[go: up one dir, main page]

MXPA99008753A - Di-aryl ethers and their derivatives as anti-cancer agents - Google Patents

Di-aryl ethers and their derivatives as anti-cancer agents

Info

Publication number
MXPA99008753A
MXPA99008753A MXPA/A/1999/008753A MX9908753A MXPA99008753A MX PA99008753 A MXPA99008753 A MX PA99008753A MX 9908753 A MX9908753 A MX 9908753A MX PA99008753 A MXPA99008753 A MX PA99008753A
Authority
MX
Mexico
Prior art keywords
hydrogen
group
alkyl
compounds
methyl
Prior art date
Application number
MXPA/A/1999/008753A
Other languages
Spanish (es)
Inventor
Daniel Tuse
Xiaoying Chen
Keith R Laderoute
Charles K Hiebert
Original Assignee
Biosource Technologies Inc
Xiaoying Chen
Charles K Hiebert
Keith R Laderoute
Olsen Cris M
Sri International
Daniel Tuse
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, Xiaoying Chen, Charles K Hiebert, Keith R Laderoute, Olsen Cris M, Sri International, Daniel Tuse filed Critical Biosource Technologies Inc
Publication of MXPA99008753A publication Critical patent/MXPA99008753A/en

Links

Abstract

The present invention relates, inter alia, to compounds that bind tubulin and exhibit anti-mitotic properties and to methods of using such compounds to inhibit abnormal cell mitosis and, in particular, to inhibit tumor cell growth. In addition, methods are presented of treating mammalian diseases associated with undesired and uncontrolled angiogenesis.

Description

ETHER OF DI-ARILO AND ITS DERIVATIVES. AS AG AGAINST _ E7 L CANCER This application is a partial continuation of the U.S. Provisional Patent Application, Serial No. 60 / 041,679, filed March 26, 1997 (Proxy File No. 017942-000200), the teachings of which are incorporated herein by reference. This application also relates to the commonly assigned, pending US Provisional Patent Application, Serial No., which bears Proxy File No. 017942-000600, filed on the same date as the present one, whose teachings are incorporated here as a reference BACKGROUND OF THE INVENTION Cell mitosis is a multi-step process, which includes cell division and duplication (Alberts, B. et al., In The Cell, pages 652-661 (1989); Stryer E. Biochemistry (1988)). Itsosis is characterized by the intracellular movement and segregation of organelles, which include mitotic spikes and chromosomes. The movement and segregation of organelles is facilitated by the polymerization of the cellular protein tubulin. Microtubules form from the polymerization tubulin a and ß and the hydrolysis of guanosine triphosphate (GTP). The formation of microtubules is important for cellular mitosis, cellular locomotion and the movement of highly specialized cellular structures, such as cilia and flagella. Unfortunately, numerous diseases are characterized by abnormal cellular mitosis. For example, uncontrolled cellular mitosis is a hallmark of cancer. Cancer is the leading cause of death, second only to heart disease, both men and women. In the fight against cancer, numerous techniques have been developed and are the subject of current research aimed at understanding the nature and cause of the disease and providing methods for controlling or curing it. Up to now, three main families of anti-tumor agents are known. Each of the families of agents is associated with a recognized mechanism of action. First, the anti-tumor agents can be alkylating agents, which are generally covalently bound to the DNA to form bifunctional lesions. These bifunctional lesions involve adjacent or nearby bases of the same cord or, alternatively, involve bases in the opposing cords, which form intertwined interlaces. Examples of alkylating agents include nitrogen mustard, cyclophosphamide and chlorambucil. Toxicities associated with the use of alkylating agents include nausea, vomiting, alopecia, hemorrhagic cystitis, pulmonary fibrosis, e € c. Second, anti-tumor agents can be anti-metabolites, which generally inhibit the enzymes involved in the synthesis or assembly of DNA. Alternatively, an anti-metabolite can serve as a fraudulent or analogous substrate for DNA processes. Examples of anti-metabolites include purine, pyrimidine and folate antagonists and plant alkaloids, such as vincristine and vinblastine. The toxicities associated with the use of anti-metabolites include alopecia, myelosuppression, vomiting, nausea, peripheral neuropathy, etc. Third, anti-tumor agents can be antibiotics, which work by intercalating the DNA helix or introducing cord breaks in the DNA. Examples of antibiotics include doxorubicin, daunorubicin and actinomycin. The toxicities associated with the use of antibiotics include myxeosuppression, anaphylactic reactions, anorexia, cardiotoxicity, pulmonary fibrosis, etc. Ionizing radiation is a well-established treatment for malignant diseases and has proven benefits for both curative and palliative purposes. However, radiotherapy can have several undesirable complications, such as mucositis, leukopenia, desquamation, spinal cord necrosis, and obliterative endarteritis. These complications frequently limit the ability to deliver the therapeutic dose of radiation or cause significant morbidity following treatment.Many chemotherapeutic agents are also toxic to normal tissue cells andThus, the side effects of chemotherapy are sometimes almost as devastating to the patient as the tumor itself. One approach to reduce the side effects of chemotherapy has been the attempt of targeted chemotherapeutic agents, which include radioisotopes and various plant and bacterial toxins, to tumor cells by binding them. agents to antibodies that are specific for the present antigens in a tumor cell. See, for example, U.S. Patent Nos. 4,348,376 and 4,460,559, which describe the radioimmunotherapy of solid tumors (carcinomas) using a carcinoembryonic antigen antibody, and U.S. Patent No. 5,595,721, which is directed to radioimmunotherapy of lymphoma, a more disseminated tumor. However, while there are several reports of individual successes, the results of the therapy using antibody conjugates have disappointed. Remission regimens have been low and generally not reproducible. Although thousands of potential anticancer agents have been evaluated, treatment of human cancer remains fraught with complications that often present an array of suboptimal treatment selections. As such, chemotherapeutic agents that "possess little or no toxicity, that are cheap to obtain or manufacture, that are well tolerated by the patient, and that are administered easily, would be a convenient addition to the therapeutic modalities currently available to the oncologist. Agents that selectively sensitize malignant tissue, to allow lower doses of radiation or therapy to achieve the same therapeutic effect, with less damage to healthy tissues, are also desirable, similarly, agents that prevent the occurrence of cancer or its recurrence, They are also desirable The present invention remedies these needs, by the provision of such chemotherapeutic and sensitizing agents.
COMPENDIUM OF THE INVENTION The present invention relates to (i) compounds that bind to tubulin and exhibit anti-mitotic properties; (ii) methods of using these compounds, to inhibit the abnormal cellular mitosis and, in particular, to inhibit the growth of tumor cells; (iii) compounds that inhibit the osteogenesis and vascularization of endothelial cells; (iv) methods of using these compounds to inhibit angiogenesis and vascularization of endothelial cells; (v) compounds that reduce the level of tumor necrosis factor (TNF-a) produced by a cell; (vi) methods for using these compounds to reduce TNF-a production and to treat inflammatory diseases; and (vii) pharmaceutical compositions comprising these compounds. In one embodiment, the present invention provides compounds having the general formula: or their pharmaceutically acceptable salts. In Formula I, R1 'R2, R3 and R4 are each, independently selected and are functional groups including, but not limited to: H, alkyl, S-alkyl, alkenyl, alkynyl, aryl, hydroxyl, alkoxy, halogen , N02 and NH2. R5, R6, R7 and R8 are each independently selected and are functional groups including, but not limited to: H, S-alkyl, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alkoxy and halogen. In Formula I, X, if present, is a group that includes, but is not limited to, the following: ^ - CH2 - y - c - A 'in the above formula, together with the carbons to which it is attached, forms a carbocyclic or heterocyclic ring of 3, 4, 5, or 6 m., Optionally substituted. . R9 and , In the above formula, are, independently, hydrogen, alkyl and halogen. And, in Formula I, it is a functional group that includes, but is not limited to H, alkyl and alkoxy. Z is a functional group that includes, but is not limited to, the following: and: -OírOQ Q, in the above formula, is a functional group that includes, but is not limited to, H, alkyl and S-alkyl, and Y is selected such that if O Z is \ OQ and Q is methyl, then Y is different from methoxy and ethoxy. Within the scope of Formula I, certain modalities are preferred. Examples of particularly preferred compounds include, but are not limited to, those compounds noted below. The compounds listed below and through this specification are referred to by key numbers, which are used only for convenience and are strictly arbitrary for the purposes of this invention.
The compounds of the present invention can be used in vivo or in vitro to inhibit the growth of tumor cells. The compounds of the present invention are also useful because they bind to tubulin and exhibit anti-mitotic properties and thus, can be used __ in vivo or in vitro, to inhibit cellular mitosis.
Also, the compounds of the present invention are useful because they exhibit angiogenesis and vascularization of endothelial cells. In addition, the compounds of the present invention are useful because they reduce (e.g., down-regulate) the level of TNF-α, produced by a cell. The compounds of the present invention are also useful in conjunction with other cancer therapies, including radiation therapy, chemotherapy and immunotherapy (which includes radioimmunotherapy). Within these embodiments, the compounds of the present invention are particularly useful as sensitizing agents. When administered before, concurrent with, or after treatment with cancer therapy, the compounds of the invention increase the sensitivity of the cancer cells to therapy. This results not only in an increase in the effectiveness of the therapy, but also in reducing the required dose, whereby the undesirable side effects are reduced. It has been found that the compounds of the present invention are safe, effective, non-toxic and easy to administer. As such, in one embodiment, the present invention provides a method for inhibiting the growth of tumor cells, this method comprises contacting the tumor cell with a compound having the general formula: or their pharmaceutically acceptable salts. In another embodiment, the present invention provides a method for treating cancer, this method comprises administering to the subject mammal having cancer, a therapeutically effective amount of a compound having the general formula: or their pharmaceutically acceptable salts. The compounds of the present invention are useful in inhibiting the growth of a number of tumor cells and in treating a wide variety of cancers. These tumor cells include, by way of example and not limitation, tumor cells of the lung, colon, breast, ovary, prostate and liver, as well as squamous cell carcinomas.
