MXPA00009345A - Benzoates derivatives for inhibiting angiogenesis - Google Patents

Benzoates derivatives for inhibiting angiogenesis

Info

Publication number
MXPA00009345A
MXPA00009345A MXPA/A/2000/009345A MXPA00009345A MXPA00009345A MX PA00009345 A MXPA00009345 A MX PA00009345A MX PA00009345 A MXPA00009345 A MX PA00009345A MX PA00009345 A MXPA00009345 A MX PA00009345A
Authority
MX
Mexico
Prior art keywords
group
alkyl
aryl
alkoxy
alkynyl
Prior art date
Application number
MXPA/A/2000/009345A
Other languages
Spanish (es)
Inventor
Daniel Tuse
Charles Hiebert
Keith R Laderoute
Nahid Waleh
Original Assignee
Biosource Technologies Inc
Sri International
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 Biosource Technologies Inc, Sri International filed Critical Biosource Technologies Inc
Publication of MXPA00009345A publication Critical patent/MXPA00009345A/en

Links

Abstract

The present invention relates to methods for effectively inhibiting unwanted angiogenesis. More particularly, this invention relates to methods of treating diseases that show unwanted angiogenesis and to delivering anti-angiogenic activity to a mammal. In other aspects this invention relates to methods of reducing the level of tumor necrosis factor&agr;.

