ternary radiopharmaceutical complexes
US6056940A (en) * 1993-04-08 2000-05-02 Diatide, Inc. Radiolabeled compounds for thrombus imaging
US6060510A (en) * 1995-02-07 2000-05-09 Brusilow Enterprises Llc Triglycerides and ethyl esters of phenylalkanoic acid and phenylalkenoic acid useful in the treatment of various disorders
US6083984A (en) * 1995-02-07 2000-07-04 Brusilow Enterprises Llc Triglycerides and ethyl esters of phenylalkanoic acid and phenylalkenoic acid useful in the treatment of various disorders
US5968979A (en) * 1995-02-07 1999-10-19 Brusilow Enterprises Llc Triglycerides and ethyl esters of phenylalkanoic acid and phenylalkenoic acid useful in treatment of various disorders
EP1195168A3 (en) * 1995-04-03 2007-05-30 Bristol-Myers Squibb Pharma Company Ternary radiopharmaceutical complexes
EP0820312A4 (en) * 1995-04-03 1998-03-18 Du Pont Merck Pharma Ternary radiopharmaceutical complexes
WO1996031243A1 (en) * 1995-04-03 1996-10-10 The Du Pont Merck Pharmaceutical Company Ternary radiopharmaceutical complexes
LT4391B (en) 1995-04-03 1998-10-26 Dupont Pharmaceuticals Company Ternary radiopharmaceutical complexes
EP0820312A1 (en) * 1995-04-03 1998-01-28 The Du Pont Merck Pharmaceutical Company Ternary radiopharmaceutical complexes
CN1080127C (en) * 1995-04-03 2002-03-06 杜邦药品公司 Ternary radiopharmaceutical complexes
AU719529B2 (en) * 1995-04-03 2000-05-11 Du Pont Pharmaceuticals Company Ternary radiopharmaceutical complexes
EA000636B1 (en) * 1995-04-03 1999-12-29 Дзе Дюпон Мерк Фармасьютикал Компани Ternary radiopharmaceutical complexes
EP1195168A2 (en) * 1995-04-03 2002-04-10 Dupont Pharmaceuticals Company Ternary radiopharmaceutical complexes
WO1996040637A1 (en) * 1995-06-07 1996-12-19 The Du Pont Merck Pharmaceutical Company Stable reagents for the preparation of radiopharmaceuticals
LT4380B (en) 1995-06-07 1998-09-25 Dupont Pharmaceuticals Company Stable reagents for the preparation of radiopharmaceuticals
AU718683B2 (en) * 1995-06-07 2000-04-20 Du Pont Pharmaceuticals Company Stable reagents for the preparation of radiopharmaceuticals
WO1997033627A3 (en) * 1996-03-13 1998-02-26 Du Pont Merck Pharma New ternary radiopharmaceutical complexes
WO1997033627A2 (en) * 1996-03-13 1997-09-18 Du Pont Pharmaceuticals Company New ternary radiopharmaceutical complexes
US5879659A (en) * 1996-03-13 1999-03-09 Dupont Pharmaceuticals Company Ternary radiopharmaceutical complexes
US6132697A (en) * 1996-06-10 2000-10-17 G. D. Searle & Co. Radiopharmaceutical compositions capable of localizing at sites of thrombus
WO1998014220A2 (en) * 1996-10-02 1998-04-09 Du Pont Pharmaceuticals Company TECHNETIUM-99m LABELED CHELATOR INCORPORATED CYCLIC PEPTIDES
WO1998014220A3 (en) * 1996-10-02 1998-07-02 Du Pont Merck Pharma Technetium-99m labeled chelator incorporated cyclic peptides
EP1293214A3 (en) * 1996-10-07 2003-03-26 Bristol-Myers Squibb Pharma Company LTB4 antagonists and radiopharmaceuticals for imaging infection and inflammation
EP1293214A2 (en) * 1996-10-07 2003-03-19 Bristol-Myers Squibb Pharma Company LTB4 antagonists and radiopharmaceuticals for imaging infection and inflammation
WO1999040882A3 (en) * 1998-02-06 1999-11-04 Univ Pennsylvania STEREOSELECTIVE Tc-99m LIGANDS