Such cancers include, by way of example and not limitation, carcinomas such as cancers of the pharynx, colon, rectal, pancreatic, stomach, liver, lung, breast, skin, prostate, ovary, cervical, uterine and vesicle; lympholas; gliomas; retinoblastomas and sarcomas. Also, according to the above methods, mammalian subjects include, but are not limited to, humans, laboratory animals, domestic pets and farm animals. Furthermore, it will be readily apparent to the experts that by using the compounds of Formula I, the growth of the tumor cells can be inherited in other higher order organisms, including, but not limited to, plants, insects, fish and Similar. In yet another embodiment, the present invention provides a method for treating a disease, characterized by abnormal cellular mitosis, this method comprises administering to a mammalian subject, having such a disease, a therapeutically effective amount of a compound having the general formula: or their pharmaceutically acceptable salts. The present invention also provides methods for treating an afflicted mammal with a disease characterized by abnormal cellular mitosis and, in particular, the growth of tumor cells, by administering to the mammal an effective amount of ionization or non-ionizing radiation. , or an effective amount of a chemotherapeutic or immunotherapeutic agent, in conjunction with an effective sensitizing amount of a compound of Formula I. The compounds intensify the deleterious cellular effects of exposure to ionization radiation or to a chemotherapeutic or immunotherapeutic agent, made in cells that suffer from abnormal cellular mitosis. Such effects include, for example, damage to cellular DNA, such as breakage of the DNA strand, disruption in cellular function, such as disruption of DNA function, death of cells and the like. In addition to the above, the compound of the present invention can be used in a therapy in conjunction with other known chemotherapeutic or antineoplastic agents (eg, vinca alkaloids, antibiotics, anti-metabolites, platinum coordination complexes, etc.). For example, the compounds of the present invention "can be used in therapy in conjunction with a vinca alkaloid compound, such as vinblastine, vincristine, taxol, etc., an antibiotic, such as adriamycin (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 .; and a platinum coordination complex, such as cisplatin (cis-DDP), carboplatin, etc. In addition, those skilled in the art will appreciate that the compounds of the present invention can be used in therapy in conjunction with other known chemotherapeutic or antineoplastic compounds. In another aspect, this invention relates to a method to inhibit the vascularization of endothelial cells, the method involves the contact of a cell, tissue or organ, which has endothelial cells, with an angiogenic amount of a compound of Formula I or its pharmaceutically acceptable salts. In another aspect, this invention relates to a method for effectively inhibiting unwanted angiogenesis in a tissue or organ, by administering to the mammal a compound of the Formula, or its pharmaceutical composition, in a dose sufficient to inhibit angiogenesis. . The invention also provides methods for preventing a disease, characterized by abnormal cellular mitosis. By the administration of a compound of Formula I to a mammal, which is or may be susceptible to such diseases, the development of abnormal mitosis can be prevented. Similarly, the present invention provides methods for preventing a disease, characterized by abnormal cellular mitosis from a recurrence.
In yet another aspect, the present invention provides a method for reducing the level of tumor factor A (TNF-α) produced by a cell. As As a result of its ability to reduce TNF-a, the compounds of Formula I are particularly useful for treating 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 synthetic schemes that can be used to prepare the compounds of the present invention. Figure 2 illustrates the in vitro metabolism of compounds BTO-956, 964, 966 and 967 in human leukemia cells. In this experiment, human leukemia cells (HL60) were incubated for 4 hours, in the presence of one the compounds BTO-956, BTO-964, BTO-966 or BTO-967, with (symbols closed) and without (symbols) open) the fraction S9 of the microsome of the liver of the rat. Following exposure for 4 hours to the drug and S9, the cells were rinsed and re-plated at a density of 2 x 10 5 cells per ml, and counted 3 days later.
DETAILED DESCRIPTION OF THE INVENTION AND PREFERRED MODALITIES The present invention relates to (i) compounds that bind to tubulin and exhibit anti-mitotic properties; (ii) methods for using these compounds, in the inhibition of abnormal cellular mitosis and, in particular, to inhibit the growth of tumor cells; (iii) compounds that inhibit the osteogenesis and vascularization of endothelial cells; (iv) methods of using these compounds to inhibit angiogenesis and vascularization of endothelial cells; (v) compounds that reduce the level of tumor necrosis factor (TNF-a) produced by a cell; (vi) methods for using these compounds to reduce TNF-a production and to treat inflammatory diseases; and (vii) pharmaceutical compositions comprising these compounds. A. DEFINITIONS The term "independently selected" is used here to indicate that the R groups, for example the groups R1, R2, R3 and R4, may be identical or different. (for example R1, R, R and R4 can all be hydrogen, or R1 and R4 can be hydrogen and R2 and R3 can be halogen. The term "alkyl" is used herein to refer to a monovalent, saturated hydrocarbon radical or unsaturated, straight or branched chain having 1 to 12 carbon atoms, preferably 1 to 6 carbon atoms When the alkyl group has 1 to 6 carbon atoms, it is referred to as "lower alkyl". suitable include, for example, methyl, ethyl, n-propyl, i-propyl, 2-propenyl (or allyl), n-butyl, i-butyl (or 2-methylpropyl), etc. As used herein, the term covers "substituted alkyls." "Substituted alkyl" refers to an alkyl, as justly described, that includes one or more functional groups, such as lower alkyl, aryl, acyl, halogen (i.e., haloalkyls, e.g. CF3), hydroxy, amino, alkoxy, alkylamino, akylamino, acyloxy, aryloxy, aryloxyalkyl, mercapto, hi cyclic drocarbides, both saturated and unsaturated, heterocycles and the like. These groups can be attached to any carbon atom in the alkyl part. The term "S-alkyl" is used herein to name the group -SR, where R is lower alkyl or substituted lower alkyl, as defined herein. The term "aryl" is used herein to name an aromatic substituent, which may be a simple aromatic ring or multiple aromatic rings that are fused together, covalently bonded, or linked to a common group, such as a methylene part or of ethylene. The common linker group can also be a carbonyl, as in the benzophenone. This one or more aromatic rings can include phenyl, naphthyl, biphenyl, diphenylmethyl and benzophenone, among others. The term "aryl" also embraces "arylalkyl". "Substituted aryl" refers to the aryl, just described, which includes one or more functional groups, such as lower alkyl, acyl, halogen, haloalkyls (for example CF3), hydroxy, amino, alkoxy, alkylamino, acylamine, acyloxy, mercapto and the cyclic hydrocarbons, both saturated and unsaturated, which are fused to one or more aromatic rings, cently bound or linked to a common group, such as a part of methylene or ethylene. The linker group can also be a carbonyl, such as in cyclohexyl phenyl ketone. The term "substituted aryl" embraces the "substituted arylalkyl". The term "halogen" is used herein to refer to the 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 -RR1, wherein R and R 1 may independently be hydrogen, alkyl, substituted alkyl, aryl, substituted aryl or acyl.
The term "nitro" is used herein to refer to the group -O2. 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 group, in which the alkyl, aryl, substituted aryl groups, Arylalkyl and substituted arylalkyl are 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 a branched, unsaturated straight or cyclic monovalent hydrocarbon radical having at least one carbon-to-carbon double bond. The radical can be in any conformation, cis or trans, with respect to the double bonds. Suitable alkenyl radicals include, for example, ethenyl, propenyl, isopropenyl, cyclopropenyl, butenyl, isobutenyl, cyclobutenyl, tertiary butenyl, pentenyl, hexenyl, etc. The term "carbocyclic" is used herein to refer to a ring structure of non-aromatic carbons. It may include cyclopropane, cyclobutane, cyclopentane, cyclohexane, and the like. The carbocyclic ring may be optionally substituted with one or more functional groups, such as alkyl, halogen, hydroxy, amino, alkoxy, hydroxyalkyl, and the like. The term "heterocyclic" is used herein to refer to aromatic or non-aromatic ring structures, which contain at least one heteroatom. They include oxacyclopropane, azacyclopropane, thiophene, furan, pyrrole, imidazole, pyridine and the like. The term "alkynyl" is used herein to refer to a monovalent, straight-chain or cyclic, branched, unsaturated hydrocarbon radical having at least one carbon-to-carbon triple bond. Suitable alkynyl radicals include, for example, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl, etc. The term "angiogenesis" refers to the "generation of new blood vessels in tissues, organs or tumors." The term "make contact" is used here interchangeably with the following: combined with, added to, in mixture with, passed on, incubated with, flowing on, etc. Also, 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 the compounds which retain the biological effectiveness and properties of the free bases and which are obtained by the reaction with inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluene sulfadic acid, salicylic acid and the like. The pharmaceutically acceptable salts include, for example, the alkali metal salts, such as sodium and potassium, alkaline earth metal salts and ammonium salts. "A sufficient amount", "an effective amount" or a "therapeutically effective amount" refers to an amount of a compound or composition effective to reduce, suppress or remove cell growth. 'malignant or resulting in relief of symptoms associated with cancerous diseases. The desired result can be a subjective relief of one or more symptoms or an objectively identifiable improvement in the dose recipe, a decrease in tumor size, a decrease in the amount of growth of cancer cells, as noted by the clinical observer or another qualified one. The term "anti-angiogenic" amount refers to the amount of a compound or composition effective to decrease, suppress or inhibit angiogenesis or that results in relief of symptoms associated with angiogenic diseases. The term "sensitization increase ratio" (SER) refers to the ratio of the radiation dose, dose of the chemotherapeutic agent or dose of the immunotherapeutic agent required to reduce the surviving fraction of the cancer cells to a predetermined level (eg, 1% of the control), compared to the dose required to obtain the same survival fraction with a present sensitizer. An "effective sensitizing amount" is that amount of the compound that is effective, in a single or multiple dose administration, to the cells, in increasing the severity or extent of the cellular effects detrimental to the cells. cancer cells, caused by exposure to or treatment with ionizing or non-ionizing radiation, a chemotherapeutic agent or an immunotherapeutic agent. The terms "cancer treatment", "therapy" and the like generally refer to any improvement in the mammal having cancer, wherein the improvement can be attributed to treatment with the compounds of the present 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 the subjective symptoms of improvement or response to therapy. Alternatively, the clinician may notice a decrease in tumor size * or tumor burden, based on a physical examination, laboratory parameters, tumor markers or radiographic findings. Some laboratory signs that the clinician may observe in response to therapy include standardization of tests, such as leukocyte count, erythrocyte count, platelet count, erythrocyte sedimentation rate, and various levels of enzymes. Additionally, the clinician may observe a decrease in a detectable tumor marker. Alternatively, other tests can be used to evaluate the objective improvement, such as sonogram tests, nuclear magnetic resonance test and positron emission test. The "growth inhibition of tumor cells" can be evaluated by any accepted method of measuring whether the growth of tumor cells has decreased or decreased. It includes direct observation and indirect evaluation such as subjective symptoms or objective signs, as discussed above.
