WO2004087720A1 - Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1) - Google Patents

Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1) Download PDF

Info

Publication number
WO2004087720A1
WO2004087720A1 PCT/IB2004/000574 IB2004000574W WO2004087720A1 WO 2004087720 A1 WO2004087720 A1 WO 2004087720A1 IB 2004000574 W IB2004000574 W IB 2004000574W WO 2004087720 A1 WO2004087720 A1 WO 2004087720A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
aryl
alkyl
membered
solution
Prior art date
Application number
PCT/IB2004/000574
Other languages
French (fr)
Inventor
Chuangxing Guo
Eleanor Ferronyalka Dagostino
Liming Dong
Xinjun Hou
Stephen Anthony Margosiak
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Priority to EP04713610A priority Critical patent/EP1603926A1/en
Priority to JP2006506287A priority patent/JP2006523669A/en
Priority to MXPA05009241A priority patent/MXPA05009241A/en
Priority to BRPI0408477-2A priority patent/BRPI0408477A/en
Priority to CA002517281A priority patent/CA2517281A1/en
Publication of WO2004087720A1 publication Critical patent/WO2004087720A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C305/00Esters of sulfuric acids
    • C07C305/02Esters of sulfuric acids having oxygen atoms of sulfate groups bound to acyclic carbon atoms of a carbon skeleton
    • C07C305/16Esters of sulfuric acids having oxygen atoms of sulfate groups bound to acyclic carbon atoms of a carbon skeleton being unsaturated and containing rings
    • C07C305/18Esters of sulfuric acids having oxygen atoms of sulfate groups bound to acyclic carbon atoms of a carbon skeleton being unsaturated and containing rings containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C307/00Amides of sulfuric acids, i.e. compounds having singly-bound oxygen atoms of sulfate groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C307/02Monoamides of sulfuric acids or esters thereof, e.g. sulfamic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/091Esters of phosphoric acids with hydroxyalkyl compounds with further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/094Esters of phosphoric acids with arylalkanols
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • C07F9/5728Five-membered rings condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/58Pyridine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/59Hydrogenated pyridine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6503Five-membered rings
    • C07F9/6506Five-membered rings having the nitrogen atoms in positions 1 and 3
    • C07F9/65068Five-membered rings having the nitrogen atoms in positions 1 and 3 condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/650952Six-membered rings having the nitrogen atoms in the positions 1 and 4
    • C07F9/650994Six-membered rings having the nitrogen atoms in the positions 1 and 4 condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having three nitrogen atoms as the only ring hetero atoms
    • C07F9/6518Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/65515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/65515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring
    • C07F9/65517Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/6552Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring
    • C07F9/65522Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a six-membered ring condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms
    • C07F9/655345Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms the sulfur atom being part of a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms
    • C07F9/655345Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms the sulfur atom being part of a five-membered ring
    • C07F9/655354Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having sulfur atoms, with or without selenium or tellurium atoms, as the only ring hetero atoms the sulfur atom being part of a five-membered ring condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom

Definitions

  • This invention is directed to phosphate/sulfate ester compounds that modulate and/or inhibit the activity of protein interacting NIMA (PIN1), and to pharmaceutical compositions containing such compounds.
  • the invention is also directed to the therapeutic or prophylactic use of such compounds and compositions, and to methods of treating disorders characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders, by administering effective amounts of such compounds.
  • PIN1 is a member of the parvulin family of peptidyl-prolyl isomerases (PPIase) and catalyzes rotation about the peptide bond preceding a proline residue.
  • PPIase peptidyl-prolyl isomerases
  • PIN1 is a regulator of Cdc25, which depho ⁇ phorylates Cdc2/cyclinB to drive cells into mitosis.
  • PIN1 has been identified in all eukaiyotic organisms where examined, including plants, yeast, insects, and mammals.
  • the yeast (£ss1) and Dorosophilia (dodo) PIN1 orthologues have high identity to human-expressed sequence tags, which ultimately led to the cloning of the human docfo gene called PINL
  • the Dorosophilia dodo gene is reported to be 45% identical to the yeast gene, Esss
  • NIMA is a kinase that drives cells into mitosis and is reported to be negatively regulated by PIN1. Depletion of NIMA in A. nidulans cells is reported to lead to cell cycle arrest in G 2 while overexpression is reported to promote premature mitosis. SerThr kinase Cdc2/cyclin B may be the analogous NIMA kinase in human cells although another NIMA-like pathway in human cells is postulated to exist.
  • PIN1 activity is reported to result in dramatic morphological cellular phenotypes. For example, overexpression of PIN1 in Hela cells was reported to cause a G 2 arrest while depletion caused mitotic arrest— the opposite phenotypes observed with NIMA modulation. Additionally, decreasing PIN1 protein expression by full-length antisense expression has been reported to cause cells to progress into mitosis prematurely, to contain aberrant nuclei due to premature chromosome condensation and to induce apoptosis. These data indicate that PI 1 is a negative regulator of mitosis through interactions with a mammalian functional homoiog of NIMA and is required for progression through mitosis. Further, depletion of PIN1 is also postulated to play a role in Alzheimer's disease. Lu et al., Nature, 380, 544-547 (1996).
  • PIN1 has been reported to interact with mitotic proteins also recognized by the MPM- 2 antibody.
  • the MPM-2 monoclonal antibody recognizes a phospho-Ser/Thr-Pro epitope on about approximately 50 proteins associated with mitosis, including important mitotic regulators, such as Cdc25, Wee1, Cdc27, Map 4, and NIMA. See, e.g., Davis et al., Proc. Natl. Acad. Sci. U.S.A. 80, 2926 (1983).
  • PIN1 has also been reported to interact with important upstream regulators of Cdc2/cyclin B, including Cdc25 and its known regulator, PlxL See Shen et al., Genes Dev. 12, 706 (1998).
  • PIN1 due to its enzymatic action, may remove Cdc25 and Plx1 from play by causing their degradation within the cell.
  • PIN1 biological function of PIN1 depends on a functional PPIase active site. Lu et al., Science, 283, 1325-1328 (1999). Studies also indicate that PIN1 recognizes its substrates (mitosis-specific phosphoproteins) through the WW domain.
  • the WW domain is a protein recognition motif that is prevalent throughout biology. However, the PIN1 WW domain is unique in that it requires its ligand protein to contain a phosphorylated serine. As with the PPIase domain, a functional WW domain is reported to be essential for biological functions of PINL This is consistent with the model where PIN1 recognizes its substrates through the WW domain followed by completion of its essential catalytic role.
  • Full-length PIN1 protein and the nucleotide sequence encoding full-length PIN1 are disclosed in U.S. Patent Nos. 5,952,467 and 5,972,697. Additionally, sequences for PIN1 have been deposited in GenBank under accession numbers NM006221 (mRNA) and S68520 (protein). The mRNA sequence for dodo is deposited in GenBank under accession number U35140. Mouse PI 1 mRNA sequence is deposited in GenBank under accession number NM023371.
  • Lu et al. International Publication No. WO 01/38878
  • Wulf et al. EMBO J. 20, 3459- 3472 (2001)
  • PIN1 is upregulated in human tumors and is a biomarker for cell proliferation.
  • Inhibitors of PI 1 have been described in the literature. For example, Hennig et al.
  • juglone (5-hydroxy-1 ,4-naphthoquinone) selectively inhibits several parvulins, including human PINL Noel et al. in U.S. Patent Application No. 20010016346, using data based on the crystal structure derived from full-length human PIN1, disclose compounds postulated to be inhibitors of PINL Lu et al. in International Publication No. WO 99/12962 report inhibitors that mimic the phospho-Ser/Thr moiety of the phosphoserine or phosphothreonine-proiine peptidyl prolyl isomerase substrate.
  • PIN1 plays in the regulation of the cell cycle
  • additional compounds that inhibit PIN1 are needed.
  • These compounds, along with pharmaceutical compositions thereof, can serve as effective chemotherapeutic agents for the treatment of a variety of disorders characterized by hypertension, inappropriate cell proliferation, including cancer, infectious diseases, and neurodegenerative brain disorders.
  • the invention provides such compounds that inhibit PINL Summarv Of The Invention
  • an objective of the invention is to discover compounds and methods for modulating or inhibiting PINL
  • Another objective of the invention is to provide compounds and methods for modulating or inhibiting PIN1 that can be used in pharmaceutical compositions for the treatment of disorders characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders.
  • compositions containing such agents are useful in treating diseases characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders.
  • the invention relates to compounds of the Formula I:
  • n 1 or 2;
  • R 1 is a C 3 -C 10 cycloalkyl, 4-10 membered heterocycloalkyl, C 3 -C 10 aryl, or 4-10 membered heteroaryl group, wherein R 1 is unsubstituted or substituted with 1 to 4 R 10 groups;
  • R 2 is -S(0) 2 OH, -S(0) 2 NR d R e , or -P(0)(OR 4 ) 2 , wherein R 4 is an H, C r C 10 -alkyl, C 6 -C 0 aryl, or -CH 2 -0-C(0)R e CH 3 group, R d and R e are each independently an H or C r C 6 alkyl group, and R 4 is unsubstituted or substituted with 1 to 4 R 10 groups; and
  • R 3 is OH, C r C 7 -alkyl, C r C 7 -alkoxyl, C 6 -C 10 aryl, 4-10 membered heteroaryl, C 3 -C 10 cycloalkyl, 3-10 membered heterocycloalkyl, -NH(R 5 ), or -N(R 5 ) 2 group, wherein R 5 is independently selected from H, C r C 7 alkyl, C 6 -C 10 aryl, or
  • ring B is a 5- or 6-membered heterocycloalkyl group
  • Z is a divalent C(0)Z ⁇ heteroaryl or heterocycloalkyl group wherein Z' is a divalent 0, S, NH, N(CH 3 ), C0 2 , or CH 2
  • the invention relates to compounds of Formula I, wherein n is 1 or 2; A is a divalent -NH-Y-, -NR d (CH 2 ) r Y-, or -0-Y-, and Y is C(0) or S(0) 2 ; X is a direct bond, CH 2 , O, or S; R 1 is a C 6 -C 10 aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R 10 groups; R 2 is -S(0) 2 OH, or -P(0)(0R 4 ) 2 , wherein R 4 is an H, C C ⁇ 0 alkyl, or C 6 -C 10 aryl group, and is unsubstituted or substituted with 1 to 4 R 10 groups; and R 3 is a C 6 -C ⁇ 0 aryl, 4-10 membered heteroaryl, -NH(C 6 H 5 ), or
  • ring B is a 5- or 6-membered heterocycloalkyl group
  • Z is a divalent C(0)Z', heteroaryl or heterocycloalkyl group wherein Z' is a divalent O, S, NH, N(CH 3 ), C0 2 , or CH 2
  • R 6 is H or a C ⁇ -C 10 alkyl group, wherein R 3 , B, and R 6 is unsubstituted or substituted with 1 to 4 R 10 groups; and wherein R 10 is as defined above.
  • the invention relates to compounds of Formula I, wherein n is 1 ; A is a divalent -NH-Y- or -0-Y-, wherein Y is C(O); X is a direct bond, CH 2 , or O; R 1 is a C 6 -C 10 aryl group unsubstituted or substituted with 1 to 4 R 10 groups; R 2 is -P(0)(OR 4 ) 2 , wherein R 4 is an H, CrCio alkyl, or C 6 -C ⁇ 0 aryl group, and is unsubstituted or substituted with 1 to 4 R 1 ⁇ groups; and R 3 is a C 6 -C 10 aryl, 4-10 membered heteroaryl, or
  • ring B is an unsubstituted 6-membered heterocycloalkyl
  • Z a divalent C(0)Z'
  • is a divalent
  • R 6 is a CrCio alkyl group, wherein R 3 , B and R 6 are unsubstituted or substituted with 1 to 4 R 10 groups; and wherein R 10 is as defined above.
  • the invention relates to compounds of Formula
  • R 1 is a C B - C 0 aryl group unsubstituted or substituted with 1 to 4 R 10 groups
  • R 2 is -P(0)(0R 4 ) 2 , wherein R 4 is an H or a C 1 -C 10 alkyl group that is unsubstituted or substituted with 1 to 4 R 10 groups
  • R 3 is a C 6 -C 10 aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R 0 groups; and wherein R 10 is as defined above.
  • the invention includes compounds, and pharmaceutically acceptable salts thereof, selected from the following group: -7-
  • the invention also relates to a method of inhibiting PIN1 by administering a compound of Formula I or a pharmaceutically acceptable prodrug, pharmaceutically active metabolite, or pharmaceutically acceptable salt of such compound or metabolite thereof.
  • the invention also relates to pharmaceutical compositions, each comprising a therapeutically effective amount of an agent selected from compounds, prodrugs, metabolites, and salts of compounds of Formula I, and a pharmaceutically acceptable carrier or vehicle for such agent.
  • the invention further provides methods of treating mammalian disease conditions mediated by PIN1 activity, by administering to a mammal in need thereof a therapeutically effective amount of a compound, prodrug, active metabolite or salt of a compound of Formula I.
  • the mammalian disease conditions to be treated according to the invention are associated with hypertension, inappropriate cell proliferation (e.g., cancer), infectious diseases (e.g., bacterial and fungal infections), and neurodegenerative brain disorders (e.g., Alzheimer's disease).
  • the compounds of Formula I are useful for modulating or inhibiting PINL More particularly, the compounds are useful as modulating or inhibiting the activity of PIN 1, thus providing treatments for hypertension, infectious diseases, neurodegenerative disorders, and cancer or other diseases associated with cellular proliferation.
  • the terms "comprising” and “including” are used herein in their open, non-limiting sense.
  • abnormal cell proliferation includes diseases or disorders associated with uncontrolled or abnormal cellular proliferation.
  • diseases and disorders include, but are not limited to, the following: a variety of cancers, including, but not limited to, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymph
  • HIV human papilloma virus, herpesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus
  • autoimmune diseases including, but not limited to, systemic lupus erythematosus rheumatoid arthritis, psoriasis, autoimmune mediated glomerulonephritis inflammatory bowel disease and autoimmune diabetes mellitus
  • neurodegenerative disorders including, but not limited to, Alzheimer's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, Parkinson's disease, AIDS-related dementia, spinal muscular atrophy and cerebellar degeneration
  • myelodysplastic syndromes aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including, but not limited to, chronic anemia and aplastic
  • alkyl refers to a straight- or branched-chain, saturated or partially unsaturated, alkyl group having one to twelve carbon atoms. Preferred alkyl groups have from 1-10, and more preferably from 1-7, carbon atoms. Exemplary alkyl groups include methyl (Me, which also may be structurally depicted by ), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and the like.
  • the term “lower alkyl” designates an alkyl having from 1 to 6 carbon atoms (a C C 6 alkyl).
  • aryl refers to a monocyclic, or fused or spiro polycyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) having from three to twelve ring atoms per ring, preferably 6-10 ring atoms atoms and more preferably 5-7 ring atoms.
  • aryl groups include the following moieties:
  • heteroaryl refers to a monocyclic, or fused or spiro polycyclic, aromatic heterocycle (ring structure having ring atoms selected from carbon atoms as well as nitrogen, oxygen, and sulfur heteroatoms) having from three to twelve ring atoms per ring, preferably 4-10 ring atoms and more preferably 5-7 ring atoms.
  • aryl groups include the following moieties:
  • cycloalkyl refers to a saturated or partially saturated, monocyclic or fused or spiro polycyclic, carbocycle having from three to twelve ring atoms per ring, preferably 3-10 carbon atoms and more preferably 5-7 carbon atoms.
  • Illustrative examples of cycloalkyl groups include the following moieties:
  • heterocycloalkyl refers to a monocyclic, or fused or spiro polycyclic, ring structure that is saturated or partially saturated and has from three to twelve ring atoms per ring selected from C atoms and N, O, and S heteroatoms, preferably 4-10 ring atoms and more preferably 5-7 ring atoms.
  • ring atoms selected from C atoms and N, O, and S heteroatoms, preferably 4-10 ring atoms and more preferably 5-7 ring atoms.
  • Illustrative examples of heterocycloalkyl groups include:
  • alkoxy refers to -O-alkyl. Illustrative examples include methoxy, ethoxy, propoxy, and the like.
  • halogen represents chlorine, fluorine, bromine or iodine.
  • halo represents chloro, fluoro, bromo or iodo.
  • haloalkyl refers to a loweralkyl radical in which one or more of the hydrogen atoms are replaced by halogen including, but not limited to, chloromethyl, trifiuoromethyl, 1- chloro-2-fluoroethyl and the like.
  • Heteroalkyl is an alkyl group (as defined herein) wherein at least one of the carbon atoms is replaced with a heteroatom. Preferred heteroatoms are nitrogen, oxygen, sulfur, and halogen. A heteroatom may, but typically does not, have the same number of valence sites as carbon.
  • the number of hydrogens bonded to the heteroatom may need to be increased or decreased to match the number of valence sites of the heteroatom. For instance, if carbon (valence of four) is replaced with nitrogen (valence of three), then one of the hydrogens formerly attached to the replaced carbon must be deleted. Likewise, if carbon is replaced with halogen (valence of one), then three (i.e., all) of the hydrogens formerly bonded to the replaced carbon must be deleted.
  • trifiuoromethyl is a heteroalkyl group wherein the three methyl groups of a t-butyl group are replaced by fluorine.
  • Preferred heteroalkyls of the invention have 2 to 10 member atoms, including both heteroatoms and carbon atoms.
  • substituted means that the specified group or moiety bears one or more substituents.
  • unsubstituted means that the specified group bears no substituents.
  • the compounds of the invention may exhibit the phenomenon of tautomerism. While Formula I cannot expressly depict all possible taufomeric forms, it is to be understood that Formulas I is intended to represent any tautomeric form of the depicted compound and are not to be limited merely to a specific compound form depicted by the formula drawings.
  • the compounds of Formula I may have one or more asymmetric centers designated by an asterisk as shown below in Formula I. Additional asymmetric centers may be present on the molecule depending upon the nature of the various substituents on the molecule.
  • the compounds of Formula I may exist as single stereoisomers (i.e., essentially free of other stereoisomers), racemates, and/or mixtures of enantiomers and/or diastereomers. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention.
  • the inventive compounds that are optically active are used in optically pure form.
  • ' is used in structural formulae herein to depict the bond that is the point of attachment of the moiety or substituent to the core or backbone structure.
  • an optically pure compound having one chiral center is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure.
  • the compounds of the present invention are used in a form that is at least 90% optically pure, that is, a form that contains at least 90% of a single isomer (80% enantiomeric excess ("e.e.") or diastereomeric excess (“d.e.”)), more preferably at least 95% (90% e.e. or d.e.), even more preferably at least 97.5% (95% e.e. or d.e.), and most preferably at least 99% (98% e.e. or d.e.).
  • Formula I is intended to cover solvated as sell as unsolvated forms of the identified structures.
  • Formula I includes compounds of the indicated structure in both hydrated and non-hydrated forms.
  • Other examples of solvates include the structures in combination with isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
  • the invention includes pharmaceutically acceptable prodrugs, pharmaceutically active metabolites, and pharmaceutically acceptable salts of such compounds and metabolites.
  • a pharmaceutically acceptable prodrug is a compound that may be converted under physiological conditions or by solvolysis to the specified compound or to a pharmaceutically acceptable salt of such compound.
  • a pharmaceutically active metabolite is intended to mean a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof.
  • Prodrugs and active metabolites of a compound may be identified using routine techniques known in the art. See, e.g., Bertolini et al., J. Med. Che ., 40, 2011-2016 (1997); Shan, et al., J. Pharm. Sci., 86 (7), 765-767; Bagshawe, Drug Dev.
  • a pharmaceutically acceptable salt is intended to mean a salt that retains the biological effectiveness of the free acids and bases of the specified compound and that is not biologically or otherwise undesirable.
  • a compound of the invention may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Exemplary pharmaceutically acceptable salts include those salts prepared by reaction of the compounds of the present invention with a mineral or organic acid or an inorganic base, such as salts including sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as gluc ⁇ ronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
  • an inorganic acid such as hydrochlor
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • Lu et al. International Publication No. WO 01/38878; incorporated herein by reference in its entirety disclose that PI 1 is overexpressed in a variety of cancers, including breast, colon, and prostate. Additionally, the authors disclose that P1 1 is overexpressed in proliferating cells.
  • the agents of the invention would have use for treating a variety of cell proliferative diseases associated with overexpression of PIN1.
  • Therapeutically effective amounts of the agents of the invention may be used to treat diseases mediated by modulation or regulation of PINL
  • An "effective amount" is intended to mean that amount of an agent that, when administered to a mammal in need of such treatment, is sufficient to effect treatment for a disease modulated or inhibited by the activity of PINL
  • a therapeutically effective amount of a compound of the Formula I, salt, active metabolite or prodrug thereof is a quantity sufficient to modulate, regulate, or inhibit the activity of PIN1 such that a disease condition which is mediated by that activity is reduced or alleviated.
  • treating refers to:
  • compositions of this invention comprise an effective modulating, regulating, or inhibiting amount of a compound of Formula I and an inert, pharmaceutically acceptable carrier or diluent.
  • efficacious levels of the inventive agents are provided so as to provide therapeutic benefits involving modulation of PINL
  • efficacious levels is meant levels in which the effects of PIN1 activity are, at a minimum, regulated.
  • An inventive agent can be administered in conventional dosage form prepared by combining a therapeutically effective amount of an agent (e.g., a compound of Formula I) as an active ingredient with appropriate pharmaceutical carriers or diluents according to conventional procedures. These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation.
  • the pharmaceutical carrier employed may be either a solid or liquid. Exemplary of solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary of liquid carriers are syrup, peanut oil, olive oil, water and the like.
  • the carrier or diluent may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate and the like.
  • a variety of pharmaceutical forms can be employed.
  • a solid carrier used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge.
  • the amount of solid carrier may vary, but generally will be from about 25 mg to about 1 g.
  • a liquid carrier is used, the preparation will be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or non-aqueous liquid suspension.
  • a pharmaceutically acceptable salt of an inventive agent is dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3M solution of succinic acid or citric acid.
  • an organic or inorganic acid such as 0.3M solution of succinic acid or citric acid.
  • the agent may be dissolved in a suitable cosolvent or combinations of cosolvents.
  • suitable cosolvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, gylcerin and the like in concentrations ranging from 0-60% of the total volume.
  • a compound of Formula I is dissolved in DMSO and diluted with water.
  • compositions may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution. It will be appreciated that the actual dosages of the agents used in the compositions of this invention will vary according to the particular complex being used, the particular composition formulated, the mode of administration and the particular site, host and disease being treated.
  • compositions of the invention may be manufactured in manners generally known for preparing pharmaceutical compositions, e.g., using conventional techniques such as mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing.
  • Pharmaceutical compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers, which may be selected from excipients and auxiliaries that facilitate processing of the active compounds into preparations, which can be used pharmaceutically.
  • the agents of the invention may be formulated into aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient io be treated.
  • Pharmaceutical preparations for oral use can be obtained using a solid excipient in admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, polyvinyl pyrrolidone, Carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate, and, optionally, stabilizers.
  • the active agents may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of gelatin for use in an inhaler or insufflator and the like may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit-dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active agents may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g, containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation.
  • Such long-acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion-exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • An exemplary pharmaceutical carrier for hydrophobic compounds is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be a VPD co-solvent system.
  • VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the VPD co-solvent system (VPD:5W) contains VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • co-solvent component may be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may be substituted for dextrose.
  • other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • the pharmaceutical compositions also may comprise suitable solid- or gel-phase carriers or excipients. Examples of such carriers or excipients include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • compositions of the invention may be provided as salts with pharmaceutically compatible counter ions.
  • Pharmaceutically compatible sails may be formed with many acids, including hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Sails lend to be more soluble in aqueous or other protonic solvents than are the corresponding free-base forms.
  • inventive agents may be prepared using the reaction routes and synthesis schemes as described below, employing the general techniques known in the art using starting materials that are readily available.
  • the preparation of preferred compounds of the present invention is described in detail in the following examples, but the artisan will recognize that the chemical reactions described may be readily adapted to prepare a number of other PIN1 inhibitors of the invention.
  • the synthesis of non-exempiified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions.
  • other reactions disclosed herein or generally known in the art will be recognized as having applicability for preparing other compounds of the invention. Examples
  • NMR spectra were recorded on a Bruker or Varian instrument operating at 300 MHz and 13 C-NMR spectra were recorded operating at 75 MHz. NMR spectra were obtained as CDCl 3 solutions (reported in ppm), using chloroform as the reference standard (7.27 ppm and 77.00 ppm) or CD 3 OD (3.4 and 4.8 ppm and 49.3 ppm), or internally tetramethylsilane (0.00 ppm) when appropriate. Other NMR solvents were used as needed.
  • Mass spectrometry was conducted with various techniques. Mass spectra were obtained using liquid chromatograph electrospray ionization mass spectrometry, MS (ESP). Matrix- Assisted Laser Desorption/lonization (MALDI) Fourier Transform Mass Spectrometry was performed on an lonSpec FTMS mass spectrometer. The following compounds of the invention were made according to the general synthetic pathways shown in Schemes 1-10 and the detailed experimental procedures that follow thereof.
  • Example 3a 1-(2-Phenyl-1-sulfooxymethyl-ethylsulfamoyl)-pyrrolidine-2S-carboxylic acid benzyl ester
  • Example 3b1 1-(2-Phenyl-1-sulfooxymethyl-ethylsulfamoyl)-piperidine-2S-carboxylic acid 4- phenyl-butyl ester
  • Example 3b3 1-(2-Phenyl-1-sulfooxymethyI-ethylsulfamoyl)-piperidine-2S-carboxylic acid 4- phenyl-1 -(3-phenyl-propyl)-butyl ester
  • Example 3b4 3,3-Dimethyl-1-(2-phenyl-1-sulfooxymethyl-ethylsulfamoyl)-piperidine-2R- carboxylic acid benzyl ester
  • Example 555 Phosphoric acid mono- ⁇ (R)-2-[(S)-2-(5-ben2yl-[1,3 l 43o ⁇ adia2 ⁇ l-2-yl)-p!peridine- 1 -suIfonylamino3-3-phenyl-propyl ⁇ ester
  • amine 8b 0.265 g, 1 mmol was added to a methylene chloride solution (5 mL) of phosgene (0.544 mL, 20% in toluene, 1.1 mmol) and triethylamine (0.5 mL). The solution was slowly warmed up to 25 °C over 30 min, and was cooled to 0 °C again. The pipecolate ester 6b (0.05 g, 0.143 mmol) was introduced at once. The mixture was stirred at 25 °C for 20 h, diluted with EtOAc (50 mL), washed with concentrated NaHCOs solution (1x50 mL), dried (Na 2 S0 4 ) and concentrated.
  • Example 14b 1-(2-Phenyl-1-sulfooxymethyl-ethylcarbamoyi)-piperidine-2-carboxylic acid 3- (7-nitro-benzo[1 ,2,5]oxadiazol-4-ylamino)-propyl ester
  • Example 14a 1-(2-Phenyl-1-sulfooxymethyl-ethylcarbamoyl)-piperidine-2S-carboxylic acid 4- phenyl-1-(3-phenyl-propyI)-butyl ester
  • Example 16a 1-(2-Phenyl-1-phosphonooxymethyI-ethylcarbamoyl)-piperidine-2S-carboxylic acid 4-phenyl-1-(3-phenyl-propyl)-butyl ester
  • Example 23d Phosphoric acid mono- ⁇ 2-p-(3,5-dimethoj y-phenyl)-ureido3-3-phenyl-propyl ⁇ ester
  • Example 25-1 Phosphoric acid mono- ⁇ 3-phen I-2- (1-phenyl-methanoyl)-®mino3-propylJ ester
  • E ⁇ ampl ⁇ 25-2' Phosphoric acid mono-(2- ⁇ [1-(1-oxo-foenzo[bJthiophen-2-yl)- aminoH-p e ⁇ yi ⁇ propyl) ester
  • Example 25-4 Phosphoric acid mono-[2-( ⁇ 1-[5-(3,5-dichloro-pheno ⁇ y)-furan-2-yl]-methanoyl ⁇ - amino)-3-ph ⁇ nyl-propyl] ⁇ ter
  • Example 25-6 Phosphoric acid mono-(2- ⁇
  • Example 25-8 Phosphoric acid mono-(2- ⁇ 1-(3-chloro-phenyl)-methanoyl3-amino ⁇ -3-phen l- propyl) ester
  • Example 25-11 Phosphoric acid mon ⁇ - ⁇ 3-phenyl-2-j;(1-quino ali ⁇ -2-yl-methanoyl)-amino3- propyl ⁇ ester
  • Example 25-13 Phosphoric acid mono-(2- ⁇ [1-(2-hydroxy-phenyl)-methanoyl]-amino ⁇ -3- phenyl-propyl) ester
  • Example 25-16 Phosphoric acid mono-[(R)-2-(2,2-dimethyl-propanoylamino)-3-phenyl-propyl] ester
  • Example 25-19 Phosphoric acid mono- ⁇ 2-I(1-benzofuran-2-yl-methanoyl)-amino3-3-phenyl- propyl ⁇ ⁇ ster
  • Example 25-22 Phosphoric acid mono-(2- ⁇ [1-(3-hydroxy-naphthalen-2-yI)-methanoyl]-amino ⁇ - 3-phenyl-propyl) ester
  • Example 25-23 Phosphoric acid mono- ⁇ (R)-2-[(1H-benzoimidazole-5-carbonyl)-amino]-3- phenyl-propyl ⁇ ester
  • Example 25-26 Phosphoric acid mono- ⁇ 2-[(1-benzo[b]thiophen-2-yl-methanoyl)-amino]-3- phenyl-propyl ⁇ ester
  • Example 25-28 Phosphoric acid mono- ⁇ 3-(3-fluoro-phenyl)-2-[(1-naphthalen-2-yl-methanoyl)- amino]-propyl ⁇ ester
  • Example 25-30 Phosphoric acid mono-[(R)-2-(2,2-dimethyl-propanoylamino)-3-(3-fluoro- phenyl)-propyl] ester
  • Example 25-31 Phosphoric acid mono-[(R)-2- ⁇ [1-(1-bromo-naphthalen-2-yl)-methanoyl]- amino ⁇ -3-(3-fluoro-phenyl)-propyl] ester
  • Example 25-32 Phosphoric acid mono-((R)-3-(3-fluoro-phenyl)-2- ⁇ [1-(6-methoxy-naphthalen- 2-yl)-methanoyl]-amino ⁇ -propyl) ester
  • Example 25-33 Phosphoric acid mono-[(R)-2- ⁇ [1-(diethylamino-oxo-2H-chromen-3-yl)- methanoyl]-amino ⁇ -3-(3-fluoro-phenyl)-propyl] ester
  • Example 25-34 Phosphoric acid mono- ⁇ 2- [1-(1-bromo-naphthalen-2-yI)-methano l3- aminoJ-3-m-tolyl-propyl) ester
  • Example 25-35 Phosphoric acid mono-((R)-2- ⁇ [1-(6-methoxy-naphthalen-2-yl)-methanoyl]- amino ⁇ -3-m-tolyl-propyl) ester
  • Example 25-36 Phosphoric acid mono- ⁇ 2-[(1-n ⁇ phthalen-2-yl-methanoyI)-arnino3-3-m-tolyl- propyl] ester
  • Example 25-30 Phosphoric acid mono- ⁇ 2-[(1-naphthalen-2-yl-methanoyI)-aminoJ-3-pyridin-3- yl-propyl ⁇ ester
  • Example 42 Phosphoric acid mono-[(R)-2-(5-dimethylamino-naphthalene-1-sulfonylamino)-3- (3-fluoro-phenyI)-propyl] ester
  • D-3-Fluorophenylalanine (7.98 g, 43.8 mmol) was dissolved in 1 M sulfuric acid solution (140 mL). To the solution at 0 °C was slowly added 6 M NaN0 2 solution (36 mL, 216 mmol) and 3.2 M sulfuric acid (36 mL). The mixture was stirred at 0 °C for 3 h and then at 25 °C for 0.5 h. The solution was extracted with EtOAc (7x75 mL). The combined organic layers were dried and concentrated. Recrystalization from EtOAc/hexanes afforded 5.36 g (67% yield) of the compound ⁇ - hydroxycarboxylic acid.
  • Example 54 Phosphoric acid mono- ⁇ 2-[(1-benso[b3 hiophen-2-yI-methanoyl)-amino3-3-(3- fIuoro-phenyl)-propyl] ester
  • Example 58 Phosphoric acid mono-f[me ⁇ hyl-(1-nap halen-2-yl-methanoyI)-amino3-phenyl- propyl ⁇ ester
  • Example 66 Phosphoric acid mono- ⁇ 3-(2,3-difTuoro-phenyI)-2-[(1-naphthalen-2-yl-methanoyl)- amino3-propyl ⁇ ester
  • Alcohol 69 (1.77 g, 5.14 mmol) was partially dissolved in CH 2 CI 2 (50 mL) and cooled to 0 °C.
  • Example 72 Sulfamic acid 2-[(1-naphthalen-2-yl-methanoyl)-aminoJ-3-phenyJ-propyl ester
  • Alcohol 47a was prepared as described in the synthesis of compound 47.
  • hydroxyl carboxylic acid 760 mg, 4.13 mmol
  • 2-naphthoyl chloride 866 mg, 4.54 mmol
  • triethylamine 2.9 mL
  • 1 M borane in THF 4.73 mL
  • column chromatography 40% EtOAc in hexanes
  • Benzyl Ester 48a was prepared as described in the synthesis of 48 using the alcohol 47s (137 mg, 0.423 mmol), 1H-tetrazole (68 mg, 0.973 mmol), dibenzyl N,N-diisopropylphosphoramidite (0.213 mL, 0.634 mmol), and MCPBA (380 mg, 77% pure, 1.69 mmol). After column chromatography purification (10% to 20% EtOAc in hexanes), the compound 48a was obtained as crude oil (330 mg), which was carried forward to the next step.
  • Example 74 Naphthalene-2-carboxylic acid (R)-1 -(3-fluoro-ben ⁇ yl)-2-phosphonooxy-ethyl ester
  • Example 74 was prepared as described in the synthesis of 49 using the crude benzyl ester (330 mg) and 10% palladium on carbon (70 mg). Preparative HPLC purification gave 70 mg of the title compound 74 (31% yield from the alcohol).
  • Example 81 Acetic acid acetoxymethoxy-(2- ⁇ [1-(1-bromo-naphthalen-2-yI)-methanoyl]- amino ⁇ -3-phenyl-propoxy)-phosphoryloxymethyl ester
  • Example 81 was prepared as described in the synthesis of Example 80 using 25-24 (23 mg,
  • Example 82 was prepared as described in the synthesis of Example 80 using 23b (22 mg, 0.0498 mmol), bromomethyl acetate (24 ⁇ L, 0.249 mmol) and diisopropylethylamine (0.05 mL). Purification by Et 3 N deactivated column chromatography (50% EtOAc in hexanes) gave 9 mg (31 % yield) of the title compound 82.
  • Example 83 Acetic acid acetoj ymetho ⁇ y-[2-( ⁇ 1-[5-(3 s 5-dichIoro-phenoJ y)-furan-2-yl3- methanoyi ⁇ -amino)-3-phenyl-propo ⁇ 5y]-phosphorylo2 ⁇ ymeth l ester
  • Example 84 Acetic acid acetoxymethoxy- ⁇ (R)-3-(3-fluoro-phenyl)-2-[(1-naphthalen-2-yI- methanoyl)-amino]-propoxy ⁇ -phosphoryloxymethyl ester
  • Example 84 was prepared as described in the synthesis of Example 80 using 25-28 (50 mg, 0.124 mmol), bromomethyl acetate (61 ⁇ L, 0.620 mmol) and diisopropylethylamine (0.13 mL). Purification by Et 3 N deactivated column chromatography (30-50% EtOAc in hexanes) gave 17 mg (25% yield) of the title compound 84.
  • Example ⁇ 5 Ace ⁇ ic acid aceto ⁇ cymeth ⁇ 2 ⁇ y-[(R)-2-[(1-ben2 ⁇ [b]thiophen-2-yl-methanoyi)-amino3- 3-(3-fluoro-phenyl)-propoxy]-phosphoryioxymethyl ester
  • Example 85 was prepared as described in the synthesis of Example 80 using 25-29 (45 mg,
  • Example 89 was prepared as described in the synthesis of Example 86 using the alcohol 88 (40 mg, 0.122 mmol), Et 3 N (0.1 mL), DMAP (4 mg) and diphenyl chlorophosphate (0.29 ⁇ L, 37.6 mg, 0.14 mmol). Column chromatography purification (40% EtOAc in hexanes)' gave 62 mg (97% yield) of the title compound 89.
  • Example 90 1-[1-Bis-acetoxymethoxy-phospgoryloxymethyJ)-2-phenyl-ethylsulfamoyI)3- piperidine-28-carboxylie acid 4-phenyl-foutyl ester
  • Example 90 was prepared as described in the synthesis of Example 80 using 5b1 (20 mg, 0.036 mmol), bromomethyl acetate (36 ⁇ L, 0.36 mmol) and diisopropylethylamine (0.1 mL). Purification by Et 3 N deactivated column chromatography (40% EtOAc in hexanes) gave 25 mg (100% yield) of the title compound 90.
  • Example 91 was prepared as described in the synthesis of Example 80 using 16d (60 mg, 0.155 mmol), bromomethyl acetate (0.15 ⁇ L, 1.55 mmol) and diisopropylethylamine (0.4 mL, 2.33 mmol). Purification by Et 3 N deactivated column chromatography (40% EtOAc in hexanes) gave 45 mg (55% yield) of the title compound 91.
  • Example 92 Acetic acid aceto2iymetho ⁇ , -[(R)-2-[(7-diethylam ⁇ no-2-o ⁇ o-2H-chromene-3- carbonyl)-amino]-3-(3-fluoro-phenyl)-propoxy3-phosphoryloxymethyl ester
  • Example 92 was prepared as described in the synthesis of Example 80 using Example 25-33
  • Example 93 Acetic acid acetoxymethoxy-[3-[(benzo[6]thiophene-2-carbonyl)-amino3-4-(3- fIuoro-phenyl)-butylJ-phosphinoyloxymethyl ester
  • Example 93 was prepared as described in the synthesis of Example 80 using Example 71 (22 mg, 0.0541 mmol), bromomethyl acetate (0.05 mL, 0.52 mmol) and diisopropylethylamine (0.13 mL, 0.77 mmol). Purification by flash column chromatography (100% EtOAc in hexanes) gave 23 mg (77% yield) of the title compound 93.
  • Example 99 Acetic acid acetoxymethoxy-[2-[benzo[ ⁇ ]thiophene-2-carbonyl)-amino]-3-(3- fluoro-phenyl)-propyl]-phosphinoyloxymethyl ester
  • Example 100 2,2-Dimethyl-propionic acid [3-[(foen2:oE&3thiophene-2-carfoonyI)-amino3-4-(3- fluoro-phenyl)-butyl3-(2,2-dimethyl-propionyl ⁇ 5cymethoxy)-phosphinoylo ⁇ ymethyl ester
  • Example 71 To an acetonitrile solution (5 mL) of Example 71 (50 mg, 0.123 mmol) was added tetrabutylamonium iodide (5 mg), diisopropylethylamine (0.2 mL), and chloromethyl pivalate (132 ⁇ L, 0.92 mmol) at 0 °C. The solution was heated at 60 °C for 4 h and concentrated in vacuo. The residue was purified by column chromatography (35% EtOAc in hexanes), affording 20 mg (26% yield) of the title compound 100.
  • PIN1 is a phosphorylation dependent peptidyl-prolyl isomerase.
  • the PIN1 assay is a spectrophotometric assay based on the coupled chymotrypsin or subtilisin catalyzed, cis-trans conformation dependent cleavage of a para-nitroanaline containing peptide substrate. This improved general rotamase assay was first described by Kofron, et al. (Biochemistry, 30, 6217-6134 (1991)) and applied to PIN1 isomerase activity by Yaffe, et al. (Science, 278, 1957-1960 (19g7)).
  • the peptide substrate Upon dilution into an aqueous assay mixture containing PIN1 , the peptide substrate undergoes PIN1 catalyzed isomerization to the trans conformation.
  • Chymotrypsin or subtilisin subtilisin (subtilisin Carisberg protease, available from Sigma, catalog number P-5380) cleaves the trans product to form free para-nitroanaline.
  • reactions are performed at 15 °C.
  • a typical reaction contains 25 mM MOPS pH 7.5, 0.5 mM TCEP, 2% DMSO, 5 ⁇ l of a 25 mg/ml solution of subtilisin Carisberg, 50 nM PINI-PPiase, and 100 ⁇ M Suc-AEPF-pNA peptide substrate. Reactions are cooled to 15 °C and initiated with the addition of Suc-AEPF-pNA. The absorbance at 390 nm is monitored continuously until all substrate has been converted to the cleaved product. This data, the progress curve, is then fitted to an exponential equation to determine a rate constant k for the reaction.
  • the rate constant k is linearly proportional to the concentration of active enzyme present in the assay mixture once the rate constant for the spontaneous isomerization is subtracted.
  • the m for this substrate is much higher than 100 ⁇ M ([S3 «Km). Therefore, during inhibition experiments, the IC 50 , for non-tight binding inhibitors, is essentially the inhibition constant K
  • the Ki data reported under the PIN1-CD heading corresponds to testing with PIN1 peptide containing the catalytic peptidyl-prolyl isomerase domain but devoid of the PIN1 WW domain.
  • the dissociation constant (Ka) data under PIN1-CD refers to testing with a peptide containing the catalytic PIN1 domain but devoid of the PIN1 WW domain.
  • the exemplary compounds described above may be formulated into pharmaceutical compositions according to the following general examples.
  • Example 1 Parenteral Composition
  • a parenteral pharmaceutical composition suitable for administration by injection 100 mg of a water-soluble salt of a compound of Formula I is dissolved in DMSO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
  • a pharmaceutical composition for oral delivery 100 mg of a compound of Formula I is mixed with 750 mg of lactose. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.

