AU6023198A - Integrin antagonists - Google Patents

Integrin antagonists

Info

Publication number
AU6023198A
AU6023198A AU60231/98A AU6023198A AU6023198A AU 6023198 A AU6023198 A AU 6023198A AU 60231/98 A AU60231/98 A AU 60231/98A AU 6023198 A AU6023198 A AU 6023198A AU 6023198 A AU6023198 A AU 6023198A
Authority
AU
Australia
Prior art keywords
alkyl
alkylene
aryl
compound
mammal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU60231/98A
Other versions
AU729869B2 (en
Inventor
Mark E. Duggan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck and Co Inc
Original Assignee
Merck and Co Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9702788.2A external-priority patent/GB9702788D0/en
Priority claimed from GBGB9725996.4A external-priority patent/GB9725996D0/en
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of AU6023198A publication Critical patent/AU6023198A/en
Application granted granted Critical
Publication of AU729869B2 publication Critical patent/AU729869B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Vascular Medicine (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Description

TITLE OF THE INVENTION INTEGRIN ANTAGONISTS
FIELD OF THE INVENTION The present invention is related to U.S. provisional applications Serial Nos. 60/035,614, filed January 17, 1997, and 60/062,594, filed October 20, 1997, the contents of which are hereby incorporated by reference.
The present invention provides novel compounds and derivatives thereof, their synthesis, and their use as vitronectin receptor ligands. More particularly, the compounds of the present invention are αvβ3 antagonists, αvβδ antagonists or dual αvβ3/ vβδ antagonists useful for inhibiting bone resorption, treating and preventing osteoporosis, and inhibiting vascular restenosis, diabetic retinopathy, macular degeneration, angiogenesis, atherosclerosis, inflammation, viral disease, and tumor growth.
BACKGROUND OF THE INVENTION
This invention relates to compounds for inhibiting bone resorption that is mediated by the action of a class of cells known as osteoclasts.
Osteoclasts are multinucleated cells of up to 400 μm in diameter that resorb mineralized tissue, chiefly calcium carbonate and calcium phosphate, in vertebrates. They are actively motile cells that migrate along the surface of bone. They can bind to bone, secrete necessary acids and proteases and thereby cause the actual resorption of mineralized tissue from the bone.
More specifically, osteoclasts are believed to exist in at least two physiological states. In the secretory state, osteoclasts are flat, attach to the bone matrix via a tight attachment zone (sealing zone), become highly polarized, form a ruffled border, and secrete lysosomal enzymes and protons to resorb bone. The adhesion of osteoclasts to bone surfaces is an important initial step in bone resorption. In the migratory or motile state, the osteoclasts migrate across bone matrix and do not take part in resorption until they attach again to bone. Integrins are transmembrane, heterodimeric, glycoproteins which interact with extracellular matrix and are involved in osteoclast attachment, activation and migration. The most abundant integrin in osteoclasts (rat, chicken, mouse and human) is the vitronectin receptor, or αvβ3, thought to interact in bone with matrix proteins that contain the RGD sequence. Antibodies to αvβ3 block bone resorption in vitro indicating that this integrin plays a key role in the resorptive process. There is increasing evidence to suggest that vβ3 ligands can be used effectively to inhibit osteoclast mediated bone resoption in vivo in mammals.
The current major bone diseases of public concern are osteoporosis, hypercalcemia of malignancy, osteopenia due to bone metastases, periodontal disease, hyperparathyroidism, periarticular erosions in rheumatoid arthritis, Paget's disease, immobilization- induced osteopenia, and glucocorticoid treatment.
All these conditions are characterized by bone loss, resulting from an imbalance between bone resorption (breakdown) and bone formation, which continues throughout life at the rate of about 14% per year on the average. However, the rate of bone turnover differs from site to site, for example, it is higher in the trabecular bone of the vertebrae and the alveolar bone in the jaws than in the cortices of the long bones. The potential for bone loss is directly related to turnover and can amount to over 5% per year in vertebrae immediately following menopause, a condition which leads to increased fracture risk. There are currently 20 million people with detectable fractures of the vertebrae due to osteoporosis in the United States. In addition, there are 250,000 hip fractures per year attributed to osteoporosis. This clinical situation is associated with a 12% mortality rate within the first two years, while 30% of the patients require nursing home care after the fracture.
Individuals suffering from all the conditions listed above would benefit from treatment with agents which inhibit bone resorption.
Additionally, αvβ3 ligands have been found to be useful in treating and/or inhibiting restenosis (recurrence of stenosis after corrective surgery on the heart valve), atherosclerosis, diabetic retinopathy, macular degeneration and angiogenesis (formation of new blood vessels), and inhibiting viral disease. Moreover, it has been postulated that the growth of tumors depends on an adequate blood supply, which in turn is dependent on the growth of new vessels into the tumor; thus, inhibition of angiogenesis can cause tumor regression in animal models. (See, Harrison's Principles of Internal Medicine. 12th ed., 1991). αvβ3 antagonists, which inhibit angiogenesis, are therefore useful in the treatment of cancer for inhibiting tumor growth. (See e.g., Brooks et al., Cell, 79:1157-1164 (1994)). Moreover, compounds of this invention can also inhibit neovascularization by acting as antagonists of the integrin receptor αvβδ. A monoclonal antibody for αvβδ has been shown to inhibit VEGF- induced angiogenesis in rabbit cornea and the chick chorioallantoic membrane model; M.C. Friedlander, et.al., Science 270, 1500-1502, 1995. Thus, compounds that antagonize αvβδ are useful for treating and preventing macular degeneration, diabetic retinopathy, and tumor growth.
In addition, certain compounds of this invention antagonize both the αvβ3 and αvβδ receptors. These compounds, referred to as "dual αvβ3/αvβδ antagonists," are useful for inhibiting bone resorption, treating and preventing osteoporosis, and inhibiting vascular restenosis, diabetic retinopathy, macular degeneration, angiogenesis, atherosclerosis, inflammation and tumor growth.
It is an object of the present invention to identify compounds which bind to the αvβ3 receptor, αvβδ receptor or both the αvβ3 and αvβδ receptors.
It is a further object of the invention to identify compounds which act as antagonists of the αvβ3 receptor. It is another object of the invention to identify αvβ3 antagonist compounds which are useful agents for inhibiting: bone resorption mediated by osteoclast cells, restenosis, atherosclerosis, inflammation, diabetic retinopathy, macular degeneration and angiogenesis in animals, preferably mammals, especially humans. Still another object of the invention is to identify αvβ3 antagonists which cause tumor regression and/or inhibit tumor growth in animals. A further object of the invention is to identify αvβ3 antagonists useful for preventing or treating osteoporosis. An additional object of the invention is to identify αvβ3 antagonists useful for treating cancer. It has now been found that the compounds of the present invention, αvβ3 ligands, are useful for inhibiting bone resorption in mammals. Thus, the compounds of the present invention are useful for preventing or reducing the incidence of osteoporosis. Additionally, the αvβ3 ligands of the present invention are also useful for treating and/or inhibiting restenosis, diabetic retinopathy, macular degeneration, viral disease, atherosclerosis and/or angiogenesis in mammals.
SUMMARY OF THE INVENTION
The present invention provides αvβ3 antagonist compounds of the formula
X-Y-Z-Aryl-A-B
wherein:
Aryl is a 6-membered aromatic ring containing 0, 1, 2 or 3 nitrogen atoms and either unsubstituted or substituted with R^ and ^;
X is selected from
NR2 NR1 NR2
1 'I 3 " 2 3
-NR1 R2, -NR1-C-R3, -C-NR2R3, -NR1-C-NR3R4,
NR1 NR2 -aryl-NR1 R2, -aryl-C-NR2R3, -aryl-NR1-C-NR3R'
a δ- or 6-membered monocyclic aromatic or nonaromatic ring system containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O or S wherein the 5- or 6-membered ring system is either unsubstituted or substituted with Rl, R2, R3 and R4, or
a 9- to 14-membered polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2, 3 or 4 heteroatoms selected from N, 0 or S, and wherein the polycyclic ring system is either unsubstituted or substituted with R1, R2, R3 and R4 ;
Y is selected from Cθ-8 alkylene,
C3-10 cycloalkyl,
Co-8 alkylene-NR10-CO-Co-8 alkylene,
Co-8 alkylene-CONR10-Co-8 alkylene,
Cθ-8 alkylene-O-Cθ-8 alkylene, Cθ-8 alkylene-NRl°-Cθ-8 alkylene,
Cθ-8 alkylene-S(0)θ-2-Cθ-8 alkylene,
Co-8 alkylene-Sθ2-NR10-Cθ-8 alkylene,
Co-8 alkylene-NR10-Sθ2-Cθ-8 alkylene,
Cθ-8 alkylene-CO-Cθ-8 alkylene, (CH2)0-6 aryl(CH2)0-6,
(CH2)o-6 aryl-CO-(CH2)0-6,
(CH2)0-6 aryl-CO-NRlO-(CH2)0-6,
(CH2)o-6 arylNRlOCO(CH2)o-6, or
OR1
I
( H2)o-8CH (CH2)o-8.
Z and A are each independently selected from
- δ O
(CH2)m, (CH2)mO(CH2)n, (CH2)rnNR yl yl(fCuH2 \)n, t(CuH2 \)m MNDR1l 1l ΓC* MNDR1-2/(CH2)n
o o o
(CH2)mC I' NR 1111 (CH2)n,(CH2)mNR 1111"C(CH2)n (CH2)mO il(CH2)n,
(CH2)mC(CH2) (CH2)mS02(CH2)n; (CH2)mS(CH2)
(CH2)mSO(CH2) (CH2)mS02NR11(CH2)
(CH2)mNR11S02(CH2)n, (CH2)mCR1 1=CR12(CH2)n) or (C\-.2)<£≡C- (CH2)n,
where m and n are each independently an integer from 0 to 6;
B is
where p is an integer from 1 to 3;
R1, R2, R3, R4, R5, R8? 9, RIO, R11 and RΪ2 are each independently selected from hydrogen, halogen, Ci-10 alkyl, aryl Cθ-8 alkyl, amino Cθ-8 alkyl, Cl-3 acylamino Cθ-8 alkyl,
Ci-6 alkylamino Cθ-8 alkyl, Ci-6 dialkylamino Cø-8 alkyl, aryl Cθ-6 alkylamino Cθ-6 alkyl, Ci- alkoxyamino Cθ-8 alkyl, hydroxy Ci-6 alkylamino Cθ-8 alkyl, Ci-4 alkoxy Cø-6 alkyl, hydroxycarbonyl Cθ-6 alkyl, Ci- alkoxy carbonyl Cθ-6 alkyl, hydroxycarbonyl Cø-6 alkyloxy, hydroxy Cι_6 alkylamino Cø-6 alkyl or hydroxy Cθ-6 alkyl;
elected from hydrogen, fluorine, Ci-8 alkyl, hydroxyl, hydroxy Ci-6 alkyl, carboxy Cθ-6 alkyl,
Ci-6 alkyloxy, Ci-6 alkylcarbonyl, aryl Cø-6 alkylcarbonyl,
Ci-6 alkylcarbonyloxy, aryl Cø-6 alkylcarbonyloxy,
Ci-6 alkylaminocarbonyloxy, C3-8 cycloalkyl, aryl Cθ-6 alkyl,
Cθ-6 alkylamino Cø-6 alkyl,
Cø-6 dialkylamino Cø-6 alkyl,
Ci-8 alkylsulfonylamino Cθ-6 alkyl, aryl Cθ-6 alkylsulfonylamino Cθ-6 alkyl,
Ci-8 alkyloxycarbonylamino Cθ-8 alkyl, aryl Cθ-8 alkyloxycarbonylamino Cθ-8 alkyl,