B. COMPOUNDS The present invention provides compounds that, inter alia, inhibit the growth of tumor cells. Also, the compounds of the present invention bind to tubulin and exhibit anti-mitotic properties. In addition, the compounds of the present invention inhibit angiogenesis and vascularization of endothelial cells. As a result of their properties, the compounds of the present invention can be used, inter alia, to inhibit the growth of tumor cells, to inhibit abnormal cell mitosis and to inhibit angiogenesis. In one embodiment, the present invention provides compounds having the general formula: or their pharmaceutically acceptable salts. In Formula I, R1 'R2, R3 and R4 are each independently selected and are functional groups including, but not limited to: H, alkyl, S-alkyl, alkenyl, alkynyl, aryl , hydroxyl, alkoxy, halogen, N ?2 and NH2-R5, R6, R7 and R8 are each, independently selected and are functional groups including, but not limited to: H, S-alkyl, alkyl, alkenyl, alkynyl , aryl, hydroxyl, alkoxy and halogen.
In Formula I, .X, if present, is a group that includes, but is not limited to, the following: - CH2 - and - C - A 'in the above formula, together with the carbons to which it is attached, forms a carbocyclic or heterocyclic ring of 3, 4, 5, or 6 members, optionally substituted. R9 and , In the above formula, are, independently, hydrogen, alkyl and halogen. And, in Formula I, it is a functional group that includes, but is not limited to H, alkyl and alkoxy. Z is a functional group that includes, but is not limited to, the following: -sfcOQ Q, in the above formula, is a functional group that includes, but is not limited to, H, alkyl and S-alkyl, and Y is selected so that if * "*" Q is methyl, so Y is different from methoxy and ethoxy. Within the scope of Formula I, certain modalities are preferred. In Formula I, a preferred embodiment is that in which X is -O-; And it's methoxy; Q is methyl; R1 and R4 are both hydrogen, R2 and R3 are both iodo; and R5, R6, R7 and R8, are all hydrogen. Another preferred embodiment is that in which X is -O-; And it's hydrogen; O Z is - C ^ \ OQ Q is methyl,; R1 and R4 are both hydrogen, R2 and R3 are both iodo; and R5, R6, R7 and R8, are all hydrogen.
Another preferred embodiment is that in which X is -O-; And it's alkyl; Q is methyl,; R and R4 are both hydrogen, R2 and R3 are both iodo; and R5, R6, R7 and R8, are all hydrogen. Still another preferred embodiment is that in which X is -0-; And it's methoxy; Q is methyl; R1 and R4 are both hydrogen, R2 and R3 are both _yode; and R5, R6, R7 and R8, are all hydrogen. Still another preferred embodiment is that in which X is -0-; And it's methoxy; Z is - CH2Q; Q is hydrogen; R1 and R4 are both hydrogen, R2 and R3 are both iodo; and R5, R6, R7 and R8, are all hydrogen. Still another preferred embodiment is that in which X is -O-; And it's methoxy; Z is - CH2Q; Q is methyl; R1 and R4 are both hydrogen, R2 and R3 are both iodo; and R5, R6, R7 and R8, are all hydrogen.
The following is a list of the compounds according to the present invention, which are particularly preferred.
BTO-964 BTO-986.
From the biological data provided herein, it is evident that a number of substituents can be added to the aromatic rings of the computer of the Formula I, without affecting the activity. Such substituents include, but are not limited to, the alkyl, halogen, nitro and amino groups, without any significant loss of biological activity. Also, although in the preferred embodiments an ether oxygen connects the two aromatic rings, it should be understood that this group may be absent or, alternatively, replaced with a variety of groups or atoms that do not confine the aromatic rings to the same plane, such as , for example, a methylene group, a carboxy group, or sulfur without significant loss of biological activity. In addition, the chemical compounds mentioned here can exhibit the phenomena of tautomerism or isomeris or conformation. As such, it should be understood that the invention encompasses any tautomeric or isomeric form of conformation, which exhibits biological or pharmacological activities similar to those of the compounds described herein. The compounds of the present invention can be synthesized in a variety of ways, using conventional techniques of synthetic chemistry. Typically, the compounds of the present invention are prepared according to the reaction scheme set forth in Figure 1, wherein A, Z, Y, X, RJ Rc R, R, R * and R 10 have the above definitions. The use of appropriate organic solvents, condition of temperature and time to carry out the reactions, are within the level of those skilled in the art. Suitable processes are illustrated by the representative examples. The necessary starting materials can be obtained by standard procedures of organic chemistry. USE OF THE COMPOUNDS OF THE PRESENT INVENTION The compounds of the present invention can be used either in vivo or in vitro, to inhibit the growth of tumor cells. The compounds of the present invention are also useful because they bind to tubulin and exhibit anti-mitotic properties and, thus, can be used in vivo or in vitro to inhibit abnormal cell mitosis. Likewise, the compounds of this invention can be used to inhibit the vascularization of endothelial cells and to inhibit angiogenesis. In addition, the compounds of this invention can be used to reduce the level of TNF-α produced by a cell. The compounds of the present invention are also useful in conjunction with other cancer therapies, including radiation therapy, chemotherapy and, in particular, immunotherapy (which includes radioimmunotherapy). Within this embodiment, the compounds of the present invention are particularly useful as sensitizing agents. When administered before, simultaneously with, or after treatment with a cancer therapy, the compounds of the invention increase the sensitivity of cancer cells to therapy. This results not only in an increase in the effectiveness of the therapy, but can also reduce the dose required, thus reducing undesirable side effects. It has been found that the compounds of the present invention are safe, effective, non-toxic and easy to administer. As such, in one embodiment, the present invention provides a method for inhibiting the growth of a tumor cell, this method comprises contacting the tumor cell with a compound having the formula: or their pharmaceutically acceptable salts. In Formula I, R1 'R2, R3 and R4 are each, independently selected and are functional groups including, but not limited to: H, alkyl, S-alkyl, alkenyl, alkynyl, aryl, hydroxyl, alkoxy, halogen , N02 and NH2. R5, R6, R7 and R8 are each independently selected and are functional groups which include, but are not limited to: H, S-alkyl, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alkoxy and halogen. In Formula I, X, if present, is a group that includes, but is not limited to, the following: - CH2 C - A in the above formula, together with the carbons to which it is attached, forms a carbocyclic or heterocyclic ring of 3, 4, 5, or 6 members, optionally substituted. R9 and R10, in the above formula, are independently hydrogen, alkyl and halogen. And, in Formula I, it is a functional group that includes, but is not limited to H, alkyl and alkoxy. Z is a functional group that includes, but is not limited to, the following: Q, in the above formula, is a functional group that includes, but is not limited to, H, alkyl and S-alkyl. With respect to the compound of Formula I, it should be noted that Z, Q and Y are selected so that when and Q is methyl, then Y is different from methoxy and ethoxy. Also, it should be noted that the above discussions relating to X, Y, Z, R1, R2, R3, R4, R5, R6, R7 and R8, and their preferred embodiments are fully applicable to the compounds used in this method herein. invention and, thus, will not be repeated. According to the above method, tumor cells include, but are not limited to, tumor cells of the lung, colon, breast, ovaries, prostate and liver, as well as squamous cell carcinomas. In a presently preferred embodiment, tumor cells are present in a mammalian subject. Mammalian subjects include, but are not limited to, humans, laboratory animals, domestic pets and farm animals. In a further preferred embodiment, the above method further comprises the step of observing the reduction in the growth of the tumor cells. In another embodiment, the present invention provides a method for treating cancer, this method comprises administering to a mammalian subject having the cancer, a therapeutically effective amount of a compound having the general formula: or a pharmaceutically acceptable salt thereof. The above discussions with respect to A, X, Y, Z, R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 'their definitions and preferred embodiments are fully applicable to the compounds used in the method for treating cancer and, thus, will not be repeated. The compounds of the present invention are useful in treating a wide variety of cancers. Such cancers include, by way of example and not limitation, carcinomas, such as pharynx, colon, rectal, pancreatic, stomach, liver, lung, breast, skin, prostate, ovarian, uterine cervical and gallbladder; leukemia; lympholas; gliomas; retinoblastomas and sarcomas. Also, according to the above method, mammalian subjects include, but are not limited to, humans, laboratory animals, domestic pets and farm animals. Furthermore, it will be readily apparent to those skilled in the art that by using the compounds of Formula I, the growth of tumor cells can be inherited in other higher order organisms, including, but not limited to, plants, insects, fish. and similar. Suitable compounds for use in the methods of the present invention can be readily identified using in vitro and in vivo classification assays. Such assays can classify the ability of a particular compound to inhibit the growth of malignant tumor cells or to abolish the tumorigenicity of malignant cells in vitro or in vivo. For example, tumor cell lines can be exposed to varying concentrations of a compound of interest, and the viability of the cells can be measured at established time points, using a Blue® alamar assay (commercially available from BioSource, International of Camarillo, California). When Blue alamar dye is added to the culture medium, this dye is reduced by cellular mitochondrial enzymes, which supply a soluble product with substantially increased fluorescence. This fluorescence can be measured with a fluorometer, whereby the signal is directly proportional to the number of cells. Using this information, the values of CI5Q (concentration of the lethal compound at 50% of the cell culture, compared to a control culture) for the compounds of interest can be easily calculated. In general, compounds useful in the methods of the present invention will exhibit an IC 50 in the approximate range of 0.1 to 20 μM, as measured by the assay described in Example IIA. As will be appreciated by those skilled in the art, many varieties of cell cultures of malignant tumors and cell lines can be used to classify the 'activity, which include, but is not limited to, the MDA, MB231 (breast), MCF-7 (breast), MDA-MB 468 (breast), Siha (squamous cell carcinoma), A549 (lung, non-small cells), HL-60 (leukemia), Ovcar-3 (ovary), etc. Of course assays in addition to in vitro and / or in vivo, to classify the anti-tumor and / or anti-cancer activity, known or used by those skilled in the art, can also be used to identify effective compounds useful in the methods of the present invention.