Abstract

Phosphate/sulfate ester compounds that modulate and/or inhibit the activity of protein interacting NIMA (PIN1), and to pharmaceutical compositions containing such compounds are described. The invention is also directed to the therapeutic or prophylactic use of such compounds and compositions, and to methods of treating disorders characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders, by administering effective amounts of such compounds.

Description

PHOSPHATE/SULFATE ESTER COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS FOR INHIBITING PROTEIN INTERACTING NIMA iPINH
Field Of The Invention
This invention is directed to phosphate/sulfate ester compounds that modulate and/or inhibit the activity of protein interacting NIMA (PIN1), and to pharmaceutical compositions containing such compounds. The invention is also directed to the therapeutic or prophylactic use of such compounds and compositions, and to methods of treating disorders characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders, by administering effective amounts of such compounds.
Background Of The Invention
PIN1 is a member of the parvulin family of peptidyl-prolyl isomerases (PPIase) and catalyzes rotation about the peptide bond preceding a proline residue. PIN1 is a regulator of Cdc25, which dephoεphorylates Cdc2/cyclinB to drive cells into mitosis.
PIN1 has been identified in all eukaiyotic organisms where examined, including plants, yeast, insects, and mammals. The yeast (£ss1) and Dorosophilia (dodo) PIN1 orthologues have high identity to human-expressed sequence tags, which ultimately led to the cloning of the human docfo gene called PINL The Dorosophilia dodo gene is reported to be 45% identical to the yeast gene, Ess
Using a yeast two-hybrid screen of a human cDNA library, human PIN1 was originally identified as a binding protein of the fungi Aspergillus πidulens protein NIMA. NIMA is a kinase that drives cells into mitosis and is reported to be negatively regulated by PIN1. Depletion of NIMA in A. nidulans cells is reported to lead to cell cycle arrest in G2 while overexpression is reported to promote premature mitosis. SerThr kinase Cdc2/cyclin B may be the analogous NIMA kinase in human cells although another NIMA-like pathway in human cells is postulated to exist.
Modulation of PIN1 activity is reported to result in dramatic morphological cellular phenotypes. For example, overexpression of PIN1 in Hela cells was reported to cause a G2 arrest while depletion caused mitotic arrest— the opposite phenotypes observed with NIMA modulation. Additionally, decreasing PIN1 protein expression by full-length antisense expression has been reported to cause cells to progress into mitosis prematurely, to contain aberrant nuclei due to premature chromosome condensation and to induce apoptosis. These data indicate that PI 1 is a negative regulator of mitosis through interactions with a mammalian functional homoiog of NIMA and is required for progression through mitosis. Further, depletion of PIN1 is also postulated to play a role in Alzheimer's disease. Lu et al., Nature, 380, 544-547 (1996).
In vitro, PIN1 has been reported to interact with mitotic proteins also recognized by the MPM- 2 antibody. The MPM-2 monoclonal antibody recognizes a phospho-Ser/Thr-Pro epitope on about approximately 50 proteins associated with mitosis, including important mitotic regulators, such as Cdc25, Wee1, Cdc27, Map 4, and NIMA. See, e.g., Davis et al., Proc. Natl. Acad. Sci. U.S.A. 80, 2926 (1983). PIN1 has also been reported to interact with important upstream regulators of Cdc2/cyclin B, including Cdc25 and its known regulator, PlxL See Shen et al., Genes Dev. 12, 706 (1998). PIN1, due to its enzymatic action, may remove Cdc25 and Plx1 from play by causing their degradation within the cell.
Studies indicate that the biological function of PIN1 depends on a functional PPIase active site. Lu et al., Science, 283, 1325-1328 (1999). Studies also indicate that PIN1 recognizes its substrates (mitosis-specific phosphoproteins) through the WW domain. The WW domain is a protein recognition motif that is prevalent throughout biology. However, the PIN1 WW domain is unique in that it requires its ligand protein to contain a phosphorylated serine. As with the PPIase domain, a functional WW domain is reported to be essential for biological functions of PINL This is consistent with the model where PIN1 recognizes its substrates through the WW domain followed by completion of its essential catalytic role.
Full-length PIN1 protein and the nucleotide sequence encoding full-length PIN1 are disclosed in U.S. Patent Nos. 5,952,467 and 5,972,697. Additionally, sequences for PIN1 have been deposited in GenBank under accession numbers NM006221 (mRNA) and S68520 (protein). The mRNA sequence for dodo is deposited in GenBank under accession number U35140. Mouse PI 1 mRNA sequence is deposited in GenBank under accession number NM023371.
The crystal structure of full-length PINl is reported in Ranganathan, R. et al., Cell, 89, 875- 886 (1997) and International Publication No. WO 99/63931. Zhang et al. provide additional analysis of the crystal structure of PIN1 in complex with Ala-Pro (Biochemistry, 41:39 11868-77 (2002)).
Lu et al. (International Publication No. WO 01/38878) and Wulf et al. (EMBO J. 20, 3459- 3472 (2001)) disclose that PIN1 is upregulated in human tumors and is a biomarker for cell proliferation. Inhibitors of PI 1 have been described in the literature. For example, Hennig et al.
{Biochemistry, 37, 5953-5960 (1998)) report that juglone (5-hydroxy-1 ,4-naphthoquinone) selectively inhibits several parvulins, including human PINL Noel et al. in U.S. Patent Application No. 20010016346, using data based on the crystal structure derived from full-length human PIN1, disclose compounds postulated to be inhibitors of PINL Lu et al. in International Publication No. WO 99/12962 report inhibitors that mimic the phospho-Ser/Thr moiety of the phosphoserine or phosphothreonine-proiine peptidyl prolyl isomerase substrate.
Given the important role that PIN1 plays in the regulation of the cell cycle, additional compounds that inhibit PIN1 are needed. These compounds, along with pharmaceutical compositions thereof, can serve as effective chemotherapeutic agents for the treatment of a variety of disorders characterized by hypertension, inappropriate cell proliferation, including cancer, infectious diseases, and neurodegenerative brain disorders. The invention provides such compounds that inhibit PINL Summarv Of The Invention
Accordingly, an objective of the invention is to discover compounds and methods for modulating or inhibiting PINL Another objective of the invention is to provide compounds and methods for modulating or inhibiting PIN1 that can be used in pharmaceutical compositions for the treatment of disorders characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders.
These and other objectives of the invention, which will become apparent from the following description, have been achieved by the discovery of phosphate/sulfate ester compounds, pharmaceutically acceptable prodrugs, pharmaceutically active metabolites, and pharmaceutically acceptable salts thereof (such compounds, prodrugs, metabolites and salts are collectively referred to as "agents") described below, which inhibit PINL Pharmaceutical compositions containing such agents are useful in treating diseases characterized by hypertension, inappropriate cell proliferation, infectious diseases, and neurodegenerative brain disorders.
In a general aspect, the invention relates to compounds of the Formula I:
Figure imgf000004_0001
Formula I wherein: n is 1 or 2; A is a divalent -CH=CH-, -(CrCralkyl)-Y-, -NRα(CH2)t -Y-, -Y-(CrC7-alkyl)-, -Y-(C,-C7 alkyl)-,
-Y-NH-, -Y-NRd(CrC6-alkyl)-, -S-, -S(0)2-, -0-Y-, -Y-0-, -Y-S-, or -S-Y-, wherein Rd is H or C,-C6 alkyl, t is an integer from 0 to 5, Y is C(O), C(S), S(O), S(0)2,or a bond; X is a direct bond, CH2, CF2, O, S, NH, C(O), or C(S);
R1 is a C3-C10 cycloalkyl, 4-10 membered heterocycloalkyl, C3-C10 aryl, or 4-10 membered heteroaryl group, wherein R1 is unsubstituted or substituted with 1 to 4 R10 groups;
R2 is -S(0)2OH, -S(0)2NRdRe, or -P(0)(OR4)2, wherein R4 is an H, CrC10-alkyl, C6-C 0 aryl, or -CH2-0-C(0)ReCH3 group, Rd and Re are each independently an H or CrC6 alkyl group, and R4 is unsubstituted or substituted with 1 to 4 R10 groups; and
R3 is OH, CrC7-alkyl, CrC7-alkoxyl, C6-C10 aryl, 4-10 membered heteroaryl, C3-C10 cycloalkyl, 3-10 membered heterocycloalkyl, -NH(R5), or -N(R5)2 group, wherein R5 is independently selected from H, CrC7 alkyl, C6-C10 aryl, or
Figure imgf000005_0001
wherein ring B is a 5- or 6-membered heterocycloalkyl group, Z is a divalent C(0)Z\ heteroaryl or heterocycloalkyl group wherein Z' is a divalent 0, S, NH, N(CH3), C02, or CH2, and R6 is H, CΓCID alkyl, aryl, CrC6 alkyl-aryl, or arylalkyl group, wherein R3, R5, B and R6 are unsubstituted or substituted with 1 to 4 R10 groups; wherein each R10 is independently selected from halo, amino, =0, =S, =NH, cyano, nitro, hydroxyl, -SH, haloalkyl, 2-10 membered heteroalkyl, CrC6 alkoxy, Cι_C10 alkyl, C2.C6 alkenyl, C2-CB alkynyl, -C(0)jRa, -OC(0)jRd, -OC(0)OC(0)Rd, -OOH, -C(NRd)NRbRc, -NRdC(NRe)NRbR° -NRdC(0)jRb, -C(0)NRbRc, -C(0)NRdCOR , -OC(0)NRbRc, -NRbRc, -NRdORc, -C(S)NR Rc -NRdC(S)NRbRc, -NRdC(0)NRbRc, -OSH, -S(0)jRb, -OS(0)jRb, -SC(0)Rb, -S(0)jC(0)OR , -SCOR' -NRdSR°, -SR , -NHS(0)jRb, -COSR , -C(0)S(0)jRb, -CSR , -CS(0)jRb, -C(SO)OH, -C(S0)20H -NRdC(S)RG, -0C(S)Rb, -OC(S)OH, -OC(SO)2Rb, -S(0)jNR R°, -SNRbR°, -S(0)NRbR°, -NRdCS(0)jRc -C(O)j(CH2)tNR -(4-10 membered heteroaryl), -C(O)j(CH2)tNRd(4-10 membered heterocycloalkyl) -(CRdRe)tCN, -(CRd Re)t(C3-C10 cycloalkyl), -(GRriRe)t(CrC10 aryl), -(CRdRΘ),(4-10 membered heterocycloalkyl), -(CRdRs)t(4-10 membered heteroaryl), -(CRdRe)qC(O)(CRdRe)t(C3-C10 cycloalkyl) -(CRdRΘ)qC(0)(CRdR8)t(Cs-C1D aryl), -(CRdRe)qC(O)(CRdR8)t(4-10 membered heterocycloalkyl) -(CRdRe)qC(O)(CRdRe),(4-10 membered heteroaryl), -(CRdR°)tO(CRdRe)q(C3-C1o cycloalkyl) -(CRdRe)(O(CRdRe)q(C6-C10 aryl), -(CRdRe),O(CRdR6)q(4-10 membered heterocycloalkyl) -(CRdRΘ)tO(CRdRe)q(4-10 membered heteroaryl), -(CRdRe)qSO2(CRdRe)t(C3-C10 cycloalkyl) -(CRdRs)qSO2(CRdReMCδ-C10 aryl), -(CRdRβ)qSO2(CRdRs),(4-10 membered heterocycloalkyl), and -(CR Re)qS02(CR Re)l(4-10 membered heteroaryl), wherein Ra is selected from the group consisting of halo, hydroxyl, -NRdRs, C Cι0 alkyl, haloalkyl, CrCB alkoxyl, Rb and Rc are independently selected from H, CrCιo alkyl, -(CRdRe)t(C3-C10 cycloalkyl), -(CRdRs)t(C3-C 0 aryl), -(CRdRε)t(4-10 membered heterocycloalkyl), and -(CRdRe)t(4-10 membered heteroaryl), Rd and Re are independently H or C C6 alkyl, j is an integer from 0 to 2, q and t are each independently an integer from 0 to 5, and 1 or 2 ring carbon atoms of the cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with =0, and the aikyl, alkenyl, alkynyl, aryl and cyclic moieties of the foregoing R1D groups are unsubstituted or substituted with 1. to 3 substituents independently selected from halo, =0, cyano, nitro, -(CRdRe),CN, haloalkyl, 2-10 membered heteroalkyl, -OR , -C(0)jRb, -NRdC(0)R , -C(0)NR Rc, -NRbRc, -NR OR c, -NRdC(0)jNRbR°, -NRdC(0)jRbRc, -OC(0)jRb, -OC(0)NRbRc, -SRd, CrC10 alkyl, C2- CB alkenyl, C2-C6 alkynyl, -(CRdRe)t(C3-C10 cycloalkyl),
Figure imgf000005_0002
aryl), -(CRdRe),(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl), -(CRdRe)t(C6-Cι0 aryl)-(C C6 alkyl); wherein t, Rb, Rc, Rd, Reare as defined above. The invention is also directed to pharmaceutically acceptable prodrugs, pharmaceutically active metabolites, and pharmaceutically acceptable salts of the compounds of Formula I and their pharmaceutically active metabolites. Advantageous methods of making the compounds of the Formula I are also described.
In a preferred embodiment, the invention relates to compounds of Formula I, wherein n is 1 or 2; A is a divalent -NH-Y-, -NRd(CH2)rY-, or -0-Y-, and Y is C(0) or S(0)2; X is a direct bond, CH2, O, or S; R1 is a C6-C10 aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R10 groups; R2 is -S(0)2OH, or -P(0)(0R4)2, wherein R4 is an H, C Cι0 alkyl, or C6-C10 aryl group, and is unsubstituted or substituted with 1 to 4 R10 groups; and R3 is a C6-Cι0 aryl, 4-10 membered heteroaryl, -NH(C6H5), or
Figure imgf000006_0001
wherein ring B is a 5- or 6-membered heterocycloalkyl group, Z is a divalent C(0)Z', heteroaryl or heterocycloalkyl group wherein Z' is a divalent O, S, NH, N(CH3), C02, or CH2, and R6 is H or a Cι-C10 alkyl group, wherein R3, B, and R6 is unsubstituted or substituted with 1 to 4 R10 groups; and wherein R10 is as defined above.
In a particularly preferred embodiment, the invention relates to compounds of Formula I, wherein n is 1 ; A is a divalent -NH-Y- or -0-Y-, wherein Y is C(O); X is a direct bond, CH2, or O; R1 is a C6-C10 aryl group unsubstituted or substituted with 1 to 4 R10 groups; R2 is -P(0)(OR4)2, wherein R4 is an H, CrCio alkyl, or C6-Cι0 aryl group, and is unsubstituted or substituted with 1 to 4 R groups; and R3 is a C6-C10 aryl, 4-10 membered heteroaryl, or
Figure imgf000006_0002
wherein ring B is an unsubstituted 6-membered heterocycloalkyl, Z a divalent C(0)Z', Σ is a divalent
0, S, or CH2, and R6 is a CrCio alkyl group, wherein R3, B and R6 are unsubstituted or substituted with 1 to 4 R10 groups; and wherein R10 is as defined above.
In a further particularly preferred embodiment, the invention relates to compounds of Formula
1, wherein n is 1; A is -NH-Y- or -0-Y-, wherein Y is C(O); X is a direct bond, CH2, or O; R1 is a CB- C 0 aryl group unsubstituted or substituted with 1 to 4 R10 groups; R2 is -P(0)(0R4)2, wherein R4 is an H or a C1-C10 alkyl group that is unsubstituted or substituted with 1 to 4 R10 groups; and R3 is a C6-C10 aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R 0 groups; and wherein R10 is as defined above.
Preferably, the invention includes compounds, and pharmaceutically acceptable salts thereof, selected from the following group:
Figure imgf000007_0001
-7-
Figure imgf000008_0001
Figure imgf000009_0001
The invention also relates to a method of inhibiting PIN1 by administering a compound of Formula I or a pharmaceutically acceptable prodrug, pharmaceutically active metabolite, or pharmaceutically acceptable salt of such compound or metabolite thereof.
The invention also relates to pharmaceutical compositions, each comprising a therapeutically effective amount of an agent selected from compounds, prodrugs, metabolites, and salts of compounds of Formula I, and a pharmaceutically acceptable carrier or vehicle for such agent. The invention further provides methods of treating mammalian disease conditions mediated by PIN1 activity, by administering to a mammal in need thereof a therapeutically effective amount of a compound, prodrug, active metabolite or salt of a compound of Formula I. The mammalian disease conditions to be treated according to the invention are associated with hypertension, inappropriate cell proliferation (e.g., cancer), infectious diseases (e.g., bacterial and fungal infections), and neurodegenerative brain disorders (e.g., Alzheimer's disease).
The compounds of Formula I are useful for modulating or inhibiting PINL More particularly, the compounds are useful as modulating or inhibiting the activity of PIN 1, thus providing treatments for hypertension, infectious diseases, neurodegenerative disorders, and cancer or other diseases associated with cellular proliferation. The terms "comprising" and "including" are used herein in their open, non-limiting sense.
As used herein, "inappropriate cell proliferation" includes diseases or disorders associated with uncontrolled or abnormal cellular proliferation. Such diseases and disorders include, but are not limited to, the following: a variety of cancers, including, but not limited to, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, neoplasms of the central nervous system (CNS), primary CNS lymphoma, spinal axis tumors, brain stem glio a, pituitary adenoma, or a combination of one or more of the foregoing cancers; a disease process which features abnormal cellular proliferation, e.g., benign prostatic hyperpiasia, familial adenomatosis polyposis, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis following angioplasty or vascular surgery, hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, endotoxic shock, and fungal infections; and defective apoptosis-associated conditions, such as cancers (including, but not limited to, those types mentioned herein above), viral infections (including, but not limited to,
HIV, human papilloma virus, herpesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals autoimmune diseases (including, but not limited to, systemic lupus erythematosus rheumatoid arthritis, psoriasis, autoimmune mediated glomerulonephritis inflammatory bowel disease and autoimmune diabetes mellitus), neurodegenerative disorders (including, but not limited to, Alzheimer's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, Parkinson's disease, AIDS-related dementia, spinal muscular atrophy and cerebellar degeneration), myelodysplastic syndromes, aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including, but not limited to, chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including, but not limited to, osteroporosis and arthritis), aspirin-sensitive rhinosinusitis, cystic fibrosis, multple sclerosis, kidney diseases, and cancer pain. The term "alkyl" as used herein refers to a straight- or branched-chain, saturated or partially unsaturated, alkyl group having one to twelve carbon atoms. Preferred alkyl groups have from 1-10, and more preferably from 1-7, carbon atoms. Exemplary alkyl groups include methyl (Me, which also may be structurally depicted by ), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and the like. The term "lower alkyl" designates an alkyl having from 1 to 6 carbon atoms (a C C6 alkyl).
The term "aryl" (Ar) refers to a monocyclic, or fused or spiro polycyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) having from three to twelve ring atoms per ring, preferably 6-10 ring atoms atoms and more preferably 5-7 ring atoms.
Illustrative examples of aryl groups include the following moieties:
Figure imgf000011_0001
The term "heteroaryl" (heteroAr) refers to a monocyclic, or fused or spiro polycyclic, aromatic heterocycle (ring structure having ring atoms selected from carbon atoms as well as nitrogen, oxygen, and sulfur heteroatoms) having from three to twelve ring atoms per ring, preferably 4-10 ring atoms and more preferably 5-7 ring atoms. Illustrative examples of aryl groups include the following moieties:
Figure imgf000011_0002
and the like.
Figure imgf000011_0003
The term "cycloalkyl" refers to a saturated or partially saturated, monocyclic or fused or spiro polycyclic, carbocycle having from three to twelve ring atoms per ring, preferably 3-10 carbon atoms and more preferably 5-7 carbon atoms. Illustrative examples of cycloalkyl groups include the following moieties:
Figure imgf000012_0001
, and the like.
Figure imgf000012_0002
A "heterocycloalkyl" refers to a monocyclic, or fused or spiro polycyclic, ring structure that is saturated or partially saturated and has from three to twelve ring atoms per ring selected from C atoms and N, O, and S heteroatoms, preferably 4-10 ring atoms and more preferably 5-7 ring atoms. Illustrative examples of heterocycloalkyl groups include:
Figure imgf000012_0003
and the like.
Figure imgf000012_0004
The term "alkoxy" refers to -O-alkyl. Illustrative examples include methoxy, ethoxy, propoxy, and the like.
The term "halogen" represents chlorine, fluorine, bromine or iodine. The term "halo" represents chloro, fluoro, bromo or iodo. As used herein, "haloalkyl" refers to a loweralkyl radical in which one or more of the hydrogen atoms are replaced by halogen including, but not limited to, chloromethyl, trifiuoromethyl, 1- chloro-2-fluoroethyl and the like.
"Heteroalkyl" is an alkyl group (as defined herein) wherein at least one of the carbon atoms is replaced with a heteroatom. Preferred heteroatoms are nitrogen, oxygen, sulfur, and halogen. A heteroatom may, but typically does not, have the same number of valence sites as carbon.
Accordingly, when a carbon is replaced with a heteroatom, the number of hydrogens bonded to the heteroatom may need to be increased or decreased to match the number of valence sites of the heteroatom. For instance, if carbon (valence of four) is replaced with nitrogen (valence of three), then one of the hydrogens formerly attached to the replaced carbon must be deleted. Likewise, if carbon is replaced with halogen (valence of one), then three (i.e., all) of the hydrogens formerly bonded to the replaced carbon must be deleted. As another example, trifiuoromethyl is a heteroalkyl group wherein the three methyl groups of a t-butyl group are replaced by fluorine. Preferred heteroalkyls of the invention have 2 to 10 member atoms, including both heteroatoms and carbon atoms. The term "substituted" means that the specified group or moiety bears one or more substituents. The term "unsubstituted" means that the specified group bears no substituents.
The compounds of the invention may exhibit the phenomenon of tautomerism. While Formula I cannot expressly depict all possible taufomeric forms, it is to be understood that Formulas I is intended to represent any tautomeric form of the depicted compound and are not to be limited merely to a specific compound form depicted by the formula drawings.
The compounds of Formula I may have one or more asymmetric centers designated by an asterisk as shown below in Formula I. Additional asymmetric centers may be present on the molecule depending upon the nature of the various substituents on the molecule.
Figure imgf000013_0001
As a consequence of these asymmetric centers, the compounds of Formula I may exist as single stereoisomers (i.e., essentially free of other stereoisomers), racemates, and/or mixtures of enantiomers and/or diastereomers. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention. Preferably, the inventive compounds that are optically active are used in optically pure form.
In accordance with a convention used in the art, ' "' is used in structural formulae herein to depict the bond that is the point of attachment of the moiety or substituent to the core or backbone structure.
As generally understood by those skilled in the art, an optically pure compound having one chiral center (i.e., one asymmetric carbon atom) is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure. Preferably, the compounds of the present invention are used in a form that is at least 90% optically pure, that is, a form that contains at least 90% of a single isomer (80% enantiomeric excess ("e.e.") or diastereomeric excess ("d.e.")), more preferably at least 95% (90% e.e. or d.e.), even more preferably at least 97.5% (95% e.e. or d.e.), and most preferably at least 99% (98% e.e. or d.e.).
Additionally, Formula I is intended to cover solvated as sell as unsolvated forms of the identified structures. For example, Formula I includes compounds of the indicated structure in both hydrated and non-hydrated forms. Other examples of solvates include the structures in combination with isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
In addition to compounds of Formula I, the invention includes pharmaceutically acceptable prodrugs, pharmaceutically active metabolites, and pharmaceutically acceptable salts of such compounds and metabolites.
"A pharmaceutically acceptable prodrug" is a compound that may be converted under physiological conditions or by solvolysis to the specified compound or to a pharmaceutically acceptable salt of such compound.
"A pharmaceutically active metabolite" is intended to mean a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof.
Prodrugs and active metabolites of a compound may be identified using routine techniques known in the art. See, e.g., Bertolini et al., J. Med. Che ., 40, 2011-2016 (1997); Shan, et al., J. Pharm. Sci., 86 (7), 765-767; Bagshawe, Drug Dev. Res., 34, 220-230 (1995); Bodor, Advances in Drug Res., 13, 224-331 (1984); Bundgaard, Design of Prodrugs (Elsevier Press 1985); Larsen, Design and Application of Prodrugs, Drug Design and Development (Krogsgaard-Larsen et al., eds., Harwood Academic Publishers, 1991); Dear et al., J. Chromatogr. B, 748, 281-293 (2000); Spraul et al., J. Pharmaceutical & Biomedical Analysis, 10, 601-605 (1992); and Pros et al., Xenobiol, 3, 103- 112 (1992).
"A pharmaceutically acceptable salt" is intended to mean a salt that retains the biological effectiveness of the free acids and bases of the specified compound and that is not biologically or otherwise undesirable. A compound of the invention may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt. Exemplary pharmaceutically acceptable salts include those salts prepared by reaction of the compounds of the present invention with a mineral or organic acid or an inorganic base, such as salts including sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-
1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybeπzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycoiates, tartrates, methane-sulfonates, propanesulfonates, naphthalene-1-sulfonates, naphthalene-2-sulfonates, and mandelates.
If the inventive compound is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucϋronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
If the inventive compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
In the case of agents that are solids, it is understood by those skilled in the art that the inventive compounds and salts may exist in different crystal or polymorphic forms, all of which are intended to be within the scope of the present invention and specified formulas.
Lu et al. (International Publication No. WO 01/38878; incorporated herein by reference in its entirety) disclose that PI 1 is overexpressed in a variety of cancers, including breast, colon, and prostate. Additionally, the authors disclose that P1 1 is overexpressed in proliferating cells.
Therefore, the agents of the invention would have use for treating a variety of cell proliferative diseases associated with overexpression of PIN1.
Therapeutically effective amounts of the agents of the invention may be used to treat diseases mediated by modulation or regulation of PINL An "effective amount" is intended to mean that amount of an agent that, when administered to a mammal in need of such treatment, is sufficient to effect treatment for a disease modulated or inhibited by the activity of PINL Thus, e.g., a therapeutically effective amount of a compound of the Formula I, salt, active metabolite or prodrug thereof is a quantity sufficient to modulate, regulate, or inhibit the activity of PIN1 such that a disease condition which is mediated by that activity is reduced or alleviated.
The amount of a given agent that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art. The term "treating" refers to:
(i) preventing a disease, disorder, or condition from occurring in an animal that may be predisposed to the disease, disorder and/or condition, but has not yet been diagnosed as having it; (ii) inhibiting the disease, disorder, or condition, i.e., arresting its development; and (iii) relieving the disease, disorder, or condition, i.e., causing regression of the disease, disorder, and/or condition.
Detailed Description Of The Invention And Preferred Embodiments
The active agents of the invention may be formulated into pharmaceutical compositions as described below. Pharmaceutical compositions of this invention comprise an effective modulating, regulating, or inhibiting amount of a compound of Formula I and an inert, pharmaceutically acceptable carrier or diluent. In one embodiment of the pharmaceutical compositions, efficacious levels of the inventive agents are provided so as to provide therapeutic benefits involving modulation of PINL By "efficacious levels" is meant levels in which the effects of PIN1 activity are, at a minimum, regulated. These compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g., parenteral or oral administration. An inventive agent can be administered in conventional dosage form prepared by combining a therapeutically effective amount of an agent (e.g., a compound of Formula I) as an active ingredient with appropriate pharmaceutical carriers or diluents according to conventional procedures. These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation. The pharmaceutical carrier employed may be either a solid or liquid. Exemplary of solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary of liquid carriers are syrup, peanut oil, olive oil, water and the like. Similarly, the carrier or diluent may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate and the like.