Ci-8 alkylcarbonylamino Cθ-6 alkyl, aryl CQ-6 alkylcarbonylamino CQ-6 alkyl, Cθ-8 alkylaminocarbonylamino Cθ-6 alkyl, aryl Cθ-8 alkylaminocarbonylamino Cθ-6 alkyl, Cθ-8 alkylaminosulfonylamino Cø-6 alkyl, aryl Cθ-8 alkylaminosulfonylamino Cø-6 alkyl, Ci-6 alkylsulfonyl Cθ-6 alkyl, aryl Cθ-6 alkylsulfonyl Cθ-6 alkyl, Cl-6 alkylcarbonyl Cθ-6 alkyl, aryl Cθ-6 alkylcarbonyl Cθ-6 alkyl, Cχ-6 alkylthiocarbonylamino Cø-6 alkyl, or aryl Cθ-6 alkylthiocarbonylamino Cθ-6 alkyl; wherein the alkyl or N atoms may be unsubstituted or substituted with R^;
R is selected from Cγ- o polycyclyl Cθ-8 alkylsulfonylamino Cθ-6 alkyl;
C7-2o polycyclyl Cθ-8 alkylcarbonylamino Cθ-6 alkyl; C7-2o polycyclyl Cθ-8 alkylaminosulfonylamino Cθ-6 alkyl; C7-2o polycyclyl Cθ-8 alkylaminocarbonylamino Cθ-6 alkyl or C7-20 polycyclyl Cθ-8 alkyloxycarbonylamino Cø-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with
Rl4, R!5, R16 and Rl ; and wherein any of the alkyl groups may be unsubstituted or subsituted with R^4 and R^^;
Rl3 is selected from hydroxy,
Ci-8 alkyloxy, aryl Cθ-6 alkyloxy,
Ci-8 alkylcarbonyloxy C±-4 alkyloxy, aryl Ci-8 alkylcarbonyloxy Ci-4 alkyloxy, Cl-6 dialkylaminocarbonylmethyloxy, aryl Cl-6 dialkylaminocarbonylmethyloxy or an L- or D-amino acid joined by an amide linkage and wherein the carboxylic acid moiety of said amino acid is as the free acid or is esterified by C_.Q alkyl; and Rl4, R!5} R16 an(j R17 are each independently selected from hydrogen, halogen, Ci-io alkyl, C3-8 cycloalkyl, oxo, aryl, aryl Ci-8 alkyl, amino, amino Ci-8 alkyl, Cl-3 acylamino, Cl-3 acylamino Ci-8 alkyl, Cl-6 alkylamino, Cl-6 alkylamino-
Ci-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino Ci-8 alkyl, Ci-4 alkoxy, Ci-4 alkoxy Cl-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cl-6 alkyl, Cl-3 alkoxycarbonyl, Cl-3 alkoxycarbonyl Cl-6 alkyl, hydroxycarbonyl - Cl-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, Cl-6 alkyloxy-
Ci-6 alkyl, nitro, cyano, trifluoromethyl, trifluoromethoxy, trifluoroethoxy, Ci-8 alkyl-S(0)q, Ci-8 alkylaminocarbonyl, Ci-8 dialkylaminocarbonyl, Ci-8 alkyloxycarbonylamino, Ci-8 alkylaminocarbonyloxy or Ci-8alkylsulfonylamino;
and the pharmaceutically acceptable salts thereof;
provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
In one embodiment of the invention is the compound wherein
Aryl is a 6-membered aromatic ring containing 0, 1 or 2 nitrogen atoms wherein Aryl is unsubstituted or substituted with R^ and R^;
X is selected from
NR1 NR2
-NR1R2, -C-NR2R3, -NR1-C-NR3R4 ,
a 5- or 6-membered monocyclic aromatic or nonaromatic ring system containing 0, 1 or 2 heteroatoms selected from N, O or S wherein the δ- or 6-membered ring system is either unsubstituted or substituted with Rl and R2, or
a 9- to 14-membered fused polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2 or 3 heteroatoms selected from N, O or S, and wherein the polycyclic ring system is either unsubstituted or substituted with Rl and R2;
Y is selected from the group consisting of Cθ-8 alkylene,
C3-10 cycloalkyl,
Co-8 alkylene-NRiO-CO-Co-δ alkylene,
Co-8 alkylene-CONRlO-Co-8 alkylene,
Cθ-8 alkylene-O-Cθ-8 alkylene, Cθ-8 alkylene-NRl0-Cθ-8 alkylene,
Cθ-8 alkylene-S(0)θ-2-Cθ-8 alkylene,
Co-8 alkylene-S0 -NR10-Cθ-8 alkylene,
Co-8 alkylene-NR10-S02-Co-8 alkylene,
Cθ-8 alkylene-CO-Co-8 alkylene, (CH2)0-6 aryl(CH2)0-6,
(CH2)0-6 aryl-CO-(CH2)0-6,
(CH2)0-6 aryl-CO-NH-(CH2)0-6, or
OR1 I (CH2)0.8CH (CH2)0_8
Z and A are each independently selected from
(CH2)m, (CH2)mO(CH2)n, o o o
(CH2)mC " NR 1111(CH2)n,(CH2)mNR 1111C» (CH2)n,(CH2)mC ! l (CH2)n,
S °
(CH2)mC(CH2)n (CH2)mS02(CH2)n; (CH2)mS(CH2)n]
(CH2)mSO(CH2)n (CH^SOgNR 1^1 ,tCHg) n,
(CH2)mNR1 1S02(CH2)n, (CH2)mCR1 1=CR12(CH2)n or (CH2)mC≡C- (CH2)n,
where m and n are each independently an integer from 0 to 4;
R1, R2, R3, R4, R5> R8, R9, RIO, R11 and R12 are each independently selected from hydrogen, halogen,
Ci-io alkyl, aryl Cθ-8 alkyl, amino Cθ-8 alkyl,
Cl-3 acylamino Cθ-8 alkyl,
Cl-6 alkylamino Cθ-8 alkyl,
Cl-6 dialkylamino Cθ-8 alkyl, aryl Cθ-6 alkylamino Cθ-6 alkyl, Cl-4 alkoxyamino Cθ-8 alkyl, hydroxy Cl-6 alkylamino Cθ-8 alkyl,
Cl-4 alkoxy Crj-6 alkyl, or
Cl-4 alkoxycarbonyl Cø-6 alkyl;
R^ is selected from hydrogen, Cl-8 alkyl or aryl Cθ-6 alkyl; R is selected from
C7-15 polycyclyl Cθ-6 alkylsulfonylamino Cθ-6 alkyl or C7-15 polycyclyl Cθ-6 alkylcarbonylamino Cθ-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with
Rl4, Rlδ? R16 and R 7; and wherein any of the alkyl groups may be unsubstituted or subsituted with Rl4 and Rl5;
and all other variables are as defined above; and the pharmaceutically acceptable salts thereof;
provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-15). In a class of the invention is the compound wherein
Aryl is a phenyl or pyridyl ring wherein the phenyl or pyridyl ring is unsubstituted or substituted with Ηβ;
X is a 9- to 14-membered fused polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2 or 3 heteroatoms selected from N, O or S, and wherein the polycyclic ring system is either unsubstituted or substituted with Rl and R2;
Y is selected from Cθ-6 alkylene,
Cθ-6 alkylene-NRlO-CO-Co-6 alkylene, Co-6 alkylene-CONRlO-Co-6 alkylene, Cθ-6 alkylene-O-Cθ-6 alkylene, Cθ-6 alkylene-NRl°-Cθ-6 alkylene, Cθ-6 alkylene-S(0)θ-2-Cθ-6 alkylene, Cfj-6 alkylene-Sθ2-NRl°-Cθ-6 alkylene, or Cθ-6 alkylene-aryl-Cθ-6 alkylene;
Z is selected from
O
(CH2)m ι (CH2)mO(CH2)n or (CH2)mNR1 1 C(CH2)n
A is
O
II 11 (CH2)mCNR11 (CH2)n .
Rl, R2, R3, R4, Rδ; R8; 9? RIO, R11 and U12 are each independently selected from hydrogen, halogen, Ci-io alkyl, aryl Cθ-8 alkyl, Cl-4 alkoxy Cθ-6 alkyl, or Cl-4 alkoxycarbonyl Cø-6 alkyl;
R^ is hydrogen;
R ' is selected from C7-10 polycyclyl Cθ-6 alkylsulfonylamino Cθ-6 alkyl or C7-10 polycyclyl Cθ-6 alkylcarbonylamino Cø-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with Rl4, Rl5, R16 ^ R17;
Rl3 is selected from hydroxy or Ci-8 alkyloxy;
Rl4, R!5? R16 and ? are each independently selected from hydrogen, halogen, Cl-6 alkyl, C3-7 cycloalkyl, oxo, amino, amino Ci-8 alkyl, C1-3 acylamino, Ci-3 acylamino Ci-8 alkyl, Cl-6 alkylamino, Ci-6 alkylamino Ci-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino Ci-8 alkyl, Cl-4 alkoxy, Cl-4 alkoxy Cl-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cl-6 alkyl, Cl-3 alkoxycarbonyl, Cl-3 alkoxycarbonyl Cl-6 alkyl, hydroxycarbonyl - Ci-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, Cl-6 alkyloxy- Ci_6 alkyl, nitro, cyano, trifluoromethyl, trifluoromethoxy, trifluoroethoxy, Ci-8 alkylsulfonyl, Ci-8 alkylaminocarbonyl,
Ci-8 alkyloxycarbonylamino, Ci-8 alkylaminocarbonyloxy or Ci-8alkylsulfonylamino;
and all other variables are as defined above; and the pharmaceutically acceptable salts thereof;
provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ). In a subclass of the invention is the compound of the formula
wherein X is selected from the group consisting of
Y is selected from Cθ-6 alkylene,
Cθ-6 alkylene-O-Cθ-6 alkylene or Cθ-6 alkylene-NRl°-Co-6 alkylene;
Z is selected from (CH2)m or (CH2)-0- (CH2)n ;
Rl4, Rl5, Rl6 and Rl? are each independently selected from hydrogen, halogen, Cl-6 alkyl, oxo or hydroxy;
and all other variables are as defined above; and the pharmaceutically acceptable salts thereof;
provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
Illustrative of the invention is the compound of the formula:
- lδ
wherein q is an integer from 0 to 2;
R!8 is selected from
and all other variables are as defined previously; and the pharmaceutically acceptable salts thereof;
provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
Exemplifying the invention is the compound selected from
and the pharmaceutically acceptable salts thereof.
An illustration of the invention is the compound of the formula
and the pharmaceutically acceptable salts thereof.
Exemplifying the invention is a pharmaceutical composition comprising any of the compounds described above and a pharmaceutically acceptable carrier. An example of the invention is a pharmaceutical composition made by combining any of the compounds described above and a pharmaceutically acceptable carrier. Another illustration of the invention is a process for making a pharmaceutical composition comprising combining any of the compounds described above and a pharmaceutically acceptable carrier.
Further illustrating the invention is a method of treating and/or preventing a condition mediated by antagonism of a vitronectin receptor in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any of the compounds described above. Preferably, the condition is selected from bone resorption, osteoporosis, restenosis, diabetic retinopathy, macular degeneration, angiogenesis, atherosclerosis, inflammation, viral disease, cancer and tumor growth. More preferably, the condition is selected from osteoporosis and cancer. Most preferably, the condition is osteoporosis.
More specifically exemplifying the invention is a method of eliciting a vitronectin antagonizing effect in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any of the compounds or any of the pharmaceutical compositions described above. Preferably, the vitronectin antagonizing effect is an αvβ3 antagonizing effect; more specifically the αvβ3 antagonizing effect is selected from inhibition of bone resorption, inhibition of restenosis, inhibition of atherosclerosis, inhibition of angiogenesis, inhibition of diabetic retinopathy, inhibition of macular degeneration, inhibition of inflammation, inhibition of viral disease, or inhibition of tumor growth. Most preferably, the αvβ3 antagonizing effect is inhibition of bone resorption. Alternatively, the vitronectin antagonizing effect is an αvβδ antagonizing effect or a dual αvβ3/αvβδ antagonizing effect. Examples of αvβδ antagonizing effects are inhibition of: restenosis, atherosclerosis, angiogenesis, diabetic retinopathy, macular degeneration, inflammation, or tumor growth. Examples of dual αvβ3/αvβδ antagonizing effects are inhibition of: bone resorption, restenosis, atherosclerosis, angiogenesis, diabetic retinopathy, macular degeneration, inflammation, viral disease, or tumor growth.
Additional examples of the invention are methods of inhibiting bone resorption and of treating and/or preventing osteoporosis in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any of the compounds or any of the pharmaceutical compositions decribed above.
Additional illustrations of the invention are methods of inhibiting tumor growth and of treating and/or preventing cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any of the compounds or any of the pharmaceutical compositions decribed above.
More specifically exemplifying the invention is any of the compositions described above, further comprising a therapeutically effective amount of a second bone resorption inhibitor; preferably, the second bone resorption inhibitor is alendronate.
More specifically illustrating the invention is any of the methods of treating and/or preventing osteoporosis and/or of inhibiting bone resoption described above, wherein the compound is administered in combination with a second bone resorption inhibitor; preferably, the second bone resorption inhibitor is alendronate.
