[go: up one dir, main page]

HK1147207A - Anti-angiogenic agents and methods of use - Google Patents

Anti-angiogenic agents and methods of use Download PDF

Info

Publication number
HK1147207A
HK1147207A HK11101281.3A HK11101281A HK1147207A HK 1147207 A HK1147207 A HK 1147207A HK 11101281 A HK11101281 A HK 11101281A HK 1147207 A HK1147207 A HK 1147207A
Authority
HK
Hong Kong
Prior art keywords
group
formula
hydrogen
compound
independently
Prior art date
Application number
HK11101281.3A
Other languages
Chinese (zh)
Inventor
屈志参
安舒‧米塔尔‧罗伊
苏布拉马尼亚姆‧阿南坦
Original Assignee
南方研究所
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南方研究所 filed Critical 南方研究所
Publication of HK1147207A publication Critical patent/HK1147207A/en

Links

Abstract

The present disclosure relates generally to treating or preventing diseases associated with angiogenesis by administering to a patient certain compounds found to inhibit or substantially reduce angiogenesis. Compounds employed according to the present disclosure exhibit good anti-angiogenic activity as well as demonstrate a prophylactic effect for preventing and substantially reducing angiogenesis. Examples of such compounds include Ritanserin, Amiodarone, Terfenadinc, Perphenazine, Bithionol, and Clomipramine.