A variety of pharmaceutical forms can be employed. Thus, if a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge. The amount of solid carrier may vary, but generally will be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation will be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or non-aqueous liquid suspension.
To obtain a stable water-soluble dose form, a pharmaceutically acceptable salt of an inventive agent is dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3M solution of succinic acid or citric acid. If a soluble salt form is not available, the agent may be dissolved in a suitable cosolvent or combinations of cosolvents. Examples of suitable cosolvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, gylcerin and the like in concentrations ranging from 0-60% of the total volume. In an exemplary embodiment, a compound of Formula I is dissolved in DMSO and diluted with water. The composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution. It will be appreciated that the actual dosages of the agents used in the compositions of this invention will vary according to the particular complex being used, the particular composition formulated, the mode of administration and the particular site, host and disease being treated.
Optimal dosages for a given set of conditions can be ascertained by those skilled in the art using conventional dosage-determination tests in view of the experimental data for an agent. For oral administration, an exemplary daily dose generally employed is from about 0.001 to about 1000 g/kg of body weight, with courses of treatment repeated at appropriate intervals. Administration of prodrugs are typically dosed at weight levels, which are chemically equivalent to the weight levels of the fully active form. The compositions of the invention may be manufactured in manners generally known for preparing pharmaceutical compositions, e.g., using conventional techniques such as mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing. Pharmaceutical compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers, which may be selected from excipients and auxiliaries that facilitate processing of the active compounds into preparations, which can be used pharmaceutically.
Proper formulation is dependent upon the route of administration chosen. For injection, the agents of the invention may be formulated into aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient io be treated. Pharmaceutical preparations for oral use can be obtained using a solid excipient in admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, polyvinyl pyrrolidone, Carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents.
Pharmaceutical preparations, which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active agents may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration intranasally or by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of gelatin for use in an inhaler or insufflator and the like may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit-dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active agents may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g, containing conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described above, the compounds may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion-exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
An exemplary pharmaceutical carrier for hydrophobic compounds is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent system may be a VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD:5W) contains VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a co- solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may be substituted for dextrose. Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed. The pharmaceutical compositions also may comprise suitable solid- or gel-phase carriers or excipients. Examples of such carriers or excipients include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Some of the compounds of the invention may be provided as salts with pharmaceutically compatible counter ions. Pharmaceutically compatible sails may be formed with many acids, including hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Sails lend to be more soluble in aqueous or other protonic solvents than are the corresponding free-base forms.
The inventive agents may be prepared using the reaction routes and synthesis schemes as described below, employing the general techniques known in the art using starting materials that are readily available. The preparation of preferred compounds of the present invention is described in detail in the following examples, but the artisan will recognize that the chemical reactions described may be readily adapted to prepare a number of other PIN1 inhibitors of the invention. For example, the synthesis of non-exempiified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or generally known in the art will be recognized as having applicability for preparing other compounds of the invention. Examples
In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius (°C) and all parts and percentages are by weight. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company or Lancaster Synthesis Ltd. and were used without further purification unless otherwise indicated. Tetrahydrofuran and N, N-dimethylformamϊde were purchased from Aldrich in Sure Seal bottles and used as received. All solvents were purified using standard methods known to those skilled in the art, unless otherwise indicated.
The reactions set forth below were done generally under a positive pressure of argon at an ambient temperature (unless otherwise stated) in anhydrous solvents, and the reaction flasks were fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried. Analytical thin layer chromatography (TLC) was performed on glass- backed silica gel 60 F 254 plates from Analtech (0.25 mm), eluted with the appropriate solvent ratios (v/v), and were denoted where appropriate. The reactions were assayed by TLC and terminated as judged by the consumption of starting material.
Visualization of the TLC plates was done with iodine vapor, ultraviolet illumination, 2% Ce(NH4)4(Sθ4)4 in 20% aqueous sulfuric acid, or p-anisaldehyde spray reagent, and activated with heat where appropriate. Work-ups were typically done by doubling the reaction volume with the reaction solvent or extraction solvent and then washing with the indicated aqueous solutions using 25% by volume of the extraction volume unless otherwise indicated. Product solutions were dried over anhydrous Na2S04 and/or Mg2SO,j prior to filtration and evaporation of the solvents under reduced pressure on a rotary evaporator and noted as solvents removed in vacua. Flash column chromatography (Still et al., J. Org. Chem., 43, 2923 (1978)) was done using Merck silica gel (47-61 μm) with a silica gel crude material ratio of about 20:1 to 50:1 , unless otherwise stated. Hydrogenolysis was done at the pressure indicated in the examples or at ambient pressure. All melting points (mp) are uncorrected.
1H-NMR spectra were recorded on a Bruker or Varian instrument operating at 300 MHz and 13C-NMR spectra were recorded operating at 75 MHz. NMR spectra were obtained as CDCl3 solutions (reported in ppm), using chloroform as the reference standard (7.27 ppm and 77.00 ppm) or CD3OD (3.4 and 4.8 ppm and 49.3 ppm), or internally tetramethylsilane (0.00 ppm) when appropriate. Other NMR solvents were used as needed. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz). Infrared (IR) spectra were recorded on a Perkin-Elmer FT-IR Spectrometer as neat oils, as
KBr pellets, or as CDCI3 solutions, and when given are reported in wave numbers (cm"1).
Mass spectrometry (MS) was conducted with various techniques. Mass spectra were obtained using liquid chromatograph electrospray ionization mass spectrometry, MS (ESP). Matrix- Assisted Laser Desorption/lonization (MALDI) Fourier Transform Mass Spectrometry was performed on an lonSpec FTMS mass spectrometer. The following compounds of the invention were made according to the general synthetic pathways shown in Schemes 1-10 and the detailed experimental procedures that follow thereof. These synthetic pathways and experimental procedures utilize many common chemical abbreviations, such as THF (tetrahydrofuran), DMF (N,N-dimethylformamide), EtOAc (ethyl acetate), DBU (1,8-diazacyclo[5.4.0]undec-7-ene), TMSCI (trimethylsilyl chloride), MCPBA (3- chloroperoxybenzoic acid), EDC (1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride), HOBT (1-hydroxybenzotriazole hydrate), DMAP (4-dimethylaminopyridine), TBDPSCI (t- butyldiphenylchlorosilane), TMSBr (bromotrimethylsilane ), DIEA (diisopropylethylamine), TBAI (tetrabutylamonium iodide), and the like.
Scheme 1
Figure imgf000021_0001
CISO3H
Et3N 5b1 m=2 R6=-(CH2)4Ph Z=-C(0)0-
Figure imgf000021_0002
3b4 R6=Bn
Alcohol 2a:
Figure imgf000021_0003
To a pyridine solution (1 mL) of the sulfamoyl chloride 1a (0.14 g, 0.5 mmol, preparation described in International Publication No. WO 0140185) was added D-phenylalaninol (0.151 g, 1 mmol) at 25 °C. After 12 h, the reaction mixture was concentrated in vacuo. The yellow residue was purified by flash column chromatography (25% ethyl acetate (EtOAc) in hexanes) followed by preparative TLC to afford 70 mg (33% yield) of the compound 2a. 1H NMR (CDCI3): δ 7.24-7.0 (1 OH, m), 4.89 (2H, AB), 4.70 (1H, d, J=8.7 Hz), 4.25 (1H, dd, J=8.7, 3.6 Hz), 3.54 (2H, m), 3.28 (1H, m), 3.04 (2H, m), 2.66 (2H, d, J=6.6 Hz), 2.33 (1H, m), 2.01 (1H, m); HRMS (FAB) caic for C21H27N2OsS (M+H+) 419.1641; found 419.1629.
Example 3a: 1-(2-Phenyl-1-sulfooxymethyl-ethylsulfamoyl)-pyrrolidine-2S-carboxylic acid benzyl ester
Figure imgf000022_0001
At -70 °C, a methylene chloride solution (2 mL) of the alcohol 2a (10 mg, 0.024 mmol) was added triethylamine (Et3N, 0.05 mL) and chlorosulfonic acid (8 mg, 5 μl, 0.068 mmol). The cooling bath was then removed and the reaction mixture was allowed to warm to 25 °C over 3 h. All solvent was evaporated in vacuo. The residue was purified by column chromatography (3% methanol (MeOH) in EtOAc) to give 8 mg (67% yield) of the title compound 3a. 1H NMR (CD3OD): δ 7.4-7.15 (10H, m), 5.16 (2H, AB), 4.22 (1H, dd, J=8.7, 3.9 Hz), 4.04 (1H, dd, J=10.2, 4.8 Hz), 3.91 (1H, dd, J=10.2, 4.5 Hz), 3.77 (1H, m), 3.38 (1H, m), 3.14 (1H, m), 2.93 (1H, dd, J=14.1, 9.1 Hz), 2.81 (1H, dd, J=14.1, 6.6 Hz), 2.18 (1H, m), 1.97-1.73 (3H,m); MS (ESP): 497 (M-H+); HRMS (FAB) caic for C21H26N208S2Na (M+Na+) 521.1028; found 521.1010.
Alcohol 2b1:
Figure imgf000022_0002
Prepared as described in the synthesis of 2a using the n-butyl-phenyl ester of sulfamoyl chloride 1a (0.15 g, 0.42 mmol, preparation described in International Publication No. WO 0140185) and D-phenylalaninol (0.18 g, 1.2 mmol). After purification by flash column chromatography (25% I
-22-
EtOAc in hexanes), the compound 2b1 (62 mg) was obtained in 31% yield. 1H NMR (CDCI3): δ 7.4- 7.1 (10H, m), 5.17 (1H, d, J=8.1 Hz), 4.68 (1H, d, J=4.5 Hz), 4.20 (2H, m), 3.75 (2H, m), 3.55 (1H, m), 3.35 (1H, br d, J=12.1 Hz), 2.95-2.73 (3H, ), 2.67 (2H, m), 2.45 (1H, br s), 2.24 (1H, d, J=13.1 Hz); HRMS (FAB) caic for CsHMNj-OsSNa (M+Na+) 497.2086; found 497.2079.
Example 3b1: 1-(2-Phenyl-1-sulfooxymethyl-ethylsulfamoyl)-piperidine-2S-carboxylic acid 4- phenyl-butyl ester
Figure imgf000023_0001
Prepared as described in the synthesis of 3® using the alcohol 2b1 (10 mg, 0.021 mmol), chlorosulfonic acid (6 mg, 4 μl, 0.055 mmol) and triethylamine (0.015 mL). The reaction mixture was diluted with EtOAc (20 mL) and washed with ice-cold 5% hydrochloric (HCI) solution (1x20 mL).
Column chromatography (8% MeOH in EtOAc) afforded 10 mg (85% yield) of the title compound 3b1. 1H NMR (CDCI3): δ 7.3-6.9 (10H, m), 6.03 (1H, br s), 4.26 (2H, m), 4.05 (2H, m), 3.93 (1H, br s), 3.58 (1H, br s), 3.13 (1H, m), 2.79 (3H, m), 2.48 (2H, m), 2.30 (1H, m), 1.88 (1H, m); HRMS
(FAB) caic for C25H33N208S2Cs2 (M-H++2Cs+) 818.9787; found 818.9756.
Alcohol 2b2:
Figure imgf000023_0002
Prepared as described in the synthesis of 2a using the sulfamoyl chloride 1b2 (0.15g, 0.47 mmol, preparation described in International Publication No. WO 0140185) and D-phenylalaninol (0.214 g, 1.4 mmol). After purification by flash column chromatography (25% to 30% EtOAc in hexanes), the compound 2b2 (66 mg) was obtained in 33% yield. 1H NMR (CDCI3): δ 7.4-7.17 (10H, m), 5.19 (2H, AB), 5.03 (1H, d, J=8.4 Hz), 4.71 (1H, br d, J=4.8 Hz), 3.8-3.6 (2H, m), 3.48 (1H, m), 3.31 (1 H, br d, J=12.6 Hz), 2.88-2.74 (3H, m), 2.34 (1H, br t, J=5.1 Hz), 2.25 (1H, m). Example 3b2: 1-(1-Benzyl-2-sulfooxy-ethylsulfamoyl)-piperidine-2S-carboxylic acid benzyl ester
Figure imgf000024_0001
Prepared as described in the synthesis of 3a using the alcohol 2b2 (30 mg, 0.069 mmol), chlorosulfonic acid (16 mg, 10 μl, 0.13 mmol) and triethylamine (0.05 mL). The reaction mixture was diluted with EtOAc (20 mL) and washed with ice-cold 5% HCI solution (1x20 mL). Column chromatography purification (5% MeOH in EtOAc) afforded 25 mg (70% yield) of the title compound 3b2. 1H NMR (CDCI3): δ 7.31-7.01 (10H, m), 6.03 (1H, br d, J=9 Hz), 5.16 (1H, d, J=12.3 Hz), 5.00 (1H, d, J=12.3 Hz), 4.43-4.30 (2H, m), 4.1 (1H, m), 3.66 (1H, m), 3.37 (3H, m), 3.14 (1H, m), 2.80 (3H, m), 1.98 (1H, br d); HRMS (FAB) caic for C22H27 2θsS2 a2 (M-H*+2Na+) 557.1004; found 557.1019.
Alcohol 2b3:
Figure imgf000024_0002
Prepared as described in the synthesis of 2a using the sulfamoyl chloride 1b3 (0.05 g, 0.105 mmol, preparation described in International Publication No. WO 0140185) and D-phenylalaninol (0.026 g, 0.17 mmol). After purification by flash column chromatography (25% to 30% EtOAc in hexanes), the compound 2b3 (30 mg) was obtained in 48% yield. 1 NMR (CDCI3): δ 7.35-7.09 (15H, m), 5.07 (1H, d, J=7.8 Hz), 5.02 (1H, m), 4.64 (1H, br d, J=4.5 Hz), 3.69 (2H, ), 3.47 (1H, m), 3.30 (1H, br d, J=12.6 Hz), 2.84 (2H, d, J=7.2 Hz), 2.80 (1H, td, J=13.5, 3.9 Hz), 2.67-2.51 (4H, m), 2.26-2.08 (2H, m). Example 3b3: 1-(2-Phenyl-1-sulfooxymethyI-ethylsulfamoyl)-piperidine-2S-carboxylic acid 4- phenyl-1 -(3-phenyl-propyl)-butyl ester
Figure imgf000025_0001
Prepared as described in the synthesis of 3a using the alcohol 2b3 (10 mg, 0.0169 mmol), chlorosulfonic acid (8 mg, 5 μl, 0.068 mmol) and triethylamine (0.05 mL). The reaction mixture was diluted with EtOAc (20 L) and washed with ice-cold 5% HCI solution (1x20 mL). Column chromatography purification (5% MeOH in EtOAc) afforded 8 mg (70% yield) of the title compound 3b3. 1H NMR (CDCI3): δ 7.36-7.00 (15H, m), 6.06 (1H, br d, J=7.5 Hz), 4.90 (1H, m), 4.31 (1H, br d), 4.20 (1H, br s), 4.06 (1H, m), 3.66 (1H, m), 3.34 (1H, br d, J=11.1 Hz), 2.85 (3H, m), 2.51 (4H, m); HRMS (FAB) caic for CsΛsNzOsSjjCsa (M-H++2Cs+) 937.0570; found 937.0557.
Synthesis of Benzyl Ester
C CBBzzCCII Na0H '
Figure imgf000025_0002
Figure imgf000025_0003
To a mixture of L-penicillamine (14.92 g), 1 ,2-dichloroethane (300 mL) and DMF (2 mL) at 0 °C was added 1,8-diazacyclo[5.4.0jundec-7-ene (DBU, 22.4 mL), followed by trimethylsilyl chloride (TMSCI, 19 mL). After stirring for 3 h, the solution was warmed to 25 °C, followed by the slow addition of DBU (29.9 mL). The reaction mixture was stirred for 17 h at 25 °C. Methanol (10 mL) was added and a precipitate formed. The precipitate was collected by filtration and was rinsed with a minimum amount of methanol. The solid was dried in vacuo at 50 °C for 6 h to give 3(R)-2,2- dimethyl-tetrahydro-2H-1,4-thiazine-3-carboxylic acid as a white powder (16 g). At 0 °C, a portion of the thiazine (0.4 g, 2.3mmo!) was dissolved in a NaOH solution (1 N, 12 mL). To the resulting mixture was added benzylchloroformate (1.4 mL, 9.2 mmol). After 15 h at 25 DC, the solution was diluted with water (20 mL) and extracted with EtOAc (2x25 mL). The extracts were dried (MgS04) and concentrated in vacuo. Flash column chromatography (15-20% EtOAc in hexanes) purification afforded 0.63 g of the title compound 3b3. 1H NMR (CDCI3): (mixture of two rotamers) δ 7.3 (10H, m), 7.11 (4H, br s), 4.87 and 4.70 (1H, s), 4.40 and 4.28 (1H, d, J=6.7 Hz), 3.72 and 3.60 (1H, m), 2.94 (1H, m), 2.37 (1H, t, J=3.9 Hz), 1.45 (3H, s), 1.34 (3H, s); MS (ESP) 400 (M+H+). Sulfamoyl Chloride 1b4:
Figure imgf000026_0001
To a methylene chloride solution (2 mL) of the benzyl ester (0.6 g, 1.5 mmol) at 0 °C was added methyl sulfide (1 mL) and BF3 • Et20 (0.2 L). After 16 h, the reaction mixture was added sat'd NaHC03 solution (5 mL) and extracted with methylene chloride (2x20 mL). Combined organic layers were washed with brine (1x25 mL) and dried (Na2S04). All solvent was removed in vacuo to give a pale yellow oil (0.35 g), which was dissolved in methylene chloride (8 mL). To the resulting solution was slowly added triethylamine (1 mL) and CIS03H (0.23 g, 1.97 mmol). The mixture was allowed to warm to about 25 °C and stirred at that temperature for about 2 h. The solution was then concentrated in vacuo after which benzene (2x15 mL) was added and evaporated to remove trace amounts of Et3N and water. To the residue was added benzene (20 mL) and PCIS (0.41 g, 1.97 mmol). The suspension was heated at reflux for about 30 minutes, then cooled to about 25 °C and poured into an ice-cold NaOH solution (5%, 40 mL). The aqueous mixture was extracted with CH2GI2 (3x30 mL), dried over sodium sulfate and concentrated. The residue was purified by flash column chromatography (3% EtOAc in hexanes) affording 0.355 g (74%) of the compound 1b4. Amine: 1H NMR (CDCI3): δ 7.4-7.3 (5H, m), 5.14 (2H, AB), 3.75 (1H, s), 3.38 (1H, m), 2.92 (2H, m), 2.27 (1H, m), 1.40 (3H, s), 1.26 (3H, s); Sulfamoyl Chloride 1b4: 1H NMR (CDCI3): δ 7.43-7.29 (5H, m), 5.22 (2H, AB), 4.53 (1H, s), 4.24-4.05 (2H, m), 3.17 (1H, dd, J=11.7, 4.5 Hz), 3.13 (1H, dd, J=11.7, 4.8 H∑), 2.56 (1H, dt, J=14.1, 2.7 Hz), 1.58 (3H, s), 1.29 (3H, s).
Alcohol 2b4:
Figure imgf000026_0002
Prepared as described in the synthesis of 2a using the sulfamoyl chloride 1b4 (0.2 g, 0.55 mmol) and D-phenylalaninol (0.166 g, 1.1 mmol). After purification by flash column chromatography
(25% to 30% EtOAc in hexanes), the compound 2b4 (10 mg) was obtained in 4% yield. 1H NMR (CDCI3): δ 7.42-7.12 (10H, m), 5.18 (2H, AB), 4.46 (2H, m), 3.68-3.59 (3H, m), 3.48 (1H,m), 3.39 (1H, m), 2.95 (1H, m), 2.76 (2H, d, J=7.2 Hz), 2.32 (1H, dt, J=13.5, 2.1 Hz), 1.87 (3H, s), 1.34 (3H, s).
Example 3b4: 3,3-Dimethyl-1-(2-phenyl-1-sulfooxymethyl-ethylsulfamoyl)-piperidine-2R- carboxylic acid benzyl ester
Figure imgf000027_0001
Prepared as described in the synthesis of 3a using the alcohol 2b4 (6 mg, 0.0126 mmol), chlorosulfonic acid (6 mg, 4 μl, 0.04 mmol) and triethylamine (0.04 mL). The reaction mixture was diluted with EtOAc (20 mL) and washed with ice-cold 5% HCI solution (1x20 mL). Column chromatography purification (5% MeOH in EtOAc) afforded 3 mg (43% yield) of the title compound 3fo4. 'H NMR (CDCI3): δ 7.34-7.06 (10H, m), 5.04 (2H, s), 4.23 (1H, s), 3.89 (1H, dd, J=9.9, 4.2 Hz), 3.82 (1H, dd, J=9.9, 5.1 Hz), 3.59 (1H, td, J=12.9, 3 Hz), 3.52-3.33 (2H, m), 2.88 (1H, dd, J=14.1 , 6.8 Hz), 2.64 (1H, dd, J=14.1, 7.5 Hz), 2.53 (1H, td, J=12.8, 4.2 Hz), 2.13 (1H, dt, J=14.1, 2.4 Hz), 1.37 (3H, s), 1.09 (3H, s); MS (ESP) 557 (M-H").
Phosphate Benzyl Ester 4b1:
Figure imgf000027_0002
To an acetonitrile solution (7 mL) of the alcohol 2b1 (25 mg, 0.0527 mmol) and 1 H-tetrazole (7.4 mg, 0.105 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (27.3 mg, 0.079 mmol) at 25 °C. After 1 h, 3-chloroperoxybeπzoic acid (MCPBA, 34 mg, 70% pure, 0.139 mmol) was added to the suspension. The solution was diluted with ether (40 mL), washed with concentrated NaHS03 solution (2x30 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by preparative TLC to give 29.5 mg of the compound 4b1 in 76% yield. H NMR (CDCI3): δ 7.30-7.05 (20H, m), 5.09 (1H, d, J=9.3 Hz), 4.99 (4H, ), 4.52 (1H, m), 4.14-3.83 (4H, m), 3.68 (1H, m), 3.20 (1H, d, J=12.9 Hz), 2.75 (2H, d, J=7.2 Hz), 2.72 (1H, m), 2.54 (2H, m), 2.10 (1H, d, J=13.5 Hz); HRMS (FAB) caic for C39H47N20BPSCS (M+CS+) 867.1845; found 867.1868. Example 5b1 : 1-(2-Phenyl-1-phosphonooxymethyl-ethylsulfamoyl)-piperidine-2S-carboxylic acid 4-phenyl-butyl ester
Figure imgf000028_0001
To a methanol solution of the phosphate benzyl ester 4b1 (29.5 mg, 0.0402 mmol) was added palladium on carbon (10%, 5mg). The suspension was kept under hydrogen (1 atm) for 1.5 h. After filtration, the filtrate was concentrated to dryness, affording 22.6 mg of the title compound 5b1 in 100% yield. 1H NMR (CDCI3): δ 7.34-6.95 (10H, m), 4.47 (1H, s), 4.17-3.82 (4H, m), 3.62 (1H, br s), 3.20 (1H, br d), 3.28 (3H, m), 2.53 (2H, m), 1.21 (1H, br d); HRMS (FAB) caic for C25H3SN208PSNa (M+Na+) 577.1749; found 577.1769.
Phosphate Benεyl Ester 4b2:
Figure imgf000028_0002
Prepared as described in synthesis of 4b1 using the alcohol 2b2 (240 mg, 0.554 mmol), 1H- tetrazole (77 mg, 1.11 mmol), dibenzyl N,N-diisopropylphosphoramidite (249 mg, 0.72 mmol). Hydrogen peroxide (30%, 2 mL) instead of MCPBA was used for the oxidation. Column chromatography purification (30% EtOAc in hexanes) provided 300 mg of the compound 4b2 in 83% yield. 1H NMR (CDCI3): δ 7.41-7.10 (20H, ), 5.11-5.00 (6H, m), 4.66 (1H, J=4.5 Hz), 4.07 (1H, m), 3.89 (1H, m), 3.72 (1H, m), 3.26 (1H, br d, J=13.2 Hz), 2.85-2.71 (3H, ), 2.22 (1H, d, J=12.9 Hz). Example 5b2: 1-(2-PhenyI-1-phosphonooxymethyl-ethylsulfamoyl)-piperidine-2-carboxylic acid
ph
Figure imgf000029_0001
Prepared as described in the synthesis of 5b1 from phosphate benzyl ester 4b2 (300 mg, 0.457 mmol). The benzyl ester of the carboxylate was also cleaved to carboxylic acid during the hydrogenation. The title compound 5b2 was obtained in 68% yield (119 mg). 1H NMR (CD3OD): δ 7.37-7.13 (5H, m), 4.46 (1H, d, J=2.1 Hz), 4.05 (2H, m), 3.64 (1H, m), 3.28 (1H, m), 3.05 (1H, m), 3.00 (1H, dd, J=13.8, 6.8 Hz), 2.82 (1H, dd, J=13.8, 7.5 Hz), 2.15 (1H, d, J=12.9 Hz); LCMS: 423 (M+H+); HRMS (FAB) caic for C15H24N20BPS (M+H+) 423.0991; found 423.0995.
Alcohol 2fo5:
Figure imgf000029_0002
Prepared as described in the synthesis of 2a using the sulfamoyl chloride 1b5 (0.07 g, 0.205 mmol, preparation described in International Publication No. WO 0140 85) and D-phenylalaninol (0.1 g, 0.662 mmol). 3,5-Lutidine was employed as the reaction solvent in place of pyridine. After purification by flash column chromatography (50% EtOAc in hexanes), the compound 2b5 (28 mg) was obtained in 30% yield. 1H NMR (CDCI3): δ 7.43-7.13 (10H, m), 5.53 (1H, d, J=8.4 Hz), 5.27 (1H, br d, J=3.3 Hz), 4.19 (2H, AB), 3.71 (2H, m), 3.48 (1H, m), 3.35 (1H, dt, J=12.9, 3.3 Hz), 2.91-2.74 (3H, m), 2.53 (1H, t, J=6.3 Hz), 2.19 (1H, dq, J=13.5, 3 Hz), 1.94 (1H, tdd, J=13.6, 5.3, 3.8 Hz). Benzyl Phosphate Ester 4b5:
Figure imgf000030_0001
Prepared as described in synthesis of 4b1 using the alcohol 2b5 (28 mg, 0.061 mmol), 1H- tetrazole (8 mg, 0.12 mmol), dibenzyl N,N-diisopropylphosphoramidite (25 mg, 0.072 mmol) and MCPBA (33 mg, 70% pure, 0.13 mmol). Column chromatography purification (30 to 60% EtOAc in hexanes) followed by preparative TLC purification (50% EtOAc in hexanes) provided 30 mg of the compound 4b5 in 69% yield. 1H NMR (CDCI3): δ 7.44-7.07 (20H, m), 5.57 (1H, d, J=8.4 Hz), 5.19 (1H, br d, J=3 Hz), 5.13-4.97 (4H, m), 4.13 (2H, AB), 4.04 (1H, m), 3.91 (1H, m), 3.75 (1H, m), 3.26 (1H, dt, J=12.6, 3 Hz), 2.89-2.59 (3H, m), 2.15 (1H, dq, J=13.8, 3 Hz), 1.89 (1H, m), 1.74 (1H, br s), 1.64 (1H, dt, J=13.2, 3.3 Hz).
Example 555: Phosphoric acid mono-{(R)-2-[(S)-2-(5-ben2yl-[1,3l43oκadia2θl-2-yl)-p!peridine- 1 -suIfonylamino3-3-phenyl-propyl} ester
Figure imgf000030_0002
Prepared as described in the synthesis of 5b1 using phosphate benzyl ester 4b5 (30 mg,
0.042 mmol) and 10% palladium on carbon (5 mg). The title compound 5b5 was obtained in quantitative yield (28 mg). 1H NMR (CD3OD): δ 7.32-7.01 (10H, m), 4.95 (1H, m), 4.12 (2H, AB), 3.92-3.76 (2H, m), 3.51 (1H, m), 3.24 (1H, br d), 2.93-2.76 (2H, m), 2.67 (1H, dd, J=13.8, 7.8 Hz), 1.92 (1H, br d), 1.79 (1H, m); MS (ESP): 559 (M+Na*); 535 (M-H) . Scheme 2
Figure imgf000031_0001
Figure imgf000031_0002
Figure imgf000031_0003
16c R2 = -P(0)(OH)2;R6 = J-Hj h
16d R2= -P(0)(OH)2; R6 = H
Figure imgf000032_0001
Figure imgf000032_0002
Figure imgf000032_0003
16c Rz ■ -P(0)(OH)2;R6 = j fH ϋ. Ph
16d R2= -P(0)(OH)2; R6 = H Alcohol 9:
Figure imgf000033_0001
To a DMF solution (5 mL) of 3-amino-1-propanol (0.207 g, 0.21 mL, 2.75 mmol) was added triethylamine (0.42 mL, 3 mmol) and 4-chloro-7-nitrobenzofurazan (0.5 g, 2.5 mmol) at 25 °C. After 20 h, the reaction mixture was poured into water (100 mL). The precipitate was collected by filtration. Recrystali∑ation from warm methanol afforded 150 mg (25% yield) of the compound 9 as a yellow solid. H NMR (CD3OD): δ 9.49 (1H, s), 8.51 (1H, d, J=8.7 Hz), 6.41 (1H, d, J=8.7 Hz), 4.65 (1H, m), 3.62-3.44 (4H, m), 1.84 (2H, p, J=6.6 Hz); MS (positive ESP): 239 (M+H+); MS (negative ESP): 237 (M-H)\
Ester 11:
Figure imgf000033_0002
To a DMF solution (2 mL) of the alcohol 8 (0.08 g, 0.336 mmol) and N-Boc-pipecolinic acid 10 (0.115 g, 0.504 mmol, preparation described in International Publication No. WO 0140185) was added triethylamine (0.2 mL), EDC (1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride) (0.097 g, 0.504 mmol) and HOBT (1-hydroxybenzotriazole hydrate) (0.068 g, 0.0504 mmol) at 25 °C. After 20 h, the reaction mixture was diluted with EtOAc (50 mL), washed with brine (3x50 mL), dried (Na2S0 ) and concentrated. The residue was purified by column chromatography (20-25% EtOAc in hexanes) affording 150 mg (100%yield) of the compound 11. 1H NMR (CDCI3): (mixture of two rotamers) δ 8.48 (1H, d, J=8.7 Hz), 7.11-6.94 (1H, br s), 6.26 (1H, d, J=8.7 Hz), 4.85 (1H, br s), 4.36 (2H, m), 4.06-3.86 (2H, m), 3.67 (2H, m), 3.12-2.81 (1H, m), 2.26-2.12 (3H, m), 1.46 (9H, s).
Amine 6b
Figure imgf000033_0003
To a methylene chloride solution (5 mL) of the ester 1 (150 mg, 0.5 mmol) at -30 °C was added trifluoroacetic acid (1 mL). The solution was warmed to 25 DC over 3 h. All solvent was removed in vacuo . o give 125 mg (100%) of the compound 6b. 1H NMR (CDCI3): δ 8.45 (1H, d, J=8.4 Hz), 6.18 (1H, d, J=8.4 Hz), 4.35 (2H, t, J=6.3 Hz), 3.66 (2H, t, J=6 Hz), 3.43 (1H, dd, J=9.9, 3.3 Hz), 3.13 (1H, br d), 2.70 (1H, br t), 2.19 (2H, p, J=6.3 Hz).
Urea 12b
Figure imgf000034_0001
To a DMF solution (5 mL) of D-phenylalaninol (1 g, 6.6 mmol) was added imidazole (0.494 g, 7.27 mmol) and t-butyldimethylchlorosilane (1 g, 7.27 mmol). After 40 h, the mixture was diluted with ether (20 mL), washed with brine (3x50 mL) and dried
Figure imgf000034_0002
All solvent was removed in vacuo to give the amine 8b (1.85 g) as a colorless oil. At -40 °C, a portion of the amine 8b (0.265 g, 1 mmol) was added to a methylene chloride solution (5 mL) of phosgene (0.544 mL, 20% in toluene, 1.1 mmol) and triethylamine (0.5 mL). The solution was slowly warmed up to 25 °C over 30 min, and was cooled to 0 °C again. The pipecolate ester 6b (0.05 g, 0.143 mmol) was introduced at once. The mixture was stirred at 25 °C for 20 h, diluted with EtOAc (50 mL), washed with concentrated NaHCOs solution (1x50 mL), dried (Na2S04) and concentrated. The residue was purified by column chromatography (50% EtOAc in hexanes) to afford 20 mg (22% yield) of the compound 12b. 1H NMR (CDCI3): δ 8.42 (1H, d, J=8.7 Hz), 7.30-7.07 (5H, m), 6.17 (1H, d, J=8.7 Hz), 5.11 (1H, d, J=8.7 Hz), 4.83 (1H, dd, J=6, 3.3 Hz), 4.37 (1H, m), 4.15 (1H, m), 4.02 (1H, m), 3.66-3.28 (4H, m), 3.19 (1H, td, J=11.7, 3.6 Hz), 2.81 (3H, m), 2.07 (3H, m), 0.88 (9H, s), 0.01 (3H, s), 0.0 (3H, s). Alcohol 13b:
Figure imgf000034_0003
To a THF solution (4 mL) of the silyl ether (20 g, 0.0313 mmol) was added tetrabutylammonium fluoride (1 mL, 1M in THF, 1 mmol). After 1 h at 25 °C, the solution was concentrated. The resulting residue was purified by column chromatography (75% EtOAc in hexanes) to give 18 mg (100%) of the compound 13b. 1H NMR (CD3OD): δ 8.49 (1 H, d, J=9 Hz), 7.3-7.1 (5H, m), 6.35 (1H, d, J=9 Hz), 4.38 (1H, m), 4.05 (1H, m), 3.92 (1H, dd, J=13.2, 4.8 Hz), 3.84- 3.59 (6H, m), 3.13 (1H, m), 3.00 (1H, dd, J=13.8, 5.4 Hz), 2.78 (1 H, m), 1.99 (3H, m).
Example 14b: 1-(2-Phenyl-1-sulfooxymethyl-ethylcarbamoyi)-piperidine-2-carboxylic acid 3- (7-nitro-benzo[1 ,2,5]oxadiazol-4-ylamino)-propyl ester
Figure imgf000035_0001
At -70 °C, a methylene chloride solution (2 L) of the alcohol 13b (10 mg, 0.019 mmol) was added triethylamine (0.05 mL) and chlorosulfonic acid (8 mg, 5 μl, 0.068 mmol). The cooling bath was then removed and the reaction mixture was allowed to warm to 25 °C over 3 h. All solvent was evaporated in vacuo. The residue was purified by column chromatography (10% MeOH in EtOAc) to give 5 mg (42% yield) of the title compound 14b. 1H NMR (CD3OD): δ 8.63 (1 H, d, J=9