In addition to the above, the compounds of the present invention can be used to treat diseases characterized by abnormal cellular mitosis. Such diseases include, but are not limited to, abnormal stimulation of endothelial cells (for example in atherosclerosis), solid tumors and tumor metastases, benign tumors, eg, hemanglomas, acoustic neuromas, neurofibromas, tracheomas and pyogenic granulomas, vascular malfunctions , scarred from abnormal wounds, inflammatory and immunological disorders. Bechet's disease, gout or gout arthritis, abnormal angiogenesis that accompanies, for example, rheumatoid arthritis, psoriasis, diabetic retinopathy and other angiogenic ocular diseases, such as retinopathy of the pre-maturity type (retrolental fibroplasty), macular degeneration, rejection of grafts of the cornea, neuroscular glaucoma and Oster Webber syndrome. As such, in another embodiment, the present invention provides a method for treating a disease characterized by abnormal cellular mitosis, this method comprising administering to a mammalian subject, having such a disease, a therapeutically effective amount of a compound having the general formula : or a pharmaceutically acceptable salt thereof. The above discussions regarding A, X, Y, Z, R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 'their definitions and preferred embodiments are fully applicable to the compounds used in this method and, Well, they will not be repeated. Compounds suitable for use in the above method of the present invention can be easily identified using the in vitro and in vivo classification assays. More particularly, a given compound can be __ easily classified for its anti-mitotic properties using, for example, the microtubule inhibition assay and / or the competitive tubulin binding assay, described in the examples. Other assays known and used by those skilled in the art can also be used to classify a given compound in its anti-mitotic properties, or its anti-angiogenic properties by inhibiting the growth of endothelial cells, such as HUUVEC (endothelial cells of the human umbilical vein) or the HMVEC (human microvascular endothelial cells) ih vitro or through the chicken chorioallantic membrane (CAM) assay, as discussed here. In another embodiment, the present invention provides methods of treating tumors in a mammal, by administering to the mammal an effective antineoplastic amount of ionizing or non-ionizing radiation, or an effective anti-neoplastic amount of a chemotherapeutic agent, in conjunction with an effective sensitizing amount of a compound of Formula I. The compounds of the invention increase the deleterious cellular effects caused by exposure to ionizing radiation or to chemotherapeutic or immunotherapeutic agents. Such effects include, but are not limited to, cellular DNA damage, such as DNA cord disruption, disruption in cellular function, such as disrupting DNA function, cell death and the like. Often, a synergistic effect is observed when a sensitizing compound is administered, in an effective amount, of Formula I in conjunction with radiation therapy, chemotherapy, immunotherapy, or other cancer treatments. As used herein, a "synergistic effect" is achieved when a greater antineoplastic effect results with a joint therapy than the use of any drug or therapy alone. An advantage of the therapy in conjunction with a synergistic effect is that of the lower doses of one or both of the drugs or therapies that can be used, so that the therapeutic index is increased and * the toxic side effects are reduced. Chemotherapeutic agents for which the compounds of Formula I are useful as sensitizers, include, but are not limited to, alkylating agents, entanglement agents and DNA intercalating agents, which interact covalently or non-covalently with the Cellular DNA, causing certain harmful cellular effects. For example, agents reactive with DNA include cisplatin, cyclophosphamide, diethylnitrosoamine ,. benzo (a) - pyrene, carboplatin, doxorubicin, mitomycin-C, and the like Techniques to determine an amount "Effective sensitizer of a compound of Formula I, are known to those skilled in the art. To determine the effective sensitizing amount or dose, a number of factors are considered, which include, but are not limited to, the following: the species of animal, its size, age and general health; the specific disease involved; the degree of or involvement or the severity of the disease; the patient's individual response; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the selected dose regimen; the use of the concomitant medication; and other relevant circumstances. When used as a sensitizing agent, the compounds of Formula I can be administered as single doses or as multiple doses and are ordinarily administered before and / or during exposure to ionizing or non-ionizing radiation or to chemotherapeutic agents. In general, when a compound of the present invention is administered in conjunction with radiation therapy, the compound of the present invention will be administered in single or multiple doses, prior to radiation therapy, followed by a program calculated to deliver the effect Maximum selective sensitizer during radiation therapy. When a compound of the present invention is administered in conjunction with an agent "chemotherapeutic, the compound of the present invention is generally administered in single or multiple doses, before and during chemotherapy, followed by a program calculated to deliver the maximum selective sensitizing effect during chemotherapy." In a particularly preferred embodiment, the compounds of Formula I are administered in combination with active immunotherapy (eg, tumor vaccine) Because the compounds of Formula I are not immunotoxic, the immune system is not significantly suppressed and, thus, active immunotherapy can be taken Advantageously, 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 (eg, a tumor vaccine). The present invention also provides methods for preventing the development of a disease, acterizada by the abnormal cellular mitosis. Thus, the compounds of the present invention are useful not only to treat a tumor that already exists, but also to prevent the development of tumors or the recurrence thereof. For example, one can administer a compound of Formula I to a mammal that is predisposed to cancer development, thereby reducing the likelihood that the cancer will eventually occur.
'Alternatively, one can administer a compound of the Formula I to a mammal that has previously had cancer, reducing the likelihood that the cancer will recur. For these applications, the compound is generally administered in multiple doses, followed by a program calculated to reduce or eliminate abnormal cell proliferation. Appropriate dose regimens may be determined by those skilled in the art, using methods routinely practiced, such as those described below.
In another embodiment, this invention relates to a method for treating mammalian diseases associated with unwanted angiogenesis and "controlled" river, the method comprising administering to a mammal an anti-angiogenic compound of Formula I, in a dose sufficient to inhibit Angiogenesis The particular dose of a compound of Formula I, required to inhibit angiogenesis and / or angiogenic diseases, according to this invention, will depend on the severity of the condition, the route of administration and the related factors that will be decided Generally, daily, accepted and effective doses will be in an amount sufficient to effectively inhibit angiogenesis and / or angiogenic diseases In yet another aspect, this invention relates to "A method for treating diseases associated with angiogenesis." The methods of treatment provided by this invention are 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, which is effective to inhibit angiogenesis and / or angiogenic diseases The term inhibition is defined including its generally accepted meanings, which comprise the prophylactic treatment of a human subject to incur angiogenesis and / or angiogenic diseases, and keep under review and / or treatment of angiogenesis and / or existing angiogenic diseases As such, the present invention includes medical and / or prophylactic therapeutic treatments, as appropriate The methods of the present invention can be used to treat a variety of Diseases associated with corneal neovascularization, which can be treated In accordance with the present invention include, but are not limited to, diabetic retinopathy, premature type retinopathy, corneal graft rejection, neovascular glaucoma and retrolental fibroplasia, epidemic keratoconjutivitis, vitamin A deficiency, excessive use of contact lenses. , atopic keratitis, upper limbic keratitis, pterygium keratitis (sic), pink acne, filectenulosis, syphilis, mycobacterial infections, • generation of lipids, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, traumas, rheumatoid arthritis, lupus systemic, polyarthritis, Wegeners sarcoidosis, scleritis, Johnson's disease of Steven, radial perifigoid keratotomy and rejection of corneal graft.