Description

DERIVATIVES OF BENZOATOS TO INHIBIT THE ANGIOGENESIS FIELD OF THE INVENTION The present invention relates to methods for effectively inhibiting unwanted angiogenesis. More particularly, this invention relates to methods for treating diseases associated with undesired angiogenesis and for delivering anti-angiogenic activity to mammals having such diseases. BACKGROUND OF THE INVENTION Angiogenesis is the development of new blood vessels from existing microvessels. The process of generating new blood vessels plays an important role in embryogenic development, in the inflammatory response, in the development of metastasis (tumor-induced angiogenesis or TIA), in diabetic retinopathy, in the formation of arthritic pannus and in psoriasis. Under normal physiological conditions, humans or animals only suffer from angiogenesis in very specific restricted situations. For example, angiogenesis is usually seen in wound healing, in fetal and embryonic development and in the formation of the corpus luteum, endometrium and placenta. The control of angiogenesis is a highly regulated system that involves angiogenic stimulators and inhibitors. It has been found that the control of angiogenesis is altered in certain disease states and, in many cases, the pathological damage associated with the disease is related to uncontrolled angiogenesis. In tumor angiogenesis, for example, capillary buds form, inducing their formation by a group of tumor cells. However, compared to blood vessels produced in normal angiogenic microenvironments, the tumor microvessels are morphologically and functionally unique. Their vascular networks typically exhibit disorganized or aberrant architecture, the luminal sizes varied and the blood flow can fluctuate chaotically. There are two main types of tumor angiogenesis in terms of the events that follow the implantation of metastasis germination on surfaces and organs. The first or primary angiogenesis is the initial vascularization of the mass of tumor cell multiplication and is considered as an essential prerequisite for the survival and further growth of a metastatic deposit. The second is a continuation or secondary angiogenesis and is the phenomenon that occurs in waves at the periphery of a growing tumor mass. This second angiogenesis is essential for the increase of new "microcirculatory territories towards the service of the expansion and infiltration of the tumor.
Persistent and unregulated angiogenesis occurs in a multiplicity of disease states, tumor metastasis and abnormal growth by endothelial cells and supports of pathological damage are observed under these conditions. The various pathological states created due to unregulated angiogenesis have been grouped as angiogenic dependents or diseases associated with angiogenesis. Therapies aimed at the control of angiogenic processes could lead to the cancellation or mitigation of these diseases. An example of a disease mediated by angiogenesis is ocular neurovascular disease. This disease is characterized by the invasion of new blood vessels in structures of the eye such as the retina or cornea. This is the most common cause of blindness and is involved in approximately twenty eye diseases. In age-related muscle degeneration, the associated visual problems are caused by an incarceration of the choroidal capillaries through defects in the Bruch membrane with proliferation of fibrovascular tissue below the retinal pigment epithelium. Angiogenic damage is also associated with diabetic retinopathy, premature retinopathy, rejection of corneal graft, neovascular glaucoma and retrolental fibroplasia. Diseases associated with retinal / choroidal neovascularization includes, but not limited to, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoid, syphilis, elastic pseudoxanthoma, and Paget's disease. Another disease in which angiogenesis is thought to be involved is rheumatoid arthritis. The blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, endothelial cells release factors and reactive oxygen species that leads to pannus growth and cartilage destruction. An important area of current research in therapeutic oncology focuses on the discovery and development of anti-angiogenic agents, which target the tumor vasculature by inhibiting or suppressing the growth of new blood vessels. Several types of compounds have been used to prevent angiogenesis. For example, Taylor, et al. they have used protamine to inhibit angiogenesis (see, Taylor, et al, Nature 297: 307 (1982)). However, the toxicity of protamine limits its practical use as a therapeutic. Folkman, et al., Have further described the use of heparin and steroids to control angiogenesis (see, Folkman, et al, Science 221: 719 (1983) and U.S. Patent Nos. 5,001,116 and 4,994,443). Steroids such as tetrahydrocortisol, which lack gluco and corticoid mineral activity, have been found to be angiogenic inhibitors. In addition, angiostatin proteins have been shown to reversibly inhibit the proliferation of endothelial cells. Angiostatin is able to inhibit diseases related to angiogenesis and modulate angiogenic processes (see, for example, WO 95/292420). In view of the foregoing, it is apparent that the need remains in the art for methods and compounds to inhibit angiogenesis, either by completely inhibiting a factor of angiogenesis or by some other mechanism. Such methods and compounds would have an adverse effect on the growth of tumors and, in addition, could be used to treat many of the other diseases stated above. The methods of the present invention fully meet these and other needs. SUMMARY OF THE INVENTION In one aspect, this invention relates to a method of inhibiting the vascularization of endothelial cells, the method comprising contacting a cell, tissue or organ having endothelial cells with an anti-angiogenic amount of an composed of Formula I. The compounds of Formula I have the following general formula: In Formula I, R1 is a functional group that includes, but is not limited to, C? -C6-alkyl. In Formula I, R2, R3, R4 and R5 are each independently selected and are functional groups including, but not limited to, hydrogen, C? -C6-alkyl, C-C3-alkenyl, C2-C6-alkynyl , aryl, hydroxyl, C? -C6-alkoxy, halogen, N02 and NH2. In Formula I, R6, R7, R8 and R9 are each independently selected and are functional groups including, but not limited to, hydrogen, C? -C6-alkyl, C2-Ce-alkenyl, C2-C6-alkynyl , aryl, hydroxyl, C? -C6-alkoxy, halogen, N02 and NH. In Formula I, if X is present, it is a functional group that includes but is not limited to the following: oxygen, sulfur, -CH2-, or carboxy.
- - In the formula I, Y is a heteroatom that includes .oxygen or sulfur. In a preferred embodiment of the invention, the compound of Formula I is methyl 3,5-diiodo-4- (4'-methoxyphenoxy) benzoate ("BTO-956"). In another aspect, this invention relates to a method for effectively inhibiting unwanted angiogenesis in a tissue or organ, the method comprising contacting the cell with a compound of Formula I, or a pharmaceutical composition thereof. , in an amount sufficient to inhibit angiogenesis. In a currently preferred embodiment, the cell is found in a mammalian subject. In yet another aspect, this invention relates to a method of treating mammalian diseases mediated or associated with unwanted and uncontrolled angiogenesis, the method comprising administering to a mammal an anti-angiogenic compound of Formula I in a dose sufficient to inhibit angiogenesis. These methods are useful for improving the effects of conditions that are characterized by abnormal or undesirable angiogenesis or endothelial cell proliferation. In another aspect this invention relates to methods that reduce the level of tumor necrosis factor-a (TNF-a) produced by a cell In yet another aspect, this invention relates to methods of using such compounds to reduce TNF production -a and to treat inflammatory diseases. Other features, objects and advantages of the invention and their preferred embodiments will become apparent from the following detailed description. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 illustrates the mean growth response as a relative tumor volume of MDA MB-231 (A) and OVCAR3 (B) tumors in nude mice exposed to BTO-956. Figure 2 illustrates that BTO-956 induces the arrest of prometaphase in MCF-7 breast carcinoma cells. Figure 3 illustrates the inhibition of microtubular array in vi tro by BTO-956 Figure 4 illustrates the disruption of microtubule networks by BTO-956 Figure 5 illustrates that BTO-956 competes with colchicine to bind to tubulin in vi tro. Figure 6 illustrates that BTO-956 inhibits the proliferation of human vascular endothelial cells. Figure 7 illustrates that BTO-956 reduces the accumulation of mRNA in TNF-α. Figure 8 illustrates the effect of different concentrations of BTO-956 on the expressions of the TNF-a gene.
- - Figure 9 illustrates that the total RNA was intact in cells treated with different concentrations of BTO-956 and colchicine. DETAILED DESCRIPTION OF THE INVENTION AND THE PREFERRED MODALITIES A. DEFINITIONS The term "angiogenesis" refers to the generation of new blood vessels in cells, tissues, organs or tumors. The term "metastasis" refers to the process by which tumor cells spread to different parts of the body. The term is also used herein to refer to a tumor that develops through the metastatic process. The term "independently selected" is used herein to indicate that the R groups, for example, R1, R2, R3 and R4 may be identical or different (for example, R1, R2, R3 and R4 can all be hydrogen or R1 and R4, can be hydrogen and R2 and R3 can be halogen, etc.) The term "alkyl" is used herein to refer to a radical of branched or unbranched, saturated or unsaturated monovalent hydrocarbon having 1-12 carbons and preferably 1-6 carbons. When the alkyl group has formed atoms of 1-6 carbons, this is referred to - as a "lower alkyl". Suitable alkyl radicals include, for example, methyl, ethyl, n-propyl, i-propyl, 2-propenyl (or allyl), n-butyl, t-butyl i-butyl (or 2-methylpropyl), etc. As used herein, the term encompasses "substituted alkyls". "Substituted alkyls" refers to alkyls as described including one or more functional groups such as the lower alkyl, aryl, acryl, halogen (ie, haloalkyls, eg, CF3), hydroxy, amino, alkoxy, alkylamine, acylamino, acyloxy , aryloxy, aryloxyalkyl, mercapto, cyclic hydrocarbons both saturated and unsaturated, heterocyclic and the like. These groups can be attached to any carbon in the alkyl residue The term "S-alkyl" is used herein to refer to the group -SR, wherein R is lower alkyl or substituted lower alkyl as defined herein. "Aryl" is used herein to refer to an aromatic substituent that may be a single aromatic ring or multiple aromatic rings that are fused together, covalently linked, or linked to a common group such as a methylene or ethylene residue The common bond group may also be a carbonyl as in the benzophenone The aromatic ring (s) may include phenyl, naphthyl, biphenyl, diphenylmethyl, and benzophenone, among others. "arylalkyl." "Substituted aryl" refers to an aryl as described, including one or more functional groups, such as the lower alkyl, acyl, halogen, haloalkyls, (eg, CF3), hydroxy, amino, alkoxy, alkylamine, acylamino, acyloxy, mercapto and cyclic hydrocarbons, both saturated and unsaturated, which are fused to the aromatic ring (s) linked covalently or linked to a common group such as a methylene or ethylene residue. The linking group can also be a carbonyl such as in the exhenyl phenyl ketone cycle. The term "substituted aryl" embraces "substituted arylalkyl". The term "halogen" is used herein to refer to fluorine, bromine, chlorine and iodine atoms. The term "hydroxy" is used herein to refer to the -OH group. The term "amino" is used to refer to the group -NRR ', where R and R' may independently be hydrogen, alkyl, substituted alkyl, aryl, substituted aryl or acyl. The term "nitro" is used herein to refer to the group -N02. The term "alkoxy" is used herein to refer to the group -OR, where R is a lower alkyl, - substituted lower alkyl, aryl, substituted aryl, arylalkyl or substituted arylalkyl, wherein the alkyl, aryl, substituted aryl, arylalkyl and substituted arylalkyl are groups as described herein. Suitable alkoxy radicals include, for example, methoxy, ethoxy, phenoxy, substituted phenoxy, benzyloxy, phenethyloxy, t-butoxy, etc. The term "alkenyl" is used herein to refer to an unsaturated, branched, straight-chain or cyclic monovalent hydrocarbon radical having at least one carbon-carbon double bond. The radical can be in any of the cis or trans configurations near the double bond (s). Suitable alkenyl radicals include, for example, ethenyl, propenyl, isopropenyl, cyclopropenyl, butenyl, isobutenyl, cyclobutenyl, tert-butenyl, pentenyl, hexenyl, etc. The term "alkynyl" is used herein to refer to an unsaturated, branched, straight-chain or cyclic monovalent hydrocarbon radical having at least one carbon-carbon triple bond. Suitable alkynyl radicals include, for example, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl, etc. The term "contacting" is used interchangeably herein with the following: combined with, added to, mixed with, passed on, incubated with, - fluid on, etc. further, the compounds of the present invention can be "administered" by any conventional method, such as, for example, parenteral, oral, topical, and inhalation routes as described herein. The term "pharmaceutically acceptable salt" refers to those salts of compounds which retain the effectiveness and biological properties of the free bases and which are obtained by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, acid phosphoric acid, methanesulfonic acid, ethane sulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. The pharmaceutically acceptable salts include, for example, alkali metal salts, such as sodium and potassium, alkaline earth salts and ammonia salts. "A sufficient amount", "an effective amount", "therapeutically effective amount" or an "anti-angiogenic amount" refers to an amount of a compound or composition effective to depress, suppress or inhibit angiogenesis or result in the improvement of the symptoms associated with an angiogenic disease. The desired result can be a subjective relief of a symptom (s) or an objectively identifiable improvement in the recipient of the dose, a decrease in the vascularization of the cells - endothelial or a decrease in the rate of angiogenesis as observed by a clinician or other qualified observer. The term "cancer treatment", "therapy" and the like generally refers to any improvement in the mammal having the cancer where the improvement can be ascribed to the treatment with the compounds of the invention. The improvement can be subjective or objective. For example, if the mammal is a human, the patient may notice improved vigor or vitality or decreased pain as subjective symptoms of improvement or response to therapy. Alternatively, the clinician may see a decrease in tumor size or tumor weight based on a physical examination, laboratory parameters, tumor markers, or radiographic findings. Some laboratory signs that clinicians can observe for response to therapy include normalization of tests such as white blood cell count, red blood cell count, platelet count, erythrocyte sedimentation rate, and level of various enzymes. Additionally, clinicians may observe a decrease in a detectable tumor marker. Alternatively, other tests may be used to evaluate the objective improvement, such as sonograms, nuclear magnetic resonance test and positron emission test.