Hz), 7.37-7.18 (5H, m), 6.49 (1H, d, J=9 Hz), 4.71-4.53 (2H, m), 4.33-4.15 (3H, m), 3.98 (1H, dd, J=12.6, 4.5 Hz), 3.84-3.71 (4H, m), 3.36-3.11 (2H, m), 2.85 (1 H, td, J=12.6, 4.5 Hz). Alcohol 13a: Method A:
Figure imgf000035_0002
To a methylene chloride solution (4 mL) of the pipecolate ester 6a (0.2 g, 0.528 mmol) and triethylamine (1 mL) was added a methylene chloride solution (1 mL) of triphosgene (0.052 g, 0.176 mmol). After 10 min, the solution was heated at reflux for 1 h, and was then cooled to 25 °C. A methylene chloride solution (1 mL) of D-phenylalaninol (0.0798 g, 0.528 mmol) was added. After 2 h, the reaction solution was diluted with Et20 (50 mL), washed with brine (2x70 mL), dried (Na2S0 ) and concentrated in vacuo. The residue was purified by flash column chromatography (20% EtOAc in hexanes) to afford 80 mg (27% yield) of the compound 13a. H NMR (CDCI3): δ 7.38-7.10 (15H, m), 4.96 (2H, m), 4.70 (1H, d, J=6.9 Hz), 4.01 (1H, ), 3.67 (1H, br d), 3.51 (1 H, dd, J=10.8, 5.1 Hz), 3.25 (2H, br t), 3.04 (1H, td, J=12.3, 3.3 Hz), 2.84 (2H, m), 2.58 (4H, m), 2.18 (1H, br d, J=12.6 Hz); MS (ESP positive): 557 (M+H+); 555 (M-H) \
Method B:
Silyl ether 17:
Figure imgf000036_0001
To a DMF solution (7 mL) of (R)-(+)-2-(t-Boc)-amino-3-phenyl-1-propanol (4.15 g, 16.5 mmol) was added imidazole (2.24 g, 33 mmol) and t-butylchlorodiphenylsilane (5.15 mL, 19.8 mmol). After 15 h at 25 °C, the mixture was diluted with ether (50 mL), washed with sat'd NH4CI solution (3x50 mL), dried (Na2S04) and concentrated in vacuo. The residue was purified by column chromatography (2-4% EtOAc in hexanes) to afford 9.19 g (100%) of the compound 17 as a white solid. MS (ESP): 512 (M+Na*). Amine 7a
Figure imgf000036_0002
To a methylene chloride solution (60 L) of the silyl ether 17 (9.19 g, 16.5 mmol) at 0 °C was added trifluoroacetic acid (20 mL). The reaction solution was stirred at 0 °C for 1 h and was then warmed to 25 °C over 30 min. The mixture was concentrated in vacuo and was redissolved in methylene chloride (50 mL). The resulting solution was washed with sat'd NaHC03 solution (2x50 mL), dried (Na2S04) and concentrated. The residue was purified by column chromatography (97.5/2.5/0.25 CH2CI2/MeOH/NH OH) to give 5.97 g (78% yield) of the compound 7a as a light yellow oil. 1H NMR (CDCI3): δ 7.56 (4H, br d), 7.36-7.23 (6H, m), 7.20-7.13 (2H, m), 7.12-7.02 (3H, m), 3.52 (1H, dd, J=9.9, 4.5 Hz), 3.43 (1H, dd, J=10.2, 6.3 Hz), 3.10-2.99 (1H, m), 2.71 (1H, dd, J=13.2, 4.8 Hz), 2.41 (1H, dd, J=13.5, 8.4 Hz), 0.97 (9H, s); MS (ESP): 390 (M+H+). Urea 12a:
Figure imgf000037_0001
To a methylene chloride solution (20 mL) of the amine 7a (3.44 g, 7.04 mmol) and triethylamine (2 mL) was added a methylene chloride solution (1 mL) of triphosgene (0.613 g, 2.07 mmol). After 2 h, the solution was heated at reflux for 1.5 h, and was then cooled to 25 °C. A methylene chloride solution (40 mL) of the amine 6a (2.67 g, 7.04 mmol) (preparation of 6a described in Guo et al. in International Publication No. WO 01/40183) was added. After 15 h, the reaction solution was diluted with CH2CI2 (100 mL), washed with brine (2x80 mL), dried (Na2S0 ) and concentrated in vacuo. The residue was purified by flash column chromatography (5-20% EtOAc in hexanes) to afford 4.85 g (77% yield) of the compound 12a. 1H NMR (CDCI3): (mixture of two rotamers) β 7.6-7.5 (4H, m), 7.39-7.23 (7H, m), 7.22-6.99 (14H, m), 5.00-4.80 (2H, m), 4.16 and 4.01 (1H, m), 3.51 (3H, m), 3.30 (1 H, m), 3.07-2.74 (4H, m), 2.55-2.43 (2H, m), 1.04 and 1.03 (9H, s); MS (ESP): 817 (M+Na+).
Alcohol 13a:
Figure imgf000037_0002
To a THF solution (30 mL) of the silyl ether 12a (4.83 g, 6.08 mmol) at 0 °C was added hydrogen fluoride-pyridine (8 mL). After 30 min at 0 °C, the mixture was warmed to 25 °C over 1 h. All solvent was removed in vacuo. The residue was dissolved in methylene chloride (50 mL) and the solution was washed with ice-cold HCI solution (2x40 mL), dried (Na2S04) and concentrated. Flash column chromatography purification provided 1.96 g (58% yield) of the compound 13a as a white solid. Example 14a: 1-(2-Phenyl-1-sulfooxymethyl-ethylcarbamoyl)-piperidine-2S-carboxylic acid 4- phenyl-1-(3-phenyl-propyI)-butyl ester
Figure imgf000038_0001
Prepared as described in the synthesis of 14b using the alcohol 13a (17 mg, 0.0306 mmol), chlorosulfonic acid (11 mg, 8 μl, 0.011 mmol) and triethylamine (0.05 mL). The reaction mixture was diluted with EtOAc (20 mL) and washed with ice-cold 5% HCI solution (1x20 mL). Column chromatography (8% MeOH in EtOAc) afforded 12 mg (62% yield) of the title compound 14a. 1H NMR (CD3OD): δ 7.36-7.06 (15H, m), 6.55 (1H, d, J=7.8 Hz), 4.99 (1H, m), 4.11 (1H, m), 3.97 (2H, m), 3.74 (1H, m), 2.99 (1H, br t), 2.87 (2H, m), 2.60 (4H, m), 2.20 (1H, br d); MS (ESP negative): 635 (M-H)"; HRMS (FAB) caic for C35H43N207S (M-H) 635.2791; found 635.2815.
Phosphate Benzyl Ester 15a:
Figure imgf000038_0002
To an acetonitrile solution (10 mL) of the alcohol 13a (73 mg, 0.131 mmol) and 1H-tetrazole (15 mg, 0.21 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (68 mg, 0.198 mmol) at 25 °C. After 1 h, MCPBA (102 mg, 70% pure, 0.4 mmol) was added to the suspension. The solution was diluted with ether (50 L), washed with concentrated NaHS03 solution (2x30 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (20-25% EtOAc in hexanes) to give 70 mg of the compound 15a in 65% yield. 1H NMR (CDCI3): δ 7.40-7.07 (25H, m), 5.67 (1H, d, J=7.8 Hz), 5.10-4.89 (6H, m), 4.14 (1H, m), 3.90 (2H, m), 3.53 (1H, br d), 3.09 (1H, br t), 2.97 (1H, dd, J=13.2, ,8 Hz), 2.69 (1H, dd, J=13.2, 9 Hz), 2.56 (4H, m), 2.18 (1H, br d). Example 16a: 1-(2-Phenyl-1-phosphonooxymethyI-ethylcarbamoyl)-piperidine-2S-carboxylic acid 4-phenyl-1-(3-phenyl-propyl)-butyl ester
Figure imgf000039_0001
To a methanol solution of the phosphate benzyl ester 15a (50 mg, 0.061 mmol) was added palladium on carbon (10%, 6 mg). The suspension was kept under hydrogen (1 atm) for 5 h. After filtration, the filtrate was concentrated to dryness, affording 40 mg of the title compound 16a in 100% yield. 1H NMR (CDCfe): δ 7.36-7.07 (15H, m), 5.02 (1H, m), 4.86 (1H, br d), 4.12 (1H, m), 3.92 (2H, m), 3.70 (1H, br d), 3.09-2.88 (2H, m), 2.83 (1H, dd, J=13.8, 7.8 Hz), 2.60 (4H, m), 2.18 (1H, br d); HRMS (MALDI) caic for C35H44N207PNa2 (M-H++2Na+) 681.2681; found 681.2691.
Alcohol 13c:
Figure imgf000039_0002
To a methylene chloride solution (10 mL) of the pipecolate ester 6c (0.5 g, 1.92 mmol, preparation described in International Publication No. WO 0140185) and triethylamine (2 mL) at -40 °C was added a toluene solution (1.04 mL) of phosgene (20%, 0.208 g, 0.176 mmol). The solution was warmed to 25 °C over 1 h and was then added D-phenylalaninol (0.29 g, 1.92 mmol). After 10 h, the reaction mixture was diluted with Et20 (100 mL), washed with 5% ice-cold HCI solution (1x50 mL) and brine (1x50 mL), dried (Na2S0 ) and concentrated in vacuo. The residue was purified by flash column chromatography (20% EtOAc in hexanes) to afford 100 mg (12% yield) of the compound 13c. 1H NMR (CDCI3): δ 7.31-7.03 (10H, m), 4.85 (1H, br d), 4.1-3.9 (2H, m), 3.64 (1H, dd, J=10.8, 3.3 Hz), 3.50 (1H, dd, J=11.4, 5.7 Hz), 3.25 (1H, br d), 2.99 (1H, td, J=12.6, 3 Hz), 2.79 (2H, m), 2.56 (2H, m). Phosphate Benzyl Ester 15c:
Figure imgf000040_0001
Prepared as described in the synthesis of 15a using the alcohol 13c (60 mg, 0.137 mmol), 1H-tetrazole (19.2 mg, 0.274 mmol), dibenzyl N,N-diisopropylphosphoramidite (0.069 mL, 71 mg, 0.205 mmol) and MCPBA (115 mg, 60% pure, 0.4 mmol). Preparative TLC purification (30% EtOAc in hexanes) provided 20 mg of the compound 15c in 21% yield. H NMR (CDCI3): δ 7.43-7.1 (20H, m), 5.64 (1H, d, J=7.5 Hz), 5.1-5.0 (4H, m), 4.97 (1H, br d), 4.21-3.82 (5H, m), 3.54 (1H, br d), 3.09 (1H, td, J=12.6, 3.3 Hz), 2.97 (1H, dd, J=13.5, 5.7 Hz), 2.70 (1H, dd, J=13.8, 9.6 Hz), 2.60 (2H, m), 2.19 (1H, d, J=13.5 Hz).
Exa le 16c: 1 -(2-Phenyl-1 -phosphonooxymethyl-ethylcarfoamoyl)-piperidine-2S-carfooκylic acid 4-phenyl-1 -butyl ester
Figure imgf000040_0002
Prepared as described in the synthesis of 16a from phosphate benzyl ester 15c (20 mg, 0.457 mmol). Ethanol instead of methanol was used as a reaction solvent. The title compound 16c was obtained in 88% yield (13 mg). 1H NMR (CD3OD): δ 7.25-6.99 (10H, m), 4.01 (3H, m), 3.83 (2H, m), 3.62 (1H, br d), 2.94-2.66 (3H, ), 2.53 (2H, ), 2.08 (1H, d, J=12.9 Hz); HRMS (MALDl) caic for
C26H35N207PNa (M+Na+) 541.2080; found 541.2106. Alcohol 13d:
Figure imgf000041_0001
Prepared as described in the synthesis of 13c using pipecolate ester 6d (0.78 g, 3.56 mmol, preparation described in International Publication No. WO 0140185), triethylamine (2 mL), phosgene (20% in toluene, 2.4 mL, 4.45 mmol) and D-phenylalaninol (1.35 g, 8.9 mmol). Flash column chromatography purification (50% EtOAc in hexanes) to afford 60 mg (4.4% yield) of the compound 13d. 1H NMR (CDCI3): δ 7.31-7.16 (5H, m), 4.42 (1H, m), 4.06 (1H, dd, J=13.2, 4.8 Hz), 3.97-3.81 (2H, m), 3.58 (1H, dd, J=12, 4.2 Hz), 3.39 (1H, dd, J=9.6, 3.6 Hz), 3.14 (2H, m), 2.75 (1H, td, J=13.8, 3.9 Hz), 2.09 (1H, m), 1.91 (1H, m), 1.67 (1H, m); MS (ESP): 397 (M+H+).
Phosphate Benzyl Ester 15d:
Figure imgf000041_0002
Prepared as described in synthesis of 15a using the alcohol 13d (89 mg, 0.225 mmol), 1H- tetrazole (32 mg, 0.45 mmol), dibenzyl N,N-diisopropylphosphoramidite (0.113 mL, 0.337 mmol) and MCPBA (136 mg, 60% pure, 0.45 mmol). Column chromatography purification (30% EtOAc in hexanes) provided 71 mg of the compound 15d in 48% yield. 1H NMR (CDCI3): δ 7.31-7.06 (20H, m), 5.62 (1H, d, J=7.8 Hz), 5.11-4.92 (2H, m), 4.13 (1H, m), 3.99-3.79 (2H, m), 3.49 (1H, br d), 3.01 (1H, td, J=12.3, 2.7 Hz), 2.89 (1H, dd, J=13.5, 5.4 Hz), 2.63 (1H, dd, J=13.8, 9.3 Hz), 2.15 (1H, br d, J=13.8 Hz); MS (ESP positive): 657 (M+H*), 679 (M+Na+). Example 16d: 1-(2-Phenyl-1-phosphonooxymethyl-ethylcarbamoyl)-piperidine-2-carboxylic acid
Figure imgf000042_0001
Prepared as described in the synthesis of 16a from phosphate benzyl ester 15d (48 mg, 0.073 mmol). Ethanol instead of methanol was used as a reaction solvent. The benzyl ester of the carboxylate was also cleaved to carboxylic acid during the hydrogenation. After HPLC purification, the title compound 16d was obtained in 4% yield (1 mg). 1H NMR (CD3OD): δ 7.46-7.11 (5H, m), 4.71-4.45 (2H, m), 4.19 (1H, m), 3.93 (1H, dd, J=13.2, 4.8 Hz), 3.69 (1H, dd, J=12, 4.2 Hz), 3.26-3.0 (2H, m), 2.79 (1H, td, J=13.2, 3.6 Hz), 2.00 (1H, m).
Scheme 3
Figure imgf000043_0001
MCPBA
Figure imgf000043_0002
Figure imgf000043_0003
The following is a list of the compounds prepared using the synthetic pathways outlined in Scheme 3, and then the detailed experimental procedures. Compounds Made Using Scheme 3
Figure imgf000044_0001
Figure imgf000044_0002
Figure imgf000045_0001
Alcohol 19a
-OBn
HN
19a
Ph. -OH To a methylene chloride solution (80 mL) of D-phenylalaninol 18a (1.15 g, 7.61 mmol) was added triethylamine (1.59 mL, 11.4 mmol) and benzyl chloroformate (1.19 mL, 8.37 mmol). The mixture was stirred for 3 h and then concentrated. The residue was dissolved in methylene chloride (50 mL) and washed with brine (1x50 mL). The solution was dried (Na2S04) and concentrated. After column chromatography purification (10 to 30% EtOAc in hexanes), the compound 19a was obtained in 73% yield (1.59 g). 1H NMR (CDCI3): δ 7.46-7.15 (10H, m), 5.11 (2H, s), 4.96 (1H, m), 3.98 (1H, m), 3.72 (1H, m), 3.63 (1H, m), 2.89 (1H, d, J=7.2 Hz); MS (ESP): 286 (M+H+); 284 (M-H)'.
Alcohol 19b:
Figure imgf000046_0001
Prepared as described in the synthesis of the alcohol 19a using L-phenylalaninol 18b (2.59 g, 17.1 mmol), triethylamine (2.6 g, 3.58 mL, 25.7 mmol) and benzyl chloroformate (2.69 mL, 18.8 mmol). After column chromatography purification (20 to 40% EtOAc in hexanes), the compound 19b was obtained in 55% yield (2.61 g). 1H NMR (CDCI3): δ 7.43-7.13 (10H, m), 5.09 (2H, s), 4.94 (1H, m), 3.95 (1H, m), 3.69 (1H, m), 3.58 (1H, m), 2.87 (1H, d, J=7.2 Hz); MS (ESP): 286 (M+H+); 284 (M- H)'.
Phosphate Benzyl Ester 20a:
Figure imgf000046_0002
To an acetonitrile solution (40 L) of the alcohol 19a (1.58 g, 5.54 mmol) and 1H-tetra∑ole (1.05 g, 15 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (3.72 mL, 11.1 mmol) at 25 DC. After 3 h, MCPBA (4.19 g, 70% pure, 13.85 mmol) was added to the suspension. The solution was diluted with EtOAc (100 mL), washed with concentrated NaHS03 solution (2x80 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (10-30% EtOAc in hexanes) to give 2.88 g of the compound 20a in 95% yield. 1H NMR (CDCI3): δ 7.47-7.05 (20H, m), 5.19-4.96 (7H, m), 4.09-3.83 (3H, m), 2.93-2.67 (2H, m); MS (positive ESP): 568 (M+Na+); MS (negative ESP): 580 (M+CI)". Phosphate Benzyl Ester 20b:
Figure imgf000047_0001
. Prepared as described in the synthesis of compound 20a using the alcohol 19a (2.61 g, 9.16 mmol), 1H-tetrazole (1.73 g, 24.7 mmol), dibenzyl N,N-diisopropylphosphoramidite (6.15 mL, 18.3 mmol) and MCPBA (6.26 g, 77% pure, 27.5 mmol). Purification by column chromatography (15-30% EtOAc in hexanes) gave 4.1 g of the compound 20b in 82% yield. 1H NMR (CDCI3): δ 7.42-7.1 (20H, m), 5.16-5.0 (7H, m), 4.09-3.84 (3H, m), 2.9-2.69 (2H, m); MS (ESP): 546 (M-H-f); 580 (M+CIV.
Aminophosphate 21a:
Figure imgf000047_0002
To an ethanol solution of the phosphate benzyl ester 20a (2.88 g, 5.28 mmol) was added palladium on carbon (10%, 300 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 4 h, and was then filtered through a pad of celite. The collected solid was washed with methylene chloride. The mixture of the solid and celite was suspended in 5% HCI solution and stirred for 20 min. After filtration, the filtrate was concentrated to dryness, affording 1.2 g of the compound 21a in 86% yield. 1H NMR (CD3OD): δ 7.49-7.25 (5H, m), 4.22-4.08 (1H, m), 4.0 (1H, m), 3.72 (1H, m), 3.03 (2H, d, J=7.5 Hz); LCMS: 232 (M+H+); 230 (M-H)"; HRMS (MALDl) caic for CBH15N04P (M+H+) 232.0733; found 232.0736.
Aminophosphate 21b:
Figure imgf000047_0003
Prepared as described in the synthesis 'of 21a using phosphate benzyl ester 20b (4.1 g, 7.5 mmol) and palladium on carbon (10%, 410 mg). After filtration and evaporation, the compound 21b was obtained in quantitative yield (2.29 g). 1H NMR (CD3OD): δ 7.48-7.24 (5H, m), 4.14 (1H, m), 3.98 (1H, m), 3.69 (1H, m), 3.00 (2H, d, J=7.4 Hz); LCMS: 232 (M+H+). Example 23a: Phosphoric acid mono-[3-phenyl-2-(3-phenyl-ureido)-propyl] ester
Figure imgf000048_0001
To a sodium carbonate solution (1 M, 1 L) was added the aminophosphate 21a (53 mg, 0.198 mmol) and phenylisocyanate (0.023 mL, 0.208 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 27 mg (42% yield) of the title compound 23a. 1H NMR (CD3OD): δ 7.36-7.17 (9H, m), 6.97 (1H, t, J=7.8 Hz), 4.19 (1H, m), 3.98 (2H, m), 2.98 (1H, dd, J=13.9, 7.1 Hz), 2.88 (1H, dd, J=13.8, 7.7 Hz); HRMS (MALDl) caic for Cι6H19N205PNa (M+Na4) 373.0924; found 373.0934.
E∑ ample 23b: Phosphoric acid mono-|2- 3-(2-phenoJcy-phenyl)-ureido3-3-phenyl-propyl} ester
Figure imgf000048_0002
Prepared as described in the synthesis of 23a using 21a (82 mg, 0.307 mmol), 1-isocyanato-
2-phenoxybenzene (67.8 mg, 0.058 mL, 0.322 mmol) and 1 M sodium carbonate solution (1 mL).
Preparative HPLC purification gave 36 mg (27% yield) of the title compound 23b. 1H NMR (CD3OD): δ 8.05 (1H, d, J=8.4 Hz), 7.36 (2H, t, J=8.1 Hz), 7.30-7.02 (7H, m), 7.02-6.91 (3H, m), 6.83 (1H, d, • J=8.1 Hz), 4.18 (1H, m), 3.95 (2H, m), 2.95 (1H, dd, J=13.8, 6.9 Hz), 2.81 (1H, dd, J=13.8, 7.8 Hz);
MS (ESP): 443 (M+H+), 465 (M+Na+); 441 (M-H)'. Example 23c: Phosphoric acid mono-{2-[3-(3-methoxy-5-methyl-phenyl)-ureido]-3-phenyI- propyl} ester
Figure imgf000049_0001
Prepared as described in the synthesis of 23a using 21a (62 mg, 0.232 mmol), 2-methoxy-5- methylphenylisocyanate (40 mg, 0.243 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 50 mg (55% yield) of the title compound 23c. H NMR (CD3OD): δ 7.76 (1H, d, J=1.8 Hz), 7.33-7.27 (4H, m), 7.22 (1H, m), 6.81 (1H, d, J=8.1 Hz), 6.74 (1H, br d), 4.19 (1H, m), 3.97 (2H, m), 2.98 (1H, dd, J=13.9, 7.0 Hz), 2.83 (1H, dd, J=14, 8.1 Hz); HRMS (MALDl) caic for C18H24N206P (M+H+) 395.1372; found 395.1383.
Example 23d: Phosphoric acid mono-{2-p-(3,5-dimethoj y-phenyl)-ureido3-3-phenyl-propyl} ester
Figure imgf000049_0002
Prepared as described in the synthesis of 23a using 21a (71 mg, 0.265 mmol), 2,4- dimethoxy-phenylisocyanate (50 mg, 0.279 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 29 mg (27% yield) of the title compound 23d. ?H NMR (CD3OD): δ 7.76 (1H, d, J=9 Hz), 7.46-7.29 (5H, m), 6.66 (1 H, d, J=2.6 Hz), 6.56 (1H, dd, J=9, 2.8 Hz), 4.30 (1H, m), 4.08 (2H, m), 3.95 (3H, s), 3.88 (3H, s), 3.09 (1H, dd, J=13.8, 6.6 Hz), 2.96 (1H, dd, J=13.8, 7.7 Hz); MS (ESP) 411 (M+H+), 433 (M+Na+); 409 (M-H)'. Example 23e: Phosphoric acid mono-(2-benzenesulfonylamino-3-phenyl-propyl) ester
Figure imgf000050_0001
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21a (66 mg,
0.246 mmol) and phenylsulfonyl chloride (0.047 mL, 0.369 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 35 mg (38% yield) of the title compound 23e. H NMR (CD3OD): δ 7.67(2H, d, J=7.5 Hz), 7.54 (1H, t, J=7.2 Hz), 7.42 (1H, t, J=7.8 Hz), 7.21-7.11 (3H, m), 7.09-7.03 (2H, m), 3.95 (1H, m), 3.84 (1H, m), 3.61 (1H, m), 2.94 (1H, dd, J=13.8, 6.6 Hz), 2.59 (1H, dd, J=13.5, 7.8 Hz); LCMS: 372 (M+H+), 394 (M+Na+); 370 (M-H)'; HRMS (MALDl) caic for
Figure imgf000050_0002
(M+Na+) 394.0485; found 394.0487.
Example 25-1: Phosphoric acid mono-{3-phen I-2- (1-phenyl-methanoyl)-®mino3-propylJ ester
Figure imgf000050_0003
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21a (70 mg, 0.262 mmol) and benzyol chloride (0.028 mL, 0.238 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 20 mg (23% yield) of the title compound 25-1. 1H NMR (CD3OD): δ 8.04 (1H, br d), 7.76 (2H, br d), 7.76 (2H, br d), 7.64-7.16 (9H, m), 4.52 (1H, m), 4.09 (2H, m), 3.08 (1H, dd, J=13.6, 6.8 Hz), 2.96 (1H, dd, J=13.5, 8.1 Hz); MS (ESP): 336 (M+H+); 334 (M-H)'. Example 25-2: Phosphoric acid mono-{(R)-2-[(1-beπzo[b]thiophen-2-yI-methanoyl)-amino]-3- phenyl-propyl} ester
Figure imgf000051_0001
Prepared as described in the synthesis of 25-1 using 21a (48 mg, 0.179 mmol), benzothiophene-2-carbonyl chloride (35 mg, 0.179 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 34 mg (48% yield) of the title compound 25-2. 1H NMR (CD3OD): δ 7.96 (1H, s), 7.90 (2H, m), 7.43 (2H, m), 7.37-7.17 (5H, m), 4.50 (1H, m), 4.10 (2H, m), 3.09 (1H, dd, J=13.9, 6.6 Hz), 3.00 (1H, dd, J=13.9, 7.8 Hz); HRMS (MALDl) caic for C18HiBN05PSNa (M+Na+) 414.0540; found 414.0536.
Eκampl© 25-2': Phosphoric acid mono-(2-{[1-(1-oxo-foenzo[bJthiophen-2-yl)- aminoH-p eπyi ■propyl) ester
Figure imgf000051_0002
To a trifluoroacetic acid solution (1 mL) of 25-2, 9 mg, 0.023 mmol) at 0 °C was added 30% hydrogen peroxide (0.0244 mL). The solution was concentrated in vacuo. The residue was purified by preparative HPLC to give 1 mg (10% yield) of the title compound 25-2'. 1H NMR (CD3OD): δ 8.01-
7.93 (2H, m), 7.78-7.6 (3H, m), 7.36-7.26 (4H, m), 7.25-7.17 (1 H, m), 4.44 (1H, m), 4.04 (2H, m),
3.02 (2H, m); MS (ESP): 408 (M+H+), 430 (M+Na+). Example 25-3: Phosphoric acid mono-{(R)-2-[(1-naphthalen-2-yl-methanoyl)-amino]-3-phenyl- propyl} ester
Figure imgf000052_0001
Prepared as described in the synthesis of 25-1 using 21a (50 mg, 0.187 mmol), 2-naphthoyl chloride (36 mg, 0.187 mmol) and 1 M sodium carbonate solution (1 L). Preparative HPLC purification gave 29 mg (40% yield) of the title compound 25-3. 1H NMR (CD3OD): δ 8.32 (1H, s), 8.1-7.88 (3H, m), 7.83 (1H, dd, J=8.4, 1.8 Hz), 7.64-7.53 (2H, m), 7.39-7.18 (5H, m), 4.57 (1H, m), 4.12 (2H, m), 3.12 (1H, dd, J=13.7, 6.8 Hz), 3.02 (1H, dd, J=13.7, 8.1 Hz); LCMS (ESP): 386 (M+H+), 408 (M+Na+); 384 (M-H)"; HRMS (MALDl) caic for C20H20NO5PNa (M+Na*) 408.0971; found 408.0986.
Example 25-4: Phosphoric acid mono-[2-({1-[5-(3,5-dichloro-phenoκy)-furan-2-yl]-methanoyl}- amino)-3-ph©nyl-propyl] ©εter
Figure imgf000052_0002
Prepared as described in the synthesis of 25-1 using 21a (76 mg, 0.284 mmol), 5-(3,5- dichlorophenoxy)-2-furoyl chloride (83 mg, 0.284 mmol) and 1 M sodium carbonate solution (1 L). Preparative HPLC purification gave 46 mg (33% yield) of the title compound 25-4. H NMR (CD3OD): δ 7.21 (1H, t, J=L7 Hz), 7.17-7.03 (5H, m), 7.00 (2H, d, J=1.8 Hz), 6.99 (1H, d, J=3.6 Hz), 5.79 (1H, d, J=3.9 Hz), 4.35 (1H, m), 3.92 (2H, m), 2.91 (1H, dd, J=13.6, 6 Hz), 2.78 (1H, dd, J=13.8, 8.7 Hz); MS (ESP): 508 (M+Na+); 484 (M-H)". Example 25-5: Phosphoric acid mono-(2-[[1-(3,4-dichloro-phenyI)-methanoyI]-amino}-3- pheπyl-propyl) ester
Figure imgf000053_0001
Prepared as described in the synthesis of 25-1 using 21a (56 mg, 0.209 mmol), 3,4- dichlorobenzoyl chloride (44 mg, 0.209 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 20 mg (24% yield) of the title compound 25-5. 1H NMR (CD3OD): δ 7.91 (1H, d, J=2.1 Hz), 7.68 (1H, dd, J=8.4, 1.8 Hz), 7.60 (1H, d, J=8.4 Hz), 7.35-7.16 (5H, m), 4.50 1H, m), 4.08 (2H, m), 3.07 (1H, dd, J=13.9, 6.8 Hz), 2.94 (1H, dd, J=13.9, 8.4 Hz); LCMS (ESP): 404 (M+H+), 426 (M+Na+); 402 (M-H)'.
Example 25-6: Phosphoric acid mono-(2-{|1-(5-chloro-4-metho2 y-thiophen-3-yl)-methanoylJ- amino}-3-phenyl-propyl) ester
Figure imgf000053_0002
Prepared as described in the synthesis of 25-1 using 21a (63 mg, 0.236 mmol), 2-chloro-3- methoxythiophene-4-carbonyl chloride (50 mg, 0.236 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 37 mg (38% yield) of the title compound 25-6. *H NMR
(CD3OD): δ 7.86 (1H, s), 7.38-7.19 (5H, m), 4.51 (1H, m), 4.09 (2H, m), 3.93 (2H, m), 3.04 (2H, m);
LCMS (ESP): 406 (M+H+), 428 (M+Na+); 404 (M-H)". Example 25-7: Phosphoric acid mono-(2-{[1-(5-methyl-2-phenyl-2H-[1,2,3ltriazol-4-yl)- methanoyl]-amino}-3-phenyl-propyl) ester
Figure imgf000054_0001
Prepared as described in the synthesis of 25-1 using 21a (46 mg, 0.172 mmol), 4-methyI-2- phenyl-1,2,3-triazoIe-5-carbonyl chloride (38 mg, 0.172 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 36 mg (50% yield) of the title compound 25-7. 1H NMR (CD3OD): δ 7.98 (1H, d, J=8.1 Hz), 7.43 (2H, br t), 7.31 (1H, br t), 7.25-7.04 (5H, m), 4.40 (1H, m), 3.98 (2H, m), 2.96 (1H, dd, J=13.7, 6.8 Hz), 2.87 (1H, dd, J=13.9, 7.9 Hz); LCMS (ESP): 417 (M+H+), 439 (M+Na+); 415 (M-H)'.
Example 25-8: Phosphoric acid mono-(2-{ϊ1-(3-chloro-phenyl)-methanoyl3-amino}-3-phen l- propyl) ester
Figure imgf000054_0002
Prepared as described in the synthesis of 25-1 using 21a (45 mg, 0.168 mmol), 3- chlorobenzoyl chloride (29 mg, 0.168 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 5 mg (8% yield) of the title compound 25-8. 1H NMR (CD3OD): δ 7.64 (1H, s), 7.57 (1H, d, J=7.5 Hz), 7.39 (1H, d, J=9 Hz), 7.30 (1H, t, J=7.8 Hz), 7.23-7.01 (5H, ), 4.33 (1H, m), 3.91 (2H, m), 2.94 (1H, dd, J=13.5, 6.9 Hz), 2.83 (1H, dd, J=13.8, 8.1 Hz); LCMS (ESP): 392 (M+Na+); 368 (M-H)".
Example 25-9:
Figure imgf000054_0003
Prepared as described in the synthesis of 25-1 using 21a (46 mg, 0.172 mmol), nicotinoyl chloride hydrochloride (31 mg, 0.172 mmol) and 1 M sodium carbonate solution (1 L). Preparative HPLC purification gave 3 mg (2% yield) of the title compound 25-9. 1H NMR (CD3OD): δ 8.89 (1H, s), 8.68 (1H, d, J=5.1 Hz), 8.36 (1H, dt, J=7.5, 1.8 Hz), 7.68 (1H, dd, J=7.8, 5.4 Hz), 7.22-7.05 (5H, m), 4.42 (1H, m), 3.98 (2H, m), 2.96 (1H, dd, J=13.5, 6.6 Hz), 2.85 (1H, dd, J=13.5, 8.1 Hz); LCMS (ESP): 337 (M+H+), 359 (M+Na+); 335 (M-H)".
Example 25-10: Phosphoric acid mono-{2-[(1-naphthalen-1-yl-methanoyl)-amino]-3-phenyl- propyl} ester
Figure imgf000055_0001
Prepared as described in the synthesis of 25-1 using 21a (62 mg, 0.232 mmol), 1-naphthoyl chloride (0.035 mL, 0.232 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 8 mg (9% yield) of the title compound 25-10. H NMR (CD3OD): δ 7.93 (1H, m), 7.88 (1H, d, J=7.8 Hz), 7.77 (1H, d, J=8.4 Hz), 7.55-7.24 (9H, m), 4.70 (1H, m), 4.16 (2H, m), 3.14 (1H, dd, J=14.1, 5.8 Hz), 2.91 (1H, dd, J=13.9, 9.4 Hz); LCMS (ESP): 408 (M+Na+), 430 (M- H+2Na÷); 384 (M-H)".
Example 25-11: Phosphoric acid monό-{3-phenyl-2-j;(1-quino aliπ-2-yl-methanoyl)-amino3- propyl} ester
Figure imgf000055_0002
Prepared as described in the synthesis of 25-1 using 21a (68 mg, 0.254 mmol), quinoxaloyl chloride (50 mg, 0.254 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 17 mg (17% yield) of the title compound 25-11. 1H NMR (CD3OD): δ 9.44 (1H, s), 8.24 (1H, m), 8.17 (1H, m), 8.01-7.90 (2H, m), 7.4-7.14 (5H, m), 4.62 (1H, m), 4.17 (2H, m), 3.10 (2H, m); LCMS (ESP): 388 (M+H+), 410 (M+Na+); 386 (M-H)". Example 25-12:
Figure imgf000056_0001
Prepared as described in the synthesis of 25-1 using 21a (63 mg, 0.236 mmol), 3-chloro- thiophene-2-carbonyl chloride (43 mg, 0.236 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 26 mg (30% yield) of the title compound 25-12. 1H NMR (CD3OD): δ 7.67 (1H, d, J=5.1 Hz), 7.37-7.18 (5H, m), 7.05 (1H, d, J=5.4 Hz), 4.50 (1H, m), 4.09 (2H, m), 3.08 (1H, dd, J=13.9, 6.9 Hz), 2.99 (1H, dd, J=13.9, 8.5 Hz); HRMS (MALDl) caic for C14H16N05PSCI (M+H+) 376.0175; found 376.0158.
Example 25-13: Phosphoric acid mono-(2-{[1-(2-hydroxy-phenyl)-methanoyl]-amino}-3- phenyl-propyl) ester
Figure imgf000056_0002
Prepared as described in the synthesis of 25-1 using 21ι (187 mg, 0.699 mmol), acetylsalicyloyl chloride (139 mg, 0.699 mmol) and 1 M sodium carbonate solution (2 mL). Preparative HPLC purification gave 25 mg (10% yield) of the title compound 25-13. 1H NMR (CD3OD): δ 7.79 (1H, dd, J=8.1 , 1.8 Hz), 7.41-7.2 (7H, m), 6.89 (2H, m), 4.53 (1H, m), 4.08 (2H, m), 3.07 (1H, dd, J=13.6, 6.8 Hz), 2.99 (1H, dd, J=13.6, 6.7 Hz); HRMS (MALDl) caic for CιeH19NOeP (M+H+) 352.0950; found 352.0960.
Example 25-14: Phosphoric acid mono-{2-[(1-furan-2-yI-methanoyl)-amino]-3-phenyl-propyl} ester
Figure imgf000056_0003
Prepared as described in the synthesis of 25-1 using 21a (85 mg, 0.318 mmol), 2-furoyl chloride (0.032 L, 0.318 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 86 mg (83% yield) of the title compound 25-14. 1H NMR (CD3OD): δ 7.65 (1H, br d), 7.33-7.17 (5H, m), 7.08 (1H, d, J=3.6 Hz), 6.57 (1H, dd, J=3.3, 1.5 Hz), 4.48 (1H, m), 4.05 (2H, m), 3.05 (1H, dd, J=13.6, 6.4 Hz), 2.94 (1h, dd, J=13.6, 8.1 Hz); HRMS (MALDl) caic for C14H17N06P (M+H+) 326.0794; found 326.0801.
Example 25-15:
Figure imgf000057_0001
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21a (106 mg,
0.397 mmol) and 2-methylpropanoic anhydride (0.16 mL, 153 mg, 0.966 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 110 mg (92% yield) of the title compound 25-15. 1H NMR (CD3OD): δ 7.29-7.08 (5H, m), 6.94 (1H, br d), 4.34 (1H, br s), 4.10 (1H, m), 3.96 (1H, m), 2.82 (2H, m), 2.41 (1H, heat, J=7.2 Hz), LOO (3H, d, J=7 Hz), 0.97 (3H, d, J=7 Hz); LCMS (ESP): 302 (M+H+); 300 (M-H)".
Example 25-16: Phosphoric acid mono-[(R)-2-(2,2-dimethyl-propanoylamino)-3-phenyl-propyl] ester
Figure imgf000057_0002
Prepared as described in the synthesis of 25-15 using 21a (110 mg, 0.412 mmol), 2,2- dimethylpropanoic anhydride (0.16 mL, 147 mg, 0.79 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 130 mg (100% yield) of the title compound 25-16. 1H NMR (CD3OD): δ 7.32-7.11 (5H, m), 6.25 (1H, br d, J=8.7 Hz), 4.37 (1H, m), 4.13 (1H, m), 3.98 (1H, m), 2.93 (1H, dd, J=14.1, 6.6 Hz), 2.80 (1H, dd, J=14.1, 8.5 Hz), 1.06 (9H, s); LCMS (ESP): 316 (M+H+); 314 (M-H)'.
Example 25-40: Phosphoric acid mono-(2-acetylamino-3-phenyl-propyl) ester
Figure imgf000058_0001
Prepared as described in the synthesis of 25-15 using 21a (120 mg, 0.45 mmol), acetic anhydride (0.1 mL, 108 mg, 1.0 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 50 mg (40% yield) of the title compound 25-40. 1H NMR (CD3OD): δ 7.64-
7.30 (5H, m), 4.47 (1H, m), 4.16 (2H, m), 3.16 (1H, dd, J=14.1, 6.6 Hz), 2.99 (1H, dd, J=13.9, 8.7
Hz), 2.10 (3H, s); LCMS (ESP): 274 (M+H+), 296 (M+Na+); 272 (M-H)'.
Example 25-17: Phosphoric acid mono-{2-[(1-cyclohexyl-methanoyl)-amino]-3-phenyl-propyl} ester
Figure imgf000058_0002
To an ether solution (5 mL) of cyclohexanecarboxylic acid (250 mg, 1.95 mmol) was added pyridine (0.5 mL) and cyclohexanecarbonyl chloride (286 mg, 0.261 mL, 1.95 mmol). After 10 h, the suspension was diluted with ether (20 mL), washed with ice-cold 5% HCI solution (1x50 mL) and concentrated NaHCOs solution (1x50 mL) and dried over Na2S0 . All solvent was removed in vacuo to give 350 mg of cyclohexanecarboxylic anhydride as a colorless oil. A portion of the cyclohexanecarboxylic anhydride (226 mg, 0.948 mmol) was added to a sodium carbonate solution (1 M, 2 mL) of the aminophosphate 21a (110 mg, 0.412 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 50 mg (36% yield) of the title compound 25-17. 1H NMR (CD3OD): δ 7.27-7.1 (5H, m), 6.67 (1H, br d), 4.34 (1H, br s), 4.08 (1H, m), 3.96 (1H, m), 2.90 (1H, dd, J=14.4, 6.6 Hz), 2.78 (1H, dd, J=14.9, 9 Hz), 2.10 (1 H, br t), 1.79-1.51 (5H, m), 1.33-1.02 (5H, m); LCMS (ESP): 342 (M+H+); 340 (M-H)'.