Description

Anti-angiogenic agents and methods of use
Technical Field
The present disclosure relates generally to the treatment or prevention of diseases associated with angiogenesis by administering to a patient certain compounds that have been found to inhibit or substantially reduce angiogenesis. The compounds employed according to the present disclosure exhibit good anti-angiogenic activity and exhibit a prophylactic effect that prevents and substantially reduces angiogenesis. Examples of the compounds include Ritanserin (Ritanserin), Amiodarone (Amiodarone), Terfenadine (Terfenadine), Perphenazine (Perphenazine), thiochlorophene (Bithionol), and Clomipramine (Clomipramine).
Background
Angiogenesis refers to the formation of new blood vessels from pre-existing capillaries and is a series of events that play a key role in many physiological and pathological processes. Normal tissue growth (e.g., during embryonic development, wound healing, and the menstrual cycle) is characterized by the reliance on neovascularization to supply oxygen and nutrients and to remove waste products. A number of different and unrelated diseases are also associated with the formation of neovasculature. Certain conditions are those in which angiogenesis is low and should be elevated to improve the disease condition. Conditions involving insufficient vascularization include peripheral and coronary ischemia and infarction, chronic wound healing failure, and ulcerations. More generally, however, excessive angiogenesis is an important feature of various pathologies, including pathologies characterized by or associated with abnormal or uncontrolled cellular proliferation. Conditions involving excessive angiogenesis include, for example, cancer (both solid and hematologic tumors), cardiovascular diseases (e.g., atherosclerosis and restenosis), chronic inflammation (rheumatoid arthritis, Crohn's disease), diabetes (diabetic retinopathy), psoriasis, endometriosis, neovascular glaucoma, and obesity. See Griffioen (Griffioen) and merlma (Molema), angiogenesis: the possibility of pharmacological Intervention in the Treatment of cancer, Cardiovascular Diseases and Chronic inflammation (Angiogenesis: opportunities for pharmacological interaction in the Treatment of cancer, Cardiovascular Diseases, and Chronic inflammation), pharmacological comments (PHARMACOL. REV.), 52, 237-.
In general, the angiogenic process requires proliferation and migration of normal quiescent endothelium, controlled proteolysis of the extracellular matrix, and synthesis of new extracellular matrix components by capillary formation. The formation of new intracellular and intercellular contacts and the differentiation of endothelial cell morphology into capillary-like tubular networks may provide support for their subsequent maturation, branching, remodeling and selective reversion to form highly ordered functional microvascular networks. Autocrine, paracrine and duplex secretory interactions of the vascular endothelium with its surrounding interstitial components, as well as with pro-and angiostatic cytokines and growth factors that coordinate physiological angiogenesis, are often tightly regulated both spatially and temporally. See, Gauss Parrini (Gasparini), The principle and future Potential of Angiogenesis Inhibitors in Neoplasia (The ratio and future patent Potential of Angiogenesis Inhibitors in neoplasma), DRUGS (DRUGS), 58 (1): 17-38(1999).
The best known anti-angiogenic agents that target endothelial cell proliferation are vascular endothelial growth factor ("VEGF") inhibitors. VEGF is a potent angiogenic growth factor that is overexpressed in most human solid tumors and retina-associated eye diseases. In many pathological angiogenic conditions, VEGF receptors are predominantly enriched in endothelial cells that transduce VEGF signals. Growth-stimulating endothelial cells are also sensitive to tyrosine kinase inhibitors targeting VEGF receptors, such as the FDA's recently approved anti-cancer drugs Sunitinib (Sunitinib) (SU11248) and Sorafenib (Sorafenib) (BAY 43-9006). anti-VEGF and VEGF receptor agents block endothelial cell proliferation and prevent new blood vessel growth. In addition to VEGF, many other growth factors, such as Fibroblast Growth Factor (FGF) and platelet-derived growth factor (PDGF), also play important roles in endothelial activation. Recently, resistance to anti-angiogenic agents targeting only VEGF signaling has emerged, presumably due to alternative signaling pathways mediated by other growth factors.
Angiogenesis has a key role in neoplastic tissue growth. Over 100 years, it has been observed that tumors have more blood vessels than normal tissue. Several experimental studies have demonstrated that neovascularization is required for both primary tumor growth and metastasis. In contrast to the well-coordinated process described above for normal tissue growth, the pathological angiogenesis required for active tumor growth is generally persistent and persistent, with the first acquisition of the angiogenic phenotype being a common mechanism leading to a variety of solid and hematologic tumor types. See foxkman (Folkman, J.), cancer medicine (CANCERMEDICINE), 132-. Tumors that are unable to recruit and maintain the vascular network often remain dormant in the form of an in situ asymptomatic lesion. Metastasis is also angiogenesis-dependent-tumor cells in order to metastasize successfully, they must generally enter the vasculature in the primary tumor, survive the circulation, arrest in the microvasculature of the target organ, leave this vasculature, grow in the target organ, and induce angiogenesis at the target site. Thus, angiogenesis appears to be required at the beginning and completion of the metastatic cascade.
The criticality of angiogenesis to tumor growth and metastasis therefore provides the best possible target for chemotherapeutic work. Suitable anti-angiogenic agents can be used to affect tumor-associated angiogenesis, either directly or indirectly, by delaying the onset of tumor-associated angiogenesis (i.e., blocking the "angiogenic switch") or by blocking the sustained focal neovascularization that is characteristic of multiple tumor types. The safety and efficacy of anti-angiogenic therapies against a variety of molecular and cellular processes and targets involved in tumor-associated endothelium and sustained pathological angiogenesis are actively evaluated in a number of clinical trials. See Delplanque (delphanque) and Harris (Harris), anti-angiogenic agents: in-Development Clinical Trial Design and therapy (Anti-angiogenic Agents: Clinical Trial Design and therapeutics in Development), European cancer journal (EUR. J. CANCER), 36: 1713-1724(2000). However, the number of safe and/or effective anti-angiogenic agents successfully discovered and/or determined to date is limited.
Disclosure of Invention
The present disclosure relates generally to treating or preventing angiogenesis-related diseases.
In one embodiment, provided are methods of inhibiting angiogenesis using compounds of formula (I) and pharmaceutically acceptable salts, solvates, and prodrugs thereof
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl. Alternatively, Alk is 1, 2-ethanediyl.
In one embodiment, the compound of formula (I) is ritanserin.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (II) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine. Or, Y and Y1Identical and selected from hydrogen, fluorine, bromine, chlorine and iodine.
In one embodiment, the compound of formula (II) is amiodarone.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (III) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is a positive integer from 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl.
In one embodiment, the compound of formula (III) is terfenadine.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (IV) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further substituted by one to four (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and includes 0 and 5.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (IV-a) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (IV-a)
Wherein
X represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3 to 8 membered heterocyclic ring;
n is a number between 0 and 5 and includes 0 and 5.
In one embodiment, the compound of formula (IV) or formula (IV-a) is perphenazine.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (V) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5.
Also provided are methods of inhibiting angiogenesis using compounds of formula (V-a) and pharmaceutically acceptable salts, solvates, and prodrugs thereof
Formula (V-a)
Wherein
Y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5.
In one embodiment, the compound of formula (V) or formula (V-a) is thiochlorophenol.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (VI) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (VI)
Wherein
X represents a group selected from ethylene-CH2CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl and N-methyl-piperidinyl (2) -yl.
In one embodiment, the compound of formula (VI) is clomipramine.
Also provided are methods of inhibiting the growth or metastasis of angiogenesis-dependent tumors using the compounds of the invention. Another embodiment relates to methods of treating diseases or disorders associated with angiogenesis, such as neoplastic diseases, restenosis, rheumatoid arthritis, crohn's disease, diabetic retinopathy, psoriasis, endometriosis, macular degeneration, neovascular glaucoma, and obesity, with a compound of the invention.
Drawings