The "inhibition of tumor cell growth" can be evaluated by any acceptable method of measurement if the growth of tumor cells has been delayed or decreased. This includes direct observation and indirect evaluation such as subjective symptoms or objective signs as discussed above. B. COMPOUNDS The present invention relates to the discovery that the compounds of Formula I are useful for inhibiting angiogenesis and / or for treating angiogenic diseases. The compounds of Formula I have the following general formula: wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, X and Y are as defined above. In a preferred embodiment of the invention, the compound of Formula I is methyl 3,5-diiodo-4- (4'-methoxyphenoxy) benzoate ("BTO-956"). The compounds used in the methods of the present invention can be made according to the procedure outlined in Borrows, et al, J. Chem. Soc. 1949, S185-190 and WO 97/46228, the teachings of which are incorporated in the present for reference. In general, the process is carried out in a series of stages. For example, starting with the case of BTO-956, a substituted phenol and a substituted benzoate are reacted to produce methyl 3, 5-dinitro-4- (4-methoxyphenoxy) benzoate. The nitro groups of the benzoate are then reduced to amines and subsequently replaced by iodine. This method for preparing BTO-956 is essentially as described in: Masuda, K., Imashiro, Y., and Okada, Y. "Synthesis of Triiodothyroformic Acid and its Derivatives" (Synthesis of Triyodotyrophoramic Acid and its derivatives), J Takeda Res. Lab., 1970, 29, 545-552, the teachings of which are incorporated herein by reference. The additional synthetic details are set forth in Example 1. Compounds suitable for use in the methods of the present invention can be readily identified using in vivo and in vitro examination analysis. Such analyzes may examine the ability of a particular compound to inhibit angiogenesis or vascularization of endothelial cells in vivo and in vi tro. For example, chick embryo chorioallantoic membrane (CAM) analysis, which is described in more detail below, can be used to examine a compound given its ability to inhibit vascularization. In the chorioallantoic membrane analyzes, the fertilized chicken embryos were removed from their shell on day 3 or 4 and a methylcellulose disk containing a compound of the formula I was implanted in the chorioallantic membrane. The embryos were examined 48 hours later and a clear avascular zone appeared around the methylcellulose disk, the diameter of that area was measured. This analysis can be used to assess the anti-angiogenic properties of the compounds of Formula I. Another test analysis useful for assessing the efficacy of the compounds of Formula I is the analysis of the angiogenesis of the corneal microcapsule (CMA). The analysis of the rat corneal microcapsule can be used to assess the ability of the compounds of Formula I to inhibit corneal angiogenesis (see, "Quantitative Angiogenesis Assays: Progress and Problems" (Quantitative Analysis of Angiogenesis: Advances and Problems) , Na t. Med., 3: 1203-1208, 1997) and "Inhibition of Tumor Angiogenesis Using a Soluble Receptor Stablishes to Role for Tie2 in Pathologic Vascular Growth" (Inhibition of Tumor Angiogenesis Using a Soluble Receptor Sets a Role for Tie2 in Pathological Vascular Growth), J. "Clin. Invest., 100: 2072-2078, 1997. In this analysis, the compound of Formula I is mixed with a polymer (eg, Hydron solution).; Sciences of the Inferieron, New Brunswick, N.J.) and implanted in a small cavity surgically created in the superficial layers of the cornea of a rat. Under normal circumstances, this wound stimulates an angiogenic response that is easily visible as the appearance of neovessels in the normally avascular cornea. If the compound of Formula I is effective, specifically as an anti-angiogenic agent, it inhibits or blocks the response. In an experimental design, a group of five animals (including a control group with only polymer implants) was tested through a range of doses of drugs that can induce tumor growth retardation. Three doses were tested in the analysis. The assessment of an anti-angiogenic response by this method is categorical. In other words, a treated eye is either positive or negative for corneal angiogenesis. This analysis determines whether the compound of Formula I is directly anti-angiogenic in an angiogenesis model of a mammal in vivo. In addition, the analysis of human microvascular endothelial cells, (HMVEC) can be used to assess the efficacy of the compounds of Formula I. The HMVEC is seeded in a 96-well plate at a concentration x of 5 x 10 3 cells / well in a volume of 100 μl / well of Endothelial Growth Medium. The - plates are then incubated at 37 ° C in 5% C02 for 24 hours and then aliquots of the compound of Formula I were added to the HMVEC preparations and the plates were then incubated at 37 ° C in 5% C02 for 3 days . The relative number of cells was determined by adding 20 μl of Alamar Blue for 3-6 hours at 37 ° C and the measurement of color changes indicates the metabolic activity when using a Fluorescence Measurement System. In this analysis, the intensity of the fluorophore signal is directly proportional to the number of cells. The HMVEC analysis is also carried out using human umbilical vein microvascular endothelial cells (HUMVEC). This analysis is done in a similar way to the previous analysis, but HUMVEC cells are used. Additionally, other assays known to those skilled in the art can be readily used to examine the compounds of the present invention for anti-angiogenic properties. It will be readily apparent to those skilled in the art that the compounds of Formula I can be administered alone, in the pharmaceutically acceptable salt form and / or in the form of a pharmaceutical composition. C. USES FOR THE COMPOUNDS OF THE PRESENT INVENTION As explained above, the present invention relates to the discovery that the compounds - of Formula I are useful for inhibiting angiogenesis and, in turn for treating diseases associated with unwanted angiogenesis. As such, in one embodiment, the present invention provides a method for inhibiting unwanted angiogenesis in a cell, the method comprising contacting the cell with an effective amount, i.e., an anti-angiogenic amount of a compound of the Formula I. In another embodiment, the present invention provides a method for inhibiting endothelial cell vascularization, the method comprising contacting a cell, tissue or organ containing the endothelial cells with an effective amount of a compound of Formula I. In a currently preferred embodiment, the cells are found in a mammalian subject. This invention relates to a method for treating diseases of mammals associated with unwanted and uncontrolled angiogenesis, the method comprising administering to a mammal an anti-angiogenic compound of Formula I in an amount, i.e., a dose sufficient to inhibit angiogenesis. . The particular dose of a compound of Formula I required to inhibit angiogenesis and / or angiogenic diseases will depend on the severity of the condition, the route of administration and the related factors that will be decided by the treating physician. In general, the accepted and effective daily dose will be sufficient to effectively inhibit angiogenesis and / or angiogenic diseases. The methods of treatment provided by this invention were practiced by administering to a mammal in need thereof, a dose of a compound of Formula I (or a pharmaceutically acceptable salt or solvate thereof) that is effective in inhibiting diseases of angiogenesis and / or angiogenesis. The term "inhibit" is used herein to include its generally accepted meaning which includes prophylactically treating a human subject who incurs angiogenesis and / or angiogenic diseases, and keeping it in check and / or existing treatment for life-threatening diseases. angiogenesis and / or angiogenesis. As such, the present invention includes both medical and prophylactic therapeutic treatments, as appropriate. The methods of the present invention can be used to treat a wide variety of diseases. Diseases associated with corneal neovascularization that can be treated using the methods of the present invention include, but are not limited to, diabetic retinopathy, premature retinopathy, corneal graft rejection, neovascular glaucoma and retrolental fibroplasia., epidemic keratoconjunctivitis, vitamin A deficiency, use of contact lenses, atopic keratitis, upper limbic keratitis, keratitic pterygium drying, sjogrens, acne rosacea, filectenulosis, syphilis, mycobacterial infections, lipid degeneration, chemical burns, bacterial ulcers, ulcers fungal infections, herpes simplex infections, herpes zoster infections, protozoan infections, kaposi sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, trauma, rheumatoid arthritis, systemic lupus, polyateritis, Wegeners sarcoidosis, scleritis, Steven-Johnson, perifigloid radial keratomy and corneal graft rejection Diseases associated with retinal / choroidal neovascularization that can be treated using the methods of the present invention include, but are not limited to, diabetic retinopathy, macular degeneration, sickle cell disease, sarcoid , syphilis, pseud elastic oxanthoma, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis / vitritis, mycobacterial infections, Lyme disease, systemic lupus erythematosus, premature retinopathy, Eales disease, Bechets disease, infections that cause a retinitis or choroiditis, presumed ocular histoplasmosis, Bests disease, myopia, optic pits, Stargarts disease, part planitis, chronic retinal detachment, hyperviscosity syndrome, toxoplasmosis, trauma and post-laser complications. Other diseases include, but are not limited to, diseases associated with rubeosis (neovascularization of the angle) and diseases caused by abnormal proliferation of fibrovascular or fibrous tissue, including all forms of proliferative vitreoretinopathy, whether or not associated with diabetes. Diseases associated with chronic inflammation can also be treated using the methods of the present invention. Diseases with symptoms of chronic inflammation include but are not limited to inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis, psoriasis, sarcoidosis and rheumatoid arthritis. Unwanted or uncontrolled angiogenesis is a key element that all these chronic inflammatory diseases have in common. Chronic inflammation depends on the continuous formation of capillary buds to maintain a flow of inflammatory cells. The flow and presence of inflammatory cells produces granulomas and, thus, maintains the state of chronic inflammation. The inhibition of angiogenesis using the compositions and methods of the present invention prevents the formation of granulomas, thus mitigating the disease. As mentioned above, the methods of the present invention can be used to treat patients with inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis. Crohn's disease occurs as a transmural chronic inflammatory disease that most commonly affects the distal ileum and colon, but can occur anywhere in the gastrointestinal tract from the mouth to the anus and perianal area. Patients with Crohn's disease usually have chronic diarrhea associated with abdominal pain, fever, anorexia, weight loss and increased abdominal volume. The prevention of angiogenesis by means of the compositions and methods of the present invention inhibit the formation of shoots and prevent the formation of granulomas. Ulcerative colitis is also a chronic, non-specific, inflammatory and ulcerative disease that originates in the colonic mucosa and is characterized by the presence of bloody diarrhea. Inflammatory bowel diseases also have extra intestinal manifestations, such as skin lesions. Such lesions are characterized by inflammation and angiogenesis and can occur at many different sites in the gastrointestinal tract. The compositions and methods of the present invention can also be used to treat these lesions by preventing angiogenesis, thus reducing the influx of inflammatory cells and the formation of lesions.
- Sarcoidosis is another chronic inflammatory disease that is characterized by a granulomatous disorder of multiple systems. The granulomas of this disease can be formed in any part of the body and thus, the symptoms depend on the site of the granulomas and where the disease is activated. Granulomas are created by angiogenic capillary buds providing a constant supply of inflammatory cells. The compounds and method of this invention can be used to treat sarcoidosis. The methods of the present invention can also be used to treat the chronic inflammatory conditions associated with psoriasis. Psoriasis A skin disease is another chronic and recurrent disease that is characterized by papules and plaques of various sizes. The prevention of the formation of the new blood vessels necessary to maintain the characteristic lesions leads to the mitigation of the symptoms. Another disease that can be treated using the methods of the present invention is rheumatoid arthritis. Rheumatoid arthritis is a chronic inflammatory disease characterized by nonspecific inflammation of the peripheral joints. It is considered that the blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. These factors involved in angiogenesis can actively contribute and help maintain the chronic inflammatory state of rheumatoid arthritis. Other diseases that can be treated using the methods of the present invention are hemangiomas, Osler-Weber-Rendu disease or hereditary hemorrhagic telangiectasia, solid or blood-bearing tumors and acquired immune deficiency syndrome. The methods of this invention are also effective in inhibiting angiogenesis associated with malignant tumor growth. This includes the growth of cancerous tumor in cells, tissues and organs. The methods of the present invention are useful in the treatment of the growth of numerous tumor cells and in treating a wide variety of cancers. Such tumor cells include, by way of example and without limitation, lung, colon, breast, ovarian, prostate and hepatic tumor cells as well as squamous cell carcinomas. Such cancers include, by way of example and without limitation, carcinomas such as cancer of the pharynx, colon, rectal, pancreatic, stomach, liver, lung, breast, skin, prostate, ovary, cervical-, uterine and bladder; leukemias, lymphomas; gliomas; retinoblastomas and sarcomas.