Example 25-18: Phosphoric acid mono-{2-[(1-1H-indol-2-yl-methanoyl)-amino]-3-phenyl- propyl} ester
Figure imgf000059_0001
To a DMF solution (1mL) of the aminophosphate 21a (78 mg, 0.292 mmol) was added imidazole (65 mg, 0.962 mmol) and t-butyldimethylchlorosilane (110 mg, 0.730 mmol). After 3.5 h, indole-2-carboxylic acid (49 mg, 0.307 mmol), EDC (70 mg, 0.365 mmol), DMAP (4- dimethylaminopyridine) (7 mg, 0.058 mmol) were added to the reaction. The mixture was stirred for 15 h and was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 1 mg (1% yield) of the title compound 25-18. H NMR (CD3OD): δ 7.48 (1H, d, J=8.5 Hz), 7.30 (1H, d, J=8.4 Hz), 7.25-6.89 (7H, m), 4.36 (1H, m), 3.93 (2H, m), 2.92 (2H, m); LCMS (ESP): 373 (M-H)".
Example 25-19: Phosphoric acid mono-{2-I(1-benzofuran-2-yl-methanoyl)-amino3-3-phenyl- propyl} ©ster
Figure imgf000059_0002
Prepared as described in the synthesis of 25-18 using aminophosphate 21a (94 mg, 0.351 mmol), imidazole (79 mg, 1.16 mmol), t-butyldimethylchlorosilane (132 mg, 0.878 mmol), 1- benzofuran-2-carboxylic acid (60 mg, 0.369 mmol), EDC (84 mg, 0.439 mmol) and DMAP (9 mg, 0.07 mmol). Preparative HPLC purification gave 16 mg (12% yield) of the title compound 25-19. 1H NMR (CD3OD): δ 7.59 (1H, d, J=7.8 Hz), 7.47 (1H, d, J=8.1 Hz), 7.39-7.29 (2H, m), 7.25-7.03 (6H, m), 4.43 (1H, m), 3.98 (2H, m), 2.96 (1H, dd, J=13.8, 6.2 Hz), 2.87 (1H, dd, J=13.8, 8.1 H∑); LCMS (ESP): 374 (M-H)'.
Example 25-20: Phosphoric acid mono-(2-{[1-(6-hydroxy-naphthalen-2-yl)-methanoyl]-amino}- 3-phenyl-propyl) ester
Figure imgf000060_0001
Prepared as described in the synthesis of 25-18 using aminophosphate 21a (152 mg, 0.568 mmol), imidazole (155 mg, 2.27 mmol), t-butyldimethylchlorosilane (214 mg, 1.42 mmol), 6- (acety)oxy)-2-naphthoic acid (131 mg, 0.569 mmol), EDC (136 mg, 0.71 mmol) and DMAP (14 mg, 0.114 mmol). Preparative HPLC purification gave 25 mg (11% yield) of the title compound 25-20. 1H NMR (CD3OD): δ 8.21 (1H, s), 7.9-7.6 (4H, m), 7.4-7.05 (6H, m), 4.57 (1H, m), 4.12 (2H, m), 3.00 (2H, m); LCMS: 400 (M-H)'.
Example 25-21:
Figure imgf000060_0002
Prepared as described in the synthesis of 25-18 using aminophosphate 21a (94 mg, 0.351 mmol), imidazole (96 mg, 1.4 mmol), t-butyldimethylchlorosilane (132 mg, 0.878 mmol), 1-hydroxy-2- naphthoic acid (66 mg, 0.351 mmol), EDC (84 mg, 0.439 mmol) and DMAP (9 mg, 0.07 mmol). Preparative HPLC purification gave 7 mg (5% yield) of the title compound 25-21. 1H NMR (CD3OD): δ 8.24 (1H, d, =8.1 Hz), 7.70 (1H, d, J=7.8 Hz), 7.64 (1H, d, J=9 Hz), 7.49 (1H, t, J=7.2 Hz), 7.41 (1H, d, J=8.1 Hz), 7.3-7.05 (6H, m), 4.50 (1H, m), 4.04 (2H, m), 2.98 (2H, m); HRMS (MALDl) caic for C20H2ιNO6P (M+H+) 402.1107; found 402.1099.
Example 25-22: Phosphoric acid mono-(2-{[1-(3-hydroxy-naphthalen-2-yI)-methanoyl]-amino}- 3-phenyl-propyl) ester
Figure imgf000060_0003
Prepared as described in the synthesis of 25-18 using aminophosphate 21a (98 mg, 0.366 mmol), imidazole (100 mg, 1.46 mmol), t-butyldimethylchlorosilane (138 mg, 0.915 mmol), 3-hydroxy-
2-naphthoic acid (69 mg, 0.366 mmol), EDC (88 mg, 0.458 mmol) and DMAP (9 mg, 0.0732 mmol). Preparative HPLC purification gave 2 mg (1% yield) of the title compound 25-22. 1H NMR (CD3OD): δ 8.31 (1H, s), 7.72 (1H, d, J=8.4 Hz), 7.55 (1H, d, J=8.7 Hz), 7.36 (1H, t, J=7.8 Hz), 7.28-7.02 (7H, m), 4.47 (1H, m), 4.00 (2H, m), 2.96 (2H, m); LCMS (ESP): 402 (M+H+), 424 (M+Na+).
Example 25-23: Phosphoric acid mono-{(R)-2-[(1H-benzoimidazole-5-carbonyl)-amino]-3- phenyl-propyl} ester
Figure imgf000061_0001
Prepared as described in the synthesis of 25-18 using aminophosphate 21a (100 mg, 0.374 mmol), imidazole (76 mg, 1.12 mmol), l-butyldimethylchlorosilane (141 mg, 0.935 mmol), 6- (acetyloxy)-2-naphthoic acid (72.7 mg, 0.449 mmol), EDC (86 mg, 0.449 mmol) and DMAP (10 mg, 0.081 mmol). Before the acidification the reaction mixture was treated with 10% NaOH solution (1 mL) for 10 h. Preparative HPLC purification gave 30 mg (21% yield) of the title compound 25-23. 1H NMR (CD3OD): δ 9.44 (1H, s), 8.23 (1H, s), 7.99 (1H, d, J=8.7 H∑), 7.87 (1H, d, J=8.7 H∑), 7.37-7.13 (5H, m), 4.55 (1H, m), 4.13 (2H, m), 3.09 (1 H, dd, J=13.5, 6.9 Hz), 2.99 (1H, dd, J=13.5, 8.1 Hz); LCMS (ESP): 376 (M+H+); 374 (M-H)".
Example 25-24: Phosphoric acid mono-(2-{[1-(1-bromo-naphthaIen-2-yl)-methanoyI|-amϊrιo$- 3-phenyI-propyl) ester
Figure imgf000061_0002
To a methylene chloride solution (2 mL) of 1-bromo-2-naphthoic acid (182 mg, 0.725 mmol) was added oxalyl chloride (0.19 mL, 2.18 mmol) and 2 drops of DMF. The mixture was stirred for2 h and concentrated in vacuo. To the residue was added sodium carbonate solution (1 M, 2 mL), the aminophosphate 21a (194 mg, 0.725 mmol) and 1 mL of acetonitrile. After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 103 mg (31% yield) of the title compound 25-24. 1H NMR (CD3 D): δ 8.32 (1H, d, J=8.7 Hz), 7.92 (2H, m), 7.65 (2H, m), 7.41-7.20 (6H, m), 4.60 (1H, m), 4.14 (2H, m), 3.12 (1H, dd, J=13.9, 6 Hz), 2.94 (1H, dd, 13.8, 8.7 Hz); HRMS (MALDl) caic for C20H20NO5PBr (M+H+) 464.0262; found 464.0271.
Example 25-25: Phosphoric acid mono-(2-{[1-(6-methoxy-naphthalen-2-yl)-methanoyI3- amino}-3-phenyl-propyl) ester
Figure imgf000062_0001
Prepared as described in the synthesis of 25-24 using 6-methoxy-2-πaphthoic acid (255 mg, 1.26 mmol), oxalyl chloride (0.33 mL, 3.78 mmol), DMF (2 drops), sodium carbonate solution (1 M, 2 mL) and the aminophosphate 21a (337 mg, 1.26 mmol). Preparative HPLC purification afforded 44 mg (13% yield) of the title compound 25-25. 1H NMR (CD3OD): δ 8.24 (1H, s), 7.86 (1H, d, J=9.3 H∑), 7.81 (2H, m), 7.41 (7H, m), 4.56 (1H, m), 4.13 (2H, m), 3.95 (3H, s), 3.11 (1H, dd, J=14.1, 7.2 Hz), 3.01 (1H, dd, J=13.6, 8.5 Hz); LCMS (ESP): 414 (M-H)"; Elemental Analysis for (C2ιH22NOsP 0.25H2O) caic: C 60.07, H 5.40, N 3.34; found: C 59.66, H 5.33, N 3.74.
Example 25-26: Phosphoric acid mono-{2-[(1-benzo[b]thiophen-2-yl-methanoyl)-amino]-3- phenyl-propyl} ester
Figure imgf000062_0002
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21b (68 mg,
0.254 mmol) and beπzothiophene-2-carbonyl chloride (50 mg, 0.254 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 20 mg (20% yield) of the title compound 25-26. 1H NMR (CD3OD): δ 7.99-7.86 (3H, m),
7.53-7.15 (7H, m), 4.49 (1H, m), 4.10 (2H, m), 3.04 (2H, m); LCMS (ESP): 414 (M+Na+); 390 (M-H)'
Example 25-27: Phosphoric acid mono-{2-[(1-naphthalen-2-yl-methanoyl)-amino]-3-pheπyl- propyl} ester
Figure imgf000063_0001
Prepared as described in the synthesis of 25-26 using 21b (70 mg, 0.262 mmol), 2-naphthoyl chloride (50 mg, 0.262 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 20 mg (20% yield) of the title compound 25-27. H NMR (CD3OD): δ 8.32 (1H, s),
8.08-7.78 (4H, m), 7.66-7.5 (2H, m), 7.39-7.17 (5H, m), 4.56 (1H, m), 4.11 (2H, ), 3.12 (1H, dd,
J=13.7, 6.8 Hz), 3.02 (1H, dd, J=13.8, 7.7 Hz); LCMS (ESP): 386 (M+H+), 408 (M+Na+); 384 (M-H)".
Aminoalcohol 18c:
Figure imgf000063_0002
To a THF solution (30 mL) of D-3-fIuorophenyialanine (5 g, 27.3 mmol) at 0 °C was added borane in THF (1 M, 68.3 mL, 68.3 mmol). After 5 h at 25 ΘC, to the solution was added sat'd NaHC03 solution (10 mL), which was stirred for 15 h. The mixture was concentrated and extracted with methylene chloride (3x50 mL). Combined organic layers were dried (Na2S04) and concentrated. The residue was purified by column chromatography (CH2CI2/MeOH/NH4OH 95/5/0.5 to 90/10/1) to give 1.94 g (42% yield) of the compound 18c as a white solid. 1H NMR (CD3OD): δ 7.32-7.21 (1H, m), 7.00-6.98 (3H, m), 3.63 (1H, dd, J=11.1, 4.2 Hz), 3.38 (1H, dd, J=10.5, 6.9 Hz), 3.12 (1H, m), 2.79 (1H, dd, J=13.5, 5.1 Hz), 2.54 (1H, dd, J=13.5, 8.4 Hz); LCMS (ESP): 170 (M+H+).
Alcohol 19c:
Figure imgf000063_0003
To a methylene chloride solution (30 mL) of aminoalcohol 18c (1.94 g, 11.5 mmol) was added triethylamine (2.8 L, 20 mmol) and benzyl chloroformate (2.05 mL, 14.3 mmol). The mixture was stirred for 15 h and then diluted with methylene chloride (50 mL). The solution was washed with brine (1x50 mL), dried (Na2S0 ) and concentrated. After column chromatography purification (20 to 30% EtOAc in hexanes), the compound 19c was obtained in 64% yield (2.22 g). 1H NMR (CDCI3): δ 7.4-7.19 (6H, m), 7.02-6.86 (3H, m), 5.08 (2H, s), 4.95 (1H, br s), 3.93 (1H, ), 3.69 (1H, dd, J=10.2, 3 Hz), 3.58 (1H, dd, J=11.4, 4.8 Hz), 2.87 (2H, d, J=7.5 Hz); LCMS (ESP): 326 (M+Na+); Elemental Analysis for (C17H18FN03) caic: C 67.31, H 5.98, N 4.62; found: C 67.31, H 5.98, N 4.62.
Phosphate Benzyl Ester 20c:
To an acetonitrile solution (40 mL) of the alcohol 19c (2.21 g, 7.29 mmol) and 1H-tetrazole
(1.37 g, 19.6 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (4.9 mL, 14.6 mmol) at 25 °C. After 5 h, MCPBA (5.72 g, 77% pure, 25.5 mmol) was added to the suspension. The solution was diluted with methylene chloride (100 mL), washed with concentrated NaHS03 solution (2x80 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (15-30% EtOAc in hexanes) to give 4.39 g of the compound 20c in 100% yield. 1H NMR (CDCI3): δ 7.44-7.12 (16H, m), 6.96-6.76 (3H, m), 5.18-4.96 (7H, m), 4.04-3.78 (3H, m), 2.87- 2.63 (2H, m). Aminophosphate 21c:
Figure imgf000064_0002
To an ethanol solution of the phosphate benzyl ester (20c, 4.37 g, 7.76 mmol) was added palladium on carbon (10%, 870 mg). The suspension was kept under hydrogen atmosphere (1 at ) for 15 h and was then added 5% HCI solution (10 mL). The mixture was filtered through a pad of celite. The filtrate was concentrated to dryness, affording 2.30 g of a yellowish solid. Preparative HPLC purification gave 1.2 g (62% yield) of the compound 21c as a white solid. H NMR (CD3OD): δ 7.45-7.35 (1H, m), 7.18-7.01 (3H, m), 4.10 (1H, m), 3.94 (1H, ), 3.70 (1H, m), 3.03 (2H, m); LCMS (ESP): 250 (M+H*); 248 (M-H)'.
Example 25-28: Phosphoric acid mono-{3-(3-fluoro-phenyl)-2-[(1-naphthalen-2-yl-methanoyl)- amino]-propyl} ester
Figure imgf000065_0001
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21c (110 mg, 0.385 mmol) and 2-naphthoyl chloride (110 mg, 0.578 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 160 mg (100% yield) of the title compound 25-28. 1H NMR (CD3OD): δ 8.31 (1H, s), 8.0-7.88 (3H, m), 7.82 (1H, dd, J=8.7, 1.8 Hz), 7.58 (2H, m), 7.31 (1H, m), 7.14 (2H, m), 6.95 (1H, br td), 4.59 (1H, m), 4.14 (2H, m), 3.13 (1H, dd, J=13.8, 6 Hz), 3.02 (1H, dd, J=13.9, 8.7 Hz); HRMS (MALDl) caic for C2oH20N05PF (M+H+) 404.1063; found 404.1078.
Example 25-29:
Figure imgf000065_0002
Prepared as described in the synthesis of 25-28 using 21c (110 mg, 0.38S mmol), 1- benzothiophene-2-carbonyl chloride (114 mg, 0.578 mmol) and 1 M sodium carbonate solution (1 mL). Preparative HPLC purification gave 145 mg (92% yield) of the title compound 25-29. 1H NMR (CD3OD): δ 7.95 (1H, s), 7.93-7.85 (2H, m), 7.4 (2H, m), 7.30 (1H, m), 7.2-7.04 (2H, m), 6.94 (1H, br td), 4.51 (1H, m), 4.2-4.0 (2H, m), 3.11 (1H, dd, J=13.9, 6.4 Hz), 3.01 (1H, dd, J=13.9, 8.3 Hz); HRMS (MALDl) caic for C18HN05PFS (M+H+) 410.0627; found 410.0639.
Example 25-30: Phosphoric acid mono-[(R)-2-(2,2-dimethyl-propanoylamino)-3-(3-fluoro- phenyl)-propyl] ester
Figure imgf000065_0003
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21c (65 mg, 0.228 mmol) and 2,2-dimethylpropanoic anhydride (0.08 mL, 0.392 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 43 mg (57% yield) of the title compound 25-30. 1H NMR (CD3OD): δ 7.19 (1H, m), 7.0-6.77 (3H, m), 4.23 (1H, m), 3.89 (2H, m), 2.91 (1 H, dd, J=13.8, 5.5 Hz), 2.74 (1H, dd, J-13.8, 9.2 Hz), 1.00 (9H, s); HRMS (MALDl) caic for C14H22N05PF (M+H*) 334.1220; found 334.1223.
Example 25-31: Phosphoric acid mono-[(R)-2-{[1-(1-bromo-naphthalen-2-yl)-methanoyl]- amino}-3-(3-fluoro-phenyl)-propyl] ester
Figure imgf000066_0001
To a methylene chloride solution (3 mL) of 1-bromo-2-naphthoic acid (161 mg, 0.643 mmol) was added oxalyl chloride (0.168 mL, 1.93 mmol) and 2 drops of DMF. The mixture was stirred for 2 h and concentrated in vacuo. To the residue was added sodium carbonate solution (1 M, 2 mL), the aminophosphate 21c (80 mg, 0.28 mmol) and 1 mL of acetonitriie. After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 70 mg (52% yield) of the title compound 25-31. 1H NMR (CD3OD): 6 8.22 (1H, d, J=8.7 Hz), 7.84 (1H, m), 7.56 (2H, m), 7.27 (1H, m), 7.18 (1H, d, J=8.7 Hz), 7.09 (1H, d, J=7.8 Hz),7.03 (1H, m), 6.91 (1H, td, J=8.7, 3 Hz), 4.51 (1H, m), 4.04 (2H, m), 3.05 (1H, dd, J=14.1, 5.7 Hz), 2.84 (1H, dd, 14.1 , 9.3 Hz); LCMS (ESP): 481 (M-H)'; Elemental Analysis for (C2oH18BrFN05P) caic: C 49.81, H 3.76, N 2.90; found: C 49.63, H 3.73, N 2.92.
Example 25-32: Phosphoric acid mono-((R)-3-(3-fluoro-phenyl)-2-{[1-(6-methoxy-naphthalen- 2-yl)-methanoyl]-amino}-propyl) ester
Figure imgf000066_0002
Prepared as described in the synthesis of 25-31 using 6-methoxy-2-naphthoic acid (143 mg, 7.07 mmol), oxalyl chloride (0.185 mL, 2.12 mmol), DMF (2 drops), sodium carbonate solution (1 M, 2 mL) and the aminophosphate 21c (88 mg, 0.353 mmol). Preparative HPLC purification afforded 50 mg (39% yield) of the title compound 25-32. 1H NMR (CD3OD): δ 8.25 (1H, s), 7.9-7.73 (3H, m), 7.37-7.06 (5H, m), 6.95 (1H, t, J=8.1 Hz), 4.57 (1H, m), 4.12 (2H, m), 3.95 (3H, s), 3.13 (1H, dd, J=13.8, 6.6 Hz), 3.01 (1H, dd, J=14.3, 7.7 Hz); LCMS (ESP): 432 (M-H)'; Elemental Analysis for (C21H2ιFNO6P 0.25H2O) caic: C 57.60, H 4.95, N 3.20; found: C 57.58, H 4.97, N 3.27.
Aminoalcohol 18d:
Figure imgf000067_0001
To a THF solution (10 mL) of D-3-methylphenylalanine (1 g, 5.58 mmol) at 0 °C was added borane in THF (1 M, 22.4 mL, 22.4 mmol). After 48 h at 25 °C, the solution was added sat'd NaHCOj solution (2 L) and stirred for 3 h. The mixture was concentrated and extracted with methylene chloride (3x20 mL). Combined organic layers were dried (Na2S04) and concentrated. The residue was purified by column chromatography (CH2CI2/MeOH/NH4θH 95/5/0.5 to 90/10/1) to give 490 mg (53% yield) of the compound 18d as a white solid. H NMR (CD3OD): δ 7.20 (1H, t, J=7.8 Hz), 7.08-6.94 (3H, m), 3.63 (1H, dd, J=10.5, 3.9 Hz), 3.37 (1H, dd, J=10.5, 7.5 H∑), 3.11 (1H, m), 2.76 (1H, dd, J=13.5, 5.7 Hz), 2.49 (1H, dd, J=13.8, 8.4 Hz), 2.34 (3H, s); LCMS (ESP): 166 (M+H*).
Alcohol 19d:
Figure imgf000067_0002
To a methylene chloride solution (5 mL) of aminoalcohol 18d (150 mg, 0.909 mmol) was added triethylamine (0.3 mL) and benzyl chloroformate (0.21 mL, 1.5 mmol). The mixture was stirred for 15 h and then diluted with methylene chloride (20 mL). The solution was washed with brine (1x20 mL), dried (Na2S04) and concentrated. After column chromatography purification (15 to 30% EtOAc in hexanes), the compound 19d was obtained in 63% yield (170 mg). 1H NMR (CDCI3): δ 7.40-7.27 (5H, m), 7.18 (1H, t, J=7.8 Hz), 7.08-6.95 (3H, m), 5.09 (2H, s), 4.94 (1H, br s), 3.94 (1H, m), 3.69 (1H, br ), 3.58 (1H, dd, J=14.5, 5.4 Hz), 2.82 (2H, d, J=6.9 Hz), 2.32 (3H, s); LCMS (ESP): 322 (M+Na+).
Phosphate Benzyl Ester 20d
Figure imgf000068_0001
To an acetonitrile solution (8 mL) of the alcohol 19d (160 mg, 0.535 mmol) and 1H-tetrazole (101 mg, 1.44 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (0.36 mL, 1.07 mmol) at 25 °C. After 5 h, MCPBA (0.42 g, 77% pure, 1.87 mmol) was added to the suspension. The solution was diluted with methylene chloride (35 mL), washed with concentrated NaHS03 solution (2x25 L), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (15-30% EtOAc in hexanes) to give 0.287 g of the compound 20d in 96% yield. H NMR (CDCI3): δ 7.39-7.2 (15H, m), 7.12 (1H, t, J=7.8 Hz), 7,01 (1H, d, J=7.8 Hz), 6.93 (2H, m), 5.14-4.98 (7H, m), 4.05-3.82 (3H, m), 2.78 (1H, brdd), 2.69 (1H, dd, J=13.8, 7.8 Hz), 227 (3H, s).
Aminophosphate 21 d:
Figure imgf000068_0002
To an ethanol solution of the phosphate benzyl ester 20d (0.287 g, 0.513 mmol) was added palladium on carbon (10%, 58 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then added 5% HCI solution (5 mL). The mixture was filtered through a pad of celite. The filtrate was concentrated to dryness, affording 134 mg (92% yield) of 2 d a yellowish solid. 1H NMR (CD3OD): δ 7.26 (1H, t, J=7.5 Hz), 7.18-7.06 (3H, m), 4.12 (1H, br d), 3.99 (1H, m), 3.68 (1H, m), 2.98 (2H, d, J=7.8 Hz), 2.36 (3H, s).
Example 25-33: Phosphoric acid mono-[(R)-2-{[1-(diethylamino-oxo-2H-chromen-3-yl)- methanoyl]-amino}-3-(3-fluoro-phenyl)-propyl] ester
Figure imgf000069_0001
25-33
To a sodium carbonate solution (1 M, 2 mL) was added the aminophosphate 21c (50 mg, 0.176 mmol) and 7-diethylaminocoumarin-3-carboxylic acid, succinimidyl ester (50 mg, 0.140 mmol). After 15 h, it was acidified to pH~1 by addition of 1 M HCI solution at 0 °C. Preparative HPLC purification afforded 43 mg (57% yield) of the title compound 25-33. H NMR (CD3OD): δ 8.69 (1H, s), 7.54 (1H, d, J=9.04 Hz), 7.31 (1H, dd, J=14.12, 7.91 Hz), 7.12 (2H, dd, J=17.33, 9.80 Hz), 6.94 (1H, t, J=8.86 Hz), 6.83 (1H, dd, J=9.04, 2.44 Hz), 6.95 (1H, d, J=2.26 Hz), 4.50 (1H, m), 4.07 (2H, t, J=4.71 Hz), 3.54 (4H, m), 3.14-2.92 (2H, m), 1.25 (6H, t, J=7.16 Hz); HRMS (MALDl) caic for C23H26FN207PH (M+H+) 493.1549; found 493.1540.
Example 25-34: Phosphoric acid mono- 2- [1-(1-bromo-naphthalen-2-yI)-methano l3- aminoJ-3-m-tolyl-propyl) ester
Figure imgf000069_0002
To a methylene chloride solution (3 mL) of 1-bromo-2-naphthoic acid (123 mg, 0.49 mmol) was added oxalyl chloride (0.128 mL, 1.47 mmol) and 2 drops of DMF. The mixture was stirred for 3.5 h and concentrated in vacuo. To the residue was added sodium carbonate solution (1 M, 2 mL), the aminophosphate 21d (69 mg, 0.245 mmol) and 2 mL of acetonitrile. After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 32 mg (27% yield) of the title compound 25-34. 1H NMR (CD3OD): δ 8.22 (1H, d, J=8.4 Hz), 7.82 (2H, m), 7.7.6-7.46 (2H, m), 7.2-6.9 (5H, m), 4.48 (1H, m), 4.03 (2H, m), 2.98 (1H, dd, J=13.9, 6.2 Hz), 2.80 (1H, dd, J=13.9, 8.7 Hz), 2.26 (3H, s); HRMS (MALDl) caic for C21H22N05PBr (M+H*) 478.0419; found 478.0438.
Example 25-35: Phosphoric acid mono-((R)-2-{[1-(6-methoxy-naphthalen-2-yl)-methanoyl]- amino}-3-m-tolyl-propyl) ester
Figure imgf000070_0001
Prepared as described in the synthesis of 25-32 using 6-methoxy-2-naphthoic acid (99 mg,
0.49 mmol), oxalyl chloride (0.128 mL, 1.47 mmol), DMF (2 drops), sodium carbonate solution (1 M, 2 mL) and the aminophosphate 21 d (69 mg, 0.245 mmol). Preparative HPLC purification afforded 76 mg (51% yield) of the title compound 25-35. 1H NMR (CD3OD): δ 8.24 (1H, s), 7.86 (1H, d, J=8.7 Hz), 7.82 (2H, m), 7.30 (1H, d, J=2.4 Hz), 7.24-7.09 (4H, m), 7.03 (1H, d, J=7.5 Hz), 4.53 (1H, m), 4.10 (2H, m), 3.95 (3H, s), 3.08 (1H, dd, J=13.9, 6.8 Hz), 2.96 (1H, dd, J=14, 8.3 Hz), 2.31 (3H, ε); HRMS (MALDl) caic for C22H25N06P (M+H+) 430.1420; found 430.1436.
Alcohol 26a:
Figure imgf000070_0002
To a sodium carbonate solution (1 M, 2 mL) was added acetonitrile (1 mL), the aminoalcohol 18d (180 mg, 1.09 mmol) and 2-naphthoyl chloride (250 mg, 1.31 mmol). After 10 h, the mixture was extracted with methylene chloride (2x20 mL). Combined organic layers were dried (MgS04) and concentrated. The residue was purified by column chromatography (40% EtOAc in hexanes) to afford 350 mg (100% yield) of the compound 26a. 1H NMR (CD3OD): δ 8.07 (1H, s), 7.82-7.73 (3H, m), 7.65 (1H, dd, J=9, 1.5 Hz), 7.54-7.41 (2H, ), 7.16 (1H, t, J=7.2 Hz), 7.08-6.97 (3H, m), 6.46 (1H, br d, J=7.2 Hz), 4.33 (1H, m), 3.77 (1H, dd, J=11.1, 3.3 Hz), 3.68 (1H, dd, J=11.1, 5.1 Hz), 2.93 (2H, d, J=7.2 Hz), 2.27 (3H, s). Alcohol 26b:
Figure imgf000071_0001
Prepared as described in the synthesis of the alcohol 26a using the aminoalcohol 18d (131 mg, 0.794 mmol), 1-benzothiophene-2-carbonyl chloride (195 mg, 0.992 mmol) in a co-solvent of sodium carbonate solution (1 M, 1.5 mL) and acetonitrile (1.5 mL). Purification by column chromatography (50% EtOAc in hexanes) afforded 230 mg (89% yield) of the compound 26b. 1H NMR (CD3OD): δ 7.96-7.81 (3H, m), 7.50-7.36 (2H, m), 7.21-7.04 (3H, m), 7.01 (1H, d, J=7.5 Hz), 4.32 (1H, m), 3.67 (1H, d, J=5.4 Hz), 3.00 (1H, dd, J=13.1, 6.6 Hz), 2.87 (1H, dd, J=13.1, 8.7 Hz), 2.29 (3H, s); LCMS (ESP): 326 (M+H+), 348 (M+Na+); 324 (M-H)".
Phosphate Benzyl Ester 27a:
Figure imgf000071_0002
To an acetonitrile solution (10 L) of the alcohol 26a (348 mg, 1.09 mmol) and 1H-fetrazole (191 mg, 2.728 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (0.92 mL, 941 mg, 2.728 mmol) at 25 °C. After 5 h, MCPBA (0.938 g, 60% pure, 3.27 mmol) was added to the suspension. The solution was diluted with methylene chloride (35 mL), washed with concentrated NaHS03 solution (2x25 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (30% EtOAc in hexanes) to give 0.400 g of the compound 27a in 63% yield. 1H NMR (CDCI3): δ 8.34 (1H, s), 7.95-7.82 (3H, m), 7.55 (2H, m), 7.41-7.28 (11H, m), 7.17 (1H, t, J=7.5 Hz), 7.10-6.99 (3H, m), 5.15-4.97 (5H, m), 4.50 (1H, m), 4.17-3.96 (2H, m), 3.04 (1H, dd, J=13.5, 5.4 Hz), 2.81 (1H, dd, J=13.5, 9 Hz), 2.31 (3H, s). Phosphate Benzyl Ester 27b:
Figure imgf000072_0001
Prepared as described in the synthesis of compound 27a using the alcohol 26b (221 mg,
0.68 mmol), 1H-tetrazole (129 mg, 1.84 mmol), dibenzyl N, N-diisopropylphosphoramidite (0.46 mL, 1.36 mmol) and MCPBA (0.533 g, 77% pure, 2.38 mmol). Purification by column chromatography (30% EtOAc in hexanes) gave 0.50 g of the compound 27b in 100% yield. 1H NMR (CDCI3): δ 7.89- 7.78 (3H, m), 7.5-7.29 (13H, m), 7.16 (1H, t, J=7.8 Hz), 7.08-6.95 (3H, m), 5.16-4.97 (4H, m), 4.41 (1H, m), 4.04 (2H, m), 3.05 (1H, dd, J=13.5, 5.4 Hz), 2.77 (1H, dd, J=13.5, 9.6 Hz), 2.31 (3H, s).
Example 25-36: Phosphoric acid mono-{2-[(1-nøphthalen-2-yl-methanoyI)-arnino3-3-m-tolyl- propyl] ester
Figure imgf000072_0002
To an ethanol solution of the phosphate benzyl ester 27a (0.35 g, 0.604 mmol) was added palladium on carbon (10%, 35 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then filtered through a pad of celite. All solvent was removed in vacuo. The residue was purified by preparative HPLC to give 45 mg (19% yield) of the title compound 25-36. 1H NMR (CD3OD): δ 8.21 (1H, s), 7.9-7.76 (3H, m), 7.72 (1H, dd, J=8.4, 1.8 Hz), 7.48 (2H, m), 7.13-7.0 (3H, m), 6.93 (1H, d, J=6.9 Hz), 4.45 (1H, m), 4.03 (2H, m), 2.97 (1H, dd, J=13.7, 6.8 Hz), 2.87 (1H, dd, J=13.5, 8.1 Hz); HRMS (MALDl) caic for C21H23N05P (M+H+) 400.1314; found 400.1314. Example 25-37: Phosphoric acid mono-{2-[(1-benzo[b]thiophen-2-yl-methanoyI)-amino]-3-m- tolyl-propyl} ester
Figure imgf000073_0001
Prepared as described in the synthesis of 25-36 using the phosphate benzyl ester (27b,
0.398 g, 0.68 mmol) was added palladium on carbon (10%, 100 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 72 h and was then filtered through a pad of celite. All solvent was removed in vacuo. The residue was purified by preparative HPLC to give 120 mg (44% yield) of the title compound 25-37. 1H NMR (CD3OD): δ 8.0-7.8 (3H, m), 7.44 (2H, m), 7.23-7.0 (4H, m), 4.47 (1H, m), 4.09 (2H, m), 3.04 (1H, dd, J=13.8, 6.8 Hz), 2.95 (1H, dd, J=14.1, 7.7 Hz), 2.30 (3H, s); LCMS (ESP): 404 (M-H)"; Elemental Analysis for (C 9H20NO5PS) caic: C 56.29, H 4.97, N 3.46; found: C 56.03, H 4.94, N 3.39.
Alcohol 28:
Figure imgf000073_0002
To a THF solution (10 mL) of Boc-D-Ala (3-pyridyl)-OH (4.39 g, 16.5 mmol) at 0 °C was added borane in THF (1 M, 40 mL, 40 mmol). After 20 h at 25 °C, saturated NaHC03 solution (5 mL) was added to the solution and stirred for 3 h. The mixture was concentrated and extracted with methylene chloride (3x50 mL). Combined organic layers were dried (Na2S04) and concentrated. The residue was purified by column chromatography (50% to 100% EtOAc in hexanes) to give 1.51 g (36% yield) of the compound 28 as a white solid. 1H NMR (CDCI3): δ 8.47 (2H, s), 7.84 (1H, d, J=7.8 Hz), 7.44 (1H, dd, J=7.8, 5.7 Hz), 4.82 (1H, m), 3.87 (1H, m), 3.72 (1H, dd, J=10.5, 3.6 Hz), 3.62 (1H, dd, J=10.5, 4.2 Hz), 2.95 (2H, m), 1.39 (9H, m); LCMS (ESP): 253 (M+H+), 275 (M+Na+); HRMS (MALDl) caic for C13H21N203 (M+H+) 253.1552; found 253.1564.
Phosphate Benzyl Ester 29:
Figure imgf000073_0003
To an acetonitrile solution (15 mL) of the alcohol 28 (997 mg, 3.96 mmol) and 1H-tetrazole (1.11 g, 15.8 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (2.66 mL, 7.91 mmol) at 25 °C. After 3 h, MCPBA (2.67 g, 77% pure, 11.9 mmol) was added to the suspension. The solution was diluted with methylene chloride (60 mL), washed with concentrated NaHS03 solution (2x25 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (50% to 100% EtOAc in hexanes) to give 0.481 g of the compound 29 in 24% yield. H NMR (CDCI3): δ 8.46 (1H, br d), 8.39 (1H, br s), 7.48 (1H, d, J=6.6 Hz), 7.36 (11H, m), 5.06 (4H, m), 4.81 (1H, m), 4.0-3.80 (2H, m), 2.72 (2H, d, J=6.3 Hz), 1.37 (9H, s); LCMS (ESP): 513 (M+H+); 535 (M+Na*).
Example 30: (1-Phosphoπooxymethyl-2-pyridin-3-yl-ethyI)-carbamic acid tert-butyl ester
Figure imgf000074_0001
To an ethanol solution (3 mL) of the benzyl ester 29 (48 mg, 0.094 mmol) was added palladium on carbon (10%, 15 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h. After filtration, the filtrate was concentrated to dryness affording 36 mg (100% yield) of the title compound 30 . 1H NMR (CD3OD): δ 8.58 (1H, s), 8.49 (1H, br d), 8.19 (1H, d, J=7.5 H∑), 7.69 (1H, br t), 3.87 (1H, m), 3.79 (2H, m), 3.06 (1H, dd, J=13.9, 5.3 Hz), 2.82 (1H, dd, J=13.8, 8.5 Hz), 1.23 (9H, s); HRMS (MALDl) caic for C13H22N206P (M+H+) 333.1216; found 333.1218.
Aminophosphate 21 e:
Figure imgf000074_0002
To a methylene chloride solution (2.5 mL) of the benzyl ester 29 (278 mg, 0.543 mmol) at 0 °C was added trifiuoroacetic acid (0.75 mL). After 45 min, the solution was concentrated in vacuo to give 385 mg of amine 31 as a colorless oil. The crude amine 31 was dissolved in ethanol (5 mL) and added palladium on carbon (10%, 75 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 48 h. After filtering, the filtrate was concentrated to dryness affording the compound 21e as a solid (225 mg, 100%). 1H NMR (CD3OD): δ 8.68 (1H, br s), 8.27 (1H, br s), 7.48 (1H, m), 7.25 (1H, m), 4.83 (1H, d), 4.0-3.6 (2H, m), 3.26-3.01 (2H, m); LCMS (ESP): 233 (M+H+), 465 (2M+H+); 231 (M-H)". Example 25-38: Phosphoric acid mono-{2-[(1-benzo[b]thiophen-2-yl-methanoyl)-amino]-3- pyridin-3-yl-propyI} ester
Figure imgf000075_0001
To a sodium carbonate solution (1 M, 1 mL) was added the aminophosphate 21e (110 mg, 0.474 mmol), 1-benzothiophene-2-carbonyl chloride (93 mg, 0.474 mmol). After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 25 mg (14% yield) of the title compound 25-38. 1H NMR (CD3OD): δ 8.83 (1H, s), 8.71-8.63 (1H, m), 8.58-8.46 (10H, m), 8.02-7.83 (4H, m), 7.44 (2H, m), 4.60 (1H, m), 4.17 (2H, m), 3.42-3.13 (2H, m); HRMS (MALDl) caic for C17H18N205PS (M+H*) 393.0674; found 393.0681.
Example 25-30: Phosphoric acid mono-{2-[(1-naphthalen-2-yl-methanoyI)-aminoJ-3-pyridin-3- yl-propyl} ester
Figure imgf000075_0002
Prepared as described in the synthesis of 25-34 using 21e (97 mg, 0.418 mmol), 2-naphthoyl chloride (80 mg, 0.418 mmol) and 1 M sodium carbonate solution (2 mL). Preparative HPLC purification gave 25 mg (16% yield) of the title compound 25-39. 1H NMR (CD3OD): δ 8.85 (1H, s), 8.70 (1H, m), 8.58 (1H, d, J=8.4 Hz), 8.323 (1H, s), 8.0-7.85 (4H, m), 7.79 (1H, dd, J=8.4, 1.8 Hz), 7.58 (2H, m), 4.71 (1H, m), 4.22 (2H, m), 3.40 (1H, dd, J=14.0, 5.3 Hz), 3.22 (1H, dd, J=14.3, 9.6 Hz); HRMS (MALDl) caic for C19H20N2O5P (M+H+) 387.1110; found 387.1117. Example 33: Phosphoric acid mono-{(R)-3-cyclohexyl-2-[(1-naphthalen-2-yl-methanoyl)- aminoj-propyl} ester
Figure imgf000076_0001
To an aminophosphate 21a (540 mg, 2.02 mmol) solution in acetic acid (70% aqueous solution, 7 mL) was added 5% rhodium on alumina (306 mg). The suspension was kept under hydrogen atmosphere (60 psi) for 15 h. After filtration, the filtrate was concentrated to dryness to give 500 mg of compound 32. A portion of 32 (96 mg, 0.416 mmol) was dissolved in 1 M sodium carbonate solution (2 mL). 2-Naphthoyl chloride (79 mg, 0.416 mmol) was added to the solution. After 15 h, it was acidified to pH~1 by addition of concentrated HCI solution at 0 °C. Preparative HPLC purification afforded 4 mg (2% yield) of the title compound 33. 1H NMR (CD3OD): δ 8.40 (1H, s), 8.09-7.87 (4H, m), 7.60 (2H, m), 4.48 (1H, m), 4.06 (2H, m), 1.93 (1H, m); LCMS (ESP): 390 (M- H)'.
Aminoalcohol 34:
Figure imgf000076_0002
To a THF solution (30 mL) of D-homophenylalanine (5 g, 27.9 mmol) at 0 °C was added borane in THF (1 M, 55.8 L, 55.8 mmol). After 48 h at 25 °C, saturated NaHC03 solution (5 mL) was added to the solution and stirred for 4 h. The mixture was concentrated and extracted with methylene chloride (3x50 mL). Combined organic layers were dried (Na2S04) and concentrated. The residue was purified by column chromatography (CH2CI2/MeOH/NH OH 95/5/0.5) to give 1.2 g (26% yield) of the compound 34 as a yellowish oil. 1H NMR (CDCI3): δ 7.39-7.13 (5H, m), 4.01 (1H, dd,