WO1999040882A2 (en) * 1998-02-06 1999-08-19 Trustees Of The University Of Pennsylvania STEREOSELECTIVE Tc-99m LIGANDS
WO2000021980A1 (en) * 1998-10-13 2000-04-20 Du Pont Pharmaceuticals Company Chelator incorporated arg-gly-asp (rgd) mimetic synthetic disintegrins as imaging agents
WO2000057787A3 (en) * 1999-03-26 2001-02-22 Du Pont Pharm Co Method for localization of blood clots
US6808698B1 (en) 1999-03-26 2004-10-26 Bristol-Myers Squibb Pharma Company Method for localization of blood clots
WO2000057787A2 (en) * 1999-03-26 2000-10-05 Du Pont Pharmaceuticals Company Method for localization of blood clots
US6254852B1 (en) 1999-07-16 2001-07-03 Dupont Pharmaceuticals Company Porous inorganic targeted ultrasound contrast agents
US10183002B2 (en) 2011-09-30 2019-01-22 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US10183003B2 (en) 2011-09-30 2019-01-22 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9254278B2 (en) 2011-09-30 2016-02-09 Horizon Therapeutics, Inc. Methods of therapeutic monitoring of nitrogen scavenging drugs
US10617665B2 (en) 2011-09-30 2020-04-14 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9326966B2 (en) 2011-09-30 2016-05-03 Horizon Therapeutics, Inc. Methods of therapeutic monitoring of nitrogen scavenging drugs
US9095559B2 (en) 2011-09-30 2015-08-04 Horizon Therapeutics, Inc. Methods of therapeutic monitoring of nitrogen scavenging drugs
US10183005B2 (en) 2011-09-30 2019-01-22 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9962358B2 (en) 2011-09-30 2018-05-08 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9962359B2 (en) 2011-09-30 2018-05-08 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9999608B2 (en) 2011-09-30 2018-06-19 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US10045958B1 (en) 2011-09-30 2018-08-14 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US10045959B1 (en) 2011-09-30 2018-08-14 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US10183006B2 (en) 2011-09-30 2019-01-22 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US10183004B2 (en) 2011-09-30 2019-01-22 Horizon Therapeutics, Llc Methods of therapeutic monitoring of nitrogen scavenging drugs
US9561197B2 (en) 2012-04-20 2017-02-07 Horizon Therapeutics, Llc Methods of therapeutic monitoring of phenylacetic acid prodrugs
US9289406B2 (en) 2012-11-21 2016-03-22 Horizon Therapeutics, Inc. Methods of administering and evaluating nitrogen scavenging drugs for the treatment of hepatic encephalopathy
WO2015016616A1 (en) 2013-07-30 2015-02-05 연세대학교 산학협력단 Saxatilin-fc fusion protein and use thereof
US10208097B2 (en) 2013-07-30 2019-02-19 Industry-Academic Cooperation Foundation, Yonsei University Method for treating vascular stenosis or occlusive disease due to thrombi by administering a saxatilin-fc fusion protein
US9914692B2 (en) 2016-05-25 2018-03-13 Horizon Therapeutics, Llc Procedure for the preparation of 4-phenyl butyrate and uses thereof
US10329236B2 (en) 2016-05-25 2019-06-25 Horizon Therapeutics, Llc Procedure for the preparation of 4-phenyl butyrate and uses thereof
US11014870B2 (en) 2016-05-25 2021-05-25 Horizon Therapeutics, Llc Procedure for the preparation of 4-phenyl butyrate and uses thereof
US10668040B2 (en) 2017-09-11 2020-06-02 Horizon Therapeutics, Llc Treatment of urea cycle disorders in neonates and infants