In a preferred embodiment, the present invention relates to methods of administering the compounds of Formula I in combination with active immunotherapy (eg, tumor vaccination). Because the compounds of Formula I are not immunotoxic, the immune system is not significantly depressed and, thus, active immunotherapy can be advantageously carried out in combination with chemotherapy. When used in conjunction with immunotherapy, the compound of Formula I can be administered before and / or during the administration of the immunotherapeutic agent (e.g., tumor vaccine). In yet another embodiment, the present invention provides a method for reducing the level of TNF-α produced by the cell. TNF-a and its various modes of action are generally described by Abbas, et al., Cellular and Molecular Immunology, Abbas, et al. , 2nd Ed., W.B. Saunders Company, 1994, pp. 244-249, the teachings of which are incorporated herein for reference. TNF-a plays an integral role in the destruction of tumors mediated by responses to tissue injury and by protecting the hosts from infections by various microorganisms. However, its activity seems to be excessive in some states of disease and inflammatory reactions such as in rheumatoid arthritis, cachexia and septic shock. The excess of TNF-a results in a - - exaggerated immune response exemplified by the over stimulation of interleukin-6 and the secretion of the granulocyte / macrophage colony stimulation factor (GM-CSF), increased cytotoxicity of polymorphonuclear neutrophils and prolonged expression of cell adhesion molecules, all which can have detrimental effects. Contacting the cells with the compounds of Formula I results in decreased levels of TNF-α. The reduced levels of TNF-α can result from any of several mechanisms including, for example, down-regulation of the expression of a gene encoding TNF-α, a reduction in the translation efficiency or stability of TNF-α mRNA, decreased stability of the TNF-α polypeptide, and reduced secretion of TNF-α from a cell. The reduced levels of TNF-a can be measured in a cell, biological sample or in the bloodstream. As a result of their ability to inhibit TNF-a, the compounds of Formula I can be used to treat inflammatory diseases. Such diseases include but are not limited to the set of previously established inflammatory diseases (eg, chronic inflammation, chronic disease, inflammatory bowel disease, sarcoidosis, psoriasis, rheumatoid arthritis, and the like). Using the analysis set forth in Example VIII, the - Compounds of Formula I can be easily selected for their ability to reduce TNF-a levels. TNF-a is noted for its pro-inflammatory actions that result in tissue lesions, such as the induction of procoagulant activity in vascular endothelial cells, increased adherence of neutrophils and lymphocytes, and stimulation of the release of platelets that activate the macrophage factor, neutrophils and vascular endothelial cells. As such, determination residues that target those cells and that are conjugated to liposomes or other drug delivery systems comprising the compounds of Formula I are preferred embodiments of the invention. For example, in a preferred embodiment, monoclonal antibodies to TNF-α (Tracey, et al., Na ture 1987, 330.662-664; Silva, et al., J. Infect. Sis. 1990. 162, 421-427. , and Williams, et al., Proc. Na ti, Acad. Sci. 1992, 89, 9784-9788) are conjugated to the liposomes comprising the compounds of Formula I. In addition, according to the above methods, the subjects Mammals include but are not limited to humans, laboratory animals, domestic pets and farm animals. D. PHARMACEUTICAL FORMULATIONS / ROUTES OF ADMINISTRATION - In the methods of the present invention, the compounds of Formula I can be administered or administered to a mammal, for example, a human patient, alone, in the form of a pharmaceutically acceptable salt, or in the form of a pharmaceutical composition wherein the The compound is mixed with suitable carriers or excipient (s) in a therapeutically effective amount, for example, at doses effective to depress, suppress or inhibit angiogenesis or which results in improvement of the symptoms associated with angiogenic diseases. The compounds of Formula I, which are used in the methods of the present invention, can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds of Formula I can be formulated into pharmaceutical compositions when combined with suitable pharmaceutically acceptable carriers or excipients and can be formulated into solid, semisolid, liquid or gaseous forms, such as tablets, capsules, pills, powders, granules, dragees, gels, mixtures, ointments, solutions, suppositories, injections, inhalants and aerosols. As such, the administration of the compounds can be achieved in various forms, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intratracheal, etc. administration. In addition, the compound can be administered - in a locally preferably systemic manner, for example by injection of the compound directly into the solid tumor, often in a depot or sustained release formulation. In addition, the compounds can be administered in a targeted drug delivery system, for example in a liposome coated with tumor-specific antibodies. Such liposomes will be directed and selectively absorbed by the tumor. In addition, the compounds of Formula I can be formulated with common excipients, diluents or vehicles and compressed into tablets, or formulated as elixirs or solutions for convenient oral administration or administered intramuscularly or intravenously. The compounds can be administered transdermally, and can be formulated in sustained release dosage forms and the like. The compounds of Formula I can be administered alone, in combination with each other or can be used in combination with other known compounds (eg, other drugs anti-cancer or other drugs such as AZT, anti-inflammatories, antibiotics, corticosteroids, vitamins, etc.). For example, the compounds of Formula I can be used in conjunctive therapy with other known anti-angiogenic chemotherapeutic agents, antineoplastic agents (eg, vinca alkaloids, antibiotics, antimetabolites, platinum coordination complexes, etc.).
For example, the compounds of Formula I can be used in conjunctive therapy with a vinca alkaloid compound, such as vinblastine, vincristine, taxol, etc .; an antibiotic such as adrimycin, (doxorubicin), dactinomycin (actinomycin D), daunorubicin (daunomycin, rubidomycin), bleomycin, plicamycin, (mitramycin) and mitomycin (mitomycin C), etc .; an antimetabolite, such as methotrexate, cytarabine (AraC), azauridine, azaribine, fluorodeoxyuridine, deoxicoformycin, mercaptopurine, etc .; or a platinum coordination complex, such as cisplatin (cis-DDP), carboplatin, etc. In addition, those of skill in the art will appreciate that the compounds of the present invention can be used in conjunctival therapy with other anti-angiogenic, chemotherapeutic or antineoplastic compounds. In the pharmaceutical dosage forms, the compounds may be administered in the form of their pharmaceutically acceptable salts, or may also be used alone or in appropriate association, as well as in combination with other pharmaceutically active compounds. Formulations suitable for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company, Philadelphia, PA, 17th Ed. (1985)) which is incorporated herein by reference. In addition, for a brief review of methods for delivering drugs, see Langer, Science 249: 1527-1533 (1990), which is incorporated herein by reference. The pharmaceutical compositions described herein can be made in a manner that is known to those skilled in the art, ie by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, trapping or lyophilizing processes. The following methods and excipients are only exemplary and not by way of limitation. For injection, the compounds may be formulated into preparations upon dissolving, suspending or emulsifying them in an aqueous or non-aqueous solvent, such as vegetable or other similar oils, synthetic aliphatic glyceride acids, higher aliphatic acid esters or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. Preferably, the compounds of the invention can be formulated in aqueous solutions, preferably in physiologically compatible regulators such as Hanks' solution, Ringer's solution or physiological saline regulator. For transmucosal administration, the appropriate penetrants for the barrier to be permeated, are - - used in the formulation. Such penetrants are generally known in the art. For oral administration, the compounds of Formula I can be formulated easily, when combined with pharmaceutically acceptable carriers that are well known in the art. Such vehicles allow the compounds to be formulated as tablets, pills, dragees, capsules, emulsions, lipophilic and hydrophilic suspensions, liquids, gels, syrups, mixtures, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by mixing the compounds with a solid excipient, optionally grinding the resulting mixture, and processing the granule mixture, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol or sorbitol; cellulose preparations such as, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth gum, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and / or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as degraded polyvinyl pyrrolidone, agar or alginic acid or a salt thereof such as sodium alginate.
The dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol and / or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the coatings of tablets or dragees for identification or to characterize different combinations of active compound doses. Pharmaceutical preparations that can be used orally include soft-coating capsules made of gelatin, as well as sealed soft capsules made of gelatin and a plasticizer such as a glycerol or sorbitol. Soft-coating capsules may contain the active ingredients in the mixture with fillers such as lactose, binders such as starches and / or lubricants such as talc or magnesium stearate and optionally stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffins or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in doses suitable for such administration.
- - For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. For administration by inhalation, the compounds to be used according to the present invention are conventionally delivered in the form of an aerosol spray presentation from pressurized packets or a nebulizer, with the use of a suitable propellant, for example, dichlorodifluoromethane , trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas, or from dry powder inhalers without propellants. In case of a pressurized aerosol, the dosage unit can be determined by providing a valve to supply a measured quantity. Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator may be formulated containing a powder mixture of the compound and a suitable powder base such as lactose or starch. The compounds can be formulated for parenteral administration by injection, for example by bolus injection or continuous infusion. Formulations for injection may be presented in the form of unit doses, for example in ampules or in multi-dose containers, with an added preservative. The compositions can take forms such as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulating agents such as suspending, stabilizing and / or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water soluble form. Additionally, suspensions of the active compounds can be prepared in oily injectable suspensions as appropriate. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injectable suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain stabilizers or suitable agents that increase the solubility of the compounds to allow the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in the form of powder for constitution with a suitable vehicle, eg, with sterile, pyrogen-free water, before use. The compounds can also be formulated in rectal compositions such as suppositories or retention enemas, for example, containing suppository bases - conventional, such as, cocoa butter, carboceras, polyethylene glycols or other glycerides, all of which melt at body temperature, but are solid at room temperature. In addition to the formulations previously described, the compounds can also be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or with ion exchange resins or as poorly soluble derivatives, for example as a poorly soluble salt. Alternatively, other delivery systems for the hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well-known examples of carriers or carriers of delivery for hydrophobic drugs. In a presently preferred embodiment, liposomes of prolonged circulation, i.e., surreptitious, are used. Such liposomes are generally described in Woodle et al. , Patent of E.U. No. 5,013,556, the teachings of which are incorporated herein by reference. Monoclonal antibodies optionally conjugated to liposomes and directed against a tumor marker, TNF-a, or a TNF-a receptor, is another strategy that can be employed. In addition, the determination of a marker in an abnormal tumor vasculature can be employed. The residue determined when coupled to a drug or toxic radioisotope will act to concentrate the drug where necessary. Ligands for markers associated with the tumor can also be used. For example, a cell adhesion molecule that binds to a surface marker of the tumor vascular element can be used. Liposomes and other drug delivery systems can also be used, especially if their surface contains a ligand to direct the vehicle preferably towards the tumor vasculature. Liposomes offer the added advantage of protecting the drug from most normal tissues, thus reducing the inherent toxicity of many compounds. When coated with polyethylene glycol (PEG) (ie, surreptitious liposomes), to minimize absorption by phagocytes and with a specific determination residue of tumor vasculature, liposomes offer a longer plasma half-life, lower tissue toxicity objective and increased efficacy on non-targeted drugs. Other determination strategies include but are not limited to ADEPT (therapy - enzyme drug directed to the antibody), GDEPT (EPT directed to the gene) and VDEPT (EPT directed to the virus). In the ADEPT, the determination of an inactive pro-drug for a tumor mass is carried out by means of an antibody against a marker associated with the tumor. The medium of the enzyme in or near the tumor transforms the pro-drug into an active toxic agent that then acts on the tumor tissue. Similarly, differential gene expression or viral determination at the tumor site is used to activate a pro-drug in its active toxic form in GDEPT and VDEPT, respectively. Other strategies include the determination of genes, enzymes or differentially expressed surface markers that appear in the vasculature associated with the tumor, to effect the control of tumor growth. Using the aforementioned methods, the compounds of Formula I can be determined to the tumor vasculature to effect control of tumor progression or to other sites of interest (eg, endothelial cells). Certain organic solvents such as dimethylsulfoxide may also be employed, but usually at the expense of increased toxicity. Additionally, the compounds can be delivered using a sustained release system, such as semi-permeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various types of sustained release materials have been established and are well known to those skilled in the art. Sustained-release capsules can. , depending on its chemical nature, release the compounds for a few weeks to up to 100 days. The pharmaceutical compositions may also comprise suitable solid phase or gel carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin and polymer such as polyethylene glycols. Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in a therapeutically effective amount. The amount of composition administered will, of course, depend on the subject to be treated, the subject's weight, the severity of the condition, the manner of administration and the judgment of the prescribing physician. The determination of an effective amount is within the ability of those skilled in the art, especially, in light of the detailed disclosure provided herein. For any compound used in the method of the invention, a therapeutically effective dose can initially be estimated from the cell culture assays. For example, a dose can be formulated in models - of animal to achieve a range of circulation concentration that includes the IC50 as determined in the cell culture (ie, the concentration of the test compound that is lethal to 50% of a cell culture) or the ICioo as determined in cell culture (ie, the concentration of the compound that is lethal up to 100% of a cell culture). Such information can be used to more accurately determine useful doses in humans. Initial doses can also be estimated from in vivo data. In addition, the toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for example by determining the LDS0 (the lethal dose up to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD50 and ED50. Compounds that exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in the formulation of a dose range that is non-toxic for use in humans. The dose of such compounds is preferably within a range of circulating concentrations that includes ED50 with little or - no toxicity The dose may vary within this range depending on the dosage form used and the route of administration used. The exact formulation, the route of administration and the dose can be chosen by the individual physician in view of the patient's conditions (see for example, Finge et al., 1975, In: The Pharmacological Basis of Therapeutics, (The Pharmacological Bases of the Therapeutics) Ch. 1, pl). The amount and range of the dose can be adjusted individually to provide plasma levels of the active compound that are sufficient to maintain the therapeutic effect. Normal doses of the patient for oral administration range from about 50-2000 mg / kg / day, commonly from about 100-1000 mg / kg / day, preferably from about 150-700 mg / kg / day, and more preferably from about 250-500 mg / kg / day. Preferably, therapeutically effective serum levels will be achieved by administering multiple doses each day. In case of local administration or selective absorption, the effective local concentration of the drug may not be related to the concentration of the plasma. Someone who is experienced in the technique will be able to optimize therapeutically effective local doses without undue experimentation. F. USES The invention will be described in more detail by means of specific examples. The following examples are offered for illustrative purposes and are not intended to limit the invention in any way. Those of skill in the art will readily recognize a variety of non-critical parameters that can be changed or modified to produce essentially the same result. EXAMPLE I This example illustrates the synthesis of methyl 3,5-diiodo-4- (4-methoxyphenoxy) benzoate (BTO-956). The synthesis of BTO-956 is carried out in a series of stages, first producing methyl 3, 5-dinitro-4- (4-methoxyphenoxy) benzoate, the nitro groups of which are then reduced to amines and subsequently replaced by iodo. The method for preparing BTO-956 is essentially as described in Masuda, K., Imashiro, Y., and Okada, Y. The synthesis of triiodothyrophoric acid and its derivatives. J *. Takeda Res. Lab. 1970, 29, 545-552. I. Methyl 3, 5-dinitro-4- (4-methoxyphenoxy) benzoate - M.W.124.14 M.W.260.59 M.W.348.77 Methyl 3, 5-dinitro-4-chlorobenzoate 20.2 g; 77.5 mmol 4-Methoxyphenol 10.0 g; 80.6 mmol Hydroxide d-potassium 4.7 g; 82.0 mmol Water 20 ml; solvent To a 100 ml round bottom flask containing potassium hydroxide (4.7 g; 82.0 mmol) dissolved in water (20 ml) were successively added 4-methoxyphenol (100 g, 80.6 mmol) and methyl 4-chloro-3,5-dinitrobenzoate (20.2 g, 77.5 mmol). The flask was adapted with a reflux condenser and the reaction was heated to 150 ° C (oil bath) for 3 hours. After cooling to room temperature, the reaction mixture was transferred to a large mortar and triturated with cold 2N NaOH (100ml) to remove the unreacted phenol. The solid was collected by filtration and dried with air to give 21.5 g of crude product. Crystallization from absolute ethanol gave 17.7 g (65.6%) of pure methyl 3,5-dinitro-4- (4-methoxyphenoxy) benzoate as light yellow needles. 300 MHz R NMR (CDC13) d 3.77 (s, 3 H, OCH 3), 4.02 (s, 3 H, OCH 3), 6.82 (m, 4 H, Ar H), 8.70 (s, 2 H, Ar H). 2. Methyl 3, 5-diamino-4- (4-methoxyphenoxy) benzoate CH3C - M.W. 348.27 M.W. 288.27 Methyl 3, 5-dinitro-4- (-methoxyphenoxy) benzoate 20.2 g; 77.5 mmol 10% palladium on carbon 0.7 g; Glacial acetic acid catalyst 80 mL; solvent To a Parr shaker bottle containing a suspension of methyl 3, 5-di-nitro-4- (4-methoxyphenoxy) benzoate (20.2 g, 77.5 mmol) in glacial acetic acid (80 ml) was added 10% palladium on carbon (0.7 g). The bottle was stirred under a hydrogen atmosphere (3 atm) until no more hydrogen was consumed. The catalyst was filtered and the resulting solution was concentrated to approximately 10 ml.
The residue was dissolved in acetone (50 ml) and heated in a steam bath while water (100 ml) was added in portions. After cooling, the medium brown needles formed which were collected by suction filtration and dried to give 7.1 g (86%) of methyl 3, 5-diamino-4- (4-methoxyphenoxy) benzoate. 300 MHz ^? NMR (CDC13) d 3.73 (s, 3H, OCH3), 3.80 (bs, 4H, ArNH2), 3.86 (s, 3H, 0CH3), 6.84 (m, 4H, ArH), 6.91 (s, 2H, ArH). 3. Methyl 3, 5-diiodo-4- (4-methoxyphenoxy) benzoate M.W. 288.27 M.W. 510.06 Methyl 3, 5-diamino-4- (4-methoxyphenoxy) benzoate 4.3 g; 14.2 mmol Sodium nitrite 2.6 g; 37.4 mmol Glacial acetic acid 80 ml; solvent Sulfuric acid 26 ml; solvent Potassium iodide 20 g; 120.0 mmol Water 30 ml; solvent Sulfuric acid (26 ml) was placed in a three-necked flask equipped with a mechanical stirrer and cooled in an ice bath. Sodium nitrite (2.58 g, - 37.4 mmol) was added in small portions and the mixture was stirred for 20 minutes to form a thick solution. To this was added a mixture of methyl 3, 5-diamin-4- (4-methoxyphenoxy) benzoate (4.30 g, 14.22 mmol) in glacial acetic acid (80 ml), dropwise over a period of 30 minutes, keeping the temperature below 10 ° C with the ice bath. The reddish-brown solution was stirred below 10 ° C for 45 minutes, after which it was slowly emptied into an aqueous solution (30 ml) of potassium iodide (20 g) at room temperature, with vigorous stirring. A thick suspension formed and it was stirred at room temperature for 1 hour. The reaction mixture was then heated in an oil bath at 80 ° C (internal temperature) for 15 minutes and then allowed to cool to room temperature. The solution was filtered and the black gummy residue dissolved in - 300 ml of acetone. The dark filtrate, when refrigerated overnight, deposited a dark residue which was collected by decanting the supernatant and the residue was dissolved in 100 ml of acetone. The combined acetone solution was filtered on a basic alumina pad (5 cm) in a 150 ml sintered glass funnel to remove some colored impurities. The alumina pad was rinsed with 100 ml of acetone and the filtrate was evaporated to dryness to give a dark solid as a crude product. This was purified by flash chromatography on silica gel, eluting with hexanes: CH2C12 (60:40). The initial fractions containing the pure product were pooled and evaporated to yield 1.67 g of the desired product as a white, off-white solid. This compound gave a single clean site on TLC (Hexanes: CH2C12; 1: 1; Rf 0.35). The impure fractions were pooled and triturated with absolute EtOH for 16 hours at room temperature. The solid was filtered and dried to yield another 0.3 g of the product as a creamy solid, containing ~5% of the slow-moving impurity as evidenced by TLC (Rf 0.29). The total yield of the product was 27%, 300MHz 1H NMR (CDC13) d 3.78 (s, 3H, OCH3), 3.94 (s, 3H, COOCH3), 6.70 and 6.83 (two d, AA'XX ', 4H, p -subs. Ar-H), 8.51 (s, 2H, Ar-H).
EXAMPLE II This example illustrates the anti-angiogenic properties of methyl 3, 5-diiodo-4- (4-methoxyphenoxy) benzoate in chick chorioallantic membrane (CAM) analysis. The final point of the CAM analysis was a quantitative determination of the biosynthesis of the base membrane by measuring the incorporation of "C-proline into the Type IV collagen protein." A. Approach CAM analysis includes the development of live chicken embryos in Petri dishes under special sterile conditions., only limited numbers of embryos can be used for the evaluation of compounds in a single experiment. For this reason, two separate analyzes were conducted to test the three Biofuente compounds at three concentrations per compound. In this analysis, the known inhibitor of angiogenesis, 2-methoxyestradiol (2 -ME), was used as the positive control and human fibroblast growth factor (hFGF) was used to induce angiogenesis in the CAM. B. Materials Fertilized eggs were supplied by Melody Ranch, Aptos, CA. The proline L- [U-14C] (specific activity, 290 mCi / mmol) was purchased from New England Nuclear, Boston, MA. Collagenase and 2 -ME were obtained from Sigma Chemical Co., St. Louis, MO. The silicone ring cups were obtained by cutting silicone tubes (3 mm diameter) into small "0" rings 1 mm thick. ~ These silicone ring cups can be reused many times if they are sterilized before each test. Plastic Petri dishes (20 x 100 mm) were purchased from Baxter Diagnostics, Inc., Hayward, CA. HFGF-B was obtained from Clonetics Corporation, San Diego, CA. For the test, a minimum amount of acetone-methanol (1: 1) was added to the test compounds for sterilization. The acetone-methanol mixture was then evaporated to dry in a sterile hood. The compounds were first dissolved in dimethyl sulfoxide (DMSO) and then diluted with saline containing methyl cellulose. The final concentrations were 2% DMSO and 0.5% methylcellulose. All test solutions were added to each CAM in 20-ml aliquots. C. Development of CAM to Measure Inhibition of Angiogenesis The method of Folkman, et al. , Dev. Biol. 41: 391-394 (1974), with some modifications, to culture the chicken embryos as follows: Fresh fertile eggs were incubated for three days in a standard egg incubator. On Day 3, the eggs were fractured under sterile conditions and the embryos were placed in 20 x 100 plastic Petri dishes and cultured at 37 ° C in an embryo incubator with a water reservoir on the lower shelf. Air was continuously bubbled into the water reservoir by using a small pump, so that moisture was kept constant in the incubator. Observations were made daily to ensure that all embryos were healthy. Dead or unhealthy embryos were removed from the incubator immediately to avoid contamination. On Day 9, a sterile silicone ring cup was placed in each CAM and 0.5 mCi of 14C-proline with or without the test compound, plus 2.5 ng of hFGF dissolved in saline containing 0.5% methylcellulose was supplied in each Ring cup sterile bell, were tested in parallel 2 -ME to serve as a reference compound. After the addition of the test materials, the embryos were returned to the incubator and the culture continued. On Day 12, all embryos were transferred to ~ a cold room at 4-10 ° C. The anti-angiogenic effect of each test compound was determined by using the collagenase analysis (Maragoudakis, et al., J. Pharm. Exp. Ther. 251; 679-682 (1989)) to measure the incorporation of 14C-proline. in the collagen protein. D. Collagenase Analysis for the Measurement of the Incorporation of 14C-Proline into the Protein - of Collagen With the embryos placed on ice, a piece of CAM of 10 mm diameter was cut under each ring cap and placed in a separate tube. 1.0 ml of phosphate-buffered saline (PBS, pH 7.3) containing 0.11 mg of cycloheximide and 0.17 mg of dipyridyl was added to each tube. The tubes were placed in a boiling water bath for 10 minutes and then cooled to room temperature. The PBS was discarded in each tube after centrifugation at 3000 x g for 10 minutes. The CAM residue was rinsed once with • 3 ml of 15% TCA and then three times- with 3 ml of 5% TCA. The centrifugation was carried out as described above between each wash. At this point all non-radioactively bound proteins were removed and the CAM containing the newly synthesized 14C-collagen protein was suspended in 0.9 ml of 0.1 N NaOH and 1.1 ml of HEPES buffer at pH 7.4. The pH of the sample was neutralized with 0.8 N HCl, using the phenol network according to the indicator. To digest the 14C-collagen protein, 7.5 units of collagenase and 500 mmol of calcium chloride in 40 ml of HEPES buffer were added to the above samples and the mixtures were incubated at 37 ° C for 4 hours. The reaction was stopped by adding 1.0 ml of 20% TCA containing 5 mg of thermal acid in each tube. The samples were configured after vortex mixing at 3000 x g for 10 min. An aliquot of the clear supernatant was taken for scintillation counting to quantitate the radiolabeled tripeptides corresponding to base membrane collagen and other collagen materials synthesized by CAM from 14C-proline. The CAM pellets in each tube were solubilized in 0.5 ml of 1.0 N NaOH upon boiling in a water bath for 5 minutes. An aliquot of the dissolved CAM was used for the determination of the protein using the method provided by Pierce Chemical Co. (Manual instruction for protein analysis using bchoninic acid (BCA) Pierce Chemical Co., Rockford, IL.). The radioactivity per milligram of protein from the CAM treated with a test compound in relation to that of the control CAM, gave the percentage of inhibition of angiogenesis. E. Results Tables 1 and 2 summarize the results of the two separate experiments. BTO-956 showed statistically significant inhibitory effects on angiogenesis induced by hFGF-B. At 75 mg / CAM, the inhibition caused by BTO-956 was 30%, compared to 38% caused by the same concentration of the known antiangiogenic agent, 2-methoxyestradiol. The results of these two experiments suggest that the antiangiogenic effect of BTO-956 in CAM analysis is on the same order as that of 2-methoxyestradiol, a drug that is under development as an antiangiogenic agent. TABLE 1 INHIBITORY EFFECTS OF THE COMPOUNDS IN ANGIOGENESIS INDUCED BY hFGF-B 14C-Proline Incorporated in the Collagen Protein (cpm / mg- Protein dose) (± D., E. Compound (μg / CAM) mean)% Inhibition BTO-956 75 43741651 31 25 5098 + 785 19 8.3 6884 + 135 0 2 -Metoxystradiol 75 38751891 38 25 5068 + 1609 20 8.3 5711 + 1469 9 Control - 63001696 TABLE 2 INHIBITORS EFFECTS OF COMPOUNDS ON ANGIOGENESIS INDUCED BY bFGF-B 14C-Proline Incorporated in the Collagen Protein (cpm / mg- Protein dose) (± D., Comp. (Μg / CAM) medium)% Inhibition BTO- 956 75 753411099a 32 25 9100 + 1664 18 8. 3 10138 + 1625 10 2-Methoxy-estradiol 75 6984 + 1022b 38 25 7303 + 1423 35 8. 3 10499 + 1372 6 Control - 11200 + 829 ~ ^ Significantly less than the control, P < 0.02 b Significantly less than the control, P < 0.01 EXAMPLE III This example illustrates the ID50 determination of BTO-956 in the proliferation of human microvascular endothelial cells (HMVEC). Human microvascular endothelial cells (HMVEC) (Clonetics Corporation # CC-2505) were seeded in 96-well plates at a concentration of 5 x 10 3 cells / well in a 100 μl volume / well cavity.