J=1 L4, 3.6 Hz), 3.69 (1H, dd, J=11.7, 6.3 Hz), 2.94 (1H, m), 2.71 (2H, t, J=7.8 Hz), 2.09 (1H, m), 1.89 (1H, ); LCMS (ESP): 166 (M+H+). Alcohol 35:
Figure imgf000077_0001
To a sodium carbonate solution (1 M, 2 mL) was added acetonitrile (2 mL), the aminoalcohol 34 (300 mg, 1.82 mmol) and 2-naphthoyl chloride (347 mg, 1.82 mmol). After 10 h, the mixture was extracted with methylene chloride (3x20 mL). The combined organic layers were dried (MgS04) and concentrated. The residue was purified by column chromatography (50% EtOAc in hexanes) to afford 220 mg (38% yield) of the compound 35. 1H NMR (CDC!3): δ 8.16 (1H, s), 7.93-7.82 (3H, m), 7.74 (1H, dd, J=8.1 , 1.8 Hz), 7.55 (2H, m), 7.34-7.15 (5H, m), 6.40 (1H, d, J=7.2 Hz), 4.29 (1H, m), 3.87 (1H, dd, J=11.1, 3.9 Hz), 3.78 (1H, dd, J=10.8, 5.1 Hz), 2.81 (2H, t, J=7.5 Hz), 2.11-2.0 (2H, m); LCMS (ESP): 342 (M+Na+); 318 (M-H)"; Elemental Analysis for (C21H21N02) caic: C 78.97, H 6.63, N 4.39; found: C 79.07, H 6.62, N 4.20.
Phosphate Benzyl Ester 36:
Figure imgf000077_0002
To an acetonitrile solution (6 mL) of the alcohol 35 (206 mg, 0.646 mmol) and 1H-tetrazole (122 mg, 1.74 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (0.433 mL, 1.29 mmol) at 25 °C. After 5 h, MCPBA (0.3 g, 77% pure, 2.26 mmol) was added to the suspension. The solution was diluted with methylene chloride (35 mL), washed with concentrated NaHS03 solution (2x25 mL), dried over MgS0 and concentrated in vacuo. The residue was purified by column chromatography (30% EtOAc in hexanes) to give 0.311 g of the compound 36 in 83% yield. 1H NMR (CDCI3): δ 8.28 (1H, s), 7.95-7.78 (4H, m), 7.55 (2H, m), 7.37-7.13 (16H, m), 6.92 (1H, d, J=8.1 Hz), 5.11-4.93 (4H, m), 4.41 (1 H, m), 4.21-4.07 (1 H, m), 2.72 (2H, t, J=7.8 Hz), 1.97 (2H, m). -78-
by column chromatography (50% EtOAc in hexanes) to give 308 mg (28% yield for 3 steps) of the desired compound 40. 1H NMR (CDCI3): δ 8.43 (1H, d, J=8.4 Hz), 8.14 (1H, dd, J=7.8, 0.9 Hz), 8.04 (1H, d, J=8.7 Hz), 7.40 (2H, m), 7.07 (1H, d, J=7.5 Hz), 6.78 (1H, m), 6.55-6.46 (2H, m), 6.38 (1H, dt, J=9, 1.2 Hz), 5.25 (1H, d, J=10.5 Hz), 3.56 (1H, dd, J=10.8, 3.9 Hz), 3.47-3.31 (3H, m), 2.81 (6H, s), 2.61 (1H, dd, J=13.8, 6.6 Hz), 2.49 (1H, dd, J=13.8, 7.8 Hz).
Phosphate Benzyl Ester 41
Figure imgf000078_0001
To an acetonitrile solution (1 mL) of the alcohol 35 (53 mg, 0.132 mmol) and 1H-tetra∑ole (28 mg, 0.394 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (91.2 mg, 0.264 mmol) at 25 °C. After 4 h, 50% hydrogen peroxide (1 mL) was added to the suspension at 0 °C. The solution was diluted with methylene chloride (25 mL), washed with concentrated NaHS03 solution (2x25 mL), dried over MgS0 and concentrated in vacuo. The residue was purified by column chromatography (35% EtOAc in hexanes) to give 0.06 g of the compound 41 in 69% yield. 1H NMR (CDCI3): δ 8.48 (1H, d, J=8.4 Hz), 8.15 (1H, dd, J=7.5, 0.9 Hz), 8.11 (1H, d, J=8.7 Hz), 7.45 (2H, m), 7.38-7.30 (10H, m), 7.12 (1H, d, J=7.5 Hz), 6.83 (1H, m), 6.59 (1H, td, J=8.7, 2.7 H∑), 6.51 (1H, d, J=7.5 Hz), 6.42 (1H, dt, J=9.6, 1.8 Hz), 5.42 (1H, d, J=8.1 Hz), 5.08-4.94 (4H, m), 3.95-3.84 (2H, m), 3.50 (1H, m), 2.86 (6H, s), 2.59 (1H, dd, J=13.8, 7.2 Hz), 2.46 (1H, dd, J=13.8, 7.2 Hz).
Example 42: Phosphoric acid mono-[(R)-2-(5-dimethylamino-naphthalene-1-sulfonylamino)-3- (3-fluoro-phenyI)-propyl] ester
Figure imgf000078_0002
-77-
Example 37: Phosphoric acid mono-{2-[(1-naphthalen-2-yl-methanoyl)-amino]-4-phenyl-butyl} ester
Figure imgf000079_0001
To an ethanol solution of the phosphate benzyl ester 36 (0.305 g, 0.527 mmol) was added 10% palladium on carbon (60 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then filtered through a pad of celite. All solvent was removed in vacuo to give 210 mg (100%) of the title compound 37. 1H NMR (CD3OD): δ 8.39 (1H, s), 8.1-7.9 (4H, m), 7.59 (2H, m), 7.3-7.18 (5H, m), 4.37 (1H, m), 4.12 (2H, m), 2.79 (2H, m), 2.06 (2H, m); HRMS (MALDl) caic for C2ιH23N05P (M+H4) 400.1314; found 400.1312.
Carboj∑ylic Acid 38:
Figure imgf000079_0002
To a methylene chloride solution (8.5 mL) of D-3-fluorophenylalanine (0.5 g, 2.73 mmol) was added triethylamine (1 mL) and dansyl chloride (0.737 g, 2.73 mmol). After 12 h, the mixture was concentrated in vacuo. The residue was dissolved in EtOAc (10 mL) and added to a 5% HCI solution (10 mL). The resulting precipitate 38 was collected by filtration. The solid 38 was dissolved in MeOH (15 mL). After cooling the solution to -20 °C, thionyl chloride (1.5 mL) was added. After 15 h at 25 °C, the solution was concentrated in vacuo and the residue was dissolved in EtOAc (50 mL). The EtOAc solution was washed with ice-cold sat'd sodium carbonate solution (1x50 mL), dried over MgS0 and concentrated to dryness affording 506 mg of the methyl ester 39. The solid 39 was dissolved in THF (10 mL) and added LiBH4 (76 mg, 3.5 mmol). After 15 h, the reaction was quenched by slow addition of sat'd NH4CI solution (1.5 mL). The mixture was extracted by EtOAc (3x25 mL), washed with brine (1x20 L), dried (MgS04) and concentrated. The residue was purified To an ethanol solution of the phosphate benzyl ester 41 (0.06 g, 0.0906 mmol) was added 10% palladium on carbon (10 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was purified by HPLC to give 20 mg (46% yield) of the title compound 42. 1H NMR (CD3OD): δ 8.46 (1H, d, J=8.7 Hz), 8.39 (1H, d, J=9 Hz), 8.20 (1H, d, J=7.5 Hz), 7.77 (1H, d, J=7.5 Hz), 7.72- 7.59 (2H, m), 6.69 (1H, m), 6.59 (1H, d, J=8.1 Hz), 6.48-6.34 (2H, m), 4.14 (1H, m), 3.97 (1H, m), 3.62 (1H, m), 3.32 (6H, s), 2.90 (1H, dd, J=14.4, 3.9 Hz), 2.49 (1H, dd, J=10.2, 14.1 Hz); LCMS (ESP): 483 (M+H+).
Alcohol 43:
Figure imgf000080_0001
To a sodium carbonate solution (1 M, 5 mL) was added acetonitrile (5 mL), ethanolamine (0.173 mL, 3.12 mmol) and 2-naphthoyl chloride (594 mg, 3.12 mmol). After 15 h, the mixture was extracted with methylene chloride (3x50 mL). The combined organic layers were dried (MgS04) and concentrated. The residue was purified by column chromatography (EtOAc) to afford 386 mg (58% yield) of the compound 43. 1H NMR (CDCI3): δ 8.31 (1H, s), 7.96-7.78 (4H, m), 7.55 (2H, m), 6.76 (1H, br s), 3.89 (2H, t, J=5.7 Hz), 3.70 (2H, q, J=5.4 Hz); LCMS (ESP): 216 (M+H+), 238 (M+Na+); Elemental Analysis for (C13H13N02) caic: C 72.54, H 6.09, N 6.51; found: C 72.68, H 6.09, N 6.51.
Phosphate Benzyl Ester 44:
Figure imgf000080_0002
To an acetonitrile solution (10 mL) of the alcohol 43 (321 mg, 1.49 mmol) and 1H-tetrazole
(282 mg, 4.02 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (1 mL, 2.99 mmol) at 25 °C. After 3 h, MCPBA (77% pure, 900 mg, 5.22 mmol) was added to the suspension at 0 °C. The solution was diluted with methylene chloride (75 mL), washed with concentrated NaHS03 solution (2x50 mL), dried over MgS0 and concentrated in vacuo. The residue was purified by column chromatography (50% to 75% EtOAc in hexanes) to give 0.699 g of the compound 44 in 99% yield. H NMR (CDCI3): δ 8.34 (1H, s), 7.96-7.81 (4H, m), 7/55 (2H, m), 7.37-7.15 (11H, m), 5.04 (4H, m), 4.19 (2H, m), 3.73 (2H, q, J=5.1 Hz); LCMS (ESP): 498 (M+Na+).
Phosphoric acid 45
Figure imgf000081_0001
To an ethanol solution of the phosphate benzyl ester 44 (0.672 g, 1.41 mmol) was added 10% palladium on carbon (134 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 2.5 h and was then filtered through a pad of celite. The filtrate was concentrated to dryness in vacuo to give 415 mg (100% yield) of the compound 45 as a white solid. 1H NMR (CD3OD): δ 8.42 (1H, s), 8.03-7.87 (4H, m), 7.59 (2H, m), 4.20 (2H, q, J=5.7 Hz), 3.73 (2H, t, J=5.4 Hz); LCMS (ESP): 296 (M+H+), 318 (M+Na+); 294 (M-H)", 589 (2M-H)"; Elemental Analysis for (C13H14N05P 0.2H2O) caic: C 52.25, H 4.86, N 4.69; found: C 52.21, H 4.95, N 4.60.
Alcohol 47:
Figure imgf000081_0002
To a methylene chloride solution (5 mL) of D-phenyllactic acid (0.5 g, 3 mmol) at 0 °C was added triethylamine (1 mL) and 2-naphthoyl chloride (0.629 g, 3.3 mmol). After 12 h, the mixture was diluted with EtOAc (80 mL), washed with 5% ice-cold HCI solution (1x50 mL), dried and concentrated. The residue 46 (0.9 g) was dissolved in THF (10 mL). The solution was cooled to -20 °C and 1 M borane in THF solution (3 L, 3 mmol) was added. After stirring the mixture at 25 °C for 6.5 h, MeOH (15 mL) was added. After 15 min, all of the solvent was removed in vacuo. The residue was dissolved in MeOH (15 mL) and stirred for 1 h. The solution was diluted with EtOAc (100 mL), washed with ice-cold 1M Na2C03 solution (2x50 mL) and brine (1x50 L), dried and concentrated. The residue was purified by column chromatography (35% EtOAc in hexanes) to give 300 mg (33% yield) of the compound 47. 1H NMR (CDCI3): δ 8.57 (1H, s), 8.02 (1H, dd, J=8.7, 1.8 Hz), 7.94 (1H, br d, J=7.8 Hz), 7.90-7.79 (2H, m), 7.57 (2H, m), 7.35-7.18 (5H, m), 5.41 (1H, m), 3.90 (1H, dd, J=12.3, 3.3 Hz), 3.80 (1H, dd, J=12.3, 3.6 Hz), 3.12 (2H, m); LCMS (ESP): 329 (M+Na+); 305 (M-H)'. Phosphate Benzyl Ester 48:
Figure imgf000082_0001
To an acetonitrile solution (10 mL) of the alcohol 43 (60 mg, 0.196 mmol) and 1H-tetrazole (70 mg, 1 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (260 mg, 0.75 mmol) at 25 °C. After 3 h, MCPBA (77% pure, 570 mg, 3 mmol) was added to the suspension at 0 °C. The solution was diluted with methylene chloride (30 mL), washed with concentrated NaHS03 solution (2x30 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (1% THF in methylene chloride) to give 80 mg of the compound 48 in 73% yield. 1H NMR (CDCI3): δ 8.57 (1H, s), 8.02 (1H, dd, J=8.4, 1.2 Hz), 7.93-7.77 (3H, m), 7.63-7.46 (2H, m), 7.40-7.17 (15H, m), 5.48 (1H, m), 5.10-4.93 (4H, m), 4.19 (2H, m), 3.11 (1H, dd, J=13.5, 6.3 Hz), 3.01 (1H, dd, J=13.5, 7.2 Hz).
Example 49: Ptaphihalene-2-earbo?ϊyIie acid (R)-2-phenyl-1-phosphonooxymethyl-®thyl ester
Figure imgf000082_0002
To an ethanol solution of the phosphate benzyl ester 48 (80 mg, 0.141 mmol) was added 10% palladium on carbon (25 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was purified by HPLC to give 30 mg (55% yield) of the title compound 49. 1H NMR (CD3OD): δ 8.53 (1H, s), 7.99-7.80 (4H, m), 7.53 (2H, p, J=6.9 Hz), 7.31-7.08 (5H, m), 5.44 (1H, m), 4.24-4.05 (2H, m), 3.08 (2H, d, J=6.9 Hz); LCMS (ESP): 385 (M-H)'; HRMS (MALDl) caic for C20H20O6P (M+H+) 387.0998; found 387.1016. α-Hydroxycarboxylic acid:
Figure imgf000083_0001
D-3-Fluorophenylalanine (7.98 g, 43.8 mmol) was dissolved in 1 M sulfuric acid solution (140 mL). To the solution at 0 °C was slowly added 6 M NaN02 solution (36 mL, 216 mmol) and 3.2 M sulfuric acid (36 mL). The mixture was stirred at 0 °C for 3 h and then at 25 °C for 0.5 h. The solution was extracted with EtOAc (7x75 mL). The combined organic layers were dried and concentrated. Recrystalization from EtOAc/hexanes afforded 5.36 g (67% yield) of the compound α- hydroxycarboxylic acid. 1H NMR (CDCI3): δ 7.29 (1H, m), 7.13-7.0 (2H, m), 6.95 (1H, td, J=8.4, 2.4 Hz), 4.36 (1H, dd, J=7.8, .2 Hz), 3.12 (1H, dd, J=14.1, 4.5 Hz), 2.93 (1H, dd, J=13.8, 7.8 Hz); LCMS (ESP): 183 (M-H)".
Diol 51:
Figure imgf000083_0002
To a THF solution (15 mL) of α-hydroxycarboxylic acid 50 (2.04 g, 11.1 mmol) was added 1 M borane in THF solution (16.6 L, 16.6 mmol). The mixture was stirred at 25 °C for 16 h. The reaction was quenched by addition of MeOH (15 mL). After 1 h, all of the solvent was removed in vacuo. The residue was dissolved in methylene chloride (15 mL). Saturated NaHC03 solution (15 mL) was added to the solution, which was stirred vigorously for 3 h. The mixture was extracted with methylene chloride (2x50 mL). The combined organic layers were dried over Na2S04 and concentrated. The residue was purified by column chromatography (35% to 50% EtOAc in hexanes) to give 969 mg (51% yield) of the compound 51. 1H NMR (CDCI3): δ 7.34-7.22 (1H, m), 7.05-6.89 (3H, ), 3.96 (1H, m), 3.71 (1H, dd, J=11.1 , 3.3 Hz), 3.53 (1H, dd, J=11.7, 6.9 Hz), 2.78 (2H, m).
Phosphate Benzyl Ester 52:
fcs uαπ
Figure imgf000083_0003
To a methylene chloride solution (4 mL) of the diol 51 (215 mg, 1.26 mmol) was added pyridine (1 mL), 4-dimethylaminopyridine (DMAP) (10 mg) and 10% dibenzyl phosphoryl chloride in benzene (8.8 mL, 2.78 mmol). After 17 h, the solution was diluted with methylene chloride (25 mL), washed with 5% HCI solution (1x30 L), dried and concentrated. The residue was purified by column chromatography (35% EtOAc in hexanes) to give 68 mg (13% yield) of the compound 52. 1H NMR (CDCIs): δ 7.43-7.29 (10H, m), 7.28 (1H, m), 7.00-6.84 (2H, m), 5.15-4.98 (4H, m), 4.03-3.78 (3H, m), 2.71 (2H, m); LCMS (ESP): 431 (M+H+), 453 (M+Na+).
Ester 53:
Figure imgf000084_0001
To a methylene chloride solution (2 L) of the alcohol 52 (67 mg, 0.158 mmol) was added triethylamine (1 L) and 1-benzothiophene-2-carbonyl chloride (62 mg, 0.316 mmol). After 3 h, the mixture was concentrated in vacuo. The residue was dissolved in methylene chloride (20 L). The solution was washed with brine (1x20 mL), dried over Na2S04 and concentrated. The residue was purified by column chromatography (5 to 15% EtOAc in hexanes) affording 23 mg (24% yield) of the compound 53. 1H NMR (CDCI3): δ 8.01 (1H, s), 7.84 (2H, d, J=8.4 H∑), 5.43 (2H, m), 7.33-7.17 (12H, m), 7.03-6.85 (3H, m), 5.37 (1H, m), 5.02 (4H, m), 4.12 (2H, m), 3.04 (1H, dd, J=13.8, 6.9.Hz), 2.96 (1H, dd, J=14.1, 6.6 Hz).
Example 54: Phosphoric acid mono- 2-[(1-benso[b3 hiophen-2-yI-methanoyl)-amino3-3-(3- fIuoro-phenyl)-propyl] ester
Figure imgf000084_0002
53 54
To an ethanol solution of the phosphate benzyl ester 53 (23 mg, 0.038 mmol) was added
10% palladium on carbon (5 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 15 h and was then filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was purified by HPLC to give 10 mg (64% yield) of the compound 54. 1H NMR (CD3OD): δ 8.17 (1H, s), 8.00 (2H, m), 7.53 (2H, m), 7.35 (1H, m), 7.24-7.11 (2H, m), 7.01 (1H, td, J=8.7, 2.7 Hz), 5.53 (1H, m), 4.24 (2H, m), 3.20 (2H, m); HRMS (MALDl) caic for C18H1706PS (M+H+) 411.0468; found 411.0480.
Aminoalcohol 55:
Figure imgf000085_0001
To a THF solution (20 mL) of N-methyl-D-phenylalanine (2.5 g, 14 mmol) was added 1 M borane solution in THF (20.9 mL, 20.9 mmol). After 15 h, sat'd NaHC03 solution (5 mL) was added to the solution. The mixture was stirred vigorously for 48 h and then extracted with methylene chloride (3x25 mL). The combined organic layers was dried over MgS04 and concentrated. The residue was purified by column chromatography (95/5/0.5 to 90/10/1 CH2CI2/CH3OH/NH4OH) to give 710 mg (31% yield) of the compound 55. 1H NMR (CDCI3): δ 7.41-7.09 (5H, m), 3.64 (1H, dd, J=10.8, 3.6 Hz), 3.34 (1H, dd, J=10.8, 4.5 Hz), 2.86-2.68 (3H, m), 2.41 (3H, ε).
Alcohol 56:
Figure imgf000085_0002
To a sodium carbonate solution (1 M, 4 mL) was added acetonitrile (4 mL), the aminoalcohol
55 (377 mg, 2.28 mmol), and 2-naphthoyl chloride (435 mg, 2.28 mmol). After 15 h, the mixture was extracted with methylene chloride (3x25 mL). The combined organic layers were dried (MgS04) and concentrated. The residue was purified by column chromatography (50-100% EtOAc in hexanes) to afford 574 mg (79% yield) of the compound 56. 1H NMR (CD3OD): (mixture of two rotamers) δ 3.16 and 2.81 (3H, s); LCMS (ESP): 320 (M+H+), 342 (M+Na+); Elemental Analysis for (C21H21N02) caic: C 78.97, H 6.63, N 4.39; found: C 79.00, H 6.76, N 4.47. Phosphate Benzyl Ester 57:
To an acetonitrile solution (8 mL) of the alcohol 56 (322 mg, 1.01 mmol) and 1H-tetra∑ole
(191 mg, 2.72 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (0.678 mL, 2.02 mmol) at 25 °C. After 3 h, MCPBA (0.523 g, 77% pure, 3.03 mmol) was added to the suspension. The solution was diluted with methylene chloride (35 mL), washed with concentrated NaHS03 solution (2x25 mL), dried over MgS04 and concentrated in vacuo. The residue was purified by column chromatography (30 to 50% EtOAc in hexanes) to give 0.23 g of the compound 57 in 40% yield. 1H NMR (CDCIs): (mixture of two rotamers) δ 5.06 (4H, m), 3.06 and 2.72 (3H, s); LCMS(ESP): 602 (M+Na+).
Example 58: Phosphoric acid mono-f[meϊhyl-(1-nap halen-2-yl-methanoyI)-amino3-phenyl- propyl} ester
Figure imgf000086_0002
To an ethanol solution of the phosphate benzyl ester 57 (224 mg, 0.387 mmol) was added 10% palladium on carbon (45 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 3 h and was then filtered through a pad of celite. The filtrate was concentrated to dryness in vacuo, affording 155 mg (100% yield) of the title compound 58. 1H NMR (CD3OD): (mixture of two rotamers) δ 3.18 and 2.87 (3H, s); LCMS (ESP): 398 (M-H)'. Scheme 4
Figure imgf000087_0002
3-fIuorophenylalanine
Figure imgf000087_0001
Figure imgf000087_0003
Alcohol 59:
Figure imgf000087_0004
3-fluorophenylalanine
Figure imgf000087_0005
To a 3-fluorophenylalanine (2.2 g, 12 mmol) solution in methanol (12 mL) at -30 °C was added thionyl chloride (1 mL). After stirring at 25 °C for 15 h, the reaction mixture was concentrated in vacuo to give 2.2 g of a solid, which was then dissolved in DMF (25 mL). To the solution was added benzimidazole-6-carboxyIic add (2.18 g, 13.44 mmol), EDC (3.22 g, 16.8 mmol), and DMAP (0.237 g, 2.24 mmol). After 15 h, the mixture was diluted with EtOAc (100 mL), washed with ice-cold 5% NaOH solution (1x80 mL) and brine (3x 80 mL), dried (MgS0 ) and concentrated. The resulting crude oil (2 g) was dissolved in THF (10 mL) and the solution was slowly added to LiBH4 (0.52 g, 24 mmol). After 15 h at 25 °C, a solution of NH4CI (1 mL) was added slowly to quench the reaction. The suspension was extracted with EtOAc (3x20 mL). The combined organic layers were washed with brine (2x40 mL), dried (MgS0 ) and concentrated. The residue was purified by column chromatography (5% MeOH in CH2CI2), providing 400 mg (11 % yield) of the compound 59. LCMS: 314 (M+H+, ESP Positive); 312 (M-H', ESP negative). Phosphate Benzyl Ester 60:
Figure imgf000088_0001
To an acetonitrile solution (6 mL) of the alcohol 59 (188 mg, 0.601 mmol) and 1H-tetrazole
(84 mg, 1.20 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (0.302 mL, 0.901 mmol) at 25 °C. After 3 h, MCPBA (404 mg, 77% pure, 1.80 mmol) was added to the suspension. The solution was diluted with EtOAc (20 mL), washed with 5% NaHS03 solution (1x20 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography (98/2/0.2 CH2CI2/CH3OH/NH4OH) and further purified by preparative HPLC to give 132 mg of the compound 60 in 38% yield. 1H NMR (CDCI3): δ 8.82 (1H, s), 7.89 (1H, d, J=8.10 Hz), 7.75 (1H, d, J=7.54 Hz), 7.47 (1 H, d, J=8.67 Hz), 7.37-7.19 (1H, m), 7.00 (1H, d, J=7.35 Hz), 6.91 (2H, d, J=8.85 Hz), 5.14- 5.00 (4H, m), 4.53 (1H, m), 4.23-4.01 (2H, m), 3.08-2.96 (1H, dd, J=13.76, 6.97 Hz), 2.91-2.80 (1H, dd, J=13.94, 8.86 Hz); LCMS: 574 (M+H+); 596 (M+Na+).
Example 61: Phosphoric acid mono-[(R)-2-[(1-3H-benzoimidazol-5-yl-methanoyl)-amino3-3-(3- fluoro-phenyl)-propyl] ester
Figure imgf000088_0002
To a methanol solution of the phosphate benzyl ester 60 (132 mg, 0.230 mmol) was added palladium on carbon (10%, 26 mg). The suspension was kept under hydrogen atmosphere (1 atm) overnight, and was then filtered through a pad of celite. Preparative HPLC purification afforded 11 mg (12% yield) of the title compound 61. 1H NMR (CD3OD): δ 9.46 (1H, s), 8.26 (1H, s), 8.06-7.86 (1H, dd, J=38.24, 8.85 Hz), 7.30 (1H, m), 7.20-7.04 (2H, m), 6.95 (1H, t, J=10.36 Hz), 4.57 (1H, m), 4.13 (2H, m), 3.18-2.94 (2H, m); LCMS: 394 (M+H+); 392 (M-H)"; HRMS (MALDl) caic for Cι7H17FN305PH (M+H+) 394.0962; found 394.0968. Scheme 5
Figure imgf000089_0001
2, 3-Difluoro-DL- 62 phenylalanine 63
Figure imgf000089_0002
Alcohol 62:
Figure imgf000089_0003
62
To a tetrahydrofuran solution (25 mL) of 2,3-difluoro-DL-phenylalanine (2.93 g, 14.6 mmol) was slowly added 1 M borane in tetrahydrafuran (36.5 mL, 36.5 mmol) at 0 °C. The mixture was warmed to room temperature and stirred overnight. Methanol (20 mL) was added and the solution was stirred vigorously for 1 h. The solvent was evaporated and the procedure was repeated. The residue was dissolved in methylene chloride (50 mL) and stirred vigorously with 1M NaHC03 (30 ml) overnight. The mixture was extracted with methylene chloride (3x50 mL). The combined methylene chloride extract was washed with brine (50 mL), dried with Na2S04 , and concentrated. After column chromatography purification (95/5/0.5 CH2CI2/CH30H/NH40H), the compound 62 was obtained in 47% yield (1.28 g). 1H NMR (CH3OD): δ 7.20-7.02 (3H, m), 3.54 (1H, dd, J=10.93, 4.52 Hz), 3.44- 3.35 (1H, m), 3.14-3.05 (1H, ), 2.93-2.84 (1H, m), 2.75-2.65 (1H, m); LCMS: 18 8.0 (M+H+).
Alcohol 63:
Figure imgf000090_0001
63
To a methylene chloride solution (30 mL) of 62 (1.28 g, 6.84 mmol) was added triethylamine
(1.9 mL, 13.7 mmol) and benzyl chloroformate (1.47 L, 10.3 mmol). The mixture was stirred overnight and then concentrated. The residue was dissolved in methylene chloride (30 mL) and washed with brine (1x30 mL). The solution was dried (Na2S0 ) and concentrated. After column chromatography purification (10 to 30% EtOAc in hexane), the compound 63 was obtained in 59% yield (1.30 g). 1H NMR (CDCI3): δ 7.27 (5H, m), 7.03-6.86 (3H, m), 5.00 (2H, s), 3.90 (1H, m), 3.70- 3.47 (2H, m), 2.88 (1H, d, J=6.80 Hz); MS (ESP): 322.1 (M+H+); 344.1 (M+Na+).
Phosphate Benzyl Ester 64:
Figure imgf000090_0002
To an acetonitrile solution (20 mL) of the alcohol 63 (1.30 g, 4.05 mmol) and 1H-tetrazole (765 mg, 10.9 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (2.72 mL, 8.10 mmol) at 25 °C. After 3 h, MCPBA (3.18 g, 77% pure, 14.2 mmol) was added to the suspension. The solution was diluted with EtOAc (80 mL), washed with 5% NaHS03 solution (2x80 mL), dried over MgS0 and concentrated in vacuo. The residue was purified by column chromatography (10-30% EtOAc in hexane) to give 2.16 g of the compound 64 in 92% yield. 1H NMR (CDCI3): δ 7.51-7.38 (15H, m), 7.18-6.93 (3H, m), 5.18-5.08 (6H, m), 4.21-3.93 (3H, m), 2.95 (2H, d, J=6.42 Hz); LCMS: 604.2 (M+Na+). Amino Phosphate 65:
HCI
Figure imgf000091_0001
To a methanol solution of the phosphate benzyl ester 64 (2.16 g, 3.72 mmol) was added palladium on carbon (10%, 300 mg). The suspension was kept under hydrogen atmosphere (1 atm) for 4 h, and was then filtered through a pad of celite. The collected solid was washed with methylene chloride. The mixture of the solid and celite was suspended in 5% HCI solution and stirred for 20 min. After filtration, the filtrate was concentrated to dryness, affording 910 mg of the compound 65 in 92% yield. 1H NMR (DMSO): δ 6.48-6.32 (3H, m), 3.38-3.28 (1H, m), 3.24-3.13 (1H, m), 3.00-2.88 (1H, m), 2.32 (2H, d, J=7.18 Hz); LCMS: 268.0 (M+H*); 266.1 (M-H)".
Example 66: Phosphoric acid mono-{3-(2,3-difTuoro-phenyI)-2-[(1-naphthalen-2-yl-methanoyl)- amino3-propyl} ester
Figure imgf000091_0002
To a sodium carbonate solution (1 M, 5 mL) was added the aminophosphate 65 (226 mg, 0.745 mmol) and 2-naphthyl chloride (142 mg, 0.745 mmol). After 15 h, it was acidified to pH~1 by the addition of 1 M HCI solution at 0 °C. Preparative HPLC purification afforded 135 mg (43% yield) of the title compound 66. 1H NMR (CD3OD): δ 8.31(1H, s), 7.98-7.91 (3H, m), 7.82 (1H, d, J=1.72 Hz), 7.60 (2H, m), 7.20-7.06 (3H, m), 4.82 (1H, m), 4.18 (2H, m), 3.24 (1H, dd, J=13.93, 5.49 Hz), 3.07 (1H, dd, J=13.69, 8.84 Hz); LCMS: 420.2 (M-H)"; Elemental Analysis for (C20H18F2NO5P) caic: C 57.01, H 4.31, N 3.32; found: C 56.80, H 4.55, N 3.27. Example 67: Phosphoric acid mono-[2-[(1-benzo[b]thiophen-2-yl-methanoyl)-amino]-3-(2,3- difluoro-phenyl)-propyI] ester
Figure imgf000092_0001
To a sodium carbonate solution (1 M, 5 mL) was added the aminophosphate 65 (i 9 mg, 0.656 mmol) and benzo[b]thiophene-2-carbonyl chloride (129 mg, 0.656 mmol). After 15 h, it was acidified to pH~1 by the addition of 1 M HCI solution at 0 °C. Preparative HPLC purification afforded 75 mg (27% yield) of the title compound 67. 1H NMR (CD3OD): δ 7.73-7.62 (3H, m), 7.23-7.15 (2H, m), 6.95-6.78 (3H, m), 4.42-4.29 (1H, m), 3.97-3.82 (2H, m), 2.98 (1H, dd, J=13.97, 4.91 Hz), 2.80 (1H, dd, J=13.98, 10.20 Hz); LCMS: 426.0 (M-H"); Elemental Analysis for (C18H16F25PS. 0.20H2O) caic: C 50.16, H 3.84, N 3.25; found: C 50.04, H 3.85, N 3.32.
Scheme 6
Figure imgf000093_0001
(R)-3-Amino-4-(3-fluoro-phenyl)- 68 butyric acid hydrochloride
Figure imgf000093_0002
69 70
Figure imgf000093_0003
Amine 68:
Figure imgf000093_0004
To a stirred solution of lithium borohydride (3 eq) in THF (45 mL) was added chlorotrimethylsilane (10.09 mL, 0.080 mol, 6 eq). The solution was stirred five minutes at room temperature. (R)-3-Amino-4-(3-fluoro-pheπyl)-butyric acid hydrochloride (3.10 g, 13.24 mmol) was added portion wise, and the reaction was stirred overnight at room temperature. The reaction was quenched with methanol, and the solvents were removed in vacuo. The residue was diluted with H20, and the pH was brought to 12 with aqueous NaOH. The product was extracted with chloroform, and the organic phase was separated, washed sequentially with water and brine, dried (MgS04), and the solvent evaporated to give 2.21 g (91%) of a clear oil 68. 1H NMR (benzene-d6): δ 1.10-1.30 (m, 2H), 1.20-1.60 (br, 3H), 1.91 (dd, 1H, J=8.3, 13.3 Hz), 2.18 (dd, 1H, J=5.2, 13.3 Hz), 2.54-2.62 (m, 1H), 3.62-3.70 (m, 1H), 3.73-3.79 (m, 1H), 6.48 (d, 1H, J=7.5 Hz), 6.55-6.59 (m, 1H), 6.69-6.76 (m, 1H), 6.83-6.90 (m, 1H); IR (neat) 1588, 1487, 1449, 1251, 1141, 1065, 783 cm"1; Rf = 0.18 (5% methanolic ammonia/CHCI3); LCMS 184 (M+H).
Alcohol 69:
Figure imgf000094_0001
69
Amine 68 (1.05 g, 5.72 mmol) was dissolved in CH2CI2 (60 mL). Triethylamine (0.88 mL, 6.30 mmol) was added, followed by benzo[b]thiophene-2-carbonyl chloride (1.12 g, 5.72 mmol). The reaction was stirred at room temperature for one hour, and the solvent was removed in vacuo. The product was purified by flash column chromatography eluting with a gradient of 30-60% EtOAc/hexanes (Rf = 0.24 (50% EtOAc/hexanes) to give 1.87 g (91%) of a white solid 69. 1H NMR (DMSO- s): δ 1.68-1.75 (m, 2H), 2.87 (d, 2H, J=7.0 Hz), 3.43-3.52 ( , 2H), 4.20-4.27 (m, 1H), 4.42 (t, 1H, J=5.1 Hz), 6.94-7.00 (m, 1H), 7.03-7.08 (m, 2H), 7.25-7.32 (m, 1H), 7.39-7.46 ( , 2H), 7.90- 8.01 (m, 2H), 8.05 (s, 1H), 8.50 (d, 1H, J=8.5 Hz); LCMS 344 (M+H).
Bromide 70:
Figure imgf000094_0002
Alcohol 69 (1.77 g, 5.14 mmol) was partially dissolved in CH2CI2 (50 mL) and cooled to 0 °C.
Triethyl phosphite (1.77 mL, 10.29 mmol) was added, followed by CBr4 (3.41 g, 10.29 mmol). The ice bath was removed, and the reaction was allowed to warm to room temperature over 5 hours. The reaction was poured into CH2CI2/H20. The organic phase was separated, washed with brine, dried (MgS04) and evaporated. The crude product was purified by flash column chromatography eluting with a gradient of 3-50% EtOAc/hexanes (Rf = 0.18 (10% EtOAc/hexanes) to give 0.45 g (21%) of a white solid 70. H NMR (DMSO-d6): δ 2.05-2.16 (m, 2H), 2.88 (d, 2H, J=6.9 Hz), 3.50-3.59 (m, 2H),
4.25-4.29 (m, 1H), 6.96-7.08 (m, 3H), 7.26-7.33 (m, 1H), 7.40-7.47 (m, 2H), 7.92-8.01 (m, 2H), 8.06
(s, 1H), 8.57 (d, 1H, J=8.5 Hz); LCMS 406, 408 (M+H). Example 7 : ([(R)-3-t(Benzo[b]thiophene-2-carbonyl)-aminoJ-4-(3-fiuoro-phenyl)-butyl]- phosphonic acid ):
Figure imgf000095_0001
Bromide 70 (0.38 g, 0.93 mmol) was suspended in triethyl phosphite (5 mL) and placed in a microwave apparatus for 15 minutes at 150 °C upon which the reaction goes clear. The solvent was removed in vacuo. The crude product was chromatographed by flash silica gel chromatography eluting with a gradient of 0-2% MeOH/CHCI3 giving 0.11 g of the diethyl phosphonate as a clear oil (Rf = 0.24, 3% MeOH/CHCIs). The oil was dissolved in CH2CI2 (3 mL) and treated with bromotrimethylsilane (0.15 L, 1.2 mmol). The reaction was stirred overnight at room temperature. The solvent was removed, and the residue triturated with water. The resuliing white precipitate was filtered, washed with water, and dried to give 0.07 g (17%) of the title compound as a white solid 71. 1H NMR (DMSO-ds): δ 1.49-1.80 (m, 4H), 2.80-2.93 (m, 2H), 4.14-4.19 (m, 1H), 6.94-7.00 (m, 1H), 7.07-7.09 (m, 2H), 7.24-7.32 (m, 1H), 7.40-7.46 (m, 2H), 7.91-8.00 (m, 2H), 8.08 (s, 1H), 8.60 (d, 1H, J=8.5 Hz); HRMS calculated for C19H20NO4PSF 408.0835 (M+H), found 408.0830; Anal. (C19H19N04PSF) C, H, N.
Scheme 7
Synthesis of Examples 72 and 73
Figure imgf000096_0001
Synthesis of Example 74
Figure imgf000096_0002
Alcohol 19a:
Figure imgf000096_0003
To a CH2CI2 solution (40 mL) of D-phenylalaninol (2.26 g, 14.9 mmol) at 0 °C was added triethylamine (3.11 mL, 22.4 mmol) and 2-naphthoyl chloride (3.13 g, 16.4 mmol). After 15 h at 25 °C, the mixture was diluted with CH2CI2 (50 mL), washed with brine (3x50 mL), dried and concentrated. The residue was purified by column chromatography (2% MeOH in CH2CI2) to give 1.33 g (30% yield) of the compound 19a as a white solid. H NMR (CDCI3): δ 8.16 (1H, s), 7.93-7.83 (3H, m), 7.73 (1H, dd, J=8.5, 1.7Hz), 7.55 (2H, m), 7.39-7.23 (5H, m), 6.48 (1H, d, J=9 Hz), 4.43 (1H, m), 3.86 (1H, dd, J=11.1, 3.6 Hz), 3.77 (1H, dd, J=11, 4.9 Hz); Elemental Analysis for (C20H19NO2) caic: C 78.66, H 6.27, N 4.59; found: C 78.41, H 6.37, N 4.52.