Figure 1 shows a dose response curve for cell proliferation assays performed using ritanserin plotted against concentration.
Figure 2 shows the results of endothelial cell migration analysis performed using ritanserin and plots the percentage of migrated cells relative to control.
Figure 3 shows the results of an endothelial tube formation assay performed using ritanserin and plots the percentage of tubule formation relative to control.
Figure 4 shows the results of in vitro angiogenic CAM analysis performed using ritanserin and plotting the vascular density index.
Figure 5 plots the results of microvascular density in a matrix plug analysis in mice performed using ritanserin.
Figure 6 shows the results of xenograft mouse model analysis performed using ritanserin and plots changes in tumor volume over time.
Figure 7 shows a dose response curve of a cell proliferation assay performed with amiodarone hydrochloride plotted against concentration.
Fig. 8 shows the results of endothelial cell migration analysis performed using amiodarone hydrochloride, and the percentage of migrated cells relative to the control was plotted.
Figure 9 shows the results of an endothelial tube formation assay performed using amiodarone hydrochloride and plots the percentage of tubule formation relative to the control.
Figure 10 shows the results of in vitro angiogenic CAM analysis performed using amiodarone and plotting the vascular density index.
Figure 11 shows a dose response curve for cell proliferation analysis performed using terfenadine plotted against concentration.
Figure 12 shows the results of endothelial cell migration analysis performed using terfenadine, and plots the percentage of migrated cells relative to control.
Figure 13 shows the results of an endothelial tube formation assay performed using terfenadine, and plots the percentage of tubule formation relative to control.
Figure 14 shows the results of in vitro angiogenic CAM analysis performed using terfenadine, and plotting the vascular density index.
Fig. 15 shows a dose response curve for cell proliferation assays performed using perphenazine plotted against concentration.
Fig. 16 shows the results of endothelial cell migration analysis performed using perphenazine, and the percentage of migrated cells relative to control was plotted.
Fig. 17 shows the results of an endothelial tube formation assay performed using perphenazine, and plots the percentage of tubule formation relative to control.
Figure 18 shows a dose response curve for cell proliferation analysis performed using thiochlorophenol plotted against concentration.
Fig. 19 shows the results of endothelial cell migration analysis performed using thiochlorophenol, and the percentage of migrated cells relative to the control was plotted.
Figure 20 shows the results of an endothelial tube formation assay performed using thiochlorophenol, and plots the percentage of tubule formation relative to control.
Figure 21 shows the results of in vitro angiogenic CAM analysis performed using thiochlorophenol and plotting the vascular density index.
Figure 22 shows a dose response curve for cell proliferation analysis performed with clomipramine plotted against concentration.
Fig. 23 shows the results of endothelial cell migration analysis performed using clomipramine, and the percentage of migrated cells relative to the control was plotted.
Figure 24 shows the results of an endothelial tube formation assay performed with clomipramine and plots the percentage of tubule formation relative to control.
Figure 25 shows the results of in vitro angiogenic CAM analysis performed using clomipramine and plots the vascular density index.
Detailed Description
The present disclosure relates generally to treating or preventing angiogenesis-related diseases in a patient by administering to a patient in need thereof certain compounds that have been found to inhibit or substantially reduce angiogenesis.
The following terms used above and throughout this disclosure should be understood to have the following meanings, unless otherwise indicated.
"alkyl" refers to a straight, branched, or cyclic aliphatic hydrocarbon group having from 1 to about 10 carbon atoms in the chain, and all combinations and subcombinations of the ranges therein. "branched" means that lower alkyl groups such as methyl, ethyl, or propyl are attached to the linear alkyl chain. In certain embodiments, alkyl is C1-C4Alkyl, i.e., branched or straight chain alkyl having from 1 to about 4 carbons. In other embodiments, alkyl is C1-C3Alkyl, i.e., branched or straight chain alkyl having from 1 to about 3 carbons. Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl, heptyl, octyl, nonyl, and decyl.
"aryl" refers to an aromatic carbocyclic group containing from about 6 to about 10 carbons, and all combinations and subcombinations of the ranges therein. Aryl groups may be optionally substituted with one or two or more substituents. Exemplary aryl groups include monocyclic groups such as phenyl and bicyclic groups such as naphthyl.
"heteroaryl" refers to aromatic carbocyclic groups containing from about 4 to about 10 atoms, and all combinations and subcombinations of ranges therein, wherein one or more of the atoms is a non-carbon element, such as nitrogen, oxygen, or sulfur. Exemplary heteroaryl groups include monocyclic groups such as pyridyl and bicyclic groups such as indolyl.
"arylalkyl" refers to an alkyl group substituted with an aryl group. For example, "C1-C4Arylalkyl "having C substituted by aryl1-C4An alkyl group.
"heteroarylalkyl" refers to an alkyl group substituted with a heteroaryl group. For example, "C1-C4Heteroarylalkyl "having C substituted by heteroaryl1-C4An alkyl group.
The term "cycloalkyl", used alone or as part of a larger moiety, shall include non-aromatic cyclic C's that are fully saturated or contain one or more units of unsaturation3-C10A hydrocarbon. The cycloaliphatic radical is usually C3-C10More typically C3-C7
The term "non-aromatic heterocyclic ring" used alone or as part of a larger moiety (as in "heterocycloalkyl") refers to a non-aromatic ring system typically having 5 to 14 atoms, preferably 5 to 10 atoms, in which one or more ring carbons, preferably 1 to 4 ring carbons, are each replaced with a heteroatom such as N, O or S.
By "effective amount" is meant an amount of a compound described herein that is therapeutically effective to treat a disease or disorder associated with angiogenesis. The exact amount of these compounds required may vary depending on the following factors: the particular compound used, the age and condition of the subject to be treated, and the nature and severity of the condition. However, the effective amount can be determined by one skilled in the art using only routine experimentation.
"pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound pharmaceutical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
"pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds which are modified by making acid or base salts of the parent compound. The compounds of the present invention form acid and base addition salts with a wide variety of organic and inorganic acids and bases and include the physiologically acceptable salts commonly used in medicinal chemistry. Said salts are also part of the present invention. Typical inorganic acids used to form the salts include hydrochloric, hydrobromic, hydroiodic, nitric, sulfuric, phosphoric, hypophosphoric, and the like. Salts derived from organic acids such as aliphatic mono-and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic acids and hydroxyalkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids may also be used. Thus, such pharmaceutically acceptable salts include acetate, phenylacetate, trifluoroacetate, acrylate, ascorbate, benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-ethoxybenzoate, naphthalene-2-benzoate, bromide, isobutyrate, phenylbutyrate, beta-hydroxybutyrate, butyne-1, 4-dioate, hexyne-1, 4-dioate, caprate, octanoate, chloride, cinnamate, citrate, formate, fumarate, glycolate, heptanoate, hippurate, lactate, malate, maleate, hydroxymaleate, malonate, mandelate, methanesulfonate, nicotinate, isonicotinate, nitrate, oxalate, ascorbate, fluoroxybenzoate, dihydroxybenzoate, nicotinate, nitrate, oxalate, dihydroxybenzoate, dihydro, Phthalates, terephthalates, phosphates, monohydrogen phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, propiolates, propionates, phenylpropionates, salicylates, sebacates, succinates, suberates, sulfates, bisulfates, pyrosulfates, sulfites, bisulfites, sulfonates, benzene-sulfonates, p-bromobenzenesulfonates, chlorobenzenesulfonates, ethanesulfonates, 2-hydroxyethanesulfonates, methanesulfonates, naphthalene-1-sulfonate, naphthalene-2-sulfonate, p-toluenesulfonate, xylenesulfonates, tartrates, and the like.
Bases commonly used to form salts include ammonium hydroxide and alkali and alkaline earth metal hydroxides, carbonates, as well as aliphatic amines and primary, secondary and tertiary amines, aliphatic diamines. Bases that are particularly useful for preparing addition salts include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, methylamine, diethylamine, and ethylenediamine.
By "patient" is meant an animal, including a mammal, preferably a human.
"metabolite" refers to any substance resulting from chemical changes involved in the growth and repair processes of a living organism, including anabolic and catabolic processes.