Endoltelial Growth (EGM-2-MV, Clonetics Corporation # CC-3162). Plates were incubated at 37 ° C in 5% C02 for 24 hours and then covered with 100 μl / well of EGM-2-WV. BTO-956 was diluted to 20 mM in dimethyl sulfoxide (DMSO) and further diluted with EGM-2-MV to 2x the concentrations reported below. Then 100 μl aliquots of BTO-956 / EGM-2-MV dilutions were added to the HMVEC preparations and the plates were incubated at 37 ° C in 5% C02 for 3 days. The relative number of cells was determined by adding 20 μl / ml of Alamar Blue (BioSource International # DAL-1025) for 3-6 hours at 37 ° C and the measurement of color changes indicating metabolic activity when using a Millipore Cytofluor 2350 Fluorescent Measurement System at an excitation wavelength of 530 nm and an emission wavelength of 590 nm. In this analysis, the intensity of the fluorescent signal is directly proportional to the number of cells. A wide range curve was first established for BTO-956 to determine the concentrations to be used for a narrow range curve. A narrow-range curve was then generated to find a region around the ID50 point at which the curve is linear. The ID50 was calculated by using the linear portion of the curve in the intermediate area around the dose giving 50% inhibition and using the equation y = ax + b, where x is the ID50 - calculated and is 50% of the maximum optical density (OD), and a and b are constant. To establish an exact ID50 for BTO-956, a reduced concentration range in HMVECs was tested (from 450 nM to 50 nM). Maximum inhibition was determined by using an upper limit of additional concentration of 10 μM. The baseline value was determined by using the medium without the compound. The ID50 was determined to be 201 nM on Day 3.
EXAMPLE IV A. BTO-956 is orally bioavailable and has minimal normal tissue toxicity Probe administration of BTO-956 to nude mice at doses as high as 1,000 mg / kg-day for 60 to 180 days produced only a slight and transient increase in liver and kidney weights. No other major pathologies were observed. No mortality was observed at one - daily administration of 4,000 mg / kg-day of BTO-956 for 3 weeks. Due to a common toxic effect of the anti-proliferative chemotherapeutic agents, myelosuppression is caused by the depletion of stem cells from the bone marrow, the possibility was proved that BTO-956 could have bone marrow toxicity. Swiss Webster mice were exposed to the drug at 500 mg / kg-day by gavage for a period of 4 weeks and the bone marrow was examined histologically. The bone marrow profiles of the treated animals did not differ of those of control animals exposed only to the carrier vehicle and no hematopoietic abnormalities were found. Both myeloid and erythroid lineages were observed in various stages of maturation. These results demonstrate that oral administration of BTO-956 is well tolerated by animals exposed to the drug for periods of up to six months and has no obvious myelosuppressive activity after one month of daily administration at a therapeutic dose. Preliminary studies (not shown) indicate that oral therapeutic doses of BTO-956 are within the range of 500 to 1, 000 mg / kg-day. B. BTO-956 has strong growth retardation effects on foreign grafts of human breast and ovarian carcinoma 5 Female nude mice nude (nu / nu) Swiss NCr implanted with foreign grafts of human breast carcinoma MDA MB-231 showed a strong growth retardation response to oral treatment with BTO-956 (Figure 1). Cyclophosphamide administered intraperitoneally (CTX; Citoxan; 150 mg / kg), which was used in some regions for adjuvant in breast cancer therapy was used as a positive control. The growth of tumors in the group treated with BTO-956 was suppressed during the six weeks of therapy, while in those control animals treated with the vehicle, it increased approximately 5 times on average (relative mean tumor volume; FIG. 1A) . BTO-956 was also effective in inhibiting the growth of the OVCAR-3 human ovarian carcinoma foreign graft (FIG. IB). Using the same schedule and dose as those described for the MDA MB-231 breast tumor study, but with shorter study duration, the relative mean tumor volume in 5 weeks of the animals exposed to BTO-956 was again approximately 5 times less than that of the controls treated with the vehicle. Two important conclusions of this study are the following: (1) BTO-956 is effective as a cytostatic / cytotoxic antitumor drug when administered orally; (2) BTO-956 is essentially non-toxic even in very high doses administered orally.
C. BTO-956 induces a mitotic disruption in MCF-7 human breast carcinoma cells. Having demonstrated the significant antitumor effects of BTO-956 in vivo, we investigated its cytotoxicity towards human breast carcinoma cells in vi tro. The IC50 (concentration for the 50% reduction of the number of cells relative to those of the control treated with the vehicle) of BTO-956 in the cell cultures MDA MB-231 and MCF-7 treated for 48 hours was 0.3 μM 0.6 μM respectively. Investigation of the effects of BTO-956 (1 μ for 48 hours) on the nuclear morphology of these cells showed that a significant number had fragmented or condensed nucleic features of apoptotic cell death. To test whether BTO-956 exerts its cytotoxicity by disturbing the cell cycle, MCF-7 cultures were exposed to the drug at 1 μM for 24 hours (Figure 2). A large fraction of these cells showed chromosomal condensation characteristics of an prometaphase disruption (Figure 2B). Consistent with this conclusion, treatment of MCF-7 cells with the antimitotic drug colchicine (1 μm) for 24 hours produced identical chromosomal condensation patterns (Figure 2C). After 48 hours of exposure to 1 μm of BTO-956, the MCF-7 cells showed substantial cytotoxicity based on the criterion of loss of membrane integrity measured by trypan blue exclusion analysis. This result is consistent with that of the tumor growth retardation study described above and showed that cell death induced by BTO-956 occurs subsequent to an interruption of prometaphase in cultures of human breast carcinoma cells. To investigate the mitotic disruption caused by BTO-956 in greater detail, flow cytometry was used to determine the effect of the drug on the cell cycle progression of MCF-7 cells. These experiments showed that at least 60% of these cells were disrupted in G2 / M within 24 hours of treatment with 1 μM of BTO-956. The same finding was obtained for MDA MB-231 cells exposed to the drug under the same conditions. These results demonstrate that BTO-956 causes human breast carcinoma cells to accumulate at the interface of the G2 / M cell cycle, presumably by activating a checkpoint for ordering the mitotic axis. Such disrupted cells eventually suffered cell death that can occur by an apoptotic mechanism. D. BTO-956 inhibits dynamic microtubules both in vivo and in cultured cells. Microtubules are the major cytoskelatal components composed of heterodimer bonds of a- and β-tubulin with GTP. Antimitotic drugs such as colchicine, vinca alkaloids and paclitaxel (Taxol) disturb the intrinsic dynamic instability of microtubules that arise from the hydrolysis of GTP by directly binding to tubulin and causing inhibition of axis formation with consequent mitotic disruption. To determine if the antimitotic effect of BTO-956 exposure could be explained by tubulin binding activity, two studies were performed. The effect of the drug on microtubule ordering was directly investigated by using a cell-free fluorescence analysis that included the visualization of microtubule formation from tubulin labeled with rhodamine and the microtubule was seeded in the presence of GTP. In this analysis, 15 μM tubulin was rapidly polymerized to form large filaments of bright microtubules in the absence of a tubulin binding agent (Figure 3A), while in the presence of 1 μM colchicine the microtubule formation was completely inhibited (Figure 3A). figure 3B). The same effect was observed in the presence of 10 μM of BTO-956 (Figure 3D). At a concentration of 1 μM of BTO-956 it was less potent but nevertheless produced structures much shorter than those visible in the control experiment (figure 3C). In a second study, the ability of BTO-956 to disrupt the ordering of cellular microtubules was investigated by using HeLa cells (Figure 4) which also - were interrupted in mitosis when exposed to the drug (Figure 4) showed that HeLa cells exposed to 10 μM of BTO-956 for 1 hour had aberrant microtubule networks that resemble those created upon exposure to • 1 μM of colchicine. Taken together, these findings demonstrate that BTO-956 can interact directly with tubulin in vi tro and in cells to inhibit the formation of microtubules, as well as the antimitotic drug colchicine. E. BTO-956 competes with colchicine for binding to tubulin in vi tro The antimitotic drugs that bind to tubulin interact with the protein at various sites. The colchicine linkages to tubulin heterodimers soluble in a single high affinity site (the binding site of the colchin-) to form a kinetically inert complex. The vinblastine μnions to one or two identical sites of high affinity in tubulin (vinca alkaloid binding sites) that are different from the colchicine site. To determine if the effect of BTO-956 on microtubule ordering is mediated by a specific site on tubulin, the ability of the drug labeled to compete with colchicine or vinblastine to bind purified tubulin was measured as a function of the concentration of colchicine or vinblastine. Figure 5 demonstrates that colchicine inhibits the binding of BTO-956 to tubulin, while vinblastine has no effect. This finding indicates that BTO-956 interacts directly or indirectly with the colchicine site but not with the high affinity binding sites of the vinca alkaloid of tubulin in vi tro. EXAMPLE V This example demonstrates that BTO-956 can down regulate the expression of cytokines in murine macrophages stimulated by lipopolysaccharides. The following is an analysis to measure the ability of the Compounds of Formula I to reduce levels of Tumor Necrosis Factor (TNF-a). A. Cell Line The murine macrophage cell line PU5-1.8 was purchased from the American Type Culture Collection ATCC, Rockville, MD). The cells were grown in DMEM medium supplemented with 100 mM sodium pyruvate, 0.1 mM non-essential amino acids, 2 mM glutamine and 5% bovine fetal serum (Life Technologies, Staten Island, NY). The cells were kept in a humid atmosphere of 5% C02-95% air at 37 ° C. The cells were passed twice a week to finally tap the side of the flask to dislodge the adherent cells. Both adhered and non-adhered cells passed the test. Cells that grew exponentially were seeded at 5 x 10 5 / ml per 60 mm vessels for 24 hours before the experiment. The test compounds were supplied in 1 ml volumes of medium added to each container at the beginning of the experiment. All vessels were incubated at 37 ° C in 5% C02- 95% air for 3 hours. B. Reagents The cDNA Tumor Necrosis Factor (TNF-a) was obtained from the ATCC (Rockville, MD). [α-32 P] -dCTP (250 μCi) and nylon membranes (Hybond N) were obtained from Amersham (Arlington Heights, II.). The mattress (used as control) was purchased from Sigma Chemical Company (St. Louis MO). The lipopolysaccharides (LPS) of Escherichia Coli were purchased from DIFCO Laboratories (Detroit, MI). All plastic supplies were from VWR Scientific produets (San Francisco, CA). C. Northern Spotting Total was isolated by the guanidinium-cesium chloride method as described in N.S. Waleh, J. Gallo, T.D. Grant, B.J. Murphy, R.H. Kramer and R.M. Sutherland. (1994) "Selective Downregulation of integrin receptors in spheroids of squamous cell carcinoma" (Selective down regulation of the integral receptors in squamous cell carcinoma spheroids) Cancer Res. , 54: 838-843. Five to 10 μg of the total was examined in electrophoresis in 1% agarose gels containing 6% formaldehyde.
After electrophoresis, the gels were stained with ethidium bromide to visualize the positions of 28S and 18S RNA. The RNAs were then transferred to nylon membranes (Amersham Hybond N) by capillary staining and fixed to the filter by exposure to UV light. The spots were tested with TNF-α-32P-tagged cDNA sequences obtained from the American Type Culture Collection (ATCC). The TNF-α cDNA was a 1.1 kb PstI fragment of plasmid pE4 in E. Coli MM294 (ATCC 39894). Hybridizations were carried out at 42 ° C in 50% formamide, 5 X SSC, 5X Denhardt's solution, 0.1% SDS, and 0.3 mg / ml salmon sperm DNA. The filters were washed by 1 X SSC, 0.1% SDS, twice at room temperature for 15 minutes and once at 55 ° C in 0.1 X SSC, 0.1% SDS for 1 hour. The filters were exposed to X-ray film at -70 ° C using an intensification screen (Coronex Hi-Plus). The hybridized bands were quantified by analyzing the images obtained by using a video densitometer (Applied Imaging Corporation, Santa Clara CA). The film densities were calibrated using a prism of optical density. D. RESULTS Treatment of murine macrophages of PUS-1.8 with LPS (100 ng / ml) for 3 hours resulted in an increase - significant (> 7 fold) at the level of TNF-a mRNA as determined by Northern blot analysis (see figure 1). Treatment of the cells with only BTO-956 or colchicine at a concentration of 10 μM had no effect on the expression of TNF-α mRNA. However, the addition of colchicine or BTO-956 at 10 μM to the cultures treated with LPS, resulted in the substantial reduction of TNF-α mRNA accumulation. The levels of inhibition were 68% for colchicine and 61% for BTO-956 respectively. To establish a concentration-effect relationship, macrophages were exposed to various concentrations of BTO-956 in the presence of LPS stimuli for 3 hours. Figure 2 illustrates that the amounts of TNF-a mRNA declined with the increased concentrations of BTO-956. The maximum effect was observed at concentrations of 10 μM of BTO-956. The total RNA was intact in the cells treated with different concentrations of BTO-956 and colchicine, suggesting that the toxicity was not problematic. The results indicate that BTO-956 at 10 μM partially suppresses but does not completely inhibit the expression of TNF-α mRNA in the murine macrophage cell line PU5-1.8. This indicates that BTO-956 down-regulates the responses stimulated by LPS by affecting the microtubule-dependent signaling pathways.
This finding is of clinical interest because BTO-956 can be used as a new class of compounds to avoid the excessive production of TNF-a mediated by LPS and its undesirable side effects. This is especially attractive, because BTO-956 has been shown to be a safe and well tolerated drug when administered orally to animals. It should be understood that the foregoing description is intended to be illustrative and not restrictive. Many modalities will be apparent to those experts in the field when reading the above description. Therefore, the scope of the invention should not be determined with reference to the foregoing description, but instead should be determined with reference to the appended claims, together with the full scope of equivalents to which such claims are entitled. Exhibits of all articles and references, including patent applications and publications, are incorporated herein for reference for all purposes.