Additional illustrations of the invention are methods of treating hypercalcemia of malignancy, osteopenia due to bone metastases, periodontal disease, hyperparathyroidism, periarticular erosions in rheumatoid arthritis, Paget's disease, immobilization- induced osteopenia, and glucocorticoid treatment in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any of the compounds or any of the pharmaceutical compositions described above. More particularly exemplifying the invention is the use of any of the compounds described above in the preparation of a medicament for the treatment and/or prevention of osteoporosis in a mammal in need thereof. Still further exemplifying the invention is the use of any of the compounds described above in the preparation of a medicament for the treatment and/or prevention of: bone resorption, tumor growth, cancer, restenosis, artherosclerosis, inflammation, viral disease, diabetic retinopathy, macular degeneration and/or angiogenesis.
Another illustration of the invention is a drug which is useful for treating and/or preventing osteoporosis in a mammal in need thereof, the effective ingredient of the said drug being any of the compounds descibed above. More specifically illustrating the invention is a drug which is useful for treating and/or preventing: bone resorption, tumor growth, cancer, restenosis, artherosclerosis, inflammation, viral disease, diabetic retinopathy, macular degeneration and/or angiogenesis in a mammal in need thereof, the effective ingredient of the said drug being any of the compounds described above.
Additional illustrations of the invention are methods of treating tumor growth in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound described above and one or more agents known to be cytotoxic or antiproliferative, e.g., taxol and doxorubicin. Further illustrative are methods of treating tumor growth wherein the compounds of the present invention are administered in conjunction with radiation therapy.
Further illustrative are methods of inhibiting angiogenesis comprising administering a compound as described above in combination with a VEGF (a vascular endothethial growth factor) inhibitor compound. Such combinations are useful for treating disease states such as macular degeneration, diabetic retinopathy, and cancer.
DETAILED DESCRIPTION OF THE INVENTION
Representative compounds of the present invention are αvβ3 antagonists which display submicromolar affinity for the human αvβ3 receptor. Compounds of this invention are therefore useful for treating mammals suffering from a condition caused or mediated by the αvβ3 receptor, who are in need of such therapy. Pharmacologically effective amounts of the compounds, including pharmaceutically acceptable salts thereof, are administered to the mammal, to inhibit: the activity of mammalian osteoclasts, restenosis, tumor growth, artherosclerosis, inflammation, macular degeneration, diabetic retinopathy and angiogenesis.
The compounds of the present invention are administered in dosages effective to antagonize the αvβ3 receptor where such treatment is needed, as, for example, in the prevention or treatment of osteoporosis or cancer. For use in medicine, the salts of the compounds of this invention refer to non-toxic "pharmaceutically acceptable salts." Other salts may, however, be useful in the preparation of the compounds according to the invention or of their pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid. Representative salts include the following: Acetate, Benzenesulfonate, Benzoate, Bicarbonate,
Bisulfate, Bitartrate, Borate, Bromide, Calcium, Camsylate, Carbonate, Chloride, Clavulanate, Citrate, Dihydrochloride, Edetate, Edisylate, Estolate, Esylate, Fumarate, Gluceptate, Gluconate, Glutamate, Glycollylarsanilate, Hexylresorcinate, Hydrabamine, Hydrobromide, Hydrochloride, Hydroxynaphthoate, Iodide, Isothionate, Lactate, Lactobionate, Laurate, Malate, Maleate, Mandelate, Mesylate, Methylbromide, Methylnitrate, Methylsulfate, Mucate, Napsylate, Nitrate, N-methylglucamine ammonium salt, Oleate, Oxalate, Pamoate (Embonate), Palmitate, Pantothenate, Phosphate/diphosphate, Polygalacturonate, Salicylate, Stearate, Sulfate, Subacetate, Succinate, Tannate, Tartrate, Teoclate, Tosylate, Triethiodide and Valerate. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts.
The compounds of the present invention, may have chiral centers and occur as racemates, racemic mixtures and as individual diastereomers, or enantiomers with all isomeric forms being included in the present invention. Therefore, where a compound is chiral, the separate enantiomers, substantially free of the other, are included within the scope of the invention; further included are all mixtures of the two enantiomers. Also included within the scope of the invention are polymorphs and hydrates of the compounds of the instant invention. The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs," ed. H. Bundgaard, Elsevier, 198δ. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu. The term "therapeutically effective amount" shall mean that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by a researcher or clinician.
The term "bone resorption," as used herein, refers to the process by which osteoclasts degrade bone.
The term "alkyl" shall mean straight, branched or monocyclic alkanes, alkenes or alkynes of the specified number of carbon atoms. Preferably, the term "alkyl" refers to straight or branced chain alkanes of Cl-10 carbon atoms, or any number within this range
(i.e., methyl, ethyl, 1-propyl, 2-propyl, n-butyl, s-butyl, t-butyl, etc.).
The term "alkenyl" shall mean straight or branched chain alkenes of two to ten total carbon atoms, or any number within this range.
The term "alkynyl" shall mean straight or branched chain alkynes of two to ten total carbon atoms, or any number within this range. The term "cycloalkyl" shall mean cyclic rings of alkanes of three to eight total carbon atoms, or any number within this range (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl).
The terms "polycyclic" or "polycyclyl," as used herein, refer to unsubstituted or substituted fused or bridged polycyclic systems containing from 7 to 20 carbon atoms and which can contain one or more degrees of unsaturation. Preferably, the term "polycyclyl" refers to unsubstituted or substituted fused or bridged bi- or tri-cyclic systems containing from 7-lδ carbon atoms and which can contain one or two degrees of unsaturation. More preferably, the term "polycyclyl" refers to unsubstituted or substituted fused or bridged bi- or tri-cyclic systems containing from 7-10 carbon atoms and which can contain one or two degrees of unsaturation. Examples of prefered polycyclyl systems include, but are not limited to, decaline, camphor, adamantyl and norbornyl.
The term "cycloheteroalkyl," as used herein, shall mean a 3- to 8-membered fully saturated heterocyclic ring containing one or two heteroatoms chosen from N, O, or S. Examples of cycloheteroalkyl groups include, but are not limited to piperidinyl, pyrrolidinyl, azetidinyl, morpholinyl, piperazinyl.
The term "alkoxy," as used herein, refers to straight or branched chain alkoxides of the number of carbon atoms specified (e.g., Cl-5 alkoxy), or any number within this range (i.e., methoxy, ethoxy, etc.). The term "alkylene" shall include both straight and branched chain alkylenes (e.g., -CH2-, -CH(CH3)-, -CH(CH3)-CH2-, etc.).
The term "aryl," as used herein, refers to a monocyclic or polycyclic system composed of δ- and 6-membered rings, such that the system comprises at least one fully unsaturated (i.e., aromatic) ring, wherein the rings contain 0, 1, 2, 3 or 4 heteroatoms chosen from N, O or S, and either unsubstituted or substituted with one or more groups independently selected from hydrogen, halogen, Cl-10 alkyl, C3-8 cycloalkyl, aryl, aryl Ci-8 alkyl, amino, amino Cl-8 alkyl, Cl-3 acylamino, Cl-3 acylamino Cl-8 alkyl, Cl-6 alkylamino, Cl-6 alkylamino Cl-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino-Ci-8 alkyl, Cl-4 alkoxy, Cl-4 alkoxy Cl-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cι_6 alkyl, Ci- 5 alkoxycarbonyl, Cl-3 alkoxycarbonyl Cl-6 alkyl, hydroxycarbonyl Cl-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, cyano, trifluoromethyl, oxo or Ci-
5 alkylcarbonyloxy. Examples of aryl include, but are not limited to, phenyl, naphthyl, pyridyl, pyrazinyl, pyrimidinyl, imidazolyl, benzimidazolyl, indolyl, thienyl, furyl, dihydrobenzofuryl, benzo(l,3) dioxolane, oxazolyl, isoxazolyl and thiazolyl, which are either unsubstituted or substituted with one or more groups independently selected from hydrogen, halogen, Cl-10 alkyl, C3-8 cycloalkyl, aryl, aryl Cl-8 alkyl, amino, amino Cl-8 alkyl, Cl-3 acylamino, Cl-3 acylamino Cl-8 alkyl, Cl-6 alkylamino, Cl-6 alkylamino-Ci-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino Cl-8 alkyl, Cl-4 alkoxy, Cl-4 alkoxy Ci-
6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cl-6 alkyl, Cl-5 alkoxycarbonyl, Cl-3 alkoxycarbonyl Cl-6 alkyl, hydroxycarbonyl Cl-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, cyano, trifluoromethyl, oxo or Ci- 5 alkylcarbonyloxy. Preferably, the aryl group is unsubstituted, mono-, di-, tri- or tetra-substituted with one to four of the above-named substituents; more preferably, the aryl group is unsubstituted, mono-, di- or tri-substituted with one to three of the above-named substituents; most preferably, the aryl group is unsubstituted, mono- or di-substituted with one to two of the above-named substituents.
Whenever the term "alkyl" or "aryl" or either of their prefix roots appear in a name of a substituent (e.g., aryl Cθ-8 alkyl) it shall be interpreted as including those limitations given above for "alkyl" and "aryl." Designated numbers of carbon atoms (e.g., Ci- 10) shall refer independently to the number of carbon atoms in an alkyl or cyclic alkyl moiety or to the alkyl portion of a larger substituent in which alkyl appears as its prefix root.
The terms "arylalkyl" and "alkylaryl" include an alkyl portion where alkyl is as defined above and to include an aryl portion where aryl is as defined above. The Co-m or Ci-m designation where m may be an integer from 1-10 or 2-10 respectively refers to the alkyl component of the arylalkyl or alkylaryl unit. Examples of arylalkyl include, but are not limited to, benzyl, fluorobenzyl, chlorobenzyl, phenylethyl, phenylpropyl, fluorophenylethyl, chlorophenylethyl, thienylmethyl, thienylethyl, and thienylpropyl. Examples of alkylaryl include, but are not limited to, toluene, ethylbenzene, propylbenzene, methylpyridine, ethylpyridine, propylpyridine and butylpyridine. When substituent Rl, R2, R , R4, R5, R6, R7, R8? R9, RIO,
Rll, Rl2, Rl3 or Ri4 includes the definition Co (e.g., aryl Cθ-8 alkyl), the group modified by Co is not present in the substituent. Similarly, when any of the variables m, q, r or s is zero, then the group modified by the variable is not present; for example, when s is zero, the group "-(CH2)s C≡CH" is "-C≡CH".
The term "halogen" shall include iodine, bromine, chlorine and fluorine.
The term "oxy" means an oxygen (0) atom. The term "thio" means a sulfur (S) atom. The term "oxo" shall mean a bivalent oxygen atom (=0).