A "prodrug" is a compound that is converted in vivo to an active form having a medicinal effect. Prodrugs can be used when the active drug may be too toxic for systemic administration, poorly absorbed by the digestive tract, or the body breaks down the active drug before it reaches the target. Methods for making PRODRUGS are disclosed in Hans bandeger (Hans Bundgaard), pharmaceutical DESIGN (desk OF produgs) (Elsevier Science Publishers, b.v., 1985), which is incorporated herein by reference in its entirety.
"solvate" refers to a compound formed by the interaction of a solvent with a solute and includes hydrates. Solvates are generally crystalline solid adducts containing solvent molecules in stoichiometric or non-stoichiometric proportions within the crystal structure.
As used herein, the term "comprising" (and grammatical variants thereof) is used in an inclusive sense of "having" or "including" and not in an exclusive sense of "consisting only of … …". The term "consisting essentially of … …" as used herein is intended to include what is specifically recited and to not materially affect the basic and novel characteristics of the recited or specified material.
As used herein, the terms "a" and "an" and "the" are to be construed to cover the plural as well as the singular, unless the context clearly dictates otherwise.
Certain acidic or basic compounds of the present invention may exist in zwitterionic form. All forms of the compounds, including free acids, free bases, and zwitterions, are intended to be encompassed within the scope of the present invention.
In the formulae described and claimed herein, when any symbol occurs more than one time in a particular formula or substituent, its meaning at each occurrence is intended to be independent of each other.
In one embodiment, provided are methods of inhibiting angiogenesis using compounds of formula (I) and pharmaceutically acceptable salts, solvates, and prodrugs thereof
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl. Alternatively, Alk is 1, 2-ethanediyl.
In one embodiment, the compound of formula (I) is ritanserin.
Methods of synthesizing compounds of formula (I) are well known in the art and are disclosed in U.S. patent No. 4,533,665 to Kennis (Kennis) et al and U.S. patent No. 4,485,107 to Kennis et al, both of which are incorporated herein by reference in their entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (II) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine. Or, Y and Y1Identical and selected from hydrogen, fluorine, bromine, chlorine and iodine.
In certain embodiments, NR3Selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-pyrrolidinyl, and N-morpholinyl; y and Y1The same or different and selected from the group consisting of: hydrogen, iodine, and bromine.
In one embodiment, the compound of formula (II) is amiodarone.
Methods of synthesizing compounds of formula (II) are well known in the art and are disclosed in U.S. patent No. 3,248,401 to tondel (toncur) et al, which is incorporated herein by reference in its entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (III) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl.
In one embodiment, the compound of formula (III) is terfenadine.
Methods of synthesizing compounds of formula (III) are well known in the art and are disclosed in U.S. patent No. 3,878,217 to Carr et al and U.S. patent No. 4,254,129 to Carr et al, both of which are incorporated herein by reference in their entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (IV) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further substituted by one to four (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and includes 0 and 5.
Methods of synthesizing compounds of formula (IV) are well known in the art and are disclosed in U.S. patent No. 2,645,640 to schwareluwa (Choisy-le-Roi), which is incorporated herein by reference in its entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (IV-a) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (IV-a)
Wherein
X represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3 to 8 membered heterocyclic ring;
n is a number between 0 and 5 and includes 0 and 5.
In one embodiment, the compound of formula (IV) or formula (IV-a) is perphenazine.
Methods of synthesizing compounds of formula (IV-a) are well known in the art and are disclosed in U.S. patent No. 2,860,138 to charlock et al, which is incorporated herein by reference in its entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (V) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is an integer between 0 and 5, including 0 and 5.
In one embodiment, the compound of formula (V) or formula (V-a) is thiochlorophenol.
Also provided are methods of inhibiting angiogenesis using compounds of formula (V-a) and pharmaceutically acceptable salts, solvates, and prodrugs thereof
Formula (V-a)
Wherein
Y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5.
Methods of synthesizing compounds of formula (V) and formula (V-a) are well known in the art and are disclosed in U.S. Pat. No. 3,506,720 to Basel et al and U.S. Pat. No. 2,849,494 to Cupressa (Cooper) et al, both of which are incorporated herein by reference in their entirety.
In another embodiment, provided are methods of inhibiting angiogenesis with compounds of formula (VI) and pharmaceutically acceptable salts, solvates, and prodrugs thereof:
formula (VI)
Wherein
X represents a group selected from ethylene-CH2CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl and N-methyl-piperidinyl (2) -yl.
In one embodiment, the compound of formula (VI) is clomipramine.
Methods of synthesizing compounds of formula (VI) are well known in the art and are disclosed in U.S. patent No. 3,467,650 to Riehen et al, which is incorporated herein by reference in its entirety.
In another embodiment, a method of inhibiting angiogenesis with a compound selected from the group consisting of: ritanserin, amiodarone hydrochloride, terfenadine, perphenazine, thiochlorophene, thiobis-dichlorophenol sulfoxide, clomipramine hydrochloride, Fexofenadine (Fexofenadine), and combinations thereof.
Also provided are methods of inhibiting the growth or metastasis of angiogenesis-dependent tumors using the compounds of the invention. Another embodiment relates to methods of treating diseases or disorders associated with angiogenesis, such as neoplastic diseases, restenosis, rheumatoid arthritis, crohn's disease, diabetic retinopathy, psoriasis, endometriosis, macular degeneration, neovascular glaucoma, and obesity, with a compound of the invention.
In certain embodiments, the present disclosure relates to methods of inhibiting angiogenesis and/or inhibiting tumor growth or metastasis. The term "inhibit" as used herein means that the amount of tumor growth or metastasis and/or the incidence of angiogenesis may desirably be reduced in a patient who has received a compound described herein as compared to a patient who has not received the compound. Thus, in one form, the method of inhibition of the present disclosure comprises administering to a patient an effective amount of an anti-angiogenic agent. The term "anti-angiogenic agent" as used herein refers to a compound that inhibits angiogenesis.
In other embodiments, the present disclosure relates to methods of treating diseases or disorders associated with angiogenesis. These methods may include the step of identifying patients suffering from such a disease, including patients who may benefit from the treatment methods described herein. Diseases or disorders associated with angiogenesis include, for example, the following conditions: wherein angiogenesis has a role in the pathology or progression of the condition such that inhibition of angiogenesis in a patient suffering from such a condition may delay or prevent further progression of the condition, or result in the alleviation or regression of the disease state. The condition is typically characterized by or associated with abnormal cell proliferation and includes, for example, neoplastic disease. The term "treating a disease or disorder" as used herein refers to the administration of an agent that is intended to limit the extent, progression and/or severity of a condition in a patient as compared to a patient not undergoing such treatment. The term "neoplastic disease" as used herein refers to any condition characterized by the presence of abnormal growth of abnormal cells or tissues, including, but not limited to, all benign or malignant cancers and tumors. By "treating neoplastic disease" is meant administration of a chemotherapeutic agent that inhibits further growth or metastasis of any neoplastic tissue that may be present in a patient and/or stimulates the tumor to recover, including reducing the size and/or number of the tumor and/or inducing the death of neoplastic cells.
Prodrug forms of compounds having various nitrogen functional groups (amino, hydroxyamino, amide, etc.) may include the following derivative types: wherein each R group may be independently hydrogen, substituted or unsubstituted alkyl, aryl, alkenyl, alkynyl, heterocycle, alkylaryl, arylalkyl, arylalkenyl, arylalkynyl, cycloalkyl or cycloalkenyl as defined above.
(a) Formamide, -NHC (O) R
(b) Carbamate, -NHC (O) OR
(c) (acyloxy) alkyl carbamates, NHC (O) OROC (O) R
(d) Enamine, -NHCR (═ CHCO)2R) or-NHCR (═ CHCONR2)
(e) Schiff Base (Schiff Base), -N ═ CR2
(f) Mannich bases (from the formide compound) RCONHCH2NR2
The preparation of such prodrug derivatives is discussed in a variety of literature sources (examples are: Alexander et al, journal of medicinal chemistry (J.Med.chem.), 1988, 31, 318; Aliges-Martin et al, PCX WO pp/41531, page 30). The nitrogen functionality converted in the preparation of these derivatives is one (or more) of the nitrogen atoms in the compounds of the present invention.