Diseases associated with neovascularization, which can be treated in accordance with the present invention include, but are not limited to, diabetic retinopathy, macular degeneration, sick cell anemia, sarcide syphilis, pseudoxanthoma elasticum, Pagets disease, occlusion of vain, occlusion of arteries, obstructive carotid disease, chronic uveitis / vitritis, mycobacterial infections, Lyme disease, systemic lupus, premature retinopathy, Eales disease, Bechets disease, infections that cause retinitis or choroiditis, ocular histoplasmosis suspected, Bests disease, myopia, optic cavities, Stargarts disease, pars planitis, chronic retinal detachment, hyperviscosity syndrome, toxoplasmosis, trauma and complications "post-laser." Other diseases include, but are not limited to, the diseases associated with redness. (neovascularization of the optic angle) and diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue, which include all forms of proliferative vitreoretinopathy, associated or not 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. Uncontrolled unwanted angiogenesis is a key element that these chronic inflammatory diseases have in common. Chronic inflammation depends on the continuous formation of capillary buds to maintain an influx of inflammatory cells. It is influenced and the presence of inflammatory cells produces granulomas and thus maintain the chronic inflammatory state. The inhibition of angiogenesis using the method compositions of the present invention prevents the formation of granulomas, thus relieving 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. The disease Crohn's disease occurs as a chronic transmural inflammatory disease, which most commonly affects the distal ileum and colon, but can also occur anywhere in the gastrointestinal tract, from the mouth to the anus and the perianal area. Patients with Crohn's disease usually have chronic diarrhea associated with abdominal pain, fever, anorexia, weight loss and abdominal swelling. Ulcerative colitis is also a non-specific, chronic inflammatory and ulcerative disease that arises in the colonic mucosa and is characterized by the presence of bloody diarrhea. ~~ r Crohn's disease and ulcerative colitis are characterized by chronic inflation and angiogenesis in various sites of the gastrointestinal tract. Crohn's disease is characterized by chronic granulomatous inflammation through the gastrointestinal tract, consisting of new capillary buds surrounded by a cylinder of inflammatory cells. The prevention of angiogenesis by the compositions and methods of the present invention, inhibits the formation of the shoots and prevents the formation of the granulomas. Inflammatory bowel diseases also exhibit extra intestinal manifestations, such as skin lesions. Such lesions are characterized by inflammation and angiogenesis and can occur at any site in addition to the gastrointestinal tract. The compositions and methods of the present invention can also be used to treat these lesions, preventing angiogenesis, thereby reducing the influx of inflammatory cells and the formation of injuries. Sarcoidosis is another chronic inflammatory disease, which is characterized as a multisystem granulomatous disorder. The granulomas of this disease can be formed anywhere in the body and thus the symptoms depend on the site of the granulomas and if the disease is active. The gyrimelomas are created by the angiogenic capillary bud that provides a constant supply of inflammatory cells. The compounds and methods of this invention can be used to treat sarcoidosis. The methods of the present invention can also be used to treat chronic inflammatory conditions associated with psoriasis. Psoriasis, a disease of the skin, is another recurrent chronic disease, characterized by papules and plaques of various sizes. The prevention of the formation of new blood vessels, necessary to maintain characteristic lesions, leads to the relief of 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 thought that the blood vessels in the synovial covering of the joints suffer from angiogenesis .. In addition to forming new vascular networks, the endothelial cell release factors and reactive oxygen species that lead to panniculus growth and cartilage destruction. The factors involved in angiogenesis can contribute to, and help maintain, the chronically inflamed 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 tumors or those that carry blood and acquired immunodeficiency syndrome. Compounds suitable for use in the above methods of the present invention can be readily identified using in vitro and in vivo classification assays. Such assays can classify the ability of a particular compound to inhibit angiogenesis or vascularization of endothelial cells in vitro and in vivo. For example, the chicken embryo chorioallantoic membrane (CAM) assay, which is described in more detail below, can be used to classify a given compound in its ability to inhibit vascularization. In the trial of the corialanthic membrane, the fertilized chicken embryos are removed from their cover on day 3 or 4, and a methylcellulose disk containing a compound of Formula I is implanted in the corialanthic membrane. The embryos are examined 48 hours later and, if a clear avascular zone appears around the methylcellulose disk, the diameter of that zone is measured. This assay can be used to assess the anti-angiogenic properties of the compounds of Formula I. Another classification assay useful for evaluating the efficacy of the compounds of Formula I is the angiogenesis assay of the corneal micro-bag. (CMA) The rat corneal micro-bag assay can be used to assess the ability of the compounds of Formula I to inhibit corneal angiogenesis (see "Quantitative Angiogenesis Assays: Progress and Problems", Nat. Med. , 3: 1203-1208 (1997) and "Inhibition of Tumor Angiogenesis Using a Soluble Receptor, Establishes a Role for Tie2 in Pathologic Vascular Growth," J. Clin.Investy., 100: 2072-2078 (1997).) In this test, the compound of Formula I is mixed with a polymer (e.g., a solution of Hydron: Interferon Sciences, New Brunswick, NJ) and is implanted in a small bag 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 according to the appearance of new vessels in the normally avascular cornea. If the compound of Formula I is effective, specifically as an anti-angiogenic agent, it will inhibit or block this response. In an experimental design, a group of five animals (which include a control group with only polymer implants) was tested in a range of drug doses, which can induce the retardation of tumor growth. Three doses were tested in the trial. The evaluation 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 assay determines whether a compound of Formula I is directly anti-angiogenic in an in vivo mammalian model of angiogenesis. In addition, the human microvascular endothelial cell assay (HMVEC) can be used to evaluate the efficacy of the compounds of Formula I. The HMVECs are seeded in a 96-well plate at a concentration of 5 x 10 3 cells / well in a volume of 100 μl / cavity of the Endothelial Growth Medium. The plates are then incubated at 37 ° C in 5% CO2 for 24 hours and then aliquots of the compound of Formula I are added to the preparation of the HMVEC and the plates are incubated at 37 ° C in 5% CO2 for 3 days. The relative number of cells is determined by adding 20 μl / ml of the Alamar Blue dye for 3-6 hours at 372C and measuring the color changes, which indicate the metabolic activity, using a Fluorescence Measurement System. In this assay, the intensity of the fluorophore signal is directly proportional to the number of cells.
The HMVEC assay can also be carried out using the microvascular endothelial cells of the human umbilical vein (HUMVEC). The assay is carried out similarly to the previous one, but the HUMVEC cells are used. In yet another embodiment of the present invention, a method for reducing the level of TNF-α is provided, produced by a cell. This TNF-a and its diverse modes of action are generally described by Abbas, et al, in Cellular and Moplecular Immunology ?, Abbas, et al, 2ß Ed. W. B. Saunders Company, 1994, pages 244-249, whose teachings are incorporated herein by reference. TNF-a plays an integral role in destroying tumors, mediating responses to tissue lesions and protecting hosts from infections by various microorganisms. However, its activity appears to be excessive in some disease states and inflammatory reactions, such as rheumatoid arthritis, cachexia and septic shock. Excessive TNF-α results in an exaggerated immune response, exemplified by an over stimulation of interleukin-6 and the secretion of granulocyte stimulus factor / macrophage colony (GM-GF), increased cytotoxicity of polymorphonuclear neutrophils and the prolonged expression of cell adhesion molecules, all of which can have detrimental effects.
Contact of the cells with the compounds of Formula I results in decreased levels of TNF-α. Without attempting to be bound by any theory, reduced levels of TNF-a can result from any of the possible mechanisms, including, but not limited to, the down-regulation of the expression of a gene encoding TNF-α, a reduction in TNFa mRNA stability or translation efficiency, decreased stability of TNF-α polypeptide, and reduced secretion of TNFα from a cell. The reduced levels of TNF-a can be measured in a cell, biological sample or 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 inflammatory diseases noted above (eg, chronic inflation, chronic diseases, inflammatory bowel disease, sarcoidosis, psoriasis, rheumatoid arthritis, and the like). Using the assay set forth in Example VIII, the compounds of Formula I can be easily classified in their ability to reduce TNF-α. TNF-a is noted for its pro-inflammatory actions, resulting in tissue damage, such as the induction of procoagulant activity in vascular endothelial cells, increased adherence of neutrophils and lymphocytes and stimulation for "release of the activation factor of platelets of macrophages, neutrofiles and vascular endothelial cells, as such, the target parts that are discussed in these cells and that are conjugated to liposomes or other drug delivery systems, which comprise the compounds of Formula I, are Preferred embodiments of this invention For example, in a preferred embodiment, monoclonal antibodies to TNF-α (Tracey et al, Nature 1987, 330, 662-664; Silva et al., J. Infect. Sis. 1990, 162, 421-427; and Williams et al, Proc. Nati Acad. Sci. 1992, 89, 9784-9788) are conjugated to the liposomes comprising the compounds of Formula I. Also, according to the above methods, mammalian subjects include, but are not limited to, human, laboratory animals, domestic pets. and farm animals.
D. PHARMACEUTICAL FORMULATIONS / ADMINISTRATION ROUTES The compounds of the present invention can be administered to mammals, for example a human patient, alone, in the form of a pharmaceutically acceptable salt or in the form of a pharmaceutical composition wherein the compound is mixed with suitable carriers or excipients, in a therapeutically effective amount, for example at effective doses to decrease or suppress the growth of malignant cells or which result in relief of symptoms associated with cancerous diseases. The compounds of this invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds of the present invention can be formulated into pharmaceutical compositions by combination with appropriate pharmaceutically acceptable carriers or diluents, and can be formulated into preparations, in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, pills, powders, dragee granules, gels, watery pastes, ointments, solutions, suppositories, injections, inhalants and aerosols. As such, the administration of the compounds can be achieved from several 'manners, which include oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intraqueal, etc. Also, the compound can be administered in a local rather than systemic manner, for example by injection of the compound directly into a solid tumor, often in a reservoir or in a sustained release formulation. In addition, the compounds can be administered in a target drug delivery system, for example, in a liposome coated with a tumor-specific antibody. Such liposomes will be objective and will be selectively taken by the tumor. The compounds of the present invention can be administered alone, in combination with each other, or can be used in combination with other known compounds (for example, other anti-cancer drugs or other drugs, such as AZT, anti-inflammatories, antibiotics, corticosteroids , vitamins, etc.) More particularly, the compounds of the present invention can be used in therapy in conjunction with other known chemotherapeutic or antineolplastic agents (eg, vinca alkaloids, antibiotics, anti-metabolites, platinum coordination complexes, etc. For example, the compounds of the present invention can be used in therapy in conjunction with an alkaloid vinca compound, such as vinblastine, vincristine, 'taxol, etc .; an antibiotic, such as adriamycin (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 skilled in the art will appreciate tthe compounds of the present invention can be used in therapy in conjunction with other known chemotherapeutic or antineoplastic compounds. In the pharmaceutical dosage forms, the compounds may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in an 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 here as a reference. Also, for a brief review of methods of drug delivery, see Langer, in Science 249: 1527-1533 (1990), which is incorporated herein by reference. The pharmaceutical compositions described herein can be manufactured in a manner known to those skilled in the art, ie by means of mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. The following methods and excipients are merely exemplary and are not in any way limiting. For injection, the compounds may be formulated in dissolution, suspension or emulsion preparations in an aqueous or non-aqueous solvent, such as vegetable oils or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycolide. 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 the Hanks solution, Ringer's solution, or a physiological saline regulator For transmucosal administration, appropriate penetrants of the barrier to be percused are used in the formulation Such penetrants are generally known in the art. oral administration, the compounds can be easily formulated by their combination with pharmaceutically acceptable carriers, which are well known in the art.