The term "L- or D-amino acids" means naturally occurring L- or D-amino acids, for example, those naturally occurring L-amino acids present in humans, e.g. protein amino acids,, including L- alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L- glutamine, L-glutamic acid, L-glycine, L-histidine, L-isoleucine, L- leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, and L-valine, and those naturally occurring D-amino acids which are non-protein amino acids, such as those found, for example, in antibiotic substances produced by bacteria and fungi, including D-valine, D-asparagine, D-glutamate, D-ornithine, D-phenylalanine, D-leucine, D-cysteine, and D-aspartate. (see Zubay "BIOCHEMISTRY" Addison-Wesley Publishing Company, Inc. (Reading, MA) 1983 pp. 867-870 and Stryer "BIOCHEMISTRY" W.H. Freeman and Company (New York, NY) 3rd Edition 1988 pp. 16-21). The term "substituted" shall be deemed to include multiple degrees of substitution by a named substitutent. Where multiple substituent moieties are disclosed or claimed, the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties, singly or plurally.
2δ Under standard nonmenclature used throughout this disclosure, the terminal portion of the designated side chain is described first, followed by the adjacent functionality toward the point of attachment. For example, a Ci-5 alkylcarbonylamino Cl-6 alkyl substituent is equivalent to
O -C1 -6 alkyl-NH-C-C1 -5 alkyl .
The present invention is also directed to combinations of the compounds of the present invention with one or more agents useful in the prevention or treatment of osteoporosis. For example, the compounds of the instant invention may be effectively administered in combination with effective amounts of other agents used in the treatment of osteoporosis such as bisphosphonate bone resorption inhibitors; preferably, the bone resorption inhibitor is the bisphosphonate alendronate, now sold as FOSAMAX®. Preferred combinations are simultaneous or alternating treatments of an αvβ3 receptor antagonist of the present invention and FOSAMAX®. In accordance with the method of the present invention, the individual components of the combination can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms. The instant invention is therefore to be understood as embracing all such regimes of simultaneous or alternating treatment and the term "administering" is to be interpreted accordingly. It will be understood that the scope of combinations of the compounds of this invention with other agents useful for treating αvβ3 related conditions includes in principle any combination with any pharmaceutical composition useful for treating osteoporosis.
Further exemplifying the invention are compositions further comprising an active ingredient selected from the group consisting of a.) an organic bisphosphonate or a pharmaceutically- acceptable salt or ester thereof, b.) an estrogen receptor modulator, and c.) a cytotoxic/antiproliferative agent, and mixtures thereof. Nonlimiting examples of such bisphosphonates include alendronate, etidronate, pamidronate, risedronate, and pharmaceutically acceptable salts and esters thereof. A particularly preferred bisphosphonate is alendronate, especially alendronate monosodium trihydrate.
Nonlimiting examples of estrogen receptor modulators include estrogen, progesterin, estradiol, raloxifene, and tamoxifene.
Nonlimiting examples of cytotoxic/antiproliferative agents are taxol and doxorubicin.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
The compounds of the present invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixers, tinctures, suspensions, syrups and emulsions. Likewise, they may also be administered in intravenous (bolus or infusion), intraperitoneal, topical (e.g., ocular eyedrop), subcutaneous, intramuscular or transdermal (e.g., patch) form, all using forms well known to those of ordinary skill in the pharmaceutical arts. An effective but non-toxic amount of the compound desired can be employed as an αvβ3 inhibitor.
The dosage regimen utilizing-the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician, veterinarian or clinician can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition. Oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably, from about 1 mg to about 100 mg of active ingredient. Intravenously, the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion. Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittant throughout the dosage regimen.
In the methods of the present invention, the compounds herein described in detail can form the active ingredient, and are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as 'carrier' materials) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices. For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral administration in liquid form, the oral drug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta- lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
The compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
Compounds of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxy- ethylaspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
In the schemes and examples below, various reagent symbols and abbreviations have the following meanings:
AcOH (HOAc): Acetic acid. BH3-DMS: Borane«dimethylsulfide.
BOC(Boc): t-Butyloxycarbonyl .
BOP: Benzotriazol-l-yloxytris(dimethylamino)- phosphonium hexafluorophosphate.
CBZ(Cbz): Carbobenzyloxy or benzyloxycarbonyl.
CDI: Carbonyldiimidazole.
CH2C12: Methylene chloride.
CHC13: Chloroform.
DEAD: Diethyl azodicarboxylate.
DIAD: Diisopropyl azodicarboxylate.
DIBAH or
DIBAL-H: Diisobutylaluminum hydride.
DIPEA: Diisopropylethylamine.
DMAP: 4-Dimethylaminopyridine.
DME: 1 ,2-Dimethoxyethane .
DMF: Dimethylformamide.
DMSO: Dimethylsulfoxide .
DPFN: 3,δ-Dimethyl-l-pyrazolylformamidine nitrate.
EDC: l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide
Et: Ethyl.
EtOAc: Ethyl acetate.
EtOH: Ethanol.
HOBT: 1-Hydroxybenzotriazole.
LDA: Lithium diisopropylamide.
MeOH: Methanol.
NEt3: Triethylamine.
NMM: N-methylmorpholine.
PCA-HC1: Pyrazole carboxamidine hydrochloride.
Pd/C: Palladium on activated carbon catalyst.
Pd(OAc)2: Palladium(II) acetate.
Ph: Phenyl. pTSA: p-Toluene sulfonic acid.
TEA: Triethylamine.
TFA: Trifluoroacetic acid. THF: Tetrahydrofuran.
TLC: Thin Layer Chromatography.
TMEDA: N,N,N',N'-Tetramethylethylenediamine.
TMS: Trimethylsilyl. TsCl: Tosyl chloride.
The novel compounds of the present invention were prepared according to the procedure of the following schemes and examples, using appropriate materials and are further exemplified by the following specific examples. The most preferred compounds of the invention are any or all of those specifically set forth in these examples. These compounds are not, however, to be construed as forming the only genus that is considered as the invention, and any combination of the compounds or their moieties may itself form a genus. The following examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. All temperatures are degrees Celsius unless otherwise noted. The following Schemes and Examples describe procedures for making representative compounds of the present invention. Moreover, by utilizing the procedures described in detail in PCT International Application Publication Nos. WO9δ/32710, published 7 December 199δ, and W09δ/17397, published 29 June 199δ, in conjunction with the disclosure contained herein, one of ordinary skill in the art can readily prepare additional compounds of the present invention claimed herein.
More specifically, procedures for preparing the N-terminus of the compounds of the present invention are described in WO 9δ/32710. Additionally, for a general review describing the synthesis of β-alanines which can be utilized as the C-terminus of the compounds of the present invention, see Cole, D.C., Recent Stereoselectiυe Synthetic Approaches to β-Amino Acids, Tetrahedron, 1994, δO, 9δl7-9δ82; Juaristi, E, et al., Enantioselective Synthesis of β-Amino Acids, Aldrichemica Ada, 1994, 27, 3. In particular, synthesis of the 3-methyl β-alanine is taught in Duggan, M.F. et al., J. Med. Chem., 1995, 38, 3332-3341; the 3-ethynyl β- alanine is taught in Zablocki, J.A., et al., J. Med. Chem., 1995, 38, 2378- 2394; the 3-pyrid-3-yl β-alanine is taught in Rico, J.G. et al., J. Org. Chem., 1993, 58, 7948-7951; and the 2-amino and 2-toslylamino β- alanines are taught in Xue, C-B, et al., Biorg. Med. Chem. Letts., 1996, 6, 339-344.
Scheme 1
1 -1 1 -2
BOC20, NEt3, CH3CN
1 -4 1-3
1 -6 Scheme 1 (continued)
Pd(OAc)2, CH3CN, 100°C
L-proline, EtOH, reflux
Scheme 1 (continued)
1 -1 1
1 -14. R = Et
NaOH, C2H5OH 1 -15. R = H
Ethyl 2(S)-Nfγ-Cbz-2.3-diaminopropionate hydrochloride (1-2)
1-1 (δ g, 21 mmol; Bachem) was dissolved in 100 mL EtOH and cooled to 0°C. SOC12 (9.2 mL, 126 mmol) was added followed by removal of the cooling bath. After 6 hours, the reaction was concentrated to provide lr2 as a white solid. iH NMR (300 MHz, CD3OD) δ 7.35 (m, 5H), 5.14 (s, 2H), 4.44 (m, IH), 4.22
(q, J=7Hz, 2H), 3.43 (m, IH), 3.20 (m, IH), 1.25 (t, J=7Hz, 3H). Ethyl 2(S)-Nrv-Cbz-Nβ-Boc-2.3-diaminopropionate (1-3) (2 g, 6.6 mmol) was dissolved in 60 mL CH3CN. NEt3 (1 mL, 7.2 mmol) was added followed by BOC2O (1.58 g, 7.3 mmol). After two hours, the reaction was concentrated, diluted with EtOAc, washed with sat. NaHC03, 10% KHSO4 and brine, dried (MgSθ4), filtered and concentrated to provide 1__ as a clear oil. TLC Rf 0.87 (silica, 80% EtOAc/hexanes).
IH NMR (300 MHz, CDCI3) δ 7.3δ (s, 5H), δ.7δ (bs, IH), δ.12 (s, 2H), 4.81 (bs, IH), 4.39 (m, IH), 4.19 (m, 2H), 3.δ6 (m, 2H), 1.42 (s, 9H), 1.29 (q, J=7Hz, 3H).
Ethyl 2(S)-N -Boc-2.3-diaminopropionate (1-4) (2.4 g, 6.6 mmol) with 10% Pd/C (240 mg) in EtOAc (35 mL) was stirred under a H2 atmosphere for 20 hours. The reaction was filtered through a celite pad and concentrated to provide _Λ as a clear oil.
TLC Rf 0.13 (silica, 80% EtOAc/hexanes). iH NMR (300 MHz, CDCI3) δ 5.00 (bs, IH), 4.19 (m, 2H), 3.5δ (m, 2H), 3.2δ (m, IH), 1.44 (s, 9H), 1.29 (q, J=7Hz, 3H).
Ethyl-2(S)-Nα-(KS)10-camphorsulfonylamino)-Nβ-Boc-2,3-diamino- propionate (1-δ)
Amine _Λ (760 mg, 3.27 mmol) was dissolved in 3δ mL CH2CI2 and cooled to 0°C. NMM (7δδ mL, 6.87 mmol) and 10(+) camphorsulfonyl chloride (1.23 g, 4.9 mmol; Aldrich) were added. After stirring at 0°C for one hour, the reaction was concentrated, then diluted with EtOAc, washed with H2O, sat. NaHCθ3, 10% KHSO4 and brine, dried (MgSθ4), and concentrated to an oil. Flash chromatography
(silica, 2δ-40% EtOAc/hexanes) provided __5 as a clear oil. TLC Rf 0.66 (silica, δ0% EtOAc/hexanes). iH-NMR (300 MHz, CDCI3) δ 6.37 (d, J=8Hz, IH), 4.99 (bt, IH), 4.32 (m, IH), 4.23 (q, J=8Hz, 2H), 3.δ6 (m, 3H), 3.0 (d, J=15 Hz, IH), 2.4 (m, IH), 2.0δ (m, 4H), 1.43 (s, 9H), 1.30 (t, J=7 Hz, 3H), 1.00 (s, 3H), 0.91 (s, 3H). Ethyl-2(S)-N -(l(S)10-camphorsulfonylamino)-2,3-diaminopro- pionate hvdrochlori.de (1-6)
Ester __5 (900 mg, 2.18 mmol) was dissolved in lδ mL EtOAc and cooled to 0°C. HC1 (g) was bubbled through the reaction mixture for lδ minutes. The reaction was removed from the cooling bath and purged with Ar (g) for 20 minutes followed by concentration to provide __. 6 as a foamly solid.