Prodrug forms of the carboxy-containing compounds of the present invention include esters (-CO)2R), wherein the R group corresponds to any alcohol with pharmaceutically acceptable levels released in vivo by enzymatic or hydrolytic processes. Another prodrug derived from the carboxylic acid form of the present invention can be the quaternary structure described in Border et al, journal of medicinal chemistry, 1980, 23, 469
It is of course to be understood that the compounds of the present invention relate to all optical isomers and stereoisomers at each possible atom in the molecule.
Pharmaceutically acceptable salts of the compounds of the present invention include those derived from pharmaceutically acceptable inorganic or organic acids. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, trifluoroacetic and benzenesulfonic acids. Salts derived from suitable bases include alkali metal salts such as sodium and ammonium salts.
The compounds of the present invention can be synthesized by those skilled in the art without undue experimentation once they have become aware of the present disclosure. Procedures suitable for preparing the desired sugar or nucleoside are available in the chemical literature. See Chua Zousina (Choi, Jong-Ryoo) for these principles; jinzhengmin (Kim, Jeong-Min); lukinyin (Roh, Kee-Yoon); zhao Dong Qu (Cho, Dong-Gyu); gold slaughter (Kim, Jae-Hong); yellow slaughter (Hwang, Jae-Taeg); zhaoyuan (Cho, Woo-Young); zhang Tang (Jang, Hyun-Sook); plum, and (Lee, Chang-Ho); zeitacin (Choi, Tae-Saeng); quiet (Kim, Chung-Mi); jinyong ancestor (Kim, Yong-Zu); forsythia (Kim, Tae-Kyun); zhao liter (Cho, Seung-Joo); jinjing yuan (Kim, Gyoung-Won), international patent application (PCT int.appl.) (2002), page 100, WO 0257288 a 120020725. Huoli, Antonin (Holy, Antonin); walibba, ivon (votuba, Ivan); tropsowa, Ewa (Tloustevova, Eva); masojikowa, Milena, Czechosvak Chemical exchanges (Collection of Czechosvak Chemical Communications) (2001), 66(10), 1545-. Leiman, dumini (Rejman, Dominik); masoji kowa, milena; deklier, Eric (De Clercq, Eric); rosenberg, Evan (Rosenberg, Ivan), Nucleosides, Nucleotides and Nucleic Acids (Nucleotides & Nucleic Acids) (2001), 20(8), 1497-1522; laughover (Ubasawa, Masaru); guangxiao Yi (Sekiya, Kouichi), International patent application (2001), page 39, WO 0164693A 120010907. Ottoman, miloslav (Otmar, Miroslav); maxojeff kowa, mielina (Masojfdkova, miltana); walibos, ivon; helley, antonin, Czechslerovak chemical exchange (2001), 66(3), 500-. Micheal (Michal); hocquengk, michael (Hocek, Michal); helley, antonin, Czechslelovap chemical exchange (2000), 65(8), 1357-. Jeffery (a.l.); killing the golden eggs; wilmo (Wiemer, D.F.), Tetrahedron (2000), 56(29), 5077-. Huoli, Antonin; junter, Jaroslav (Guenter, Jaroslav); dove baby, hanna (Dvorakova, Hana); masoji kowa, milena; anderle, glaschela (Andrei, Graciela); snooker, Robert (Snoeck, Robert); bazarini, poplar (Balzarini, Jan); dekrael, eric, Journal of Medicinal Chemistry (1999), 42(12), 2064-. Janeba, zaltork (Janeba, Zlatko); huoli, Antonin; masojikowa, Milena, Czeislavag chemical exchange (2001), 66(9), 1393-. Huoli, Antonin; junte, yarrowia; dove child, hanna; masoji kowa, milena; anderle, gla xira; snooker, robert; bazarini, populus; dekrel, ehek, journal of medicinal chemistry (1999), 42(12), 2064-. Party, group (Dang, Qun); erlilong, Mark D. (Erion, Mark D.); radi m. Robinson, Edward D. (robinson, Edward D.); carscholtra, snevaau (Kasibhatla, Srinivas Rao); leydi, k.lagra (Reddy, k.raja), international patent application (1998), page 126, WO 9839344 a 119980911. Arimly, mortier N. (arigili, Murty N.); kandy, kenniss C (Cundy, Kenneth C); doldi, Joseph P. (Dougherty, Joseph P.); gold god (Kim, chong U.); oriya, rezha (Oliyai, Reza); stella, Valentino J. (Stella, Valentino J.), International patent application (1998), page 74, WO 9804569. Guangxiao I; highlands english (Takashima, Hideaki); rectus plantaris (Ueda, Naoko); gorgon fruit (Kamiya, Naohiro); sourishi (Yuasa, Satoshi); tenmura (Fujimura, Yoshiyuki); laozheng, journal of medicinal chemistry (2002), 45(14), 3138-. The consumptive and lustrous peptides are winy; guangxiao I; gao dao ying Zhao; planting field straight seeds; light clever soup; gorguba, European patent application (Eur. Pat. appl.) (1997), page 56, EP 785208A 119970723. Hokkark, michel; masoji kowa, milena; helley, Antonin, Czechhollovap chemical exchange (1997), 62(1), 136-. Huoli, Antonin; walibos, ivon; tros Towa, Ewa; masoja Korowa, Milena, Czeislavag chemical exchange (2001), 66(10), 1545-. Huoli, Antonin; dekliel, eric, diesel, Alice (Desire Alice), international patent application (1996), page 57, WO 9633200a 119961024. Leiman, dumini; rosenberg, Ilv, Czewslovace chemical exchange (1996), 61 (Spec. issue), S122-S123. Huoli, Antonin; dove child, hanna; jindri, jindri (Jindrich ); masoji kowa, milena; bujeikenski, Milos (budesinky, Milos); bazarini, populus; anderle, glaehara (Andrei, Graciella); de cleel, Ehrick, journal of medicinal chemistry (1996), 39(20), 4073-. Guan Di, Jusseppe (Guanti, Giuseppe); mero, valencia (Merlo, valria); narisolor, Eleca (Narisano, Enrica), tetrahedron (1995), 51(35), 9737-46. Gao dao ying Zhao; uphole straighteners (Inoue, Naoko); the consumptive and lustrous peptides are winy; guangxiao I; buerger (Yabuuchi, Shingo), European patent application (1995), page 88, EP 632048A 119950104. Alexander, Petr; huoli, Antonin; masoji kowa, Milena, Czeislavag chemical exchange (1994), 59(8), 1853-69. Alexander, peter; huoli, Antonin; masoji kowa, milena; czechslevak chemical exchange (1994), 59(8), 1853-69. Goddri, goddri; huoli, Antonin; devocharcot Wawa, Hanna, Czesler Lovack chemical exchange (1993), 58(7), 1645-67. Heli, Antonin, Czechslerovak chemical exchange (1993), 58(3), 649-74. Guanti, Zhu saipei; mylo, vallia; nallisolol, ereeca; tetrahedron (1995), 51(35), 9737-46. Ezetimibe, prussian (emischetti, Purushotham); brodfuehrer, Paul R.); hauer, Henry G. (Howell, Henry G.); sapino, Chester Jr. (Sapino, Chester, Jr.), international patent application (1992), page 43, WO 9202511 a 119920220. Glazier, Arnold (Glazier, Arnold), international patent application (1991), page 131, WO 9119721. Gold Zhongyuan; luobiu (Luh, Bing Yu); miesco, Peter F. (Misco, Peter F.); brownson, geoani j. (Bronson, joane J.); hilbert, Michael J.M (Hitchcock, Michael J.M.); kazuri, ismeil (Ghazzouli, Ismail); martin, John c. (Martin, John C), journal of medicinal chemistry (1990), 33(4), 1207-13. Rosenberg, ivon; huoli, Antonin; masojikowa, Milena, Czeislavack chemical exchange (1988), 53(11B), 2753-77. Rosenberg, ivon; huoli, Antonin; masojikowa, Milena, Czeislavack chemical exchange (1988), 53(11B), 2753-77.
The following non-limiting examples illustrate and describe aspects of the present disclosure. The examples show and describe only limited embodiments, but it is to be understood that the disclosure is capable of use in various other combinations, modifications, and environments and is capable of changes or modifications within the scope of the concept as described herein and commensurate with the teachings and/or skill or knowledge in the relevant art.
Example 1
Cell proliferation assay
Human primary cell lines (arterial endothelia such as HAEC and HPAEC; venous endothelium HUVEC (obtained from Cambrex Bioscience Rockland, Inc.) and lung fibroblasts LL47 (obtained from American Type Culture Collection)) were cultured according to the instructions and used to assess the different activities of target compounds against human endothelium versus fibroblasts by cell titration-Green luminescence cell viability assayCloning and testing of the cytotoxic effects of the compounds. At about 5X 103Cells/well cells were seeded in growth medium in 96-well plates. After 24 hours, various doses of the compound of interest were added to the cultures, each dose being repeated four times. After 72 hours of treatment, cell titer-gruu reagent was added to the culture and the luminosity was measured according to the manufacturer's instructions. Control groups were given DMSO vehicle only. Determination of IC of target Compounds on their proliferation in endothelial and fibroblast cell culture media based on dose-response curves50And plotted against concentration range.
Example 2
Endothelial cell migration assay
Endothelial cell migration is a key step in the angiogenic process, which has a crucial role in the in situ recruitment response of angiogenesis. Endothelial cell migration analysis was performed using a permeable membrane cell filter/insert chamber of the bio-coated (Biocoat) endothelial cell migration angiogenesis system (BD biosciences), a 24-permeable membrane cell plate containing a 3- μm pore size insert coated with human fibronectin. The inserts were incubated with endothelial cell basal medium containing 0.1% bovine serum albumin for 1 hour at 37 ℃. Endothelial Cells (HUVECs) were starved for 4 to 5 hours with 0.1% bovine serum albumin in endothelial cell basal medium, after which the cells were harvested and subsequently seeded (1 × 10)5Per well) in the upper chamber of a permeable membrane well plate, where various treatments were performed in 100 μ l of 0.1% bovine serum albumin in endothelial cell basal medium. Complete growth medium containing various chemoattractants was added to the lower chamber. Cells were allowed to migrate for 22 + -1 hours at 37 deg.C. The non-migrated cells inside the insert were carefully removed with a Q-shaped cotton swab. Migrating cells in the lower part of the permeable membrane chamber insert were then fixed with 4% paraformaldehyde, stained with Hoechst (Hoechst)33342, and photographed under a fluorescent microscope. The number of migrated cells was analyzed in three microscopic fields per filter/insert. Triplicate filter/insert chamber cultures were taken for each concentration tested and control for analysis. Data are presented as each 10 × magnification micrographMean number of migrated cells in the field, and IC was calculated based on dose curves50