Claims (25)

    -
  1. CLAIMS 1. A method for inhibiting angiogenesis in a tissue or organ, the method comprising contacting the tissue or organ with an anti-angiogenic amount of a compound having the formula: or a pharmaceutically acceptable salt thereof; wherein: R1 is C? -C6-alkyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C3-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C6-alkenyl, C2-C3 alkynyl, aryl, hydroxyl, C-Cg-alkoxy, halogen, N02 and NH2; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl. -
  2. 2. A method according to claim 1, wherein: R1 is methyl or ethyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C4-alkyl, C2-C4-alkenyl, C2-C4 alkynyl, aryl, hydroxyl, C? -C -alkoxy, halogen , N02 and NH2; R6, and R7 are each independently selected from the group consisting of hydrogen, C? -C4-alkyl, C2-C4-alkenyl, C2-C4 alkynyl, aryl, hydroxyl, C3.-C4-alkoxy, halogen, N02, and NH2; R8, and R9 are iodine; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  3. 3. A method according to claim 1, wherein the compound is methyl 3,5-diiodo-4- (4-methoxyphenoxy) benzoate.
  4. 4. A method according to claim 1, wherein the tissue or organ is a mammalian subject.
  5. 5. A method according to claim 1, wherein the compound is formulated in a pharmaceutically acceptable form with an excipient or vehicle.
  6. 6. A method according to claim 1, wherein the compound is formulated in a liposome
  7. 7. A method according to claim 6, wherein the liposome is conjugated to a target residue that is specific for endothelial cells.
  8. 8. A method for treating a mammalian disease associated with undesirable and uncontrolled angiogenesis, the method comprising administering to a mammal an anti-angiogenic amount of a compound having the formula or a pharmaceutically acceptable salt thereof; Wherein: R1 is C? -C3-alkyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C6-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; Rs, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C3-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  9. 9. A method according to claim 8, wherein: R1 is methyl or ethyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C4-alkyl, C2-C4-alkenyl, C-C4 alkynyl, aryl, hydroxyl, C? -C4-alkoxy, halogen , N0 and NH2; R6, and R7 are each independently selected from the group consisting of hydrogen, C? -C4-alkyl, C2-C4-alkenyl, C2-C4 alkynyl, aryl, hydroxyl, C? -C4-alkoxy, halogen, N02 and NH2; R8, and R9 are iodine; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  10. 10. A method according to claim 8, wherein the compound is methyl 3,5-diiodo-4- (4-methoxyphenoxy) benzoate.
  11. 11. A method according to claim 8, wherein the mammalian disease is a member selected from the group consisting of arthritis, arteriosclerotic plaques, diabetic retinopathy, neovascular glaucoma, trachoma and corneal graft neovascularization, psoriasis, scleroderma, hemangioma, hypertrophic scarring, vascular adhesions and angifibroma.
  12. 12. A method according to claim 8, further comprising the step of determining the inhibition of undesirable and non-controlled angiogenesis by tissue biopsy.
  13. 13. A method of inhibiting endothelial cell vascularization, the method comprising contacting a tissue or organ containing the endothelial cells with an anti-angiogenic amount of a compound having the formula: or a pharmaceutically acceptable salt thereof; wherein: R1 is C? -C6-alkyl; R2, R3, R4, and R5 each independently selected from the group consisting of hydrogen, Ci-Css-alkyl, C2-C6-alkenyl, C2-C3 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen, N02 and NH2; R6, R7, R8, and R9 each independently selected from the group consisting of hydrogen, C? -C3-alkyl, C2-C6-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C 6 -alkoxy, halogen , N02 and NH2; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  14. 14. A method according to claim 8, wherein: R1 is methyl or ethyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C1-C4-alkyl, C2-C-alkenyl, C2-C4 alkynyl, aryl, hydroxyl, C? -C4-alkoxy, halogen, N02 and NH2; Rs, and R7 are each independently selected from the group consisting of hydrogen, C1-C4-alkyl, C2-C4-alkenyl, C2-C4 alkynyl, aryl, hydroxyl, C C4-alkoxy, halogen, N02 and NH2; R8, and R9 are iodine; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  15. 15. A method according to claim 13, wherein the compound is methyl 3,5-diiodo-4- (4-methoxy-nyl) -benzoate.
  16. 16. A method according to claim 13 wherein the endothelial vascularization is a non-cancerous tissue or organ.
  17. 17. A method according to claim 13 wherein the compound is formulated in a liposome and this liposome is conjugated to a target residue that is specific for endothelial cells.
  18. 18. A method for inhibiting the growth of a tumor in a mammal, the method comprising: (a) administering to the mammal an anti-angiogenic amount of a compound having the formula: or a pharmaceutically acceptable salt thereof; wherein: R1 is C? -C6-alkyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C6-alkenyl, C2-C3 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; Rd, R7, R8, and R9 each independently selected from the group consisting of hydrogen, C? C6 alkyl, C2-C3-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C 6 -alkoxy, halogen , N02 and NH2; X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl; and (b) histologically observe the vascularization of the tumor, thereby determining the inhibition of tumor growth.
  19. 19. A method for inhibiting the growth of a tumor in a mammal according to claim 18 wherein the administration to the mammal is carried out with immunotherapy.
  20. 20. A method for inhibiting the growth of a tumor in a mammal according to claim 19 further comprising the step of administering to the mammal a tumor vaccine.
  21. 21. A method for reducing the level of tumor necrosis factor (TNF-α) produced by a cell, the method comprises contacting the cell with a compound having the formula: or a pharmaceutically acceptable salt thereof; wherein: R1 is C? -C6-alkyl; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C6-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; R6, R7, R8, and R9 each independently selected from the group consisting of hydrogen, C? C6 alkyl, C2-C6-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C 6 -alkoxy, halogen , N02 and NH2; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
  22. 22. A method according to claim 21, wherein the compound is methyl 3,5-diiodo-4- (4-methoxyphenoxy) benzoate.
  23. 23. A method according to claim 21, wherein the compound is formulated in a liposome
  24. 24. A method according to claim 21, wherein the compound is formulated in a liposome and the liposome is conjugated to a target residue. which is specific for tumor necrosis factor-a (TNF-a) or a receptor for tumor necrosis factor-a.
  25. 25. A method for treating an inflammatory disease, the method comprises a therapeutically effective amount of a compound having the formula or a pharmaceutically acceptable salt thereof; wherein: R1 is C? -C6-alkyl ?; R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, C? -C6-alkyl, C2-C6-alkenyl, C2-C3 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen , N02 and NH2; R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, C? -C3-? Alkyl, C2-C6-alkenyl, C2-C6 alkynyl, aryl, hydroxyl, C? -C6-alkoxy, halogen, N02 and NH2; and X, if present, is selected from the group consisting of oxygen, sulfur, -CH2- and carboxyl.
MXPA/A/2000/009345A 1998-03-25 2000-09-25 Benzoates derivatives for inhibiting angiogenesis MXPA00009345A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/079,313 1998-03-25