"Such carriers make it possible to formulate the compounds as in tablets, pills, dragees, capsules, emulsions, lipophilic and hydrophilic suspensions, liquids, gels, syrups, aqueous pastes, suspensions and the like, for oral ingestion by a patient who is leaving. 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 mixture into granules, after adding the appropriate auxiliaries, if desired, to obtain tablets or Dragee cores Suitable excipients are, in particular, fillers, such as sugars, which include lactose, sucrose or mannitol or sorbitol, cellulose preparations, such as, for example, corn starch, wheat starch, starch rice, potato starch, gelatin, tragacanth gum, methylcellulose, hydroxypropylmethyl cellulose, sodium camoxymethylcellulose and / or polyvinylpyrrolidone (PVP) If desired, disintegrating agents can be added, such as the interlaced polyvinyl pyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, 'carbopol gel, polyethylene glycol and / or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyes or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of the doses of the active compound. Pharmaceutical preparations that can be used orally include the appropriate stimulation capsules, made of gelatin, as well as sealed, soft capsules, made of gelatin and a plasticizer, such as glycerol or 61 sorbitol. Suitable pulse capsules contain the active ingredients in admixture with a filler, 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 paraffin or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in doses suitable for each administration. For buccal administration, the compositions may take the form of tablets or lozenges, formulated in a conventional manner. For administration by inhalation, the compounds used in accordance with the present invention are conveniently delivered in the form of an aerosol spray penetration from pressurized packets or a nebulizer, with the use of a suitable impeller, for example, dichlorodifluoromethane, trichlorofluoromethane. , dislorotetrafluoroethane, carbon dioxide or other suitable gas, or from dry powder inhalers, exempt from the impeller. In the use of a pressurized aerosol, the dose unit can be determined by supplying a valve to deliver a dosed amount. Capsules and cartridges 62, for example gelatin, for use in an inhaler or insufflator, can be formulated containing a mixture of powder 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 condom. The composition may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents, such as suspending, stabilizing and / or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water soluble forms. Additionally, suspensions of the active compounds can be prepared as suitable oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous suspensions for injection may contain substances which increase the viscosity of the suspension, such as Sodium carboxymethyl cellulose, sorbitol, or dextrene, Optionally, the suspension may also contain suitable stabilizers or agents that feed the solubility of the compounds, to allow the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, eg, sterile, pyrogen-free water, before use. The compounds can also be formulated in rectal compositions, such as suppositories or retention enemas, for example containing conventional suppository bases, such as cocoa butter, carbo-waxes, polyethylene glycols or other glyceride, all of which fuse to the body temperature, but they are solid at room temperature. In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example 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 ion exchange resins, or as derivatives 64 sparingly soluble, for example, as a sparingly soluble salt. Alternatively, other delivery systems for the hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well-known examples of vehicles or delivery carriers for hydrophobic drugs. Certain organic solvents, such as dimethylsulfoxide, can also be used, although usually at a higher toxicity cost. Additionally, the compounds can be delivered using a sustained release system, such as semipermeable 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 their chemical nature, release the compounds for a few weeks to more than 100 days. In addition, in the presently preferred embodiment, the compounds of Formula I can be administered in a target drug delivery system, for example in a liposome coated with a tumor-specific antibody. Such liposomes will be the target a and are selectively taken by the site of interest (e.g., the tumor cell). Liposomes and emulsions are examples 65 well-known vehicles or delivery carriers for hydrophobic drugs. In a presently preferred embodiment, of long circulation, ie demurely, the liposomes are used. Such liposomes are generally described in Woodle et al., U.S. Patent No. 5, 013, 556, the teachings of which are incorporated herein by reference. Generally, such liposomes or other drug delivery systems typically have an objective part, i.e., a ligation, conjugated there, that is specific to the target site of interest (eg, the tumor cell). For example, some property (biochemical, architectural or genetic) of the tumor, which is different from normal tissue, can be exploited to concentrate the compounds of Formula I, in, or at least close to, the target tumor. The vasculature of the tumor, which is composed primarily of endothelial cells, is inherently different from the normal differentiated vasculature. For example, the architecture of the tumor vasculature is different, that is, the vessels are known as leaking, and the flow of blood through them is intermittent at most, with periods of perfusion and periods of occlusion and with the subsequent hypoxia. This aberrant micro-environment can be caused by and, in turn, leads to, an additional differential gene expression in the tumor vasculature relative to the normal vasculature. 66 This abnormal architecture and function, at the molecular level, is characterized by differences in the surface markers in the micro-vessels * of the tumor relative to normal vessels and such differences can be exploited at the liposome target or other delivery system. drug to the site of interest. Monoclonal antibodies, directed against a tumor marker, TNF-α, TNFα receptor, endothelial cell vasculature, etc., is a strategy that can be employed. Other strategies that can be employed are the target of a vasculature of an abnormal tumor or marker. The target part, when coupled to a liposome or other drug delivery system, which contains a drug or a radioisotope, will act to concentrate the drug where necessary. Ligatures for vessel markers associated with the tumor can also be used. For example, a cell adhesion molecule that binds to the marker of the surface of the vascular element of the tumor can be used. Liposomes and other drug delivery systems can also be used, especially if their surface contains a ligature to direct the carrier preferably to the vasculature of the tumor. Liposomes offer the additional advantage of protecting the drug from most normal tissues, thus reducing the inherent toxicity of many compounds. When it is covered 67 with polyethylene glycol (PEG) (ie, the 'restrained' liposomes) to minimize admission by phagocytes and with a specific target part of the tumor vasculature, the liposomes offer longer plasma half-lives, less specific tissue, and increased efficacy on the non-target drug. Other target strategies include, but are not limited to, ADEPT (antibody-directed enzyme prodrug therapy), GDEPT (gene-directed EPT) and VDEPT (virus-directed EPT). In ADEPT, the target of an inactive prodrug to a tumor mass is effected by an antibody against a marker associated with the tumor. The enzyme milieu in or around the tumor transforms the prodrug into an active toxic agent that then acts on the tumor tissue. Similarly, the expression of gene "differential or viral target to the site of the tumor are used to activate a prodrug in its toxic, active form, in the GDEPT and VDEGT, respectively. Other strategies include differentially expressed target genes, enzyme or surface markers, which appear on, for example, the vasculature associated with the tumor or will control the growth of the tumor. Using the above methods, the compounds of Formula I can be targeted to the vasculature of the tumor to perform the control of tumor progression, or to other sites of 68 interest (for example, endothelial cells, TNFa, TNF-a receptor, etc.). The pharmaceutical compositions may also comprise suitable carriers or excipients, solid or in gel phase. Examples of such carriers or excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Pharmaceutical compositions suitable for use in the present invention include compositions in which the active ingredients are contained in a therapeutically effective amount. The amount of the composition will be administered, of course, depending on the subject to be treated, on the weight of the subject, the severity of the affliction, the manner of administration and the judgment of the prescribing physicians. The determination of an effective amount is well within the ability of those skilled in the art, especially in light of the detailed description provided herein. For any compound used in the method of the invention, a therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a range of concentration 69 which includes a Clsn as determined in the cell culture (i.e. the concentration of the test compound that is lethal at 50% of a cell culture culture) or the Clioor as determined in the cell culture (i.e. concentration of the compound that is 100% lethal to a cell culture). Such information can be used to more accurately determine the useful doses in humans. Initial doses can be estimated from the in vivo data. Initial doses can also be formulated by comparing the effectiveness of the compounds described herein in cell culture assays with the effectiveness of known cancer drugs, such as vincristine. In this method, an initial dose can be obtained by multiplying the ratio of the effective concentrations, obtained in the cell culture assay for a compound of the present invention and a known anti-cancer drug, by the effective dose of a known anti-cancer drug. For example, if a component of the present invention is twice as effective in a cell culture assay, such as vinicristin (ie, the CI5n of that compound is equal to half of the IC50 of vinicristin in the same assay) , an initial effective dose of the compound of the present invention would be half the known dose for vinicristin. Using these initial guidelines, a An ordinary expert in the field can determine an effective dose in humans. Also, the toxicity? * The therapeutic efficacy of the compounds described herein, can be determined by standard pharmaceutical procedures in cell cultures or in experimental animals, for example, by determining the DL5n (the dose lethal to 150% of the population) and DE5n (the therapeutically effective dose 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 D 50 and ED 50. Compounds that exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used to formulate a dose range that is non-toxic for use in a human. The dose of such compounds is preferably within a range of circulating concentrations, which include the ED5o with little or no toxicity. The dose may vary within this range, depending on the dosage form employed and the route of administration used. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, for example Fingi et al., 1975, in: The Pharmacological Basis of Therapeutics, Chapter 1, page 1) .
The amount of dose and the range can be individually adjusted to provide plasma levels of the active compound that are sufficient to maintain the therapeutic effect. Usual doses of the patient for oral administration range from about 50 to 2000 mg / kg / day and more preferably from about 250 to 500 mg / kg / day. Preferably, effective serum levels will be achieved therapeutically by the administration of multiple doses each day. In cases of local administration or selective admission, the effective local concentration of the drug may not be related to plasma concentration. A person skilled in the art will be able to optimize effective local doses therapeutically, without undue experimentation. 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 skilled in the art will readily recognize a variety of non-critical parameters that can be changed or modified to provide essentially the same results.
EXAMPLES I. COMPOUNDS A, Synthesis of methyl 3-5-diiodo-4- (4-methoxyphenoxy) enzoate: BTO-956 Synthesis of compound BTO-956 was achieved in a series of steps, first, to supply methyl 3,5-dinitro-4- (4-methoxyphenoxy) benzoate, whose nitro groups are then reduced to amines and then replaced by iodine. The method for preparing the compound BTO-956 is essentially described by: Masuda, K. Imashiro, Y., and Okada, Y. Synthesis of triiodothyrophoric acid and its derivatives. J. Takeda Res. , Lab. , 1970, 29, 545-552. The synthesis of the ethyl analog, the compound BTO-967, is an adaptation of this same procedure. 1. 3, 5-dinitro-4- (4-methoxyphenoxy) methyl benzoate M.W. 134.14 M.W. 260.59 M.W. 348J7 M.W. = * Molecular Weight 3,5-dinitro-4-chlorobenzoate methyl 20.2 g 77.5 mmoles 4-methoxyphenol 10.0 g 80.6 mmoles Potassium hydroxide "" * 4.7 g 82.0 mmoles 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), 4-methoxyphenol (10.0 g, 80.6 mmol) and 4-chloro were successively added. -3,5-dinitrobenzoate methyl (20.22 g, 77.5 mmol). The flask was equipped with a reflux condenser and the reaction mixture was heated to 1500 C (oil bath) for 3 hours. After cooling to room temperature, this reaction mixture was transferred to a large mortar and triturated with cold 2N NaOH (100 ml) to remove the phenol without rationing. The solid is It was collected by filtration and dried in the air to give 21.5 g of the crude product The crystallization of absolute ethanol gave 17. 7 g (65.6%) of the pure 3,5-dinitro-4- (4-methoxyphenoxy) benzoate, as light yellow needles. 300 MHz XH NMR (CDC13) d 3.77 (s, 3H, OCH3), 4.02 (s, 3H, 0CH3), 6.82 (m, 4H, ArH), 8.70 (S, 2H, ArH).