Example 3
Endothelial tube formation assay
Human endothelial cells are cultured in vitro on an extracellular matrix, which stimulates ligation and endothelial cell differentiation into tubules. Endothelial tube formation analysis is based on this phenomenon. Endothelial Cells (HUVEC) were seeded (1.5X 10)4/well) in 96-well plates coated with extracellular matrix and treated with different concentrations of the compound of interest and complete growth medium, performed in triplicate. Cells were allowed to form endothelial tubes at 37 ℃ over about 18 hours and photographed under an inverted light microscope. The tubule length was quantified using Image analysis software professional Image processing (Image-Pro Plus) (scientific imaging software (MediaCybernetics), inc., silver Spring (Sivler Spring), MD). Data are expressed as average tube length in three fields of view per well and three wells are taken for analysis for each treatment condition. IC50 values were calculated based on the dose curves.
Example 4
Chick chorioallantoic membrane (CAM) analysis
The chick embryo chorioallantoic membrane provides an ideal in vivo model for the physiological process of angiogenesis. Angiogenesis modulators applied to a methylcellulose disk placed on the chick embryo chorioallantoic membrane alter the development of neovasculature. See Staton (Staton) et al, Current Methods for analyzing angiogenesis in vitro and in vivo (Current Methods for analyzing angiogenesis in vitro and in vivo), journal of international experimental pathology (int.j. exp. path.), 85: 233-48(2004), which is incorporated herein by reference in its entirety. The anti-angiogenic potential of the synthetic compounds was assessed in vitro using the angiogenic chorioallantoic membrane assay. Fertilized chicken eggs were cultured at 37.5 ℃ in a humidified egg incubator with forced air circulation. On day 3 of the embryonic period, the eggs opened and the embryos were transferred to 100-mm3Development was continued in Petri (Petri) plates at 37.5 ℃ in a cell culture incubator. In the embryoAt day 5 of the fetal period, a preformed methylcellulose disk of approximately 2mm diameter is gently planted on top of the chorioallantoic membrane of an embryo, and then test compound in solution or control vehicle is applied on top of the methylcellulose disk. The embryos were cultured in the cell incubator for two more days. Chorioallantoic membranes were examined and neovascularization quantified at day 7 of embryonic life. The therapeutic effect on angiogenesis was assessed by determining the Vascular Density Index (VDI) of each CAM and live chicken embryo. VDI represents the number of intersections of a blood vessel with three equidistant concentric circles over the methylcellulose disc footprint obtained by using specialized image processing software. Data are expressed as mean VDI with standard deviation based on quantitative analysis (N > 5) for each treatment group.
Example 5
Matrix plug analysis in mice
The matrigel plug assay is an in vivo neovascularization assay that is widely used to assess the anti-angiogenic activity of synthetic compounds and recombinant proteins. Matrigel plug experiments were performed using female C57BL/6 mice of about 8-10 weeks of age and a high concentration matrigel matrix. There were four or five mice per treatment group and two matrix plugs per mouse. Matrigel was mixed with 50ng/ml VEGF, 50ng/ml FGFb, and 3ng/ml heparin as an angiogenic stimulator. Different doses of the target compound are mixed with matrigel or administered intraperitoneally or intravenously or orally. Matrigel was injected subcutaneously (500. mu.l matrigel forming one plug) at 4 ℃ to each side of the shaved mice. The injected matrigel can rapidly form a single solid gel plug. The plugs of each group were collected at about two weeks after matrigel inoculation. By inhaling CO2Mice were subjected to painless death and the skin of the mice was pulled open to expose the plug. The intact plugs were removed and fixed in 10% formalin (formalin) for histological analysis. Sections of paraffin-embedded plugs (5 μm thick) were immunostained with antibodies specific for CD31 and H&E, contrast dyeing is carried out. CD 31-positive microvessels were counted throughout the cross-section of each matrigel plug. For each group of mice, about six matrigel plugs were quantitatively analyzed to evaluate the control and compound sitesAny statistically significant difference in microvascular density between treatment groups.
Example 6
Evaluation of antitumor efficacy of Compounds by xenograft nude mouse model
Tumor growth is dependent on angiogenesis. Inhibition of tumor angiogenesis has become an effective treatment for cancer. The anti-tumor activity of potential anti-angiogenic agents was evaluated using a standard xenograft nude mouse model. Human tumor cells or fragments were implanted into 5 to 7 week old nude mice. The target compound was administered to each group of mice intraperitoneally or intravenously or orally. Tumor size and mouse body weight were monitored twice weekly. The mean tumor volume per time point is expressed as the standard deviation for tumor growth time (which is the number of days after xenograft inoculation).
The data shown in the figures and table I below were obtained using a procedure such as that described in the examples above.
Formulations
The compounds of the present invention may be administered as a single therapeutic agent or in combination with multiple therapeutic agents by any conventional means that may be used in conjunction with multiple drugs. They can be administered alone, but are typically administered with a pharmaceutical carrier selected according to the chosen route of administration and standard pharmaceutical practice. The compounds may also be administered with other therapeutic agents such as: interferon (IFN), interferon alpha-2 a, interferon alpha-2 b, Consensus Interferon (CIFN), ribavirin (ribavirin), amantadine, rimantadine (remantadine), interleukin-12, ursodeoxycholic acid (UDCA), and glycyrrhizin.
Pharmaceutically acceptable carriers (e.g., vehicles, adjuvants, excipients, or diluents) described herein are well known to those skilled in the art. Generally, pharmaceutically acceptable carriers are chemically inert to the active compound and have no deleterious side effects or toxicity under the conditions of use. Pharmaceutically acceptable carriers can include polymers and polymer matrices.
The compounds of the present invention can be administered as single therapeutic agents or in combination with multiple therapeutic agents by any conventional method that can be used in conjunction with multiple drugs.
Of course, the dosage administered may vary depending on a variety of known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; age, health, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent therapy; the frequency of treatment; and the desired effect. Daily dosages of the active ingredient in the range of about 0.001 milligrams (mg) to 1000mg per kilogram (kg) of body weight are contemplated, with dosages in the range of 0.1mg/kg to about 60mg/kg being preferred.
Dosage forms (compositions suitable for administration) contain from about 1mg to about 500mg of active ingredient per unit. In these pharmaceutical compositions, the active ingredient is generally present in an amount of about 0.5 to 95% by weight, based on the total weight of the composition.
The active ingredients can be administered orally in solid dosage forms (e.g., capsules, tablets, and powders) or in liquid dosage forms (e.g., elixirs, syrups, and suspensions). It can also be administered parenterally in sterile liquid dosage forms. The active ingredient may also be administered intranasally (nasal drops) or by inhalation of a medicinal powder spray. Other dosage forms may be used, such as transdermal administration, administration via a patch route, or administration in an ointment.
Formulations suitable for oral administration may consist of: (a) liquid solutions, for example an effective amount of the compound dissolved in a diluent such as water, saline or orange juice; (b) capsules, sachets, tablets, lozenges and troches, each containing a predetermined amount of active ingredient in solid or granular form; (c) powder preparation; (d) a suspension in a suitable liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, such as ethanol, benzyl alcohol, propylene glycol, glycerin, and polyvinyl alcohol, with or without the addition of pharmaceutically acceptable surfactants, suspending agents, or emulsifiers. The capsule form may be of the ordinary hard-or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms may include one or more of the following: lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid and other excipients, colorants, diluents, buffering agents, disintegrating agents, wetting agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms may contain the active ingredient in a flavoring agent, typically sucrose and acacia or tragacanth, and the lozenges comprise the active ingredient in an inert base such as gelatin and glycerin, or sucrose and acacia, emulsions and gels containing, in addition to the active ingredient, carriers known in the art.