Example 72: Sulfamic acid 2-[(1-naphthalen-2-yl-methanoyl)-aminoJ-3-phenyJ-propyl ester
Figure imgf000097_0001
To an acetonitrile (1 mL) solution of chlorosulfonyl isocyanate (136 μL, 1.57 mmol) at 0 °C was added water (28 μL, 1.57 mmol). After stirring for 1.5 h at 0 °C, acetonitrile (1 mL), pyridine (149 μL, 1.57 mmol), and the alcohol (255 mg, 0.836 mmol) were added to the solution. The mixture was stirred at 25 °C for 15 h, diluted with EtOAc (20 mL), and washed with ice-cold 2% HCI solution (1x20mL). Column chromatography (30-50% EtOAc in hexanes) purification gave 19 mg (6% yield) of the title compound 72. 1H NMR (CD3OD): δ 8.29 (1H, S), 8.0-7.89 (3H, M), 7.80 (1H, dd, J=8.7, 1.7 Hz), 7.58 (2H, m), 7.4-7.18 (5H, m), 4.66 (1H, m), 4.29 (2H, m), 3.08 (2H, m); LCMS (ESP): 385 (M+H+), 407 (M+Na+).
Eκample 73: Sulfuric acid mono-{2-[(1-naphthalen-2-yl-methanoyl)-amino3-3-ph@nyl-propyi} ester.
Figure imgf000097_0002
To a CH2CI2 (5 mL) solution of the alcohol (105 mg, 0.344 mmol) at -30 °C was added triethylamine (0.5 mL) and chlorosulfonic acid (116 mg, 90 μl, 1 mmol). After stirring at 25 °C for 15 h, the mixture was diluted with EtOAc (10 mL), washed with ice-cold 2% HCI solution (1x15 mL), dried and concentrated. The residue was purified by column chromatography to afford 131 mg (87% yield) of the title compound 73 as a white solid. 1H NMR (CD3OD): δ 8.34 (1H, s), 7.91 (4H, m), 7.58
(2H, m), 7.4-7.15 (5H, m), 4.59 (1H, m), 4.23 (1H, dd, J=10.5, 4.3 Hz), 4.13 (1H, dd, J=10.6, 5.5 Hz),
3.07 (2H, m); HRMS (MALDl) caic for C20H1BNO5SNa2 (M-H++2Na+) 430.0695; found 430.0676.
Alcohol 47a:
Figure imgf000098_0001
Alcohol 47a was prepared as described in the synthesis of compound 47. In the first step (preparation of 46a), hydroxyl carboxylic acid (760 mg, 4.13 mmol), 2-naphthoyl chloride (866 mg, 4.54 mmol), and triethylamine (2.9 mL) were used. In the second step, 1 M borane in THF (4.73 mL) was used. After column chromatography (40% EtOAc in hexanes) purification, the compound 47a was obtained as a crude oil (137 mg). LCMS: 325 (M+H+).
Benzyl Ester 48a:
Figure imgf000098_0002
48a
Benzyl Ester 48a was prepared as described in the synthesis of 48 using the alcohol 47s (137 mg, 0.423 mmol), 1H-tetrazole (68 mg, 0.973 mmol), dibenzyl N,N-diisopropylphosphoramidite (0.213 mL, 0.634 mmol), and MCPBA (380 mg, 77% pure, 1.69 mmol). After column chromatography purification (10% to 20% EtOAc in hexanes), the compound 48a was obtained as crude oil (330 mg), which was carried forward to the next step.
Example 74: Naphthalene-2-carboxylic acid (R)-1 -(3-fluoro-ben∑yl)-2-phosphonooxy-ethyl ester
Figure imgf000098_0003
Example 74 was prepared as described in the synthesis of 49 using the crude benzyl ester (330 mg) and 10% palladium on carbon (70 mg). Preparative HPLC purification gave 70 mg of the title compound 74 (31% yield from the alcohol). Η NMR (CD3OD): δ 8.56 (1H, s), 8.02-7.8 (4H, m), 7.53 (2H, m), 7.23 (1H, m, 7.06 (2H, m), 6.89 (1H, m), 4.40 (1H, dd, J=12, 3 Hz), 4.26 (1H, dd, J=11.8, 6 Hz), 3.12 (2H, m); LCMS (ESP Negative): 403 (M-H). HRMS (MALDl): caic for C20H19O6FP (M+H+) 405.0903; found 405.0902.
Scheme 8
TBDPS
Figure imgf000099_0001
D-3-Fluorophenylalanine
Figure imgf000099_0002
11 1 H-tetrazole
Figure imgf000099_0003
D-Phenylalaninol 18c:
Figure imgf000099_0004
18c
To a tetrahydrofuraή solution (30 mL) of D-3-FluorophenylaIanine (5.00 g, 27.3 mmol) was slowly added 1 M borane in tetrahydrofuran (68.3 mL, 68.3 mmol) at 0 °C. The mixture was warmed to room temperature and stirred overnight. Methanol (40 mL) was added and stirred vigorously for 1 h. The solvent was evaporated and the procedure was repeated. The residue was dissolved in methylene chloride (100 mL) and stirred vigorously with 1 M NaHC03 (50 ml) overnight. The mixture was extracted with methylene chloride (3x50 mL). The combined methylene chloride extract was washed with brine (50 mL), dried with Na2S04, and concentrated. After column chromatography purification (95/5/0.5 CH2CI2/CH3OH/NH4OH), the compound 18c was obtained in 28% yield (1.29 g). 1H NMR (CD3OD): δ 7.34 (1H, m), 7.09-6.92 (3H, m), 3.54 (1H, dd, J=10.74, 4.33 Hz), 3.39 (1H, dd, J=10.74, 6.60 Hz), 3.13-3.02 (1H, m), 2.83 (1H, dd, J=13.37, 6.21 Hz), 2.62 (1H, dd, J=13.38, 7.73 Hz); LCMS: 170.1 (M+H+).
Amine 75:
Figure imgf000100_0001
To a DMF solution (10 mL) of D-phenylalaninol 18c (1.30 g, 7.70 mmol) was added imidazole (1.05 g, 15.4 mmol) and t-butyldiphenylchlorosilane (TBDPSCI) (2.40 mL, 9.24 mmol). After stirring overnight, the mixture was diluted with ether (80 mL), washed with saturated ammonium chloride (1x50), brine (1x50 mL) and dried (Na2S04). The solvent was removed in vacuo. The residue was purified by column chromatography (95/5 CH2CI2/CH3OH) to afford 2.35 g (74% yield) of the compound 75. *H NMR (CDCI3): δ 7.87 (4H, d, J=6.22 Hz), 7.47-7.34 (6H, m), 7.21 (1H, t, J=7.35 Hz), 6.91 (3H, dd, J=16.58, 9.42 Hz), 3.61 (2H, dd, J=9.98, 4.71 Hz), 3.52 (1H, dd, J=9.98, 6.22 Hz), 3.17-3.08 (1H, m), 2.80 (1H, dd, J=13.55, 5.27 Hz), 2.52 (1H, dd, J=13.38, 8.29 H∑), 2.52 (1H, dd, J=13.38, 8.29 Hz), 1.08 (9H, s); LCMS: 408.2 (M+H+), 430.2 (M+Na+).
Silyl Ether 76:
Figure imgf000100_0002
To a methylene chloride solution (15 mL) of the amine 75 (2.35 g, 5.77 mmol) and triethylamine (1.53 mL, 11.0 mmol) was added a methylene chloride solution (2 mL) of triphosgene (1.03 g, 3.46 mmol). After 2 h, the solution was heated at reflux for 1.5 h, and was then cooled to 25
°C. A methylene chloride solution (25 mL) of the amine 6a (2.19 g, 5.77 mmol) was added. After 15 h, the reaction solution was diluted with CH CI2 (80 mL), washed with brine (2x80 mL), dried (Na2S04) and concentrated in vacuo. The residue was purified by flash column chromatography (5- 20% EtOAc in hexane) to afford 3.92 g (79% yield) of the compound 76. 'H NMR (CDCI3): δ 7.65- 7.58 (4H, m), 7.47-7.33 (6H, m), 7.25-7.18 (4H, m), 7.17-7.07 (7H, m), 6.88-6.82 (3H, m), 5.06 (1H, d, J=8.29 Hz), 5.01-4.94 (1H, m), 4.93-4.88 (1H, m), 4.11-3.97 (1H, m), 3.55 (2H, d, J=3.02 Hz), 3.42-3.30 (1H, m), 3.15-2.81 (3H, m), 2.63-2.49 (4H, m), 2.23-2.13 (1H, m), 1.72-1.46 (10H, m), 1.34-1.20 (4H, m), 1.10 (9H, s).
Alcohol 77:
Figure imgf000101_0001
To a THF solution (30 mL) of the silyl ether 76 (3.92 g, 4.80 mmol) was added 1 M HF/pyridine (7 mL) at 0 °C. The reaction mixture was warmed to 25 °C after 30 minutes and stirred overnight. The THF was removed by vacuum. The residue was dissolved in methylene chloride (50 mL) and washed with cold 1 M HCI (2x50). The solution was concentrated. The resulting residue was purified by column chromatography (30% EtOAc in hexane) to give 2.26 g (82% yield) of the compound 77. 1H NMR (CDCI3): δ 7.33-7.23 (4H, m), 7.22-7.12 (6H, m), 7.06-6.89 (3H, m), 4.99 (1H, m), 4.90 (1H, m), 4.05-3.94 (1H, m), 3.72 (1H, d, J=3.20 Hz), 3.68 (1H, d, J=3.02 Hz), 3.60-3.50 (1H, m), 3.39 (1H, d, J=11.68 H∑), 3.18-3.05 (1H, m), 2.99-2.78 (3H, m), 2.60 (5H, m), 2.21 (1 H, d, J=12.81 Hz), 1.81-1.51 (9H, m), 1.50-1.36 (1H, m), 1.33-1.20 (1H, m).
Phosphate Benzyl Ester 78:
Figure imgf000101_0002
To an acetonitrile solution (35 mL) of the alcohol 77 (2.26 g, 3.94 mmol) and 1H-tetrazole (552 mg, 7.88 mmol) was added dibenzyl N,N-diisopropylphosphoramidite (1.98 L, 5.91 mmol) at 25 °C. After 3 h, MCPBA (2.65 g, 77% pure, 11.8 mmol) was added to the suspension. The solution was diluted with EtOAc (100 mL), washed with concentrated NaHS03 solution (2x80 mL), dried over MgS0 and concentrated in vacuo. The residue was purified by column chromatography (10-30% EtOAc in hexane) to give 2.23 g of the compound 78 in 68% yield. 1H NMR (CDCI3): δ 7.18 (12H, m), 7.09-7.03 (3H, m), 7.03-6.91 (6H, m), 6.81-6.67 (3H, m), 5.57-5.45 (1H, m), 4.95-4.84 (5H, m), 4.84-4.73 (2H, m), 4.00-3.91 (2H, m), 3.81-3.58 (2H, m), 3.43-3.30 (1H, m), 3.00-2.87 (1H, m), 2.83- 2.73 (1H, m), 2.54-2.35 (5H, m), 2.08-1.97 (1H, m), 1.55-1.32 (5H, m), 1.13-1.06 (6H, m).
Example 79: (S)-1 -[(R)-2-(3-Fluoro-phenyl)-1 -phosphonooxymethyl-ethylcarbamoyl]- piperidine-2-carboxyJic acid 4-phenyl-1-(3-phenyl-propyl)-butyl ester
Figure imgf000102_0001
To a methanol solution of the phosphate benzyl ester 78 (400 mg, 0.479 mmol) was added palladium on carbon (10%, 80 mg). The suspension was kept under hydrogen atmosphere (1 atm) overnight, and was then filtered through a pad of celite. The filtrate was purified by preparative HPLC, affording 160 mg of the compound 79 in 51% yield. 1H NMR (CD3OD): δ 7.34-7.19 (5H, m), 7.19-7.09 (6H, m), 7.09-6.88 (3H, m), 5.05-4.95 (1H, m), 4.87-4.81 (1H, m), 4.17-4.06 (1H, m), 3.93 (2H, t, J=5.09 Hz), 3.75-3.65 (1H, m), 3.07-2.90 (2H, m), 2.88-2.76 (1H, m), 2.69-2.51 (4H, m), 2.18 (1H, d, J=13.56 Hz), 1.72-1.51 (11H, m), 1.45-1.14 (2H, m); HRMS (MALDl) caic for C9H15N04P (M+H+) 655.2954; found 655.2958.
Scheme 9: Synthetic Routes to Prodruαs
1. Synthesis of Acetoxymethyl Phospate Ester:
Figure imgf000103_0001
2. Synthesis of Phenyl Phospate Ester:
Figure imgf000103_0002
92 R 1111 = F; R 33 = Et2Nγ KWSJ° Example 80: 1-[1-(Bis-acetoxymethoxy-phosphoryloxymethyI)-2-phenyI-ethylcarbarnoyrj- piperidine-2S-carboxylic acid 4-phenyI-1-(3-phenyl-propyl)-butyl ester
Figure imgf000104_0001
To an acetonitrile solution of the phosphate 16a (10 mg, 0.0158 mmol) at 0 °C was added bromomethyl acetate (15.4 μL, 24.2 mg, 0.158 mmol) and diisopropylethylamine (0.1 mL). After 2 h at 20 °C, the solution was concentrated in vacuo. The residue was purified with column chromatography (deactivated by 1% Et3N in hexanes, eluted with 30% EtOAc in hexanes) to give 5 mg (41% yield) of the title compound 80. 1H NMR (CDCI3): δ 7.36-7.06 (15H, m), 5,64 (4H, m), 5.45 (1H, d, J=8.4 Hz), 4.96 (2H, m), 4.22 (1H, m), 4.03 (2H, m), 3.58 (1H, br d), 3.12 (1H, td, J=12.8, 3.3 Hz), 2.99 (1H, dd, J=13.8, 5.7 H∑), 2.79 (1H, dd, J=13.5, 9 H∑), 2.59 (4H, m), 2.19 (1H, br d), 2.13 (3H, s), 2.11 (3H, s); HRMS (MALDl) caic for C41H53N2011P (M+H+) 803.3279; found 803.3258.
Example 81: Acetic acid acetoxymethoxy-(2-{[1-(1-bromo-naphthalen-2-yI)-methanoyl]- amino}-3-phenyl-propoxy)-phosphoryloxymethyl ester
Figure imgf000104_0002
Example 81 was prepared as described in the synthesis of Example 80 using 25-24 (23 mg,
0.05 mmol), bromomethyl acetate (24.5 μL, 38.2 mg, 0.25 mmol) and diisopropylethylamine (0.1 mL). Purification by Et3N deactivated column chromatography (30% EtOAc in hexanes) gave 3 mg (10% yield) of the title compound 81. 1H NMR (CDCI3): δ 8.33 (1H, d, J=8.4 Hz), 7.83 (2H, m), 7.61 (2H, m), 7.41 (1H, dd, J=8.7 Hz), 7.38-7.21 (5H, m), 6.52 (1H, br d, J=9 Hz), 5.64 (4H, m), 4.68 (1H, m), 4.33 (1H, m), 4.18 (1H, m), 3.13 (1H, dd, J=13.8, 6.9 Hz), 3.02 (1H, dd, J=13.5, 8.4 Hz), 2.06 (3H, s), 2.05 (3H, s). Example 82: Acetic acid acetoxymethoxy-{(R)-2-[3-(2-phenoxy-phenyl)-ureido]-3-phenyl- propoxy}-phosphoryloxymethyl ester
Figure imgf000105_0001
Example 82 was prepared as described in the synthesis of Example 80 using 23b (22 mg, 0.0498 mmol), bromomethyl acetate (24 μL, 0.249 mmol) and diisopropylethylamine (0.05 mL). Purification by Et3N deactivated column chromatography (50% EtOAc in hexanes) gave 9 mg (31 % yield) of the title compound 82. Η NMR (CDCI3): δ 8.22 (1H, dd, J=8.1, 1.5 Hz), 7.36-7.17 (5H, m), 7.14-7.06 (3H, m), 7.01-6.88 (4H, m), 6.82 (1H, dd, J=8.4, 1.8 Hz), 5.65-5.49 (5H, m), 4.28 (1H, m), 4.17 (1H, m), 4.02 (1H, m), 3.01 (1H, dd, J=13.2, 5.7 Hz), 2.81 (1H, dd, J=13.5, 9 Hz), 2.05 (3H, s), 2.04 (3H, s); LCMS (ESP): 609 (M+Na+).
Example 83: Acetic acid acetoj ymethoκy-[2-({1-[5-(3s5-dichIoro-phenoJ y)-furan-2-yl3- methanoyi}-amino)-3-phenyl-propoϊ5y]-phosphorylo2ϊymeth l ester
Figure imgf000105_0002
Example 83 was prepared as described in the synthesis of Example 80 using 25-4 (23 mg, 0.0473 mmol), bromomethyl acetate (23 μL, 0.237 mmol) and diisopropylethylamine (0.049 mL). Purification by Et3N deactivated column chromatography (50% EtOAc in hexanes) gave 6 mg (20% yield) of the title compound 83. *H NMR (CDCfe): δ 7.37-7.17 (6H, m), 7.11 (1H, d, J=3.3 Hz), 7.00 (2H, d, J=1.5 Hz), 6.67 (1H, br d. J=8.7 Hz), 5.76 (1H, d, J=3.3 Hz), 5.62 (4H, m), 4.5 (1H, m), 4.19 (1H, m), 4.09 (1H, m), 3.02 (1H, dd, J=13.5, 6.3 Hz), 2.93 (1H, dd, J=13.8, 8.1 Hz), 2.11 (3H, s), 2.09 (3H, s). Example 84: Acetic acid acetoxymethoxy-{(R)-3-(3-fluoro-phenyl)-2-[(1-naphthalen-2-yI- methanoyl)-amino]-propoxy}-phosphoryloxymethyl ester
Figure imgf000106_0001
Example 84 was prepared as described in the synthesis of Example 80 using 25-28 (50 mg, 0.124 mmol), bromomethyl acetate (61 μL, 0.620 mmol) and diisopropylethylamine (0.13 mL). Purification by Et3N deactivated column chromatography (30-50% EtOAc in hexanes) gave 17 mg (25% yield) of the title compound 84. 1H NMR (CDCI3): δ 8.38 (1H, s), 8.00-7.81 (4H, m), 7.56 (1H, m), 7.37-7.23 (2H, m), 7.11 (1H, d, J=7.8 Hz), 7.05 (1H, d, J=9.9 Hz), 6.95 (1H, td, J=8.7, 2.7 Hz), 5.75-5.57 (4H, m), 4.63 (1H, m), 4.30 (1H, m), 4.16 (1H, td, J=10.8, 3.9 Hz), 3.15 (1H, dd, J=13.5, 6 Hz), 2.99 (13.5, 8.7 Hz), 2.12 (3H, s), 2.06 (3H, s); LCMS (ESP): 548 (M+H*), 570 (M+Na+); HRMS (MALDl) caic for C26H23N0gFP (M+H+) 548.1486; found 548.1489.
Example §5: Aceϊic acid aceto∑cymethθ2ϊy-[(R)-2-[(1-ben2θ[b]thiophen-2-yl-methanoyi)-amino3- 3-(3-fluoro-phenyl)-propoxy]-phosphoryioxymethyl ester
Figure imgf000106_0002
Example 85 was prepared as described in the synthesis of Example 80 using 25-29 (45 mg,
0.11 mmol), bromomethyl acetate (55 μL, 0.55 mmol) and diisopropylethylamine (0.115 mL). Purification by Et3N deactivated column chromatography (30-50% EtOAc in hexanes) gave 39 mg (64% yield) of the title compound 85. 1H NMR (CDCI3): δ 7.92-7.81 (3H, ), 7.48-7.27 (3H, m), 7.09 (1H, br d, J=7.5 Hz), 7.03 (1H, br d, J=9.6 Hz), 6.95 (1H, dd, J=8.7, 2.4 Hz), 5.76-5.59 (4H, m), 4.54 (1H, m), 4.27 (1H, m), 4.13 (1H, td, J=11.1, 3.9 Hz), 3.14 (1H, dd, J=13.6, 6 Hz), 2.96 (1H, dd, J=13.5, 9.3 Hz), 2.15 (3H, s), 2.09 (3H, s); LCMS (ESP): 576 (M+Na+); HRMS (MALDl) caic for C24H26N09FPS (M+H+) 554.1050; found 554.1044. Exa ple 86: Phosphoric acid (R)-2-(5-dimethylamino-naphthalene-1-sulfonylamino)-3-(3- fluoro-phenyl)-propyl ester diphenyl ester
Figure imgf000107_0001
To a THF solution (10 mL) of the alcohol 40 (300 mg, 0.746 mmol) was added Et3N (0.5 mL), DMAP (30 mg) and diphenyl chlorophosphate (0.23 mL, 301 mg, 1.12 mmol). After 15 h, the solution was diluted by EtOAc (50 mL), washed with brine (2x50 mL), dried and concentrated. The residue was purified by column chromatography to give 310 mg (66% yield) of the title compound 86. 1H NMR (CDCI3): δ 8.49 (1H, d, J=8.7 Hz), 8.18 (1H, dd, J=7.5, 1.2 Hz), 8.06 (1H, d, J=8.7 Hz), 7.45 (2H, m), 7.40-7.30 (4H, m), 7.25-7.16 (6H, m), 7.12 (1H, d, J=7.5 Hz), 6.85 (1H, m), 6.60 (1H, td, J=8.4, 2.4 Hz), 6.50 (1H, d, J=7.5 Hz), 6.40 (1H, dt, J=12.3, 1.5 Hz), 5.21 (1H, d, J=8.4 Hz), 4.19 (2H, m), 3.59 (1H, m), 2.87 (6H, s), 2.69 (1H, dd, J=13.5, 7.2 H∑), 2.51 (1H, dd, J=13.8, 7.2 H∑).
Alcohol 88:
Figure imgf000107_0002
To a 1 M Na2C03 solution (5 mL) at 0 °C was added D-3-fluorophenylalanine (0.5 g, 2.73 mmol) and 1-benzothiophene-2-carbonyl chloride (62 mg, 0.316 mmol). After 15 h at 20 °C, the mixture was acidified by addition of ice-cold 5% HCI solution (10 mL). The suspension was extracted with methylene chloride (3x25 mL). The combined organic layers were dried over MgS04 and concentrated to yield 0.6 g of 87 as a white solid. The carboxylic acid 87 was dissolved in THF (5 mL). To the THF solution at 0 °C was added 1 M borane in THF (1.31 mL). After 15 h at 25 °C, a sat'd NaHC03 solution (15 mL) was introduced. The suspension was stirred for 3 h and then extracted with methylene chloride (3x25 mL). The combined organic layers were washed with brine (2x25 mL), dried over Na2S0 , and concentrated. Purification by column chromatography (35% EtOAc in hexanes) gave 200 mg (22.5% yield) of the compound 88. H NMR (CDCI3): δ 7.84 (2H, m), 7.72 (1H, s), 7.41 (2H, m), 7.34-7.25 (1H, m), 7.1 -6.89 (3H, m), 6.41 (1H, br d, J=7.5 Hz), 4.37 (1H, m), 3.77 (2H, m), 3.03 (2H, AB), 2.33 (1H, br s).
Example 89: Phosphoric acid 2-[(1-benzo[b]thiophen-2-yl-methanoyl)-amino]-3-(3-fIuoro- pheny -propyl ester diphenyl ester
Figure imgf000108_0001
Example 89 was prepared as described in the synthesis of Example 86 using the alcohol 88 (40 mg, 0.122 mmol), Et3N (0.1 mL), DMAP (4 mg) and diphenyl chlorophosphate (0.29 μL, 37.6 mg, 0.14 mmol). Column chromatography purification (40% EtOAc in hexanes)' gave 62 mg (97% yield) of the title compound 89. 1H NMR (CDCI3): δ 7.84 (1H, d, J=7.5 Hz), 7.76 (1H, m), 7.66 (1H, s), 7.45-7.32 (5H, m), 7.30-7.13 (8H, m), 7.11-6.98 (2H, m), 6.93 (2H, m), 4.52 (1H, m), 4.40 (1H, m), 4.25 (1H, td, J=11.4, 4.8 Hz), 3.11 (1H, dd, J=13.5, 5.7 Hz), 2.85 (1H, dd, J=13.5, 9.3 Hz); HRMS (MALDl) caic for C3OH2BN05FPS (M+H+) 562.1253; found 562.1279.
Example 90: 1-[1-Bis-acetoxymethoxy-phospgoryloxymethyJ)-2-phenyl-ethylsulfamoyI)3- piperidine-28-carboxylie acid 4-phenyl-foutyl ester
Figure imgf000108_0002
Example 90 was prepared as described in the synthesis of Example 80 using 5b1 (20 mg, 0.036 mmol), bromomethyl acetate (36 μL, 0.36 mmol) and diisopropylethylamine (0.1 mL). Purification by Et3N deactivated column chromatography (40% EtOAc in hexanes) gave 25 mg (100% yield) of the title compound 90. 1H NMR (CDCI3): δ 7.35-7.12 (10H, m), 5.68 (2H, d, J=3 Hz), 5.64 (2H, d, J=2.7 Hz), 5.23 (1H, d, J=9 Hz), 4.61 (1H, d, J=3.g Hz), 4.26-4.01 (4H, m), 3.81 (1H, m), 3.31 (1H, br d), 2.92 (2H, AB), 2.80 (1H, td, J=12.9, 3.6 Hz), 2.65 (2H, m), 2.20 (1 H, br d), 2.13 (6H, s); MS (ESP): 721 (M+Na+); 733 (M+CI)'. Example 91: (S)-1-[(R)-1-(Bis-acetoxymethoxy-phosphoryloxymethyl)-2-phenyl- ethylcarbamoyl]-piperidine-2-carboxylic acid
Figure imgf000109_0001
Example 91 was prepared as described in the synthesis of Example 80 using 16d (60 mg, 0.155 mmol), bromomethyl acetate (0.15 μL, 1.55 mmol) and diisopropylethylamine (0.4 mL, 2.33 mmol). Purification by Et3N deactivated column chromatography (40% EtOAc in hexanes) gave 45 mg (55% yield) of the title compound 91. 1H NMR (CDCI3): δ 7.32-7.07 (5H, m), 5.58 (2H, d, J=1.8 Hz), 5.54 (2H, d, J=1.8 Hz), 4.68-4.50 (2H, m), 4.22 (1H, m), 3.98 (1H, dd, J=13.8, 5.1 Hz), 3.55 (1H, dd, J=12, 4.2 Hz), 3.16 (1H, dd, J=13.8, 9.3 Hz), 3.02 (1H, dd, J=13.8, 6 Hz), 2.69 (1H, td, J=13.2, 3.3 Hz), 2.10 (3H, s), 2.10 (3H, s).
Example 92: Acetic acid aceto2iymetho∑^,-[(R)-2-[(7-diethylamϊno-2-oπo-2H-chromene-3- carbonyl)-amino]-3-(3-fluoro-phenyl)-propoxy3-phosphoryloxymethyl ester
Figure imgf000109_0002
Example 92 was prepared as described in the synthesis of Example 80 using Example 25-33
(18 mg, 0.0366 mmol), bromomethyl acetate (0.03 mL, 0.3 mmol) and diisopropylethylamine (0.1 mL, 0.6 mmol). Purification by Et3N deactivated column chromatography (100% EtOAc in hexanes) gave 5 mg (25% yield) of the title compound 92. 1H NMR (CDCI3): δ 9.05 (1 H, d, J=8.3 Hz), 8.64 (1H, s), 7.41 (1H, t, J=9.1 Hz), 7.26 (1H, m), 7.08 (1H, d, J=7.5 Hz), 7.02 (1H, br d, J=9.8 Hz), 6.92 (1H, td, J=8.3, 2.1 Hz), 6.64 (1H, dd, J=9.3, 2.7 Hz), 6.49 (1H, d, J=2.2 Hz), 5.67 (1H, dd, J=13.6, 0.9 Hz), 4.55 (1H, br s), 4.17 (2H, m), 3.46 (4H, q, J=7 Hz), 3.01 (1H, d, J=7.3 Hz), 2.13 (6H, s), 1.24 (6H, t, J=7.2 Hz). Example 93: Acetic acid acetoxymethoxy-[3-[(benzo[6]thiophene-2-carbonyl)-amino3-4-(3- fIuoro-phenyl)-butylJ-phosphinoyloxymethyl ester
Figure imgf000110_0001
Example 93 was prepared as described in the synthesis of Example 80 using Example 71 (22 mg, 0.0541 mmol), bromomethyl acetate (0.05 mL, 0.52 mmol) and diisopropylethylamine (0.13 mL, 0.77 mmol). Purification by flash column chromatography (100% EtOAc in hexanes) gave 23 mg (77% yield) of the title compound 93. 1H NMR (CDCI3): δ 7.88-7.79 (3H, m), 7.41 (2H, m), 7.27 (1H, m), 7.02 (1H, d, J=7.6 Hz), 6.94 (2H, m), 6.80 (1H, d, J=8.6 Hz), 5.71-5.55 (4H, m), 4.38 (1H, br t, J=7.3 Hz), 3.05 (1H, dd, J=13.7, 6.1 Hz), 2.87 (1H, dd, J=13.7, 7.1 Hz), 2.12 (3H, s), 2.05 (3H, s).
Scheme 10
Synthesis of Examples and 99
Figure imgf000110_0002
Synthesis of Example 100
Figure imgf000111_0001
Phosphonate Ester 95:
Figure imgf000111_0002
To a dry CH2CI2 (20 mL) suspension of polymer-supported triphenylphosphine (1.69 g, 4.53 mmol) was added iodine (1.15 g, 4.53 mmol). After 15 min, imidazole (0.33 g, 5.15 mmol) was added. The suspension was stirred for another 15 min. A CH2CI2 (8 mL) solution of 19c (600 mg, 2.06 mmol) was added. The mixture was heated at reflux for 1 h. After cooling the mixture to 25 °C, the polymeric solid was filtered off. The filtrate was washed with Na2S203 solution (concentrated, 2x30 L), water (1x25 L) and brine (1x25 mL). All the solvent was removed in vacuo, affording 914 mg (100%) of the iodide 94 as a yellow solid. A portion of the iodide 94 (420 mg) was mixed with triethyl phosphite (2.5 mL) in a sealed tube. The suspension was heated at 150 °C for 30 min by microwave radiation. The triethyl phosphite was removed in vacuo. The residue was purified by column chromatography (30-50% EtOAc in hexanes) to give 80 mg (19 % yield) of the phosphonate 95 as a colorless oil. 1H NMR (CDCI3): δ 7.32-7.10 (5H, m), 6.95-6.82 (4H, m), 5.43 (1H, d, J=6 Hz), 5.01 (2H, s), 4.12-3.92 (4H, m), 3.01-2.75 (2H, m), 1.92 (2H, ), 1.28-1.13 (6H, m); LCMS (positive APCI): 424 (M+H+), 446 (M+Na+).
Amine 96:
Figure imgf000111_0003
96
To an ethanol solution (5 mL) of compound 95 (235 mg, 0.556 mmol) was added palladium on carbon (10%, 40 mg). The suspension was kept under hydrogen (1 atm) for 15 h. After filtration, the filtrate was concentrated. The residue was purified by column chromatography (MeOH/CH2CI2 5/95) to afford 148 mg (92% yield) of the compound 96 as an oil. 1H NMR (DMSO-d6): δ 7.4-7.0 (4H, m), 4.05-3.90 (4H, m), 3.25 (1H, m), 2.78 (1H, dd, J=13.4, 6.1 Hz), 2.69 (1H, dd, J=12.2, 6.1 Hz), 2.06 (2H, br s), 1.9-1.70 (2H, m). LCMS (positive APCI): 290 (M+H+), 312 (M+Na+).
Amide 97:
Figure imgf000112_0001
To a methylene chloride solution (5 mL) of the amine 96 (144 mg, 0.498 mmol) was added triethylamine (0.139 mL, 0.996 mmol), 4-(dimethylamino)pyridine (6 mg, 0.0498 mmol), and 1- benzothiophene-2-carbonyl chloride (123 mg, 0.623 mmol) at 0 °C. After 15 h at 25 °C, the mixture was diluted with methylene chloride (20 mL), washed with ice-cold HCI solution (1 M, 1x20mL), sodium carbonate solution (1 M, 1x20mL), and brine (1x20mL). The solution was then dried (Na2S04) and concentrated. The residue was purified by column chromatography (30-50% EtOAc in hexanes) to give 150 mg (67% yield) of the compound 97 as a white solid. 1H NMR (CDCI3): δ 7.85- 7.65 (3H, m), 7.33 (2H, m), 7.21-7.15 (1H, m), 7.03 (1H, d, J=7.7 Hz), 6.96 (1H, br d, J=9.9 Hz), 6.88 (1H, td, J=8.1, 1.9 Hz), 4.51 (1H, d, J=2.1 Hz), 4.16-3.97 (4H, m), 3.17 (1H, dd, J=12, 5.1 Hz), 2.91 (1H, dd, J=13.4, 8.9 Hz), 2.02 (1H, d, J=4.7 Hz), 1.98 (1H, d, J=5.3 Hz), 1.33 (3H, t, J=6.9 Hz), 1.73 (3H, t, J=7.1 H∑); LCMS (ESP): 450 (M+H*), 472 (M+Na+); 448 (M-H).
Example 98: [2-[(Benzo[b]thiophene-2-carbonyl)-amino]-3-(3-fluoro-phenyl)-propyl]- phosphonic acid
Figure imgf000112_0002
To a methylene chloride solution (2 mL) of phosphonate ester 97 (140 mg, 0.31 mmol) was added bromotrimethylsilane (1 mL). After 15 h, the solution was concentrated in vacuo. The oily residue was triturated with water (3x2mL). In the process, a white solid 98 (110 mg, 89% yield) was obtained by filtration. 1H NMR (DMSO-d6): δ 8.66 (1H, d, J=8.4 Hz), 8.1-7.92 (3H, m), 7.45 (2H, m), 7.32 (1H, q, J=8 Hz), 7.12-7.0 (3H, m), 4.42 (1H, m), 3.14 (1H, dd, J=13.7, 2.1 Hz), 2.94 (1H, dd, J=13.4, 8.1 Hz), 1.92 (2H, m); LCMS (positive APCI): 394 (M+H+), 416 (M+Na+); Elemental Analysis for (CH17N04PFS 0.3H2O) caic: C 54.21, H 4.45, N 3.51; found: C 54.15, H 4.47, N 3.47. Example 99: Acetic acid acetoxymethoxy-[2-[benzo[ύ]thiophene-2-carbonyl)-amino]-3-(3- fluoro-phenyl)-propyl]-phosphinoyloxymethyl ester
Figure imgf000113_0001
To an acetonitrile solution (1 L) of the phosphonate acid 98 (31.8 mg, 0.0809 mmol) was added diisopropylethylamine (0.127 mL, 0.728 mmol) and bromomethyl acetate (60 μL, 0.607 mmol) at 0 °C. After 15 h at 25 °C, the solution was concentrated and the resulting residue was purified by column chromatography (50-70% EtOAc in hexanes), affording 26 mg (59% yield) of the title compound 99 as a white solid. 1H NMR (CD3OD): δ 7.85-7.75 (3H, m), 7.33 (2H, m), 7.19 (1H, q, J=8 Hz), 6.99 (1H, d, J=7.5 Hz), 6.94 (1H, dt, J=7.6, 2.2 Hz), 6.84 (1H, td, J=8.3, 2.5 Hz), 5.6-5.45 (4H, m), 4.50 (1H, m), 2.93 (2H, m), 2.35-2.16 (2H, m), 1.94 (3H, s), 1.94 (2H, s); HRMS (MALDl) caic for C24H26N08FPS (M+H+) 538.1129; found 538.1101.
Example 100: 2,2-Dimethyl-propionic acid [3-[(foen2:oE&3thiophene-2-carfoonyI)-amino3-4-(3- fluoro-phenyl)-butyl3-(2,2-dimethyl-propionylθ5cymethoxy)-phosphinoyloκymethyl ester
Figure imgf000113_0002
To an acetonitrile solution (5 mL) of Example 71 (50 mg, 0.123 mmol) was added tetrabutylamonium iodide (5 mg), diisopropylethylamine (0.2 mL), and chloromethyl pivalate (132 μL, 0.92 mmol) at 0 °C. The solution was heated at 60 °C for 4 h and concentrated in vacuo. The residue was purified by column chromatography (35% EtOAc in hexanes), affording 20 mg (26% yield) of the title compound 100. 1H NMR (CD3OD): δ 7.81-7.75 (3H, ), 7.32 (2H, ), 7.16 (1H, q, J=8 Hz), 6.97 (1H, d, J=7.8 Hz), 6.92 (1H, dt, J=10.1, 2.3 Hz), 6.81 (1H, td, J=8.3, 2Hz), 5.6-5.48 (4H, m), 4.24 (1H, m), 2.84 (2H, m), 1.95-1.7 (4H, m), 1.09 (9H, s), 1.04 (9H, s); LCMS (positive APCI): 636 (M+H+); HRMS (MALDl) caic for C31H40NO8FPS (M+H+) 636.2196; found 636.2182. Bioloqical Testing: Pin1 Peptidyl-Prolyl Isomerase Assay
PIN1 is a phosphorylation dependent peptidyl-prolyl isomerase. The PIN1 assay is a spectrophotometric assay based on the coupled chymotrypsin or subtilisin catalyzed, cis-trans conformation dependent cleavage of a para-nitroanaline containing peptide substrate. This improved general rotamase assay was first described by Kofron, et al. (Biochemistry, 30, 6217-6134 (1991)) and applied to PIN1 isomerase activity by Yaffe, et al. (Science, 278, 1957-1960 (19g7)). Cleavage of the isomerized peptide releases para-nitroanaline, which can be monitored by an increase in absorbance at 390 nm. The PIN1 peptide substrate, succinyl-leucine-proline-phenylalanine- paranitroaniline (Suc-AEPF-pNA) (Bachem), is kept in a predominantly cis conformation with an anhydrous TFE LiCI solvent mixture.
Upon dilution into an aqueous assay mixture containing PIN1 , the peptide substrate undergoes PIN1 catalyzed isomerization to the trans conformation. Chymotrypsin or subtilisin (subtilisin Carisberg protease, available from Sigma, catalog number P-5380) cleaves the trans product to form free para-nitroanaline. To minimize the spontaneous isomerization of the peptide substrate, reactions are performed at 15 °C. A typical reaction contains 25 mM MOPS pH 7.5, 0.5 mM TCEP, 2% DMSO, 5 μl of a 25 mg/ml solution of subtilisin Carisberg, 50 nM PINI-PPiase, and 100 μM Suc-AEPF-pNA peptide substrate. Reactions are cooled to 15 °C and initiated with the addition of Suc-AEPF-pNA. The absorbance at 390 nm is monitored continuously until all substrate has been converted to the cleaved product. This data, the progress curve, is then fitted to an exponential equation to determine a rate constant k for the reaction. The rate constant k is linearly proportional to the concentration of active enzyme present in the assay mixture once the rate constant for the spontaneous isomerization is subtracted. The m for this substrate is much higher than 100 μM ([S3«Km). Therefore, during inhibition experiments, the IC50, for non-tight binding inhibitors, is essentially the inhibition constant K|.
In Table 1, the Ki data reported under the PIN1-CD heading corresponds to testing with PIN1 peptide containing the catalytic peptidyl-prolyl isomerase domain but devoid of the PIN1 WW domain. Similarly, the dissociation constant (Ka) data under PIN1-CD refers to testing with a peptide containing the catalytic PIN1 domain but devoid of the PIN1 WW domain.
Table 1
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
The exemplary compounds described above may be formulated into pharmaceutical compositions according to the following general examples.
Example 1: Parenteral Composition
To prepare a parenteral pharmaceutical composition suitable for administration by injection, 100 mg of a water-soluble salt of a compound of Formula I is dissolved in DMSO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
Example 2: Oral Composition
To prepare a pharmaceutical composition for oral delivery, 100 mg of a compound of Formula I is mixed with 750 mg of lactose. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration.