Methyl 3,5-diamino-4- (4-metioxyphenoxy) enzoate 3, 5-dinitro-4- (4-methoxyphenoxy) methyl benzoate 20.2 g 77.5 mmoles 10% palladium on carbon 0.7 g catalyst Glacial acetic acid 80 ml solvent To a bottle of Parr stirring, containing a suspension of 3, 5 -dinitro-methyl 4- (4-methoxyphenoxy) -benzoate (20.2 g, 77.5 mmol) in glacial acetic acid (80 ml), 10% palladium on carbon (0.7 g) was added. The bottle was shaken under a hydrogen atmosphere (3 atm) until hydrogen was no longer consumed. The catalyst was removed by filtration 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 adding water (100 ml) in portions. Upon cooling, medium brown needles were 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 lH NMR (CDC13) d 3.73 (s, 3H, OCH3), 3.80 (bs, 4H, ArNH2), 3.86 (S, 3H, OCH3), 6.84 (m, 4H, ArH), 6.91 (s, 2H, ArH ). 3. Methyl 3,5-diiodo-4- (4-methoxyphenoxy) enzoate (BTO-956) M.W. 28 &27 M.W. 510.06 3, 5-diamino-4- (4-methoxyphenoxy) methyl benzoate 4.3 g 14.2 mmoles sodium nitrite 2.6 g 37.4 mmoles Glacial acetic acid 80 ml solvent Sulfuric acid 26 ml solvent Potassium iodide 20 g 120. Ominóles 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 an aqueous paste of methyl 3,5-diamino-4- (4-methoxyphenoxy) benzoate (4.30 g, 14.22 mmol) in glacial acetic acid (80 ml), in droplets, for a period of 30 minutes, keeping the temperature below 102C with the ice bath. The reddish-brown solution was stirred below 10 ° C for 45 minutes, after which it was slowly drained into an aqueous solution (30 ml) of potassium iodide (20 g) at room temperature, with vigorous stirring. formed an aqueous suspension and allowed to stir at room temperature for 1 hour.The reaction mixture was then heated in an oil bath at 802C (internal temperature) for 15 minutes and then allowed to cool to room temperature. filtered and the black gummy residue was 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 sintered glass funnel, 150 ml, to remove some colored impurities. The alumina pad was washed with 100 ml of acetone and the red filtrate was evaporated to dryness to give a dark solid as the crude product. This was purified by evaporative chromatography on silica gel, eluting with hexanes-CH2CH (60:40). The initial fractions containing the pure product were combined and evaporated to give 1.67 g of the desired product as an almost white solid. This compound gave a simple, clean zone in TLC chromatography (Hexanes: Ch2Cl2; 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 give another 3.0 g of the product as a creamy solid, "31 total yield of the product was 27%, 300 MHz * H NMR (CDC13) d 3.78 (s, 3H, OCH3), 3.94 (s, 3H, COOH3), 6.70 and 6.83 (two d, AA'XX ', 4H, p-subs.Ar-H), 8.51 (s, 2H, Ar-H).
B. Synthesis of methyl 3,5-diiodo-4- (-ethoxyphenoxy) benzoate; BTO-957 The synthesis of BTO-957 was achieved essentially by the same method used to produce the BTO-956, described above (structures not shown), with the most notable exception being the use of 4-ethoxyphenol as a starting reagent in place of 4-methoxyphenol. The synthesis in stages first gave methyl 3, 5-dinitro-4 (4-ethoxyphenoxy) benzoate, whose nitro groups were then reduced to amines and then replaced by iodine. 1. Methyl 3,5-dinitro-4- (4-ethoxyphenoxy) benzoate A mixture of 13 g (50 mmoles) of methyl 4-chloro-3,5-dinitrobenzoate, 7.45 g (54 mmoles) of 4-ethoxyphenol and 13 ml of a solution of potassium hydroxide (3.03 g, 54 mmol) was heated under argon at 150 ° C for 4 hours. The mixture was cooled and partitioned between 130 ml of a 2N solution of sodium hydroxide and 400 ml of dichloromethane. The organic layer was washed with 2 x 100 ml of a saturated sodium bicarbonate solution, dried over magnesium sulfate and evaporated under high vacuum to give 14.5 g, which were subjected to chromatography on a column of silica gel ( 5 cm x 50 cm), eluting with 25% hexane / 75% dichloromethane to give 13.2 g (74% yield). TLC (silica gel CH2C12), Rf 0.80. NMR: 300 MHz * H NMR (CDC13) d 1.39 (3H, t, -CH3); 3.97 (2H, q, -CH2-); 4.02 (3H, s, -OCH3); 6.81 (4H, s, aromatic); and 8.70 ppm (2H, s, aromatic). 2. Methyl 3,5-diamino-4- (4-ethoxyphenoxy) benzoate A solution of 214 g (66.2 mmoles) of methyl 3, 5-4- (4-ethoxyphenoxy) benzoate in 200 ml of glacial acetic acid was hydrogenated with 2.0 g of 10% palladium on carbon, at 50 mm Hg, about 30 minutes. The catalyst was removed by filtration through Celite. The mass of the filter was washed with 20 ml of acetic acid and the combined organic solution was poured into 2 liters of water. A white precipitate formed immediately, which was filtered and dried overnight under high vacuum, to give 16.31 g (82%). TLC (silica gel CH2C12), Rf 0.2-0.4 (belt). NMR: 300 MHz iH NMR (CDCl 3) d 1.38 (3H, t, -CH 3); 3.87 (3H, s-CH3); 3.98 (2H, q, -CH2-); 6.82 (4H, m, aromatic) and 6.93 ppm (2H, s, aromatic). 3. Methyl 3, 5-diiodo-4- (4-ethoxyphenoxy) enzoate (BTO-957) To 60 ml of concentrated sulfuric acid in a 250 ml three-necked flask, equipped with an overhead agitator motor and cooled to oac in an ice bath, 6.20 g (90 mmol) of sodium nitrite was added in portions over 20 minutes, keeping the temperature of the reaction mixture below ioac. To this solution was added a solution of 10.8 g (35.9 mmoles) of methyl 3,5-diamino-4- (4-ethoxyphenoxy) benzoate in 50 ml of glacial acetic acid, in drops, over 45 minutes, while maintaining the reaction temperature below 10 c. The reaction was stirred at oac for an additional 30 minutes and was poured into 50 g (300 moles) of potassium iodide in 100 ml of water. This solution was allowed to stand for 1 hour. The mixture was heated to 8oac in 5 minutes on a steam bath and cooled to room temperature. To this solution was added 1 liter of water and a gummy precipitate was collected. The precipitate was dissolved in 400 ml of acetone and filtered through a 5 cm x 10 cm column of basic alumina. The eluate was evaporated to give a red gum, which was subjected to chromatography on a 5 cm x 50 cm silica gel column, eluting 50% hexane / 50% dichloromethane. The product was collected to give 8.5 g of a white solid. This solid was triturated with 80 ml of 95% ethanol to give 8.4 g of a white solid (36% yield). MS (El) m / e 524 (M +, 100%). TLC (50% hexane / 50% CH2C12), Rf 0.50. NMR: 300 MHz * H NMR (CDC13) d 1.39 XßK, t, -CH 3); 3.93 (3H, t, -OCH3); 3.99 (2H, q, -CH2-); 6.75 (4H, q, aromatic) and 8.51 ppm (2H, s, aromatic C. Synthesis of methyl 3,5-diiodo-4- (4 * -methoxyphenoxy) thiobenzoate (BT0-967) BTO-956 BTO- S7 To a solution of methyl 3,5-diiodo-4- (4'-methoxyphenoxy) benzoate (BTO-956) (5.10 g, 10.0 mmol) in dry xylene (10 ml), Lawensson's reagent was added,, [ 2,4-bis (methoxyphenyl) -l, 3-dithia-2,4-diphospheate-2,4-disulfide] (4.85 g, 12.0 mmol) and the reaction mixture was stirred at reflux for 24 hours. The cooled mixture was evaporated to an orange residue, applied to a pad of silica gel (20 g) and eluted with The fractions containing the product were pooled and concentrated under reduced pressure to give an orange oil. The material was further purified on a column of silica gel (26 cm x 2 cm) eluting the compound with CH2Cl2 / petroleum ether (1: 4). The product r began to crystallize spontaneously in the elution. The material was allowed to crystallize overnight and was then collected using suction filtration and air dried to give 1.36 g (26%) of methyl 3,5-diiodo-4- (4'-methoxyphenoxy) thiobenzoate (1) as a yellow crystalline solid. NMR: 300 MHz H NMR (CDC13) d 3.78 (S, 3 H), 4.29 (s, 3 H>, 6.72 (m 2 H), 6.83 (m, 2 H), 8.66 (s, 2 H).
D. Synthesis of the alcohol 3 5-diiodo-4- (4l-methoxyphenoxy) -benzyl (BTO-972) BTO-956 BTO-972 To a dry, 15 ml, 2-necked flask equipped with a serum stopper and a reflux condenser under argon, methyl 3,5-diiodo-4- (4'-methoxyphenoxy) benzoate (1.02 g) was added. 2.0 mmol) followed by 2M LiBH4 in THF (2.0 mL, 4.0 mmol). The mixture was stirred overnight at room temperature. EtOAc (10 ml) was added to consume the excess of LBH4 and the resulting mixture was stirred at room temperature under argon. After about 20 minutes, a moderate exothermic reaction was observed and the reaction mixture became cloudy. This reaction mixture was digested with saturated NH 4 Cl (20 mL), EtOAc (20 mL) was added and the resulting layers were partitioned and separated. The organic layer was washed with saturated NaCl (25 ml), then dried over Na 2 SO 4, filtered and evaporated to give a pale amber oil. The crude product was applied to a silica gel filter pad (10 g, 230-400 mesh) and s eluted are EtOAs / hexanes (3: 7). The frassions which are the product were semised and evaporated to give 592 mg (61%) of the product as a white solid. The crystallization of EtOAs / hexanes gave 347 mg of the 3,5-diiodo-4- (4'-methoxyphenoxy) benzyl alcohol (9) as a white crystalline solid.
NMR: 300 MHz lH NMR (CDC13) d 3.78 (s, 3H), 4.52 (s, 2H), 6.72 (m, 2H), 6.83 (m, 2H), 7.82 (s, 2H). TLC: Rf 0.47 (EtOAc / hexanes, 3: 7).