The compounds of the present invention may be formulated, alone or in combination with other suitable components, into aerosol formulations for administration by inhalation. These aerosol formulations can be placed in pressurized acceptable propellants, such as difluorodichloromethane, propane, and nitrogen. They may also be formulated, for example, as medicaments for non-pressurized preparations in nebulizers or nebulizers.
Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions, which may include suspending agents, solubilizers, thickeners, stabilizers, and preservatives. The compounds can be administered in the form of pharmaceutical carriers and physiologically acceptable diluents, such as sterile liquids or liquid mixtures, including water, saline, aqueous dextrose and related sugar solutions; alcohols, such as ethanol, isopropanol or hexadecanol; glycols, such as propylene glycol or polyethylene glycol (e.g., poly (ethylene glycol) 400); glycerol ketals, such as 2, 2-dimethyl-1, 3-dioxolane-4-methanol; an ether; an oil; a fatty acid; fatty acid esters or glycerides; or acetylated fatty acid glycerides with or without the addition of pharmaceutically acceptable surfactants (such as soaps or detergents), suspending agents (such as pectins, carbomers, methylcellulose, hydroxypropylmethylcellulose or carboxymethylcellulose) or emulsifying agents and other pharmaceutical adjuvants.
Oils that may be used in parenteral formulations include petroleum, animal, vegetable or synthetic oils. Specific examples of oils include peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum, and mineral oil. Suitable fatty acids for parenteral formulations include oleic acid, stearic acid, and isostearic acid. Examples of suitable fatty acid esters are ethyl oleate and isopropyl myristate. Soaps suitable for use in parenteral formulations include fatty alkali metal, ammonium and triethanolamine salts, and suitable detergents include (a) cationic detergents, such as dimethyl dialkyl ammonium halides and alkyl pyridinium halides; (b) anionic detergents such as alkyl, aryl and olefin sulfonates, alkyl, olefin, ether and monoglyceride sulfates, and sulfosuccinates; (c) nonionic detergents such as fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene polypropylene copolymers; (d) amphoteric detergents such as alkyl beta-aminopropionates and 2-alkylimidazoline quaternary ammonium salts; and (e) mixtures thereof.
Solutions of parenteral formulations typically contain from about 0.5% to about 25% by weight of the active ingredient. Suitable preservatives and buffers can be used in the formulations. To minimize or eliminate irritation of the injection site, the composition may contain one or more nonionic surfactants having a Hydrophilic Lipophilic Balance (HLB) between about 12 and about 17. The amount of surfactant in the formulation is between about 5% to about 15% by weight. Suitable surfactants include polyoxyethylene sorbitan fatty acid esters (e.g. sorbitan monooleate) and high molecular weight adducts of ethylene oxide with hydrophobic bases, high molecular weight adducts formed by condensing propylene oxide with propylene glycol.
Pharmaceutically acceptable excipients are also well known to those skilled in the art. The choice of excipient will depend in part on the particular compound and in part on the particular method used to administer the composition. Thus, there are many suitable formulations of the pharmaceutical compositions of the present invention. The following methods and excipients are merely exemplary and are not intended to limit the present invention in any way. Preferably, the pharmaceutically acceptable excipients do not interfere with the action of the active ingredient and do not cause adverse side effects. Suitable carriers and excipients include solvents (e.g., water, alcohols, and propylene glycol), solid absorbents and diluents, surface active agents, suspending agents, tablet binders, lubricants, flavoring agents, and coloring agents.
The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition in which only the addition of the sterile liquid excipient, for example water, for injections, is required immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets. It is well known to those skilled in the art that injectable compositions require an effective pharmaceutical carrier (5, 6). See, bankol (Banker) and charmers (Chalmers), pharmacy and pharmacy practice (PHARMACEUTICS AND PHARMACY PRACTICE), 238-.
Formulations suitable for topical administration include lozenge-shaped tablets containing the active ingredient in a flavoring agent, typically sucrose and acacia or tragacanth; lozenges comprising the active ingredient in an inert matrix, such as gelatin and glycerol, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier; and creams, milks and gels containing, in addition to the active ingredient, carriers known in the art.
In addition, formulations suitable for rectal administration may be presented as suppositories, which are prepared by mixing with a variety of bases such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Suitable pharmaceutical carriers are described in the standard reference text Remington's pharmaceutical Sciences, Mark Publishing Company (Mack Publishing Company) in this field.
In the context of the present invention, the dose administered to an animal (particularly a human) should be sufficient to achieve a therapeutic response in the animal within a reasonable period of time. One skilled in the art will recognize that the dosage may depend on a variety of factors, including the condition of the animal, the weight of the animal, and the severity and stage of the condition being treated.
An appropriate dose is one that produces a concentration of active agent in the patient known to achieve the desired response. The preferred dosage is that amount which provides the greatest inhibition of the condition being treated without difficulty in controlling side effects.
The size of the dose can also depend on the route, timing and frequency of administration, and the presence, nature and extent of any adverse side effects that may accompany the administration of the compound, as well as the desired physiological effect.
Pharmaceutical dosage forms useful for administering the compounds of the present invention are exemplified by:
hard shell capsule
A large number of capsule units are prepared by filling each tablet of a standard two-piece hard gelatin capsule with 100mg of powdered active ingredient, 150mg of lactose, 50mg of cellulose and 6mg of magnesium stearate.
Soft gelatin capsule
A mixture of the active ingredient in a digestible oil, such as soybean oil, cottonseed oil or olive oil, is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100mg of the active ingredient. The capsules are washed and dried. The active ingredient may be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water-miscible pharmaceutical mixture.
Tablet formulation
A large number of tablets are prepared by conventional procedures so that the dosage unit is 100mg of active ingredient, 0.2mg of colloidal silicon dioxide, 5mg of magnesium stearate, 275mg of microcrystalline cellulose, 11mg of starch and 98.8mg of lactose. Suitable aqueous and non-aqueous coatings may be applied to enhance palatability, to provide aesthetic appearance, and to stabilize or delay absorption.
Immediate release tablet/capsule
These dosage forms are solid oral dosage forms prepared by conventional and novel processes. The units can be immediately dissolved and the drug delivered without oral ingestion of the water. The active ingredient is mixed in a liquid containing ingredients such as sugar, gelatin, pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze-drying and solid state extraction techniques. The pharmaceutical compound may be compressed together with viscoelastic and thermoelastic sugars and polymer or effervescent components, resulting in a porous matrix intended for immediate release (without water).
In addition, the compounds of the present invention may be administered as nasal drops, or in metered dose and nasal or oral inhalant forms. The drug is delivered from a nasal solution in the form of a fine mist or from a powder in the form of an aerosol.
The foregoing description of the invention illustrates and describes the present invention. Further, the disclosure shows and describes only the preferred embodiments, but as mentioned above, it is to be understood that the disclosure can be used in various other combinations, modifications, and in other environments and is capable of changes or modifications within the concept described herein and commensurate with the above teachings and/or the skill or knowledge of the relevant art.
The embodiments described hereinabove are further intended to explain best modes known of practicing the invention and to enable others skilled in the art to utilize the disclosure in such, or other, embodiments and with the various modifications required by the particular applications or uses. Accordingly, the description is not intended to limit the invention to the form disclosed herein. Also, it is intended that the appended claims be construed to include alternative embodiments.
All publications, patents, and patent applications cited in this specification are herein incorporated by reference as if each individual publication, patent, or patent application were specifically and individually indicated to be incorporated by reference for all purposes.