Also Published As

Publication number Publication date
HU9502862D0 (en) 1995-11-28
FI954655A0 (en) 1995-09-29
KR960701666A (en) 1996-03-28
CA2159445A1 (en) 1994-10-13
BR9406820A (en) 1996-09-10
EP0692982B1 (en) 2000-07-05
RU2145608C1 (en) 2000-02-20
AU6524894A (en) 1994-10-24
EP0995761A2 (en) 2000-04-26
NO953886L (en) 1995-11-30
LV11106B (en) 1997-04-20
LV11106A (en) 1996-04-20
BG100036A (en) 1996-11-29
PL311037A1 (en) 1996-01-22
UA32577C2 (en) 2001-02-15
TW445267B (en) 2001-07-11
PT692982E (en) 2000-11-30
JPH08509710A (en) 1996-10-15
ATE194293T1 (en) 2000-07-15
CN1122577A (en) 1996-05-15
DE69425138D1 (en) 2000-08-10
HUT72889A (en) 1996-06-28
AU3452595A (en) 1996-03-21
JP3042887B2 (en) 2000-05-22
ES2149266T3 (en) 2000-11-01
EP0995761A3 (en) 2000-07-12
NZ263970A (en) 1996-09-25
PL178252B1 (en) 2000-03-31
AU689643B2 (en) 1998-04-02
US5879657A (en) 1999-03-09
GR3034272T3 (en) 2000-12-29
DK0692982T3 (en) 2000-10-23
BR9406055A (en) 1996-09-10
NO953886D0 (en) 1995-09-29
EP0692982A1 (en) 1996-01-24
EP0692982A4 (en) 1998-03-11
DE69425138T2 (en) 2001-03-01
NO307568B1 (en) 2000-04-25
FI954655A (en) 1995-11-02
ZA942262B (en) 1995-10-02
US6022523A (en) 2000-02-08

Similar Documents

Publication Publication Date Title
EP0692982B1 (en) RADIOLABELED PLATELET GPIIb/IIIa RECEPTOR ANTAGONISTS AS IMAGING AGENTS FOR THE DIAGNOSIS OF THROMBOEMBOLIC DISORDERS
EP0832068B1 (en) Stable reagents for the preparation of radiopharmaceuticals
CA2452923C (en) Peptide-based compounds for targeting integrin receptors
US5888970A (en) Technetium-99m labeled chelator incorporated cyclic peptides that bind to the GPIIb/IIIa receptor as imaging agents
US6524554B1 (en) Radiopharmaceuticals for imaging infection and inflammation and for imaging and treatment of cancer
JP2002527450A (en) Arg-Gly-Asp (RGD) mimetic synthetic disintegrins incorporating chelator as image forming reagent
US6251364B1 (en) Ternary ligand complexes useful as radiopharmaceuticals
US20030152512A1 (en) Imaging thrombus with glycoprotein llb/llla antagonists
RO114895B1 (en) Radiopharmaceutical product and reagent for preparing such product, method for viewing mammal platelet deposit position and radiolabeled compound
WO1995023811A1 (en) Novel carbocyclic compounds which inhibit platelet aggregation by interaction with the gpiib/iiia receptor complex
MXPA00009574A (en) Pharmaceuticals for the imaging of angiogenic disorders

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 94192010.0

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AU BB BG BR BY CA CN CZ FI GE HU JP KG KP KR KZ LK LV MD MG MN MW NO NZ PL RO RU SD SI SK TJ TT UA UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 263970

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1994912870

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2159445

Country of ref document: CA

Ref document number: 95-01701

Country of ref document: RO

WWE Wipo information: entry into national phase

Ref document number: 954655

Country of ref document: FI

WWP Wipo information: published in national office

Ref document number: 1994912870

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1994912870

Country of ref document: EP