TLC Rf O.Oδ (silica, 20% MeOH/EtOAc).
1H-NMR (300 MHz, CDCI3): δ 4.7δ (m, IH), 4.26 (q, J=7Hz, 2H), 3.δ0 (m, 4H), 2.40 (m, 3H), 1.98 (m, 4H), 1.30 (t, J=7Hz, 3H), 1.04 (s, 3H), 0.91 (s, 3H).
Ethyl 4-(3-oxo-butyl)benzoate (1-9)
3-Buten-2-ol __1 (2.15 mL, 25 mmol; Aldrich), ethyl 4- iodobenzoate __8 (δ.δ g, 20 mmol, Aldrich) and NEt3 (3.δ mL, 2δ mmol) were combined in CH3CN (6 mL) under argon in a pressure tube. Pd(OAc)2 (19 mg, 80 umol) was added and the reaction heated at 100 C for 3 hours. The reaction was cooled, then diluted with ether and washed with H20, 10% KHSO4, sat. NaHC03, and brine, dried (MgS04), and concentrated to furnish __9 as a brown solid. TLC Rf 0.23 (silica, 30% EtOAc/hexanes).
Ethyl 4-r2-(1.8-Naphthyridin-7-yl)ethyllbenzoate (1-11)
An ethanol solution (70 mL) of O (1.75 g, 14.3 mmol; JOC, 1983, 48, 3401-3408), __9 (3.15 g, 14.3 mmol, and L-proline (0.8 g, 7.0 mmol) was refluxed for 16 hours. The reaction was concentrated to dryness. Flash chromatography (silica, 60%-80% EtOAc/hexane) gave __
11 as a yellow solid.
Rf 0.21 (silica, 3/1 EtOAc/hexane). IH NMR (300 MHz, CDCI3) δ 9.1 (m, IH), 8.18 (j, J=8 Hz, IH), 8.08 (d, J=8
Hz, IH), 7.95 (d, J=8 Hz, 2H), 7.48 (m, IH), 7.30 (dd, J=8 Hz, 2H), 4.35 (q,
J=7Hz, 2H), 3.35 (m, 4H), 1.38 (t, J=7Hz, 3H). Ethyl 4-[2-(l,2,3,4-tetrahydro-l,8-naphthyridin-7-yl)ethylbenzoate
A mixture of ±11 (64δ mg, 2.1 mmol), 10% Pd/C (65 mg), and ethanol (10 mL) was stirred under a hydrogen atmosphere for 18 hour. Filtration through a celite pad followed by concentration provided crude 1-12.
Trituration of the solid with 1:1 ether/hexanes furnished 1-12 as a light yellow solid.
TLC Rf 0.75 (silica, 70% EtOAc/hexanes).
IH NMR (300 MHz, CDCI3) δ 7.94 (d, J=8Hz, 2H), 7.26 (d, J=8 Hz, 2H), 7.03 (d, J=7 Hz, IH), 6.28 (d, J=7 Hz, IH), 4.81 (s, IH), 4.35 (q, J=7 Hz, 2H), 3.40 (m, 2H), 3.03 (m, 2H), 2.84 (m, 2H), 2.69 (t, J=6 Hz, 2H), 1.93 (t, J=6 Hz, 2H), 1.38 (t, J=7 Hz, 3H).
4-r2-(1.2.3.4-Tetrahvdro-1.8-naphthyridin-7-yl)ethylbenzoic acid (1-13) The ester _-_2 (680 mg, 2.1 mmol) in 6N HC1 (10 mL) was heated to 50 C for 18 hours. Concentration provided 1-13 as a light yellow solid. iH NMR (300 MHz, CD3OD) δ 7.93 (d, J=8 Hz, 2H), 7.52 (d, J=8 Hz, IH),
7.31 (d, J=8 Hz, 2H), 5.δ4 (d, J=8 Hz, 1), 3.48 (t, J-δ Hz, 2H), 3.03 (m, 4H), 2.79 (t, J=6 Hz, 2H), 1.93 (t, J=6 Hz, 2H).
4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10- camphorsulfonylaminol β-alanine ethyl ester (1-14)
1-13 (200 mg, 0.627 mmol), amine 1=6 (240 mg, 0.69 mmol), NMM (34δ mL, 3.13 mmol) and BOP reagent (332 mg, 0.75 mmol) were combined in 5 mL CH3CN. After stirring overnight, the reaction was concentrated, then diluted with EtOAc, washed with H2O, sat. NaHCθ3 and brine, dried (MgSθ4), filtered and concentrated. Flash chromatography (silica, EtOAc) provided 1-14 as an off-white foamy solid.
TLC Rf 0.13 (silica, EtOAc). iH NMR (300 MHz, CDCI3) δ 7.70 (d, J=8Hz, 2H), 7.2δ (d, J=8Hz, 2H), 7.03 (d, J=7Hz, IH), 6.72 (t, J=δHz, IH), 6.5 (bm, IH), 6.28 (d, J=7Hz, IH), 4.79 (s, IH), 4.42 (bs, IH), 4.25 (q, J=7Hz, 2H), 4.04 (m, IH), 3.8δ (m, IH), 3.δδ (d, J=lδHz, IH), 3.41 (m, 2H), 3.00 (m, 3H), 2.82 (t, J=4Hz, 2H), 2.69 (t, J=6Hz, 2H), 2.04 (m, 8H), 1.58 (bs, 3H), 1.31 (t, J=7Hz, 3H), 1.00 (s, 3H), 0.90 (s, 3H).
4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7-yl)ethyl]benzoyl-
2(S)-ri(S)10-camphorsulfonylaminol β-alanine (1-15)
1-14 (250 mg, 0.409 mmol) was dissolved in 4 mL EtOH, 1M NaOH (1.02 mL, 1.02 mmol) was added and the reaction mixture was stirred for two hours. The reaction mixture was neutralized with IN HCl and then concentrated to a foamy solid. Flash chromatography (silica, 18:10:1:1 EtOAc/EtOH/NH4θH/H2θ) provided __l_\ as a slightly yellow solid.
TLC Rf 0.49 (silica, 12:10:1:1 EtOAc/EtOH/NH4θH/H2θ). iH NMR (400 MHz, DMSO) δ 8.48 (bt, IH), 7.72 (d, J=8Hz, 2H), 7.δδ (bs, IH), 7.28 (d, J=8Hz, 2H), 7.02 (d, J=7Hz, IH), 6.37 (s, IH), 6.26 (d, J=7Hz, IH), 4.13 (s, IH), 3.δ4 (m, 3H), 3.37 (m, 2H), 2.94 (m, 3H), 2.73 (t, J=7Hz, 2H), 2.6 (t, J=6Hz, 2H), 2.3 (m, 3H), 2.02 (m, IH), 1.89 (m, 2H), 1.75 (m, 2H), 1.49 (m, IH), 1.37 (m, IH), 1.05 (m, IH), 0.95 (s, 3H), 0.66 (s, 3H).
Structurally related analogs of 1-lδ can be prepared from commercially available starting materials utilizing the chemistry shown in Scheme 1. Several such representative compounds are examplified as compounds 2-4 depicted in Table 1. Compounds 1 and 2 are readily available through the chemistry illustrated in Scheme 1 by using 2(R)-Nα-Cbz-2,3-diaminopropionic acid 5_ (Bachem) or (lR)-(-)10- camphorsulfonyl chloride 6 (Aldrich), respectively. Compounds 3 and 4 are attained by starting with the appropriate alcohols, 1- adamantanemethanol 1_ (Aldrich) or (lR)-(-)-Nopol 8 (Aldrich), respectively. These alcohols can be converted to their corresponding sulfonyl chlorides via the standard chemistry shown in Scheme 2. Scheme 2
TsCI, pyridine
-1 2-2
NaHC03lCH3COSH. DMF
2-5
Table 1
Starting
Compound Material
Z
SCHEME 3
H, dioxane
SCHEME 3 (Cont'd)
2_ .
3-7 N-(4-Iodo-phenylsulfonylamino)-L-asparagine (3-2)
To a stirred solution of acid 34 (4.39 g, 33.2 mmol), NaOH (1.49 g, 37.2 mmol), dioxane (30 ml) and H2O (30 ml) at 0°C was added pipsyl chloride (10.34 g, 34.2 mmol). After ~5 minutes, NaOH (1.49, 37.2 mmol) dissolved in 15 ml H2O, was added followed by the removal of the cooling bath. After 2.0 h, the reaction mixture was concentrated. The residue was dissolved in H2O (300 ml) and then washed with EtOAc.
The aqueous portion was cooled to 0°C and then acidified with concentrated HCl. The solid was collected and then washed with Et2θ to provide acid 3=2 as a white solid. H NMR (300 MHz, D2O) δ 7.86 (d, 2H, J=8HZ), 7.48 (d, 2H, J=8Hz) 3.70
(m, IH), 2.39 (m, 2H).
2(S)-(4-Iodo-phenylsulfonylamino)-β-alanine (3-3) To a stirred solution of NaOH (7.14 g, 181.8 mmol) and H2O
(40 ml) at 0°C was added Br2 (1.30 ml, 24.9 mmol) dropwise over a ten minute period. After ~5 minutes, acid 3=2 (9.9 g, 24.9 mmol), NaOH (2.00 g, 49.8 mmol) and H2O (3δ ml) were combined, cooled to 0°C and then added in a single portion to the reaction. After stirring for 20 minutes at 0°C, the reaction was heated to 90°C for 30 minutes and then recooled to 0°C. The pH was adjusted to ~7 by dropwise addition of concentrated HCl. The solid was collected, washed with EtOAc, and then dried in vacuo to provide acid 3=3 as a white solid. iH NMR (300 MHz, D2O) δ 8.02 (d, 2H, J=8Hz), 7.63 (d, 2H, J=8Hz), 4.36 (m, IH), 3.51 (dd, IH, J=5Hz, 13Hz) 3.21 (m, IH).
Ethyl 2(S)-(4-iodo-phenylsulfonylamino)-β-alanine-hydrochloride (3-4)
HCl gas was rapidly bubbled through a suspension of acid 3=3 (4.0 g, 10.81 mmol) in EtOH (δO ml) at 0°C for 10 minutes. The cooling bath was removed and the reaction was heated to 60°C. After 18 h, the reaction was concentrated to provide ester 3=4 as a white solid. iH NMR (300 MHz, CD3OD) δ 7.98 (d, 2H, J=8Hz), 7.63 (d, 2H, J=8Hz),
4.2δ (q, IH, J=δHz), 3.92 (m, 2H), 3.33 (m, IH), 3.06 (m, IH), 1.01 (t, 3H, J=7Hz). Ethyl 4-r2-(2-Aminopyridin-6-yl)ethyllbenzoate (3-5)
A mixture of ester 3-δa (700 mg, 2.63 mmol), (for preparation, see: Scheme 29 of PCT International Application Publication No. WO 9δ/32710, published December 7, 1995) 10% Pd/C (350 mg) and EtOH were stirred under 1 atm H2- After 20 h, the reaction was filtered through a celite pad and then concentrated to provide ester 3=δ as a brown oil.
TLC Rf = 0.23 (silica, 40% EtOAc/hexanes) iH NMR (300 MHz, CDCI3) δ 7.9δ (d, 2H, J=8Hz), 7.26 (m, 3H), 6.43 (d, IH, J=7Hz), 6.3δ (d, IH, J=8Hz), 4.37 (m, 4H), 3.0δ (m, 2H), 2.91 (m, 2H), 1.39 (t, 3H, J=7Hz).
4-r2-(2-Aminopyridin-6-yl)ethvnbenzoic acid hvdrochloride (3-6)
A suspension of ester 3=δ (62δ mg, 2.31 mmol) in 6N HCl (12 ml) was heated to 60°C. After ~20 h, the reaction was concentrated to give acid 3=6 as a tan solid. iH NMR (300 MHz, CD3OD) δ 7.96 (d, 2H, J=8Hz), 7.80 (m, IH), 7.33 (d,
2H, J=8Hz), 6.84 (d, IH, J=9Hz), 6.69 (d, IH, J=7Hz), 3.09 (m, 4H).
Ethyl 4-[2-(2-Aminopyridin-6-yl)ethyl]benzoyl-2(S)-(4-iodo- phenylsulfonylamino)-β-alanine (3-7)
A solution of acid 3=6 (400 mg, 1.43 mmol), amine __4 (686 mg, 1.57 mmol), EDC (358 mg, 1.86 mmol), HOBT (252 mg, 1.86 mmol), NMM (632 μl, 5.72 mmol) and DMF (10 ml) was stirred for -20 h. The reaction was diluted with EtOAc and then washed with sat NaHCθ3, brine, dried (MgSθ4) and concentrated. Flash chromatography (silica, EtOAC to 5% isopropanol/EtOAc) provided amide 3=7 as a white solid. TLC Rf = 0.4 (silica, 10% isopropanol/EtOAc) iH NMR (300 MHz, CD3OD) δ 7.79 (d, 2H, J=9Hz) 7.61 (d, 2H, J=8Hz), 7.δ2 (d, 2H, J=9Hz), 7.29 (m, IH), 7.27 (d, 2H, J=8Hz), 4.20 (m, IH), 3.9δ (q, 2H, J=7Hz), 3.66 (dd, IH, J=6Hz, 14Hz), 3.49 (dd, IH, J=8Hz, 13Hz), 3.01 (m, 2H), 2.86 (m, 2H), 1.08 (t, 3H, J=7Hz).
- 4δ 4-[2-(2-Aminopyridin-6-yl)ethyι]benzoyl-2(S)-(4-iodophenyl- sulfonylamino)-β-alanine (3-8)
A solution of ester 3=7 (200 mg, 0.3213 mmol) and 6N HCl (30 ml) was heated to 60°C. After ~20 h, the reaction mixture was concentrated. Flash chromatography (silica, 20:20:1:1 EtOAc/EtOH/ NH4OH/H2O) provided acid 3=8 as a white solid. TLC Rf = 0.4δ (silica, 20:20:1:1 EtOAc/EtOH/NH4θH/H2θ) iH NMR (400 MHz, DMSO) δ 8.40 (m, IH), 8.14 (Bs, IH), 7.81 (d, 2H, J=8Hz), 7.62 (d, 2H, J=8Hz), 7.48 (d, 2H, J=8Hz), 7.27 (m, 3H), 6.34 (d, IH, J=7Hz), 6.2δ (d, IH, J=8Hz), δ.8δ (bs, 2H), 3.89 (bs, IH), 3.3δ (m, 2H), 2.97 (m, 2H), 2.79 (m, 2H).
4-[2-(2-Aminopyridin-6-yl)ethyl)benzoyl-2(S)-(4-trimethylstannyl- phenylsulfonylamino-β-alanine (3-9) A solution of iodide 3=8 (70 mg, 0.1178 mmol), (CH3Sn)2
(49 μl, 0.23δ6 mmol), Pd(PPh3)4 (δ mg) and dioxane (7 ml) was heated to
90°C. After 2 h, the reaction was concentrated and then purified by prep HPLC (Delta-Pak Cl8 lδ μM 100A°> 40 x 100 mm; 9δ:δ /£ δ:95 H2O/CH3CN) provided the trifluoroacetate salt. The salt was suspended in H2O (10 ml), treated with NH4OH (5 drops) and then lyophilized to provide amide 3=9 as a white solid. iH NMR (400 MHz, DMSO) δ 8.40 (m, IH), 8.18 (d, IH, J=8Hz), 7.67 (m, δH), 7.δ6 (d, 2H, J=8Hz), 7.29 (d, 2H, J=8Hz), 6.9δ-7.δ2 (m, 2H), 6.4δ (bs, 2H), 4.00 (m, IH), 3.δ0 (m, IH), 3.33 (m, IH), 2.97 (m, 2H), 2.86 (m, 2H).
4-[2-(2-Anιinopyridin-6-yl)ethyl]benzoyl-2(S)-4-l25iodo- phenylsulfonylamino-β-alanine (3-10)
An iodobead (Pierce) was added to a shipping vial of δ mCi of Nal25l (Amersham, IMS30) and stirred for five minutes at room temperature. A solution of 0.1 mg of 3=9 in O.Oδ mL of 10% H2Sθ4/MeOH was made and immediately added to the Nal25l/i0dobead vial. After stirring for three minutes at room temperature, approximately 0.04-0.0δ mL of NH4OH was added so the reaction mixture was at pH 6-7. The entire reaction mixture was injected onto the HPLC for purification [Vydac peptide-protein C-18 column, 4.6 x 2δ0 mm, linear gradient of 10% acetonitrile (0.1% (TFA):H2θ (0.1% TFA) to 90% acetonitrile (0.1% TFA):H2θ (0.1% TFA) over 30 minutes, 1 mlVmin]. The retention time of 3-10 is 17 minutes under these conditions. Fractions containing the majority of the radioactivity were pooled, lyophilized and diluted with ethanol to give approximately 1 mCi of 3-10. which coeluted on HPLC analysis with an authentic sample of 3-8.