Claims

What is claimed is:
1. A compound of the Formula I :
Figure imgf000117_0001
Formula I
wherein n is 1 or 2; A is a divalent -CH=CH-, -(CrC7-alkyl)-Y-, -NR _άα(,CH2), -Y-, -Y-(CrC7-aIkyl)-, -Y-(C,-C7 alkyl)-, -Y-NH-, -Y-NRd(CrC6-alkyl)-, -S-, -S(0)2-, -0-Y-, -Y-0-, -Y-S-, or -S-Y-, wherein Rd is H or CrC6 alkyl, t is an integer from 0 to 5, Y is C(O), C(S), S(O), S(0)2, or a bond; X is a direct bond, CH2, CF , O, S, NH, C(O), or C(S);
R1 is a Cs-Cto cycloalkyl, 4-10 membered heterocycloalkyl, C6-Cιo aryl, or 4-10 membered heteroaryl group, wherein R1 is unsubstituted or substituted with 1 to 4 R10 groups;
R2 is -S(0)2OH, -S(0)2NRdR°, or -P(0)(OR4)2, wherein R4 is an H, C C10-aIkyl, C6-C10 aryl, or -CH2-0-C(0)ReCH3 group, Rd and Re are each independently an H or CrC6 alkyl group, and R4 is unsubstituted or substituted with 1 to 4 R10 groups; and
R3 is OH, CrC7-aikyl, CrC7-aIkoxyl, C6-G10 aryl, 4-10 membered heteroaryl, C3-C10 cycloalkyl, 3-10 membered heterocycloalkyl, -NH(R5), or -N(R5)2 group, wherein R5 is independently selected from H, CrC7 alkyl, C6-C10 aryl, or
Figure imgf000117_0002
wherein ring B is a 5- or 6-membered heterocycloalkyl group, Z is a divalent C(0)Z', heteroaryl or heterocycloalkyl group wherein Z' is a divalent O, S, NH, N(CH3), C02, or CH2, and RB is H, C C10 alkyl, aryl, CrCβ alkyl-aryl, or arylalkyl group, wherein R3, R5, B and R6 are unsubstituted or substituted with 1 to 4 R10 groups; wherein each R10 is independently selected from halo, amino, =0, =S, =NH, cyano, nitro, hydroxyl, -SH, haloalkyl, 2-10 membered heteroalkyl, CrC6 alkoxy, Cι.C10 alkyl, C2.C6 alkenyl, C2-C6 alkynyl, -C(0)jRa, -OC(0)jRd, -OC(0)OC(0)Rd, -OOH, -C(NRd)NRfaRc, -NRdC(NRe)NRbRc, -NRdC(0)jRb, -C(0)NRbRc, -C(0)NRdCOR , -OC(0)NRbRc, -NRbRc, -NRdORc, -C(S)NRbRc, -NRdC(S)NRbRc, -NR C(0)NRbRc, -OSH, -S(0)jRb, -OS(0)jRb, -SC(0)Rb, -S(0)jC(0)ORb, -SCORd, -NRdSRc, -SRb, -NHS(0)jRb, -COSRb, -C(0)S(0)jRb, -CSRb, -CS(0)jRb, -C(SO)OH, -C(SO)2OH, -NRdC(S)Rc, -OC(S)Rb, -OC(S)OH, -OC(SO)2Rb, -S(0)jNRbRc, -SNRbRc, -S(0)NRbRc, -NRdCS(0)jRc, -C(0);(CH2)tNRd-(4-10 membered heteroaryl), -C(0)j(CH2),NRd(4-10 membered heterocycloalkyl), -(CRdRe),CN, -(CRd Re)t(C3-C10 cycloalkyl), -(CRdRe)f(C6-C10 aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), -(CR Re)t(4-10 membered heteroaryl), -(CRdRe)qC(0)(CRdRe),(C3-C1D cycloalkyl), -(CRdRe)qC(O)(CRdRe),(C6-C10 aryl),
-(CRdR6)qC(O)(CRdRe),(4-10 membered heterocycloalkyl), -(CRdRe)qC(O)(CRdRe),(4-10 membered heteroaryl), -(CRdRe)tO(CRdRe)q(C3-C10 cycloalkyl), -(CRdRe)tO(CRdRe)q(C6-C10 aryl), -(CRdRe)tO(CRdRe)q(4-10 membered heterocycloalkyl), -(CRdRe)tO(CRdRe)q(4-10 membered heteroaryl), -(CRdRe)qSO2(CRdRe),(C3-C10 cycloalkyl), -(CRdR6)qSO2(CRdRe)f(C6-C10 aryl), -(CRdRe)qSO2(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRe)qSO2(CRdRe),(4-10 membered heteroaryl), wherein Ra is selected from the group consisting of halo, hydroxyl, -NRdRe, C Cιo alkyl, haloalkyl, CrC6 alkoxyl, Rb and Rc are independently selected from H, C C10 alkyl, -(CRdRe),(C3-C10 cycloalkyl), -(CRdRe),(C6-C10 aryl), -(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRs)((4-10 membered heteroaryl), Rd and Re are independently H or C Ge alkyl, j is an integer from 0 to 2, q and t are each independently an integer from 0 to 5, and 1 or 2 ring carbon atoms of the cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with =0, and the alkyl, alkenyl, alkynyl, aryl and cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with 1 to 3 substituents independently selected from halo, =0, cyano, nitro, -(CRdRe)tCN, haloalkyl, 2-10 membered heteroalkyl, -OR , -C(0)jR , -NRdC(0)Rb, -C(0)NRbR°, -NRbRc, -NR ORc, -NR^^NR^0, -NRdC(0)jRbRc, -OC(0) R , -OC(0)NRbRc, -SRri, CrC10 alkyl, C2-C6 alkenyl, C2-C3 alkynyl, -(CRdRe)t(C3-C10 cycloalkyl), -(CRdRe)t(C3-C10 aryl), -(CRdRs),(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl), -(CRdRe)t(C6-C10 aryl)-(CrC6 alkyl); wherein t, Rb, Rc, Rd, Rsare as defined above; or a pharmaceutically acceptable prodrug of said compound, pharmaceutically active metabolite of said compound, or pharmaceutically acceptable salt of said compound or metabolite.
2. A pharmaceutically acceptable salt according to claim 1.
3. A compound or pharmaceutically acceptable salt according to claim 1 , wherein: n is 1 or 2; A is a divalent -NH-Y-, -NRd(CH2),-Y-, or -0-Y-, and Y is C(O) or S(0)2;
X is a direct bond, CH2, O, or S;
R1 is a C6-Cιo aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R1D groups;
R2 is -S(0)2OH, or -P(0)(OR4)2> wherein R4 is an H, CrC10 alkyl, or C6-C10 aryl group, and is unsubstituted or substituted with 1 to 4 R10 groups; and R3 is a C6-Cιo aryl, 4-10 membered heteroaryl, -NH(C6HS), or
Figure imgf000119_0001
wherein ring B is a 5- or 6-membered heterocycloalkyl group, Z is a divalent C(0)Z', heteroaryl or heterocycloalkyl group wherein Z' is a divalent O, S, NH, N(CH3), C02, or CH2, and R6 is H or a C C10 alkyl group, wherein R3, B, and R6 is unsubstituted or substituted with 1 to 4 R10 groups; wherein each R10 is independently selected from halo, amino, =0, =S, =NH, cyano, nitro, hydroxyl, -SH, haloalkyl, 2-10 membered heteroalkyl, Cι-C6 alkoxy, C1.C10 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, -C(0)jRa, -00(0)^", -OC(0)OC(0)Rd, -OOH, -C(NRd)NRbRc, -NRdC(NRe)NRbRc, -NRdC(0)jRb, -C(0)NRbRc, -C(0)NRdCORb, -OC(0)NRbRc, -NR Rc, -NRdORc, -C(S)NRbRc, -NRdC(S)NRbRc, -NRdC(0)NRbRc, -OSH, -S(0)jR , -OS(0)jRb, -SC(0)R , -S(0)j-C(0)OR , - SCORd, -NRdSRc, -SRb, -NHS(0)jRb, -COSR , -C(0)S(0)jR , -CSRb, -CS(0)jRb, -C(SO)OH, -C(SO)2OH, -NRdC(S)Rc, -OC(S)R , -OC(S)OH, -OC(SO)2Rb, -S(0)jNRbR°, -SNRbRc, -S(0)NR Rc, -NRdCS(0)jRc, -C(O)j(CH2)tNRd-(4-10 membered heteroaryl), -C(O)j(CH2)tNR (4-10 membered heterocycloalkyl), -(CRdRe)tCN, -(CRd Re)t(C3-C10 cycloalkyl), -(CRdRe)t(C6-C10 aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl), -(CRdRe)qC(O)(CRdRe),(C3-C10 cycloalkyl), -(CRdRe)qC(O)(CRdRe),(C6-C10 aryl), -(CRdRe)qC(O)(CRdRsM4-10 membered heterocycloalkyl), -(CR Re)qC(O)(CRdRe)t(4-10 membered heteroaryl), -(CRdRe)tO(CRdRe)q(C3-C10 cycloalkyl), -(CRdRΘ),O(CRdRs)q(C6-C10 aryl), -(CRdRe)ιO(CRdRe)q(4-10 membered heterocycloalkyl), -(CRdRe)tO(CRdRe)q(4-10 membered heteroaryl), -(CRdRe)qSO2(CRdRe)t(C3-C10 cycloalkyl), -(CRdRe)qSO2(CRdRe)t(C6-C10 aryl), -(CRdRe)qSO2(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRe)qSO2(CRdRe),(4-10 membered heteroaryl), wherein R8 is selected from the group consisting of halo, hydroxyl, -NRdRe, C1-C10 alkyl, haloalkyl, CrC6 alkoxyl, Rb and R° are independently selected from H, C C10 alkyl, -(CRdRe)t(C3-C10 cycloalkyl), -(CRdRe)t(C6-C10 aryl), -(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRe)t(4-10 membered heteroaryl), Rd and Re are independently H or CrCe alkyl, j is an integer from 0 to 2, q and t are each independently an integer from 0 to 5, and 1 or 2 ring carbon atoms of the cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with =0, and the alkyl, alkenyl, alkynyl, aryl and cyclic moieties of the foregoing R 0 groups are unsubstituted or substituted with 1 to 3 substituents independently selected from halo, =0, cyano, nitro, -(CRdRe)tCN, haloalkyl, 2-10 membered heteroalkyl, -0Rb, -C(0)jRb, -NRdC(0)R , -C(0)NRbR°, -NRbRc, -NRbORc, -NRdC(0)jNRbRc, -NRdC(0)jRbRc, -OC(0)jRb, -OC(0)NR Rc, -SRd, CrC10 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, -(CR Re),(C3-C10 cycloalkyl), -(CRdRe)t(C6-Cιo aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl), -(CR Re),(C6-C10 aryl)-(CrC_ alkyl); and wherein t, Rb, Rc, Rd, Re are as defined above.
4. A compound or pharmaceutically acceptable salt according to claim 3, wherein: n is i;
A is a divalent -NH-Y- or-O-Y-, wherein Y is C(O); X is a direct bond, CH2, or O;
R1 is a C6-Cιo aryl group unsubstituted or substituted with 1 to 4 R10 groups; R2 is -P(0)(OR )2, wherein R4 is an H, CrC10 alkyl, or C6-C10 aryl group, and is unsubstituted or substituted with 1 to 4 R1D groups; and R3 is a C6-C10 aryl, 4-10 membered heteroaryl, or
Figure imgf000120_0001
wherein ring B is an unsubstituted 6-membered heterocycloalkyl, Z a divalent C(0)Z', Z' is a divalent O, S, or CH2, and R6 is a CrC 0 alkyl group, wherein R3, B and R6 are unsubstituted or substituted with 1 to 4 R10 groups; wherein each R10 is independently selected from halo, amino, =0, =S, =NH, cyano, nitro, hydroxyl, -SH, haloalkyl, 2-10 membered heteroalkyl, C C6 alkoxy, C1.C10 alkyl, C2.Cβ alkenyl, C2-C6 alkynyl, -C(0)jRa, -OC(0)jRd, -OC(0)OC(0)Rd, -OOH, -C(NR )NRbRc, -NR C(NRe)NR Rc, -NRdC(0)jRb, -C(0)NRbRc, -C(0)NR CORb, -OC(0)NRbRc, -NRbR°, -NRdOR°, -C(S)NR Rc, -NRdC(S)NRbRc, -NRdC(0)NRbR°, -OSH, -S(0)jRb, -OS(0)jRb, -SC(0)Rb, -S(0)jC(0)ORb, - SCORd, -NRdSR°, -SRb, -NHS(0)jR , -COSRb, -C(0)S(0)jR , -CSR , -CS(0)jR , -C(SO)OH, -C(SO)2OH, -NRdC(S)Rc, -OC(S)R , -OC(S)OH, -OC(SO)2Rb, -S(0)JNR Rc, -SNRbRc, -S(0)NRbRc, -NRdCS(0)jRc, -C(O)j(CH2),NRd-(4-10 membered heteroaryl), -C(O)j(CH2),NRd(4-10 membered heterocycloalkyl), -(CRdRβ)tCN, -(CRd Re)t(C3-C10 cycloalkyl), -(CRdRe),(C6-C10 aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl), -(CRdR C(O)(CRdRe),(C3-C10 cycloalkyl), -(CRdRβ)qC(O)(CRdRe)t(CB-C10 aryl),
-(CRdRe)qC(O)(CRdRβ),(4-10 membered heterocycloalkyl), -(CRdRe)qC(O)(CRdRe)t(4-10 membered heteroaryl), -(CRdRe)tO(CRdRo)q(C3-C10 cycloalkyl), -(CRdRe)tO(CRdRe)q(C6-C10 aryl), -(CRdRe),O(CRdRe)q(4-10 membered heterocycloalkyl), -(CRdRe)tO(CRdRe)q(4-10 membered heteroaryl), -(CRdRe)qSO2(CRdRe)t(C3-C10 cycloalkyl), -(CR Re)qSO2(CRdRe),(C6-C10 aryl), -(CRdRe)qSO2(CRdRe)t(4-10 membered heterocycloalkyl), and -(CRdRe)qSO2(CRdRe),(4-10 membered heteroaryl), wherein Ra is selected from the group consisting of halo, hydroxyl, -NRdRe, CrC10 alkyl, haloalkyl, C C6 alkoxyl, Rb and Rc are independently selected from H, C C10 alkyl, -(CRdRe)t(C3-C10 cycloalkyl), -(CRdRe),(CB-C10 aryl), -(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRe)t(4-10 membered heteroaryl), Rd and Re are independently H or Cι-C6 alkyl, j is an integer from 0 to 2, q and t are each independently an integer from 0 to 5, and 1 or 2 ring carbon atoms of the cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with =0, and the alkyl, alkenyl, alkynyl, aryl and cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with 1 to 3 substituents independently selected from halo, =0, cyano, nitro, -(CRdRe)tCN, haloalkyl, 2-10 membered heteroalkyl, -ORb, -C(0)jRb, -NRdC(0)R , -C(0)NRbR°, -NRbRc, -NRb0Rc, -NRdC(0)jNRbRc, -NRdC(0)jRbR°, -OC(0)jRb, -0C(0)NRbRc, -SRd, d-Cn, alkyl, C2-C6 alkenyl, C2-C6 alkynyl, -(CRdRe),(C3-C10 cycloalkyl), -(CRdRe),(C6-C10 aryl), -(CRdRe),(4-10 membered heterocycloalkyl), -(CRdRe),(4-10 membered heteroaryl), -(CRdRB)t(CB-C10 aryl)-(CrC6 alkyl); and wherein t, Rb, Rc, R , Re are as defined above.
5. A compound or pharmaceutically acceptable salt according to claim 4, wherein: n is 1 ;
A is -NH-Y- or -0-Y-, wherein Y is C(O); X is a direct bond, CH2, or O;
R1 is a C6-C1Qaryl group unsubstituted or substituted with 1 to 4 R10 groups; R2 is -P(0)(OR4)2, wherein R4 is an H or a C1-C10 alkyl group that is unsubstituted or substituted with 1 to 4 R10 groups; and
R3 is a C3-C10 aryl or 4-10 membered heteroaryl group unsubstituted or substituted with 1 to 4 R10 groups; wherein each R1D is independently selected from halo, amino, =0, =S, =NH, cyano, nitro, hydroxyl, -SH, haloalkyl, 2-10 membered heteroalkyl, C C6 alkoxy, C^Cio alkyl, C2-C6 alkenyl,
C2-C5 alkynyl, -C(0)jRa, -OC(0)jRd, -OC(0)OC(0)Rd, -OOH, -C(NRd)NRbR°, -NRdC(NRe)NR R°,
-NRdC(0)jR , -C(0)NRbRc, -C(0)NR CORb, -OC(0)NR Rc, -NRbRc, -NRdOR°, -C(S)NRbRc, -NRdC(S)NR Rc, -NRdC(0)NRbRc, -OSH, -S(0)jRb, -OS(0)jRb, -SC(0)Rb, -S(0)jC(0)ORb, -
SCORd, -NRdSRc, -SRb, -NHS(0)jRb, -COSRb, -C(0)S(0)jR , -CSR , -CS(0)jRb, -C(SO)OH,
-C(SO)2OH, -NRdC(S)Rc, -OC(S)R , -OC(S)OH, -OC(SO)2Rb, -S(0)jNRbRc, -SNRbR°,
-S(0)NR Rc, -NRdCS(0)jRc, -C(O)j(CH2)tNRd-(4-10 membered heteroaryl), -C(O)i(CH2)tNRd(4-10 membered heterocycloalkyl), -(CRdRe)tCN, -(CRd Re)t(C3-C1D cycloalkyl), -(CR Re)t(CB-C10 aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), -(CRdRe)t(4-10 membered heteroaryl),
-(CR Re)qC(0)(CRdR6)t(C3-C1o cycloalkyl), -(CRdRe)qC(O)(CRdRe),(C6-C10 aryl),
-(CRdRe)qC(O)(CRdRe),(4-10 membered heterocycloalkyl), -(CRdRβ)qC(O)(CRdRe),(4-10 membered heteroaryl), -(CRdRe)tO(CRdRe)q(C3-C10 cycloalkyl), -(CRdRe)tO(CRdRδ)q(C6-C10 aryl),
-(CRdRe)tO(CRdRe)q(4-10 membered heterocycloalkyl), -(CRdR6)tO(CRdR°)q(4-10 membered heteroaryl), -(CRdRe)qSO2(CRdRe)t(C3-C10 cycloalkyl), -(CRdRe)qSO2(CRdRe),(C6-C10 aryl),
-(CRdRe)qSO2(CRdRe),(4-10 membered heterocycloalkyl), and -(CRdRe)qSO2(CRdRe)t(4-10 membered heteroaryl), wherein Ra is selected from the group consisting of halo, hydroxyl,
-NRdRe, C1-C10 alkyl, haloalkyl, CrC6 alkoxyl, Rb and R° are independently selected from H, Cr
C10 alkyl, -(CRdRe)t(C3-C10 cycloalkyl), -(CR Re)t(CB-C10 aryl), -(CRdRe)t(4-10 membered heterocycloalkyl), and -(CRdRe)t(4-10 membered heteroaryl), Rd and Re are independently H or CrCB alkyl, j is an integer from 0 to 2, q and t are each independently an integer from 0 to 5, and 1 or 2 ring carbon atoms of the cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with =0, and the alkyl, alkenyl, alkynyl, aryl and cyclic moieties of the foregoing R10 groups are unsubstituted or substituted with 1 to 3 substituents independently selected from halo, =0, cyano, nitro, -(CRdRB)tCN, haloalkyl, 2-10 membered heteroalkyl, -ORb, -C(0)jRb, -NRdC(0)R , -C(0)NRbRc, -NRbRc, -NR OR c, -NRdC(0)jNRbR°, -NRdC(0)jRbRc, -OC(0)jRb, -OC(0)NRbRc, -SRd, CrC10 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, -(CRdRe)t(C3-C10 cycloalkyl),
-(CRαRe)t(C6-C10 aryl), -(CR _ά°rR_,ee-)t(4-10 membered heterocycloalkyl),
Figure imgf000122_0001
membered heteroaryl), -(CR^ Ce-C™ aryl)-(CrC6 alkyl); and wherein t, Rb, Rc, Rd, Re are as defined above.
A compound selected from the group consisting of:
Figure imgf000122_0002
Figure imgf000123_0001
Figure imgf000124_0001
or a pharmaceutically acceptable salt thereof.
7. A pharmaceutical composition comprising: a therapeutically effective amount of an agent selected from the group consisting of compounds, prodrugs, meiabolites, and salts as defined in claim 1 ; and a pharmaceutically acceptable carrier.
8. A method of treating a mammalian disease condition mediated by PIN1 activity, comprising administering to a mammal in need thereof a therapeutically effective amount of a compound, pharmaceutically acceptable prodrug, pharmaceutically active metabolite, or pharmaceutically acceptable salt as defined in claim 1.
9. A method according to claim 8, wherein the mammalian disease condition is associated with hypertension, inappropriate cell proliferation, infectious diseases, or neurodegenerative brain disorders.
PCT/IB2004/000574 2003-03-10 2004-02-23 Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1) WO2004087720A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP04713610A EP1603926A1 (en) 2003-03-10 2004-02-23 Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1)
JP2006506287A JP2006523669A (en) 2003-03-10 2004-02-23 Phosphate / sulfate compounds and pharmaceutical compositions for inhibiting NIMA regulatory protein (PIN1)
MXPA05009241A MXPA05009241A (en) 2003-03-10 2004-02-23 Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1).
BRPI0408477-2A BRPI0408477A (en) 2003-03-10 2004-02-23 phosphate / sulfate ester compounds and pharmaceutical compositions for inhibiting the use of protein (pin 1) and their use
CA002517281A CA2517281A1 (en) 2003-03-10 2004-02-23 Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US45316303P 2003-03-10 2003-03-10
US60/453,163 2003-03-10

Publications (1)

Publication Number Publication Date
WO2004087720A1 true WO2004087720A1 (en) 2004-10-14

Family

ID=33131661

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2004/000574 WO2004087720A1 (en) 2003-03-10 2004-02-23 Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1)

Country Status (6)

Country Link
EP (1) EP1603926A1 (en)
JP (1) JP2006523669A (en)
BR (1) BRPI0408477A (en)
CA (1) CA2517281A1 (en)
MX (1) MXPA05009241A (en)
WO (1) WO2004087720A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005080406A2 (en) * 2003-12-19 2005-09-01 Jerini Ag Compounds for the inhibition of undesired cell proliferation and use thereof
WO2006040646A1 (en) * 2004-10-14 2006-04-20 Pfizer, Inc. Benzimidazole or indole amides as inhibitors of pin1
EP1742907A2 (en) * 2004-05-06 2007-01-17 Cytokinetics, Inc. Certain chemical entities, compositions, and methods
US7504413B2 (en) 2004-05-06 2009-03-17 Cytokinetics, Inc. N-(4-(imidazo[1,2A]pyridin-YL)phenethyl)benzamide inhibitors of the mitotic kinesin CENP-E for treating certain cellular proliferation diseases
US7795448B2 (en) 2004-05-06 2010-09-14 Cytokinetics, Incorporated Imidazoyl-benzamide anti-cancer agents
EP2845588A1 (en) * 2013-09-09 2015-03-11 Centre National de la Recherche Scientifique (C.N.R.S.) PIN1 inhibitors for use in the prevention and/or treatment of theileriosis, and related applications
WO2017063754A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds as pin1 modulators
WO2017063756A1 (en) * 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds as pin1 modulators
WO2017063757A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds
WO2017063755A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds
WO2017167995A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused hexacyclic imidazole derivatives as modulators of tnf activity
WO2017167993A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
WO2017167994A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
WO2018101329A1 (en) 2016-11-29 2018-06-07 国立大学法人広島大学 Novel ester compound and pin1 inhibitor, inflammatory disease therapeutic, and colon cancer therapeutic in which said ester compound is used
WO2018167176A1 (en) 2017-03-15 2018-09-20 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
US10202405B2 (en) 2014-10-03 2019-02-12 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives
WO2019031470A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 NOVEL AMIDE COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
WO2019031471A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 Therapeutic agent for fatty liver diseases and therapeutic agent for adiposity
WO2019031472A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 NOVEL ANTHRANILIC ACID-BASED COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
US10793578B2 (en) 2016-04-01 2020-10-06 UCB Biopharma SRL Fused pentacyclic imidazole derivatives as modulators of TNF activity
WO2021182457A1 (en) 2020-03-12 2021-09-16 国立大学法人広島大学 Novel 3,5-diaminobenzoic acid compound, and pin1 inhibitor and therapeutic agent for inflammatory diseases using same
WO2022032179A1 (en) * 2020-08-07 2022-02-10 The Regents Of The University Of California Pin1 inhibitors and uses thereof
WO2022107745A1 (en) 2020-11-17 2022-05-27 国立大学法人広島大学 Therapeutic agent or prophylactic agent for covid-19
US11440928B2 (en) 2017-01-31 2022-09-13 Gilead Sciences, Inc. Crystalline forms of tenofovir alafenamide

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3223875A1 (en) * 2021-06-25 2022-12-29 Shigeru Matsuoka Heterocycloalkyl-substituted polyheteroazole derivatives as medicaments for treating and/or preventing rs virus infections

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995004751A1 (en) * 1993-08-04 1995-02-16 Glycomed Incorporated Selectin binding glycopeptides
WO2004005315A2 (en) * 2002-07-09 2004-01-15 Pfizer Inc. Pin1 peptidyl-prolyl isomerase polypeptides, their crystal structures, and use thereof for drug design

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995004751A1 (en) * 1993-08-04 1995-02-16 Glycomed Incorporated Selectin binding glycopeptides
WO2004005315A2 (en) * 2002-07-09 2004-01-15 Pfizer Inc. Pin1 peptidyl-prolyl isomerase polypeptides, their crystal structures, and use thereof for drug design

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
B.A.SCHWEITZER ET AL.: "Design and Synthesis of beta-Carboxamido Phosphonates as Potent Inhibitors of Imidazole Glycerol Phosphate dehydratase", BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 9, no. 14, 1999, pages 2053 - 2058, XP002285312 *
BIN XU, ANDREW STEPHENS, GARY KIRSCHENHEUTER,ARTHUR F GRESLIN,XIAOQUIN CHENG,JOE SENELLO,MARCO CATTANEO ET AL: "Acyclic Analogues of Adenosine Biphosphates as P2Y Receptor Antagonists: Phosphate Substitution Leads to Multiple Pathways of Inhibition of Platelet Aggregation", J. MED. CHEM., vol. 45, 2002, pages 5694 - 5709, XP002285317 *
BROUWER A J ET AL: "AN EFFICIENT SYNTHESIS OF N-PROTECTED BETA-AMINOETHANESULFONYL CHLORIDES: VERSATILE BUILDING BLOCKS FOR THE SYNTHESIS OF OLIGOPEPTIDOSULFONAMIDES", SYNTHESIS, GEORG THIEME VERLAG. STUTTGART, DE, no. 11, 2000, pages 1579 - 1584, XP001041496, ISSN: 0039-7881 *
C.M. SUTER, W.E. TRUCE: "Sulfonation of the Phenylpropylenes", J. AM. CHEM. SOC., vol. 66, 1944, pages 1105 - 1109, XP002285314 *
CHOUNG UN KIM ET AL.: "A New Class of Acyclic Phosphonate Nucleotide Analogues: Phosphate Isosteres of Acyclovir and Gancyclovir Monophosphates as Antiviral Agents", J. MED. CHEM., vol. 34, 1991, pages 2286 - 2294, XP002285315 *
HAK SUNG KIM, DOV BARAK, T. KENDALL HARDEN, JOSÉ L. BOYER, KENNETH A. JACOBSON: "Acyclic and Cyclopropyl Analogues of Adenosine Biphosphate Antagonists of the P2Y1 Receptor: Structure-Activity Relationships and Receptor Docking", J. MED. CHEM., vol. 44, 2001, pages 3092 - 3108, XP002285318 *
JUNG DAE PARK, DONG H. KIM, SEUNG-JUN KIM, JOO-RANG WOO, SEONG EON RYU: "Sulfamide-Based Inhibitors for Carboxypeptidase A. Novel Type Transition State Analogue Inhibitors for Zinc Proteases", J. MED. CHEM., vol. 45, 2002, pages 5295 - 5302, XP002285319 *
MICHAEL R. HANDEN, RICHARD L. JARVEST, MALCOLM R. BOYD, DAVID SUTTON, R. ANTHONY VERE HODGE: "Prodrugs of the Selective Antiherpesvirus Agent 9-(4-Hydroxy-3-(hydroxymethyl)but-1-yl)guanine (BRL39123) with Improved Gastrointestinal Absorption Properties", J. MED. CHEM., vol. 32, 1989, pages 1738 - 1743, XP002285316 *
R.A.AITKEN, D.P.ARMSTRONG, R.H.B. GALT, S.T.E.MESHER: "Synthesis and Oxidation of Chiral 2-Thiazolines (4,5-dihydro-1,3-thiazoles)", J.CHEM.SOC.,PERKIN TRANS 1, no. 6, 1997, pages 935 - 943, XP002285313 *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005080406A2 (en) * 2003-12-19 2005-09-01 Jerini Ag Compounds for the inhibition of undesired cell proliferation and use thereof
WO2005080406A3 (en) * 2003-12-19 2005-12-01 Jerini Ag Compounds for the inhibition of undesired cell proliferation and use thereof
JP4805916B2 (en) * 2004-05-06 2011-11-02 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
US8772507B2 (en) 2004-05-06 2014-07-08 Cytokinetics, Inc. Imidazole-benzamide anti-cancer agents
JP2007537163A (en) * 2004-05-06 2007-12-20 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
EP1742907A4 (en) * 2004-05-06 2008-01-02 Cytokinetics Inc Certain chemical entities, compositions, and methods
US7504413B2 (en) 2004-05-06 2009-03-17 Cytokinetics, Inc. N-(4-(imidazo[1,2A]pyridin-YL)phenethyl)benzamide inhibitors of the mitotic kinesin CENP-E for treating certain cellular proliferation diseases
EP1742907A2 (en) * 2004-05-06 2007-01-17 Cytokinetics, Inc. Certain chemical entities, compositions, and methods
US7618981B2 (en) 2004-05-06 2009-11-17 Cytokinetics, Inc. Imidazopyridinyl-benzamide anti-cancer agents
US7795448B2 (en) 2004-05-06 2010-09-14 Cytokinetics, Incorporated Imidazoyl-benzamide anti-cancer agents
US8207340B2 (en) 2004-05-06 2012-06-26 Cytokinetics, Incorporated Imidazopyridinyl benzamide mitotic kinesin inhibitors
US8163919B2 (en) 2004-05-06 2012-04-24 Cytokinetics, Incorporated Imidazopyridinyl benzamide mitotic kinesin inhibitors
WO2006040646A1 (en) * 2004-10-14 2006-04-20 Pfizer, Inc. Benzimidazole or indole amides as inhibitors of pin1
US7582668B2 (en) 2005-11-09 2009-09-01 Cytokinetics, Incorporated Imidazoyl-benzamide anti-cancer agents
EP2845588A1 (en) * 2013-09-09 2015-03-11 Centre National de la Recherche Scientifique (C.N.R.S.) PIN1 inhibitors for use in the prevention and/or treatment of theileriosis, and related applications
WO2015032998A1 (en) * 2013-09-09 2015-03-12 Centre National De La Recherche Scientifique (Cnrs) Pin1 inhibitors for use in the prevention and/or treatment of theileriosis, and related applications
US10202405B2 (en) 2014-10-03 2019-02-12 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives
US11912721B2 (en) 2014-10-03 2024-02-27 UCB Biopharma SRL Fused pentacyclic imidazole derivatives
US10906919B2 (en) 2014-10-03 2021-02-02 UCB Biopharma SRL Fused pentacyclic imidazole derivatives
WO2017063754A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds as pin1 modulators
WO2017063756A1 (en) * 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds as pin1 modulators
WO2017063757A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds
WO2017063755A1 (en) 2015-10-12 2017-04-20 Polyphor Ag Conformationally constrained macrocyclic compounds
WO2017167994A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
US10793578B2 (en) 2016-04-01 2020-10-06 UCB Biopharma SRL Fused pentacyclic imidazole derivatives as modulators of TNF activity
WO2017167995A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused hexacyclic imidazole derivatives as modulators of tnf activity
WO2017167993A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
US10654861B2 (en) 2016-04-01 2020-05-19 UCB Biopharma SRL Fused pentacyclic imidazole derivatives as modulators of TNF activity
US10669286B2 (en) 2016-04-01 2020-06-02 UCB Biopharma SRL Fused pentacyclic imidazole derivatives as modulators of TNF activity
US10766906B2 (en) 2016-04-01 2020-09-08 UCB Biopharma SRL Fused hexacyclic imidazole derivatives as modulators of TNF activity
WO2018101329A1 (en) 2016-11-29 2018-06-07 国立大学法人広島大学 Novel ester compound and pin1 inhibitor, inflammatory disease therapeutic, and colon cancer therapeutic in which said ester compound is used
US11203565B2 (en) 2016-11-29 2021-12-21 Hiroshima University Ester compound and PIN1 inhibitor, inflammatory disease therapeutic, and colon cancer therapeutic in which said ester compound is used
US11440928B2 (en) 2017-01-31 2022-09-13 Gilead Sciences, Inc. Crystalline forms of tenofovir alafenamide
WO2018167176A1 (en) 2017-03-15 2018-09-20 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
US10953019B2 (en) 2017-03-15 2021-03-23 UCB Biopharma SRL Fused pentacyclic imidazole derivatives as modulators of TNF activity
KR20200037363A (en) 2017-08-07 2020-04-08 고쿠리츠다이가쿠호진 히로시마다이가쿠 New amide compounds, and Pin1 inhibitors using the same, therapeutic agents for inflammatory diseases and therapeutic agents for cancer
KR20200042906A (en) 2017-08-07 2020-04-24 고쿠리츠다이가쿠호진 히로시마다이가쿠 New anthranilic acid compounds, and Pin1 inhibitors using the same, therapeutic agents for inflammatory diseases and therapeutic agents for cancer
KR20200041336A (en) 2017-08-07 2020-04-21 고쿠리츠다이가쿠호진 히로시마다이가쿠 Treatment of fatty liver disease and treatment of obesity
US11071738B2 (en) 2017-08-07 2021-07-27 Hiroshima University Therapeutic agent for fatty liver diseases and therapeutic agent for adiposity
WO2019031472A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 NOVEL ANTHRANILIC ACID-BASED COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
WO2019031471A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 Therapeutic agent for fatty liver diseases and therapeutic agent for adiposity
WO2019031470A1 (en) 2017-08-07 2019-02-14 国立大学法人広島大学 NOVEL AMIDE COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
WO2021182457A1 (en) 2020-03-12 2021-09-16 国立大学法人広島大学 Novel 3,5-diaminobenzoic acid compound, and pin1 inhibitor and therapeutic agent for inflammatory diseases using same
KR20220152535A (en) 2020-03-12 2022-11-16 고쿠리츠다이가쿠호진 히로시마다이가쿠 Novel 3,5-diaminobenzoic acid compounds, and Pin1 inhibitors using the same and therapeutic agents for inflammatory diseases
WO2022032179A1 (en) * 2020-08-07 2022-02-10 The Regents Of The University Of California Pin1 inhibitors and uses thereof
WO2022107745A1 (en) 2020-11-17 2022-05-27 国立大学法人広島大学 Therapeutic agent or prophylactic agent for covid-19
KR20230110723A (en) 2020-11-17 2023-07-25 고쿠리츠다이가쿠호진 히로시마다이가쿠 Treatment or prevention of COVID-19

Also Published As

Publication number Publication date
JP2006523669A (en) 2006-10-19
BRPI0408477A (en) 2006-04-04
CA2517281A1 (en) 2004-10-14
MXPA05009241A (en) 2005-10-19
EP1603926A1 (en) 2005-12-14

Similar Documents

Publication Publication Date Title
EP1603926A1 (en) Phosphate/sulfate ester compounds and pharmaceutical compositions for inhibiting protein interacting nima (pin1)
EP0114027B1 (en) Analogs of mevalolactone and derivatives thereof, processes for their production, pharmaceutical compositions containing them and their use as pharmaceuticals
CA2806190C (en) N-acylsulfonamide apoptosis promoters
JP2019521082A (en) Cyclopropyl-amide compounds as LSD1 / HDAC dual inhibitors
CN102482221B (en) Bicyclic and tricyclic compounds as kat ii inhibitors
Erion et al. Structure-based design of inhibitors of purine nucleoside phosphorylase. 3. 9-Arylmethyl derivatives of 9-deazaguanine substituted on the arylmethyl group
EP1847539A1 (en) Quinazoline derivatives
MXPA06008375A (en) Novel inhibitors of chymase.
Halazy et al. Fluorophosphonate derivatives of N9-benzylguanine as potent, slow-binding multisubstrate analogue inhibitors of purine nucleoside phosphorylase
EP0092190A2 (en) Phospholipid derivatives, process for preparation thereof and pharmaceutical composition of the same
Liu et al. Enantioselective synthesis of phosphinyl peptidomimetics via an asymmetric Michael reaction of phosphinic acids with acrylate derivatives
AU8022187A (en) Azaindole and indolizine derivatives, processes for their production and their use as pharmaceuticals
AU670457B2 (en) Alpha-phosphonosulfonate squalene synthetase inhibitors and method
Mucha et al. The preparation of phosphono peptides containing a phosphonamidate bond
US20050250742A1 (en) Phosphate/sulfate ester compounds and pharmaceutical composition for inhibiting protein interacting NIMA (PIN1)
AU657554B2 (en) Dialkyl (dialkoxyphosphinyl)methyl phosphates as anti-inflammatory agents
JP4526534B2 (en) Novel phosphonic acid compounds as inhibitors of serine proteases
JPH09500131A (en) Method for producing HIV protease inhibitor
KR20210005863A (en) 1H-indazole-3-carboxamide compound as a glycogen synthase kinase 3 beta inhibitor
Abdurakhmanova et al. 1, 3-oxazole derived cytisines
KR20020046283A (en) Synthetic Routes for the Preparation of Rhinovirus Protease Inhibitors and Key Intermediates
US4743687A (en) Pyridazo (1,2-a) (1,2) diazepine derivatives
FI85862C (en) Process for the preparation of novel therapeutically useful phosphonylhydroxyacyl prolines
JP2024500391A (en) 2-(3,5-dichloro-1-methyl-indazol-4-yl)-1-[(1S,3R)-3-(hydroxymethyl)-5-(1-hydroxy-1-methyl-ethyl)- Prodrug of 1-methyl-3,4-dihydro-1H-isoquinolin-2-yl]ethanone
Yuasa et al. Diastereoselective synthesis of ω-phosphonic acid analogues of 4-arylkainoids

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2517281

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/009241

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2004713610

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006506287

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 2004713610

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0408477

Country of ref document: BR

WWW Wipo information: withdrawn in national office

Ref document number: 2004713610

Country of ref document: EP