Publications (1)

Publication Number Publication Date
MXPA00009345A true MXPA00009345A (en) 2001-07-09

Family

ID=

Similar Documents

Publication Publication Date Title
US6150407A (en) Methods for inhibiting angiogenesis
US6433012B1 (en) Method for inhibiting inflammatory disease
US6169104B1 (en) Di-aryl ethers and their derivatives as anti-cancer agents
AU2002311985B2 (en) Methods for inhibiting angiogenesis
EP0954299B1 (en) Thyroxine analogues having no significant hormonal activity for treating malignant tumours
JP6891286B2 (en) Tubulin inhibitor
JP2002534400A (en) Colhinol derivatives as vascular damaging agents
WO1997046228A9 (en) Method of treating malignant tumors with thyroxine analogues having no significant hormonal activity
AU2002311985A1 (en) Methods for inhibiting angiogenesis
JP7001599B2 (en) Dactinomycin Compositions and Methods for the Treatment of Acute Myeloid Leukemia
CA3103958A1 (en) Activators of the unfolded protein response
KR20100051837A (en) Treatment of pediatric tumors
JP2022504184A (en) Combination therapy for the treatment of melanoma of the grape membrane
US20040048808A1 (en) Methods for inhibiting angiogenesis
MXPA00009345A (en) Benzoates derivatives for inhibiting angiogenesis
WO2005072723A1 (en) Pharmaceutical composition comprising hydroxylphenyl derivatives of rosmarinic acid for anticancer
MXPA99008753A (en) Di-aryl ethers and their derivatives as anti-cancer agents
WO1998034583A2 (en) Enhanced anti-angiogenic activity of permanently charged derivatives of steroid hormones