Instrumentation: Analytical and preparative HPLC was carried out using a Waters 600E Powerline Multi Solvent Delivery System with 0.1 mL heads with a Rheodyne 7125 injector and a Waters 990 Photodiode Array Detector with a Gilson FC203 Microfraction collector. For analytical and preparative HPLC a Vydac peptide-protein C-18 column, 4.6 x 250 mm was used with a C-18 Brownlee modular guard column. The acetonitrile used for the HPLC analyses was Fisher Optima grade. The HPLC radiodetector used was a Beckman 170 Radioisotope detector. A Vydac C-18 protein and peptide column, 3.9 x 2δ0 mm was used for analytical and preparative HPLC. Solutions of radioactivity were concentrated using a Speedvac vacuum centrifuge. Calibration curves and chemical concentrations were determined using a Hewlett Packard Model 84δ2A UV/Vis Diode Array Spectrophotometer. Sample radioactivities were determined in a Packard Aδδ30 gamma counter.
The test procedures employed to measure αvβ3 binding and the bone resorption inhibiting activity of the compounds of the present invention are described below. In addition, the activity of the compounds of the present invention for treating cancer and/or inhibiting tumor growth is confirmed utilizing the nude mouse tumor xenograft assay described in Kohl et al., PNAS 91 (1994) 9141-45. BONE RESORPTION-PIT ASSAY
When osteoclasts engage in bone resorption, they will literally cause the formation of pits in the surface of bone that they are acting upon. Therefore, when testing compounds for their ability to inhibit osteoclasts, it is useful to measure the ability of osteoclasts to excavate these resorption pits when the inhibiting compound is present.
Consecutive 200 micron thick cross sections from a six mm cylinder of bovine femur diaphysis were cut with a low speed diamond saw (Isomet, Beuler, Ltd., Lake Bluff, II). Bone slices were pooled, placed in a 10% ethanol solution and refrigerated until further use.
Prior to experimentation, bone slices were ultrasonicated twice, 20 minutes each in H2O. Cleaned slices were placed in 96 well plates such that two control lanes and one lane for each drug dosage are available. Each lane represents either triplicate or quadruplicate cultures. The bone slices in 96 well plates were sterilized by UV irradiation. Prior to incubation with osteoclasts, the bone slices were hydrated by the addition of 0.1 ml Medium 199, pH 6.9 containing 15% fetal bovine serum and 1% penicillin/streptomycin.
Osteoclasts were isolated from the long bones of 1 to 3 day old rat pups (Sprague-Dawley) by modifications of Chambers et al.. (<__ Cell. Science. 66:383-399). The resulting suspension (0.75 ml/bone) was gently triturated 90-120 times using a wide bore transfer pipet. The cellular population was separated from bone fragments by a cell strainer with a 100 micron nylon mesh. 100 μl of the cell suspension was placed onto each bone slice. Test compounds were then added at the desired experimental concentrations.
Bone slices exposed to osteoclasts for 20-24 firs were processed for staining. Tissue culture media was removed from each bone slice. Each well was washed with 200 μl of H2O, and the bone slices were then fixed for 20 minutes in 2.5% glutaraldehyde, 0.1 M cacodylate, pH 7.4. After fixation, any remaining cellular debris was removed by 2 min. ultrasonication in the presence of 0.25 M NH4OH followed by 2 X 15 min ultrasonication in H2O. The bone slices were immediately stained for 6-8 min with filtered 1% toluidine blue and 1% borax. After the bone slices have dried, resorption pits were counted in test and control slices. Resorption pits were viewed in a Microphot Fx (Nikon) fluorescence microscope using a polarizing Nikon IGS filter cube. Test dosage results were compared with controls and resulting IC50 values were determined for each compound tested.
The appropriateness of extrapolating data from this assay to utility and use in mammalian (including human) disease states is supported by the teaching found in Sato, M., et al., Journal of Bone and Mineral Research. Vol. 5, No. 1, 1990. That article teaches that certain bisphosphonates have been used clinically and appear to be effective in the treatment of Paget's disease, hypercalcemia of malignancy, osteolytic lesions produced by bone metastases, and bone loss due to immobilization or sex hormone deficiency. These same bisphosphonates are then tested in the resorption pit assay described above to confirm a correlation between their known utility and positive performance in the assay.
EIB ASSAY
Duong et al., J. Bone Miner. Res.. 8:S 378, describe a system for expressing the human integrin αvβ3. It has been suggested that the integrin stimulates attachment of osteoclasts to bone matrix, since antibodies against the integrin, or RGD-containing molecules, such as echistatin (European Publication 382 4δl), can effectively block bone resorption.
Reaction Mixture:
1. 17δ μl TBS buffer (δO mM Tris-HCl pH 7.2, lδO mM NaCl, 1% BSA, 1 mM CaCl2, 1 mM MgCl2).
2. 2δ μl cell extract (dilute with 100 mM octylglucoside buffer to give 2000 cpm/2δ μl).
3. 125l-echistatin (2δ μl/δ0,000 cpm) (see EP 382 4δl).
4. 2δ μl buffer (total binding) or unlabeled echistatin (nonspecific binding). The reaction mixture was then incubated for 1 h at room temp. The unbound and the bound αvβ3 were separated by filtration using a Skatron Cell Harvester. The filters (prewet in 1.5% poly- ethyleneimine for 10 mins) were then washed with the wash buffer (50 mM Tris HCl, lmM CaC /MgC , pH 7.2). The filter was then counted in a gamma counter.
SPA ASSAY
MATERIALS:
1. Wheatgerm agglutinin Scintillation Proximity Beads (SPA): Amersham
2. Octylglucopyranoside: Calbiochem 3. HEPES: Calbiochem
4. NaCl: Fisher
5. CaCl2: Fisher
6. MgCl2: SIGMA
7. Phenylmethylsulfonylfluoride (PMSF): SIGMA 8. Optiplate: PACKARD
9. 3=1Q (specific activity δ00-1000 Ci/mmole)
10. test compound
11. Purified integrin receptor: αvβ3 was purified from 293 cells overexpressing αvβ3 (Duong et al., J. Bone Min. Res., S:S378, 1993) according to Pytela (Methods in Enzymology, 144:475,
1987)
12. Binding buffer: δO mM HEPES, pH 7.8, 100 mM NaCl, 1 mM Ca2+/Mg2+, O.δ mM PMSF
13. δO mM octylglucoside in binding buffer: δO-OG buffer
PROCEDURE:
1. Pretreatment of SPA beads: δOO mg of lyophilized SPA beads were first washed four times with 200 ml of δO-OG buffer and once with 100 ml of binding buffer, and then resuspended in 12. δ ml of binding buffer.
- δO - 2. Preparation of SPA beads and receptor mixture
In each assay tube, 2.δ μl (40 mg/ml) of pretreated beads were suspended in 97. δ μl of binding buffer and 20 ml of δO-OG buffer, δ μl (~30 ng/μl) of purified receptor was added to the beads in suspension with stirring at room temperature for 30 minutes. The mixture was then centrifuged at 2,δ00 rpm in a Beckman GPR Benchtop centrifuge for 10 minutes at 4°C. The pellets were then resuspended in δO μl of binding buffer and 2δ μl of δO-OG buffer.
3. Reaction
The following were sequentially added into Optiplate in corresponding wells: (i) Receptor/beads mixture (75 μl)
(ii) 25 μl of each of the following: compound to be tested, binding buffer for total binding or 3=8 for non-specific binding (final concentration 1 μM) (iii) 3-10 in binding buffer (25 μl, final concentration 40 pM) (iv) Binding buffer (125 μl)
(v) Each plate was sealed with plate sealer from PACKARD and incubated overnight with rocking at 4°C
4. Plates were counted using PACKARD TOPCOUNT
5. % inhibition was calculated as follows: A = total counts B = nonspecific counts C = sample counts % inhibition = [{(A-B)-(C-B)}/(A-B)]/(A-B) x 100
δl OCFORM ASSAY
Osteoblast-like cells (1.8 cells), originally derived from mouse calvaria, were plated in CORNING 24 well tissue culture plates in α MEM medium containing ribo- and deoxyribonucleosides, 10% fetal bovine serum and penicillin- streptomycin. Cells were seeded at
40,000/well in the morning. In the afternoon, bone marrow cells were prepared from six week old male Balb/C mice as follows:
Mice were sacrificed, tibiae removed and placed in the above medium. The ends were cut off and the marrow was flushed out of the cavity into a tube with a 1 mL syringe with a 27. δ gauge needle.
The marrow was suspended by pipetting up and down. The suspension was passed through >100 μm nylon cell strainer. The resulting suspension was centrifuged at 3δ0 x g for seven minutes. The pellet was resuspended, and a sample was diluted in 2% acetic acid to lyse the red cells. The remaining cells were counted in a hemacytometer. The cells were pelleted and resuspended at 1 x 10^ cells/mL. δO μL was added to each well of 1.8 cells to yield δ0,000 cells/well and l,2δ-dihydroxy-vitamin D3(D3) was added to each well to a final concentration of 10 nM. The cultures were incubated at 37°C in a humidified, δ% Cθ2 atmosphere. After 48 h, the medium was changed. 72 h after the addition of bone marrow, test compounds were added with fresh medium containing D3 to quadruplicate wells. Compounds were added again after 48 h with fresh medium containing D3. After an additional 48 h the medium was removed, cells were fixed with 10% formaldehyde in phosphate buffered saline for 10 minutes at room temperature, followed by a 1-2 minute treatment with ethanol: acetone (1:1) and air dried. The cells were then stained for tartrate resistant acid phosphatase as follows:
The cells were stained for 10- lδ minutes at room temperature with δO mM acetate buffer, pH 5.0 containing 30 mM sodium tartrate, 0.3 mg/mL Fast Red Violet LB Salt and 0.1 mg/mL Naphthol AS -MX phosphate. After staining, the plates were washed extensively with deionized water and air dried. The number of multinucleated, positive staining cells were counted in each well. EXAMPLE OF A PHARMACEUTICAL FORMULATION
As a specific embodiment of an oral composition, 100 mg of compound 1 (shown in Table 1) is formulated with sufficient finely divided lactose to provide a total amount of δ80 to δ90 mg to fill a size O hard gel capsule.
While the invention has been described and illustrated in reference to certain preferred embodiments thereof, those skilled in the art will appreciate that various changes, modifications and substitutions can be made therein without departing from the spirit and scope of the invention. For example, effective dosages other than the preferred doses as set forth hereinabove may be applicable as a consequence of variations in the responsiveness of the mammal being treated for severity of bone disorders caused by resorption, or for other indications for the compounds of the invention indicated above. Likewise, the specific pharmacological responses observed may vary according to and depending upon the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. It is intended, therefore, that the invention be limited only by the scope of the claims which follow and that such claims be interpreted as broadly as is reasonable.