Claims (44)

1. A method of inhibiting or reducing angiogenesis in a patient in need thereof, comprising administering to the patient an anti-angiogenic effective amount of a compound of:
a compound of formula (I)
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl;
a compound of formula (II)
Formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine;
a compound of formula (III)
Formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl;
a compound of formula (IV)
Formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May be taken together to form 3To 8-membered heterocyclic ring, which may optionally be further substituted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5;
a compound of formula (V)
Formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; or
A compound of formula (VI)
Formula (VI)
Wherein
X represents a member selected from the group consisting of: ethylene-CH2-CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
2. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (I):
formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
3. The method of claim 2, wherein Alk is 1, 2-ethanediyl.
4. The method of claim 2, wherein the compound of formula (I) is Ritanserin (Ritanserin).
5. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (II):
formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
6. The method of claim 5, wherein the compound of formula (II) is Amiodarone (Amiodarone).
7. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (III):
formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
8. The method of claim 7, wherein the compound of formula (III) is Terfenadine (Terfenadine).
9. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (IV):
formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further interrupted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
10. The method of claim 9, wherein the compound of formula (IV) is Perphenazine (Perphenazine).
11. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (V):
formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
12. The method of claim 11, wherein the compound of formula (V) is thiochlorophenol (bisthionol).
13. The method of claim 1, comprising administering to the patient an anti-angiogenic effective amount of a compound of formula (VI):
formula (VI)
Wherein
X represents a member selected from the group consisting of: ethylene-CH 2-CH 2-and ethenylene-CH ═ CH-;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and salts, solvates and prodrugs thereof.
14. The method of claim 13, wherein the compound of formula (VI) is Clomipramine (Clomipramine).
15. A method of inhibiting the growth or metastasis of an angiogenesis-dependent tumor in a patient in need thereof, comprising administering to the patient an effective amount of a compound of:
a compound of formula (I)
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halogen radical, hydroxy, lower alkylOxy, lower alkyl and trifluoromethyl;
a compound of formula (II)
Formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine;
a compound of formula (III)
Formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl;
a compound of formula (IV)
Formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further interrupted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5;
a compound of formula (V)
Formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; or
A compound of formula (VI)
Formula (VI)
Wherein
X represents a member selected from the group consisting of: ethylene-CH2-CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
16. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (I)
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting ofGroup (2): hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
17. The method of claim 16, wherein Alk is 1, 2-ethanediyl.
18. The method of claim 16, wherein the compound of formula (I) is ritanserin.
19. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (II):
formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
20. The method of claim 19, wherein the compound of formula (II) is amiodarone.
21. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (III):
formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
22. The method of claim 21, wherein the compound of formula (III) is terfenadine.
23. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (IV):
formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further interrupted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
24. The method of claim 23, wherein said compound of formula (IV) is perphenazine.
25. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (V):
formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
26. The method of claim 25, wherein the compound of formula (V) is thiochlorophenol.
27. The method of claim 15, comprising administering to the patient an effective amount of a compound of formula (VI):
formula (VI)
Wherein
X represents a group selected from ethylene-CH2CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and salts, solvates and prodrugs thereof.
28. The method of claim 27, wherein the compound of formula (VI) is clomipramine.
29. A method of treating a disease or disorder associated with angiogenesis in a patient in need thereof, the disease or disorder selected from the group consisting of: neoplastic disease, restenosis, rheumatoid arthritis, Crohn's disease, diabetic retinopathy, psoriasis, endometriosis, macular degeneration, neovascular glaucoma, and obesity, comprising administering to the patient an effective amount of the following anti-angiogenic compounds:
a compound of formula (I)
Formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl;
a compound of formula (II)
Formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine;
a compound of formula (III)
Formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl;
a compound of formula (IV)
Formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, or,Cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further interrupted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5;
a compound of formula (V)
Formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; or
A compound of formula (VI)
Formula (VI)
Wherein
X represents a member selected from the group consisting of: ethylene-CH2-CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
30. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (I):
formula (I)
Wherein:
r is hydrogen, hydroxy or lower alkoxy;
R1is a member selected from the group consisting of hydrogen and lower alkyl;
alk is a lower alkanediyl group;
x is selected from the group consisting of-S-, -CH2-and-C (R)2)=C(R3) -members of a group of constituents, said R2And R3Each independently is hydrogen or lower alkyl;
a is a divalent group having the formula: -CH2CH2-、-CH2CH2CH2-or
Wherein R is4And R5Each independently selected from the group consisting of: hydrogen, halo, amino and lower alkyl; and is
Ar1And Ar2Each independently selected from the group consisting of: pyridyl, thienyl and phenyl, optionally substituted with: halo, hydroxy, lower alkoxy, lower alkyl, and trifluoromethyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
31. The method of claim 30, wherein Alk is 1, 2-ethanediyl.
32. The method of claim 30, wherein the compound of formula (I) is ritanserin.
33. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (II):
formula (II)
Wherein:
R1is an alkyl group containing 1 to 6 carbon atoms;
R2selected from the group consisting of hydrogen and methyl;
NR3is a group selected from the group consisting of: dimethylamino, diethylamino, dipropylamino, N-piperidinyl, N-piperazinyl, N-pyrrolidinyl, N-morpholinyl, and N-substituted heteroaryl; and is
Y and Y1Independently selected from the group consisting of: hydrogen, fluorine, bromine, chlorine and iodine; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
34. The method of claim 33, wherein the compound of formula (II) is amiodarone.
35. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (III):
formula (III)
Wherein
R is selected from the group consisting of hydrogen or hydroxy;
R1is hydrogen; or
R and R1Together carrying R and R1Form a second bond between the carbon atoms;
n is an integer of 1 to 3; and is
Z is selected from the group consisting of: a thienyl, phenyl, or substituted phenyl, wherein the substituents on the substituted phenyl can be attached at the ortho, meta, or para positions of the substituted phenyl ring and are selected from the group consisting of: a halogen atom, a straight or branched lower alkyl chain having 1 to 4 carbon atoms, a lower alkoxy group having 1 to 4 carbon atoms, a di (lower) alkylamino group, or a saturated monocyclic heterocycle selected from the group consisting of: n-pyrrolidinyl, N-piperidinyl, N-morpholinyl, or N- (lower) alkyl N-piperazinyl; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
36. The method of claim 35, wherein the compound of formula (III) is terfenadine.
37. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (IV):
formula (IV)
Wherein
X independently at each occurrence represents hydrogen, chlorine or bromine;
a represents OH, OR, NR1R2、OC(O)R、OC(O)OR、C(O)OH、C(O)OR、C(O)NR1R2、OC(O)NR1R2、NHC(O)NR1R2Or NHC (NH) NR1R2
R represents alkyl, cycloalkyl, heterocycle, aryl, arylalkyl or heterocycloalkyl;
R1and R2Each independently represents hydrogen, alkyl, cycloalkyl, heterocycle, aryl, arylalkyl, heterocycloalkyl, or R1And R2May together form a 3-to 8-membered heterocyclic ring, which may optionally be further interrupted by 1 to 4 (CH)2)nA substituent is substituted;
n is a number between 0 and 5 and including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
38. The method of claim 37, wherein said compound of formula (IV) is perphenazine.
39. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (V):
formula (V)
Wherein the content of the first and second substances,
y represents O, S or S ═ O;
x independently at each occurrence represents halogen or OH; and is
n is independently at each occurrence an integer between 0 and 5, including 0 and 5; and pharmaceutically acceptable salts, solvates and prodrugs thereof.
40. The method of claim 39, wherein the compound of formula (V) is thiochlorophenol.
41. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (VI):
formula (VI)
Wherein
X represents a group selected from ethylene-CH2CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and salts, solvates and prodrugs thereof.
42. The method of claim 41, wherein the compound of formula (VI) is clomipramine.
43. The method of claim 29, comprising administering to the patient an effective amount of an anti-angiogenic compound of formula (VI):
formula (VI)
Wherein
X represents a group selected from ethylene-CH2CH2-and vinylidene-CH ═ CH —;
r represents a member selected from the group consisting of: methyl, ethyl, propyl, chlorine and bromine,
y represents an alkylene group having 2 to 3 carbon atoms, and
am represents a member selected from the group consisting of: lower dialkylamino, N-pyrrolidinyl, N-piperidinyl, N-piperazinyl, N-morpholinyl, and N-methyl-piperidinyl (2) -yl; and salts, solvates and prodrugs thereof.
44. The method of claim 43, wherein the compound of formula (VI) is clomipramine.
HK11101281.3A 2007-04-13 2008-04-14 Anti-angiogenic agents and methods of use HK1147207A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/911,645 2007-04-13

Publications (1)

Publication Number Publication Date
HK1147207A true HK1147207A (en) 2011-08-05

Family

ID=

Similar Documents

Publication Publication Date Title
CN101801386B (en) Anti-angiogenic agents and methods of use
AU2016213972A1 (en) Combinations of IRS/Stat3 dual modulators and anti-cancer agents for treating cancer
EP3458448B1 (en) Fasn inhibitors for use in treating non-alcoholic steatohepatitis
WO2012117396A1 (en) Tyrphostin derivative in combination with cytotoxic compounds for treating cancer
JP2023518375A (en) Method of treatment
HK1147207A (en) Anti-angiogenic agents and methods of use
AU2013201604B2 (en) Anti-angiogenic agents and methods of use
HK1136495A (en) Anti-angiogenic agents and methods of use
WO2024019661A1 (en) Labdane based compounds and uses thereof