Claims (1)

  1. WHAT IS CLAIMED IS:
    1. A compound of the formula
    X-Y-Z-Aryl-A-B
    wherein:
    Aryl is a 6-membered aromatic ring containing 0, 1, 2 or 3 nitrogen atoms and either unsubstituted or substituted with R^ and R^;
    X is selected from
    NR1 NR2
    -aryl-NR1 R2, -aryl-C-NR2R3, -aryl-NR1-C-NR3R'
    a 5- or 6-membered monocyclic aromatic or nonaromatic ring system containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O or S wherein the δ- or 6-membered ring system is either unsubstituted or substituted with Rl, R2, R3 and R4, or
    a 9- to 14-membered polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2, 3 or 4 heteroatoms selected from N, O or S, and wherein the polycyclic ring system is either unsubstituted or substituted with Rl, R2, R3 and R4 ;
    Y is selected from
    Cθ-8 alkylene, C3-10 cycloalkyl, Cθ-8 alkylene-NRl°-CO-Co-8 alkylene,
    Co-8 alkylene-CONRl0-Co-8 alkylene,
    Cθ-8 alkylene-O-Cθ-8 alkylene,
    Cθ-8 alkylene-NRl°-Cθ-8 alkylene, Cθ-8 alkylene-S(0)θ-2-Cθ-8 alkylene,
    Co-8 alkylene-S02-NRl°-Co-8 alkylene,
    Cθ-8 alkylene-NRl°-S02-Co-8 alkylene,
    Cθ-8 alkylene-CO-Cθ-8 alkylene,
    (CH2)0-6 aryl(CH2)0-6, (CH2)0-6 aryl-CO-(CH2)0-6,
    (CH2)0-6 aryl-CO-NRlO-(CH2)0-6,
    (CH2)0-6 arylNRlOCO(CH2)θ-6, or
    OR1 I (C H2)0-8 C H (C H2)0-8
    Z and A are each independently selected from
    O
    (CH2)m, (CH2)mO(CH2)n, (CH2)mNR1 1 (CH2)n,(CH2)mNR1 1CNR12(CH2)r
    O O O
    11 11 1111 IL
    (CH2)mCNR1 1 (CH2)n,(CH2)mNR1 1 C(CH2)n>(CH2)m6(CH2)n,
    S
    (CH2)mC(CH2)n> (CH2)mS02(CH2)n, (CH2)mS(CH2)
    (CH2)mSO(CH2)n] (CH2)mS02NR1 1(CH2)
    (CH2)mNR11S02(CH2)n, (CH2)mCR1 1=CR12(CH2)n, or (CH2)mC≡C- (CH2)n,
    where m and n are each independently an integer from 0 to 6;
    B is
    - δδ -
    where p is an integer from 1 to 3;
    Rl, R2, R3, R4, R5} R8 R9, RIO, R11 and Rl2 are each independently selected from hydrogen, halogen,
    Ci-io alkyl, aryl Cθ-8 alkyl, amino Cθ-8 alkyl,
    Cl-3 acylamino Cθ-8 alkyl,
    Cl-6 alkylamino Cθ-8 alkyl,
    Cl-6 dialkylamino Cθ-8 alkyl, aryl Cθ-6 alkylamino Cθ-6 alkyl, Cl-4 alkoxyamino Cθ-8 alkyl, hydroxy Cl-6 alkylamino Cθ-8 alkyl,
    Cl-4 alkoxy Cθ-6 alkyl, hydroxycarbonyl Cθ-6 alkyl,
    Cl-4 alkoxycarbonyl Cθ-6 alkyl, hydroxycarbonyl Cθ-6 alkyloxy, hydroxy Cl-6 alkylamino Cθ-6 alkyl. or hydroxy Cθ-6 alkyl;
    R6 is selected from hydrogen, fluorine, Cl-8 alkyl, hydroxyl, hydroxy Cl-6 alkyl, carboxy Cθ-6 alkyl,
    - δ6 - Cl-6 alkyloxy,
    Cl-6 alkylcarbonyl, aryl Cθ-6 alkylcarbonyl,
    Ci-6 alkylcarbonyloxy, aryl Cθ-6 alkylcarbonyloxy,
    Cl-6 alkylaminocarbonyloxy,
    C3-8 cycloalkyl, aryl Cθ-6 alkyl,
    Cø-6 alkylamino Cθ-6 alkyl, Cθ-6 dialkylamino Cθ-6 alkyl,
    Cl-8 alkylsulfonylamino Cθ-6 alkyl, aryl Cθ-6 alkylsulfonylamino Cø-6 alkyl,
    Cl-8 alkyloxycarbonylamino Cθ-8 alkyl, aryl Cθ-8 alkyloxycarbonylamino Cθ-8 alkyl, Cl-8 alkylcarbonylamino Cθ-6 alkyl, aryl Cθ-6 alkylcarbonylamino Cø-6 alkyl,
    Cθ-8 alkylaminocarbonylamino Cø-6 alkyl, aryl Cθ-8 alkylaminocarbonylamino Cø-6 alkyl,
    Cθ-8 alkylaminosulfonylamino Cø-6 alkyl, aryl Cθ-8 alkylaminosulfonylamino Cθ-6 alkyl,
    Cl-6 alkylsulfonyl Cθ-6 alkyl, aryl Cθ-6 alkylsulfonyl Cθ-6 alkyl,
    Cl-6 alkylcarbonyl Cθ-6 alkyl, aryl Cø-6 alkylcarbonyl Cθ-6 alkyl, Cl-6 alkylthiocarbonylamino Cθ-6 alkyl, or aryl Cø-6 alkylthiocarbonylamino CQ_6 alkyl; wherein the alkyl or N atoms may be unsubstituted or substituted with R°;
    elected from
    C7-20 polycyclyl Cθ-8 alkylsulfonylamino Cø-6 alkyl; C7-20 polycyclyl Cθ-8 alkylcarbonylamino Cθ-6 alkyl; C7-20 polycyclyl Cθ-8 alkylaminosulfonylamino Cø-6 alkyl; C7-20 polycyclyl Crj-8 alkylaminocarbonylamino CQ-6 alkyl or
    δ7 C7-20 polycyclyl Cθ-8 alkyloxycarbonylamino Cθ-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with
    Rl4, Rl5; R16 an(j Rl7- an(j wherein any of the alkyl groups may be unsubstituted or subsituted with Rl4 and Rl5;
    Rl3 is selected from hydroxy,
    Cl-8 alkyloxy, aryl Cθ-6 alkyloxy, Cl-8 alkylcarbonyloxy Cl-4 alkyloxy, aryl Cl-8 alkylcarbonyloxy Cl-4 alkyloxy,
    Cl-6 dialkylaminocarbonylmethyloxy, aryl Cl-6 dialkylaminocarbonylmethyloxy or an L- or D-amino acid joined by an amide linkage and wherein the carboxylic acid moiety of said amino acid is as the free acid or is esterified by Cl-6 alkyl; and
    Rl4, Rl5, Rl6 and Rl ' are each independently selected from hydrogen, halogen, Ci-io alkyl, C3-8 cycloalkyl, oxo, aryl, aryl Cl-8 alkyl, amino, amino Cl-8 alkyl, Cl-3 acylamino,
    Cl-3 acylamino Cl-8 alkyl, Cl-6 alkylamino, Cl-6 alkylamino- Ci-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino Cl-8 alkyl, Ci-4 alkoxy, Cl-4 alkoxy Cl-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cl-6 alkyl, Cl-3 alkoxycarbonyl, Cl-3 alkoxycarbonyl Ci-6 alkyl, hydroxycarbonyl-
    Ci-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, Cl-6 alkyloxy- Ci-6 alkyl, nitro, cyano, trifluoromethyl, trifluoromethoxy, trifluoroethoxy, Ci-8 alkyl-S(0)q, Cl-8 alkylaminocarbonyl, Cl-8 dialkylaminocarbonyl, Cl-8 alkyloxycarbonylamino, Cl-8 alkylaminocarbonyloxy or Ci-8alkylsulfonylamino;
    and the pharmaceutically acceptable salts thereof; provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
    2. The compound of Claim 1, wherein
    Aryl is a 6-membered aromatic ring containing 0, 1 or 2 nitrogen atoms wherein Aryl is unsubstituted or substituted with R& and R ;
    X is selected from
    NR1 NR2
    -NR1R2, -C-NR2R3, -NR1-C-NR3R4 ,
    a δ- or 6-membered monocyclic aromatic or nonaromatic ring system containing 0, 1 or 2 heteroatoms selected from N, O or S wherein the δ- or 6-membered ring system is either unsubstituted or substituted with Rl and R2, or
    a 9- to 14-membered fused polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2 or 3 heteroatoms selected from N, O or S, and wherein the polycyclic ring system is either unsubstituted or substituted with Rl and R2;
    Y is selected from the group consisting of Cθ-8 alkylene,
    C3-10 cycloalkyl,
    Co-8 alkylene-NRlO-CO-Co-8 alkylene,
    Co-8 alkylene-CONRlO-Co-8 alkylene,
    Cθ-8 alkylene-O-Cθ-8 alkylene, Co-8 alkylene-NRlO-Co-8 alkylene,
    Cθ-8 alkylene-S(0)o-2-Co-8 alkylene,
    Co-8 alkylene-S02-NRlO-Co-8 alkylene, Co-8 alkylene-NRl°-S02-Co-8 alkylene, Cθ-8 alkylene-CO-Cθ-8 alkylene, (CH2)0-6 aryl(CH2)0-6, (CH2)0-6 aryl-CO-(CH2)0-6, (CH2)0-6 aryl-CO-NH-(CH )0-6, or
    OR1
    I
    (CH2)o-8 H(CH2)o-8
    Z and A are each independently selected from
    (CH2)m, (CH2)mO(CH2)n, (CH2)mNR11(CH2)n,
    O O O
    (CH2)mC l!NR 1111(CH2)(CH2)mNR n11C11(CH2)n,(CH2)mC II(CH2)n]
    (CH2)n,
    (CH2)mSO(CH2) (CH2)mS02NR11(CH2)n;
    (CH2)mNR11S02(CH2)n> (CH2)mCR11=CR12(CH2)n or (CH2)mC≡C-(CH2)n,
    where m and n are each independently an integer from 0 to 4;
    Rl, R2, R3, R4, R5, R8, R9, RIO, R11 and Rl2 are each independently selected from hydrogen, halogen,
    Ci-io alkyl, aryl Cθ-8 alkyl, amino Cθ-8 alkyl,
    Cl-3 acylamino Cθ-8 alkyl, Cl-6 alkylamino Cθ-8 alkyl,
    Cl-6 dialkylamino Cθ-8 alkyl, aryl Cθ-6 alkylamino Cθ-6 alkyl, Cl-4 alkoxyamino Cθ-8 alkyl, hydroxy Cl-6 alkylamino Cθ-8 alkyl, Ci-4 alkoxy Cθ-6 alkyl, or Cl-4 alkoxycarbonyl Cθ-6 alkyl;
    R" is selected from hydrogen, Ci-8 alkyl or aryl Cθ-6 alkyl;
    R ' is selected from
    C7-15 polycyclyl Cθ-6 alkylsulfonylamino Cθ-6 alkyl or C7-15 polycyclyl Cθ-6 alkylcarbonylamino Cθ-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with
    Rl4, R15, R16 an_ R17- an(j wherein any of the alkyl groups may be unsubstituted or subsituted with Rl4 and Rl5;
    and the pharmaceutically acceptable salts thereof;
    provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
    3. The compound of Claim 2, wherein
    Aryl is a phenyl or pyridyl ring which is unsubstituted or substituted with R8;
    X is a 9- to 14-membered fused polycyclic ring system, wherein one or more of the rings is aromatic, and wherein the polycyclic ring system contains 0, 1, 2 or 3 heteroatoms selected from N, O or S, and wherein the polycyclic ring system is either unsubstituted or substituted with Rl and R2;
    Y is selected from Cθ-6 alkylene,
    Cθ-6 alkylene-NRlO-CO-Co-6 alkylene, Cθ-6 alkylene-CONRlO-Co-6 alkylene, Cθ-6 alkylene-O-Cθ-6 alkylene, Cθ-6 alkylene-NRl°-Cθ-6 alkylene, Cθ-6 alkylene-S(0)θ-2-Cθ-6 alkylene,
    Cθ-6 alkylene-Sθ2-NRl°-Cθ-6 alkylene, or Cθ-6 alkylene-aryl-Cθ-6 alkylene;
    Z is selected from
    (CH2)m ι (CH2)mO(CH2)n or (CH2)mNR1 .
    A is
    O (CH2)m »CNR 1111 (CH2)n .
    B is
    Rl, R2, R3, R4, R5, R8, R9, RIO, R11 and Ri2 are each independently selected from hydrogen, halogen, Ci-io alkyl, aryl Cθ-8 alkyl, Cl-4 alkoxy Cθ-6 alkyl, or
    Cl-4 alkoxycarbonyl Cθ-6 alkyl;
    R6 is hydrogen;
    R^ is selected from
    C7-10 polycyclyl Cθ-6 alkylsulfonylamino Cθ-6 alkyl or C7-IO polycyclyl Cθ-6 alkylcarbonylamino Cθ-6 alkyl; wherein the polycyclyl may be unsubstituted or substituted with Rl4, Rl^ Rlβ and Rl?;
    Rl3 is selected from hydroxy or Cl-8 alkyloxy;
    Rl4, Rl } R16 an(j R17 are each independently selected from hydrogen, halogen, Cl-6 alkyl, C3-7 cycloalkyl, oxo, amino, amino Cl-8 alkyl, Cl-3 acylamino, Cl-3 acylamino Cl-8 alkyl, Cl-6 alkylamino, Cl-6 alkylamino Cl-8 alkyl, Cl-6 dialkylamino, Cl-6 dialkylamino Ci-8 alkyl, Cl-4 alkoxy, Cl-4 alkoxy Cl-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Cl-6 alkyl, Cl-3 alkoxycarbonyl, Cl-3 alkoxycarbonyl Cl-6 alkyl, hydroxycarbonyl - Ci-6 alkyloxy, hydroxy, hydroxy Cl-6 alkyl, Cl-6 alkyloxy- Ci-6 alkyl, nitro, cyano, trifluoromethyl, trifluoromethoxy, trifluoroethoxy, Cl-8 alkylsulfonyl, Cl-8 alkylaminocarbonyl,
    Cl-8 alkyloxycarbonylamino, Cl-8 alkylaminocarbonyloxy or Cl-8alkylsulfonylamino;
    and the pharmaceutically acceptable salts thereof; provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
    4. The compound of Claim 3, of the formula:
    wherein X is selected from the group consisting of
    Y is selected from
    Cθ-6 alkylene,
    Cθ-6 alkylene-O-Cθ-6 alkylene or
    Cθ-6 alkylene-NRl°-Cθ-6 alkylene;
    Z is selected from
    (CH2)m or (CH^O- ^H,), ;
    Rl4, Rl5} R16 and R17 are each independently selected from hydrogen, halogen, Cl-6 alkyl, oxo or hydroxy;
    and the pharmaceutically acceptable salts thereof;
    provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ). The compound of Claim 3, of the formula:
    wherein q is an integer from 0 to 2;
    Rl is selected from
    and the pharmaceutically acceptable salts thereof;
    provided that the compound is not 4-[2-(l,2,3,4-Tetrahydro-l,8- naphthyridin-7-yl)ethyl]benzoyl-2(S)[l(S)10-camphorsulfonylamino] β- alanine ethyl ester (1-14) or 4-[2-(l,2,3,4-Tetrahydro-l,8-naphthyridin-7- yl)ethyl]benzoyl-2(S)-[l(S)10-camphorsulfonylamino] β-alanine (1-lδ).
    6. The compound of Claim δ, selected from
    and the pharmaceutically acceptable salts thereof.
    The compound of Claim 4, of the formula
    and the pharmaceutically acceptable salts thereof.
    8. A pharmaceutical composition comprising the compound of Claim 1 and a pharmaceutically acceptable carrier.
    9. A pharmaceutical composition made by combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
    10. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
    11. A method of eliciting a vitronectin antagonizing effect in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1.
    12. The method of Claim 11, wherein the vitronectin antagonizing effect is selected from inhibition of bone resorption, inhibition of restenosis, inhibition of angiogenesis, inhibition of artherosclerosis, inhibition of inflammation, inhibition of viral disease, inhibition of diabetic retinopathy, inhibition of macular degeneration or inhibition of tumor growth.
    13. The method of Claim 12, wherein the vitronectin antagonizing effect is the inhibition of tumor growth.
    14. A method of treating or preventing a condition mediated by antagonism of a vitronectin receptor in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1.
    15. The method of Claim 14, wherein the condition is selected from the group consisting of osteoporosis and cancer.
    16. A method of inhibiting tumor growth in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1.
    17. A method according to Claim 16 wherein said compound is administered in conjunction with radiation therapy.
    18. A method of treating cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1.
    19. A method of eliciting a vitronectin antagonizing effect in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the composition of Claim 8.
    20. A method of treating or preventing a condition mediated by antagonism of a vitronectin receptor in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the composition of Claim 8.
    21. A method of inhibiting tumor growth in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the composition of Claim 8.
    22. A method of treating cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the composition of Claim 8.
    23. A method of treating cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1 and one or more agents known to be cytotoxic or antiproliferative.
    24. A method of inhibiting angiogenesis in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of the compound of Claim 1 and a VEGF inhibitor compound.
    25. A method according to claim 24 which further comprises treating a condition selected from cancer, macular degenration, or diabetic retinopathy.
    26. A composition according to Claim 8 which further comprises an active ingredient selected from the group consisting of a.) an organic bisphosphonate or a pharmaceutically- acceptable salt or ester thereof, b.) an estrogen receptor modulator, and c.) a cytotoxic/antiproliferative agent, and mixtures thereof.
    27. A composition according to Claim 26 wherein said organic bisphosphonate or pharmaceutically-acceptable salt or ester thereof is alendronate monosodium trihydrate.
AU60231/98A 1997-01-17 1998-01-13 Integrin antagonists Ceased AU729869B2 (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US3561497P 1997-01-17 1997-01-17
US60/035614 1997-01-17
GBGB9702788.2A GB9702788D0 (en) 1997-02-11 1997-02-11 Alpha v beta 3 antagonists
GB9702788 1997-02-11
US6259497P 1997-10-20 1997-10-20
US60/062594 1997-10-20
GB9725996 1997-12-09
GBGB9725996.4A GB9725996D0 (en) 1997-12-09 1997-12-09 Intergrin antagonists
PCT/US1998/000617 WO1998031359A1 (en) 1997-01-17 1998-01-13 Integrin antagonists

Publications (2)

Publication Number Publication Date
AU6023198A true AU6023198A (en) 1998-08-07
AU729869B2 AU729869B2 (en) 2001-02-15

Family

ID=27451601

Family Applications (1)

Application Number Title Priority Date Filing Date
AU60231/98A Ceased AU729869B2 (en) 1997-01-17 1998-01-13 Integrin antagonists

Country Status (5)

Country Link
EP (1) EP1007026A4 (en)
JP (1) JP2001509176A (en)
AU (1) AU729869B2 (en)
CA (1) CA2277273C (en)
WO (1) WO1998031359A1 (en)

Families Citing this family (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19629816A1 (en) 1996-07-24 1998-01-29 Hoechst Ag New cycloalkyl derivatives as inhibitors of bone resorption and vitronectin receptor antagonists
US6211191B1 (en) 1997-12-17 2001-04-03 Merck & Co., Inc. Integrin receptor antagonists
US6048861A (en) 1997-12-17 2000-04-11 Merck & Co., Inc. Integrin receptor antagonists
US6066648A (en) 1997-12-17 2000-05-23 Merck & Co., Inc. Integrin receptor antagonists
US6017926A (en) * 1997-12-17 2000-01-25 Merck & Co., Inc. Integrin receptor antagonists
US6313119B1 (en) 1998-01-23 2001-11-06 Adventis Pharma Deutschland Gmbh Sulfonamide derivatives as inhibitors of bone resorption and as inhibitors of cell adhesion
US6372719B1 (en) * 1998-03-04 2002-04-16 Jay Cunningham ανβ3 integrin antagonists in combination with chemotherapeutic agents
GB9805655D0 (en) 1998-03-16 1998-05-13 Celltech Therapeutics Ltd Chemical compounds
US6521626B1 (en) 1998-03-24 2003-02-18 Celltech R&D Limited Thiocarboxamide derivatives
GB9814414D0 (en) 1998-07-03 1998-09-02 Celltech Therapeutics Ltd Chemical compounds
US7008645B2 (en) 1998-07-14 2006-03-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of inhibiting restenosis using bisphosphonates
US6984400B2 (en) 1998-07-14 2006-01-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of treating restenosis using bisphosphonate nanoparticles
IL125336A0 (en) * 1998-07-14 1999-03-12 Yissum Res Dev Co Compositions for inhibition and treatment of restinosis
JP2002521450A (en) * 1998-07-29 2002-07-16 メルク エンド カムパニー インコーポレーテッド Integrin receptor antagonist
DE19842415A1 (en) 1998-09-16 2000-03-23 Merck Patent Gmbh Pharmaceutical preparation for treating e.g. tumors, thrombosis or inflammation, contains cyclic pentapeptide integrin inhibitor and chemotherapeutic agent and/or angiogenesis inhibitor
GB9821061D0 (en) 1998-09-28 1998-11-18 Celltech Therapeutics Ltd Chemical compounds
GB9826174D0 (en) 1998-11-30 1999-01-20 Celltech Therapeutics Ltd Chemical compounds
CA2356748A1 (en) * 1998-12-23 2000-07-06 G.D. Searle & Co. Method of using an integrin antagonist and radiation therapy as combination therapy in the treatment of neoplasia
NZ514661A (en) 1999-04-06 2003-06-30 Sankyo Co alpha-substituted carboxylic acid derivatives and their use as therapeutic agents in pharmaceutical compositions
GB9908355D0 (en) * 1999-04-12 1999-06-09 Rhone Poulenc Rorer Ltd Chemical compounds
GB9910394D0 (en) * 1999-05-05 1999-07-07 Rhone Poulenc Rorer Ltd Chemical compounds
US6518283B1 (en) 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
EP1187592B1 (en) 1999-06-02 2007-08-01 Merck & Co., Inc. Alpha v integrin receptor antagonists
AU5379900A (en) 1999-06-07 2000-12-28 Shire Biochem Inc. Thiophene integrin inhibitors
EP1065207A1 (en) 1999-07-02 2001-01-03 Aventis Pharma Deutschland GmbH Naphthyridine derivatives, processes for their preparation, their use, and pharmaceutical compositions comprising them
EP1065208A1 (en) * 1999-07-02 2001-01-03 Aventis Pharma Deutschland GmbH Substituted purine derivatives as inhibitors of cell adhesion
BR0012683A (en) * 1999-07-21 2002-04-16 American Home Prod Selective bicyclic antagonists for the alpha-beta3 integrin
AU6316900A (en) 1999-08-05 2001-03-05 Meiji Seika Kaisha Ltd. Omega-amino-alpha-hydroxycarboxylic acid derivatives having integrin alphavbeta3antagonism
US6534513B1 (en) 1999-09-29 2003-03-18 Celltech R&D Limited Phenylalkanoic acid derivatives
US6455539B2 (en) 1999-12-23 2002-09-24 Celltech R&D Limited Squaric acid derivates
ES2295150T3 (en) 2000-04-17 2008-04-16 Ucb Pharma, S.A. ENAMINE DERIVATIVES AS CELLULAR ADHESION MOLECULES.
US6403608B1 (en) 2000-05-30 2002-06-11 Celltech R&D, Ltd. 3-Substituted isoquinolin-1-yl derivatives
US6545013B2 (en) 2000-05-30 2003-04-08 Celltech R&D Limited 2,7-naphthyridine derivatives
US6900232B2 (en) 2000-06-15 2005-05-31 Pharmacia Corporation Cycloalkyl alkanoic acids as integrin receptor antagonists
JP2004503540A (en) 2000-06-15 2004-02-05 ファルマシア・コーポレーション Dihydrostilbene alkanoic acid derivatives useful as integrin antagonists
US6638950B2 (en) 2000-06-21 2003-10-28 Bristol-Myers Squibb Pharma Company Piperidine amides as modulators of chemokine receptor activity
JP2004502762A (en) 2000-07-07 2004-01-29 セルテック アール アンド ディ リミテッド Squaric acid derivatives as integrin antagonists containing bicyclic heteroaromatic rings
EP1305291A1 (en) 2000-08-02 2003-05-02 Celltech R&amp;D Limited 3-substituted isoquinolin-1-yl derivatives
US6531494B1 (en) 2001-08-29 2003-03-11 Pharmacia Corporation Gem-substituted αvβ3 antagonists
AU2002316855B2 (en) 2001-04-24 2008-03-13 Merck Patent Gmbh Combination therapy using anti-angiogenic agents and TNFalpha
JP2005516054A (en) 2002-01-29 2005-06-02 ワイス Composition and method for modulating connexin hemichannels
US7365209B2 (en) * 2003-02-11 2008-04-29 Pharmacopeia, Inc. Nitrogen heterocycle biaryls for osteoporosis and other diseases
US10517883B2 (en) 2003-06-27 2019-12-31 Zuli Holdings Ltd. Method of treating acute myocardial infarction
MXPA06003164A (en) * 2003-09-23 2006-06-05 Novartis Ag Combinations of a vegf receptor inhibitor with other therapeutic agents.
CA2637387A1 (en) 2006-01-18 2007-07-26 Simon Goodman Specific therapy using integrin ligands for treating cancer
BRPI0710083A2 (en) 2006-03-31 2011-08-02 Janssen Pharmaceutica Nv benzoimidazol-2-yl pyrimidine and pyrazine chemical entity, pharmaceutical composition and use of these
EP2004188B1 (en) 2006-03-31 2010-09-01 Janssen Pharmaceutica NV Benzoimidazol-2-yl pyridines as modulators of the histamine h4 receptor
WO2008087025A2 (en) 2007-01-18 2008-07-24 Merck Patent Gmbh Specific therapy and medicament using integrin ligands for treating cancer
US8084466B2 (en) 2007-12-18 2011-12-27 Janssen Pharmaceutica Nv Bicyclic heteroaryl-substituted imidazoles as modulators of the histamine H4 receptor
US9371311B2 (en) 2008-06-30 2016-06-21 Janssen Pharmaceutica Nv Benzoimidazol-2-yl pyrimidine derivatives
US20120130146A1 (en) 2009-05-25 2012-05-24 Merck Patent Gmbh Continuous administration of cilengitide in cancer treatments
SI2964229T1 (en) 2013-03-06 2020-02-28 Janssen Pharmaceutica Nv Benzoimidazol-2-yl pyrimidine modulators of the histamine h4 receptor
US9993427B2 (en) 2013-03-14 2018-06-12 Biorest Ltd. Liposome formulation and manufacture
FI3929196T3 (en) 2013-09-24 2023-10-02 Fujifilm Corp Pharmaceutical composition of a nitrogen-containing compound or salt thereof, or metal complex thereof
RU2016146826A (en) 2014-05-30 2018-07-04 Пфайзер Инк. CARBONITRIL DERIVATIVES AS SELECTIVE ANDROGEN RECEPTOR MODULATORS
WO2018049068A1 (en) 2016-09-07 2018-03-15 Pliant Therapeutics, Inc. N-acyl amino acid compounds and methods of use
WO2023275715A1 (en) 2021-06-30 2023-01-05 Pfizer Inc. Metabolites of selective androgen receptor modulators

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2150550A1 (en) * 1992-12-01 1994-06-09 Melissa S. Egbertson Fibrinogen receptor antagonists
US5563158A (en) * 1993-12-28 1996-10-08 The Dupont Merck Pharmaceutical Company Aromatic compounds containing basic and acidic termini useful as fibrinogen receptor antagonists
ES2186720T3 (en) * 1994-05-27 2003-05-16 Merck & Co Inc COMPOSITIONS TO INHIBIT OSEA-CALLAST MEDIATED OSAB REABSORPTION.

Also Published As

Publication number Publication date
EP1007026A4 (en) 2002-08-07
CA2277273A1 (en) 1998-07-23
EP1007026A1 (en) 2000-06-14
JP2001509176A (en) 2001-07-10
CA2277273C (en) 2008-03-25
AU729869B2 (en) 2001-02-15
WO1998031359A1 (en) 1998-07-23

Similar Documents

Publication Publication Date Title
CA2277273C (en) Integrin antagonists
US6017925A (en) Integrin antagonists
AU722360B2 (en) Integrin antagonists
AU717283B2 (en) Integrin antagonists
US6090944A (en) Alkanoic acid derivatives as αv integrin receptor antagonists
AU747784B2 (en) Integrin receptor antagonists
US5919792A (en) Integrin antagonists
AU720758B2 (en) Alphavbeta3 antagonists
US6413955B1 (en) Integrin receptor antagonists
US5952341A (en) Integrin antagonists
AU1912899A (en) Integrin receptor antagonists
AU747503B2 (en) Benzazepine derivatives as alpha-V integrin receptor antagonists
WO2000078317A1 (en) Integrin receptor antagonists
US5925655A (en) αv β3 antagonists
US6211184B1 (en) Integrin antagonists
US6916810B2 (en) αν integrin receptor antagonists
Duggan et al. α v β 3 antagonists
CA2268916A1 (en) Integrin antagonists

Legal Events

Date Code Title Description
SREP Specification republished
FGA Letters patent sealed or granted (standard patent)