E. Synthesis of 3, 5-diiodo-4- (4'-methoxyphenoxy) enzaldehyde BTO-972 BTO-964 To a solution of 2M oxalyl chloride in CH 2 Cl 2 (0.57 mL, 1.1 mmol) was added dried CH 2 Cl 2 (2.5 mL) under argon. The solution was cooled to -55 ° C (dry ice / asetone) and DMSO (178 mL, 2.5 mmol) in CH2C1 (400 mL) was slowly added over 1 minute. After 3 minutes, a solution of 3, 5-diiodo-4- (4'-methoxyphenoxy) benzyl ester (9) (432 mg, 0.9 mmol) in CH 2 Cl 2 (800 mL) was added in one portion. After stirring the mixture for 15 minutes, triethylamine (733 ml, 5.2 mmol) was added and thereaction was stirred for a further 5 minutes at -55ac, before allowing it to warm to room temperature. The reaction was quenched with water (10 ml) and then diluted with EtOAc (10 ml). The organic layer was separated, washed sequentially with dilute HCl, saturated NaHC 3 3 and saturated NaCl, then sited over Na 2 S 4, filtered and evaporated to a golden solid. The crude product was applied to a silica gel filter pad (10 g, 230-400 mesh) and eluted with EtOAc / hexanes (3: 7). The fractions containing the product were combined and evaporated to give 360 mg (84%) of the product as a pale yellow solid. The crystallization-from EtOAc / hexanes gave 175 mg (41%) of 3,5-diiodo-4- (4-methoxyphenoxy) -benzaldehyde (8) as an almost white crystalline solid. NMR: 300 MHz XH NMR (CDC13) d 3.78 (s, 3H), 6.73 (m, 2H), 6.84 (m, 2H), 8.34 (s, 2H), 9.87 (s, 1H), TLC: Rf 0.67 ( EtOAc / hexanes, 3: 7).
F. Synthesis of 3,5-diiodop-4- (* -methoxyphenoxy) -benzyl-methyl ether (BTO-966) BTO-972 BTO-966 A solution of alsohol 3,5-diiodo-4- (4'-methoxyphenoxy) bensylism (9) (347 mg, 0.72 mmol) in dry THF, under argon, was cooled to 02c and treated with 60% sodium hydride. in mineral oil (28 mg, 0.7 mmol). The reaction was stirred at oac for 10 minutes, then allowed to warm to room temperature and stirred for a further 30 minutes. Methyl iodide (0.5 ml, 8.1 mmol) was added and the mixture was stirred at room temperature for 18 hours. The reaction was emptied into saturated NH4C1 and extracted with EtOAc (20 mL). The organic layer was washed with saturated NaCl, dried over Drierite, and evaporated to a golden oil, which solidified upon standing. The solid was dissolved in a minimum amount of CH2Cl2 (3 ml), was aplisted to a silica gel sol (30 g, 230-400 mesh) and eluted with EtOAs / hexanes (3:17). The slurries containing the product were combined and evaporated to give 231 mg of the 3,5-diiodo-4- (4'-methoxyphenoxy) benzyl-methyl ether (10), as a white solid. NMR: 300 MHz H NMR (CDC13) d 3.42 (s, 3H), 6. 70 (m, 3H), 6.81 (m 2H), 7.80 (s, 2H). TLC: Rf 0.69 (EtOAC / hexanes, 3: 7).
II. EFFICACY OF THE BTO-956 PRODUCTS. 964 AND 967 AGAINST TUMORS A. Classification of in vitro cytotoxicity against human tumor cell lines Cell lines of human tumors were continuously exposed to various concentrations of the test agents, ie the compounds BTO-956, BTO-964, BTO- 966 and BTO-967) and the viability of the cells was measured at the established time points (1, 3 and 7 days) using the Alamar Blue® dye test. When this dye was added to the culture medium, it was reduced by the mitochondrial enzymes, providing a soluble produst, they are substantially increased fluoressence. This fluorescens can be measured in a fluorimeter, so that the signal is directly proportional to the number of cells. Several tumor cell lines of human origin, representing a wide variety of Sanser phenotypes and genotypes, were exposed in vitro to the agents to evaluate the effector range of the drug. Lines of slasifised tumors include MDA MB 231 (breast), MCF-7 (breast), MDA MB 468 (breast), Siba (sarsinoma selular essamoso), A459 (non-small lung cells), HL-60 (leukemia) , Ovcar-3 (ovary). The test agents were prepared in serial dilutions of 10 mM twill solusions in DMSO providing a dose range of 60 veses are concentrations of 20 μM, 10 μM, 3 μM, 1 μM and 0.3 μM. In sada saso, the cells were maintained at 37 c under 5% CO2 in the air. All the lines were insulated in Dulbesso modified Eagle's medium (DMEM), supplemented with 10% fetal bovine serum, inactivated by heat, and the doses of BTO-956, 964, 966 or 967 were titrated. 5-fluorourasyl (5-FU) was included, and a positive test in the experiments are two of the lines. The experiments were performed on 96-well tissue culture plasters (Falcon) with an initial seed density of 1000 cells per well in aliquots of 250 μi. The next day, the medium was replaced with 100 μl of drug dilutions per triplised. After a 7-day exposure to the various agents, the Alamar Blue dye was diluted to 20% with DMEM and 100 μl was added to each well. The cells were incubated for 8 hours until obvious color changes were not sufficient quantities of reduced dye for quantification. The relative cell number was evaluated by fluorometry in a Millipore 2300 CytoFLuor fluoressender measurement system. The measurements were taken from the 96-sapid plasmas after the exsitation at 560 nm, the emission is at 590 nm. The IC50 values were salsulated vs the sontrol (untreated cells); the results are presented in Table 1. Table 1 - Cytotoxicity of Representative Compounds of 'Guide Against Cell Lines of Human Cancer * Cell Line Clso (μM) Clso (μM) Clso (μM) Clso (μM) Clso (μM) BOT -956 BOT-954 BOT-966 BOT-967 5FU A549 4. 0 > 20 > 30 5. 5 5. 5 OVCAR 3 < 0. 3 1. 8 6.9 0. 8 NDb MDA MB 231 10 2. 0 10 2. 0 10 MDA MB 468 0. 5 4. 0 7. 5 1. 5 ND MCF 7 5. 0 2. 4 7. 0 1. 0 NDb SiHa 0. 5 17 > 20 20 NDb a Cytotoxicity 1 determined 7 days after exposure b Not determined B. Vibrio metabolism of compounds BTO-956, 964, 966 and 967 in human leukemia cells In human and laboratory animals, enzyme systems in the liver they are able to metabolize a large number of chemical products to inastive forms, and some chemotherapeutic agents are inastive, unless they are metabolized into astive forms.To test any of these possibilities, the compounds, is desir BTO-956, BTO-964, BTO-966 and BTO-967, were incubated as a frassion of microsomal enzyme, referred to as S9, Human leukemia cells (HL60) were used to evaluate the metabolism of the drug. performed in the presensia and ausensia of S9, prepared from the liver of adult male rats, providing an intraperitoneal injection of Arashlor 1254 (500 mg / kg) .The S9 was supernatant of 9000 x G liver homogen 0.25 M of sasarose - 100 mM phosphate buffer (pH 7.4) (Molesular Toxisology, Ins.). Co-factors were 1 mM of NADP and 5 mM of sodium isositrate. On the day of the exposure, the cell cultures, whether or not they were metabolism, were incubated for 4 hours in RPMI, which is 5% FBS and dosed doses of BTO-956, 964, 966 or 967. Test were prepared in serial dilutions, from 10mM twill solusions in DMSO, providing a dose range of 100 μM, 50 μM, 25 μM, 10 μM and 5 μM consension and a final DMSO concentration of 1 μM. %. In each case, the cells were kept at 37 ° C under 5% CO2 in the air. The experiments were sonduced in tissue plots of 6 savory tissues (Falson) with an initial seed density of 2 x 106 cells per coat in 2 ml aliquots. The test compounds were immediately added to the medium in duplicate cultures. Treatment solvation was removed by a series of low velocity sentrifugations to pellet the cells, followed by removal of the supernatant and resuspension of cells in fresh RPMI, containing 10% FBS. The results shown in Figure 2 indicate that Compound BTO-956 is metabolized within the period of insubassion from 4 hours to an inastive form. These The results also show that the BTO-964 complex is more resistant to metabolic degradation and more toxic to the HL60 cells than the BTO-956 complex. In contrast, the compounds BTO-966 and BTO-967 appear to be activated at more sitotoxic products.
C. In vivo efficacy of Compounds BTO-964 and BTO-967 against mouse mammary tumors, Met-I Mammary fat cushions of immunodeficient Balb / s mice were implanted are 1 mm3 of breast tumor tissue. These mammary tumors originated from the sellar line of mammary tumors of the Met-I mouse and they were propagated by passing them in vivo. When palpable tumors appeared, the animals were subdivided into several treatment groups and treated daily with an intraperitoneal injection of the blood vehicle, the BST-967 (50 mg / kg) or BTO-964 (50 mg / kg) suspension. . A group of untreated sontrol animals were insubordinated in the study, due to the agents used in the vehicle for these test articles. (DMSO / ethane1 / Cremophor EL / PEG 400, 1: 0.5_ 0.5: 6) were sonded by themselves have a sitotóxisa astivity. Two times weekly, the volumes of the animal sada tumors in the group were measured to obtain information on the tumor (volume) sresimiento a function of the type of treatment, dose and time. Implanted mice are xenografts of pesho 'Met-I, showed a positive response to treatment are the somatic BTO-964 (Table 2). The primary tumors in the groups of treated animals are BTO-964 were significantly smaller (p = 0.06) than the animals treated with the control vehicle, after 4 weeks of therapy. The average volume of the tumor of the animals treated with the drug vs animals treated with the vehicle, in 4 weeks was 876 ± 126 mm3 vs 1238 ± 170 mm3, respec- tively; the difference between the treated animals is the compound BTO-964 and the untreated animals (without vehicle) was significantly higher (p = 0.01).
Table 2 - In Vivo Efficacy of a Representative Candidate Drug in the Murine Sinemic Maternal Tumor Model, Met-I Tentative Volume of Tumor Percent of Reduction (mm3) to (none) / re Vehicles) None 1570 ± 219b na Only vehicle 1238 ± 170 -21% / na BTO-964 876 ± 126 -44% / -29% * Volumes of tumors measured 4 weeks after the start of treatment b SEM = Standard error of the average III. ANTI-ANGIOGENIC PROPERTIES A. Chorioallantoic membrane test of chicken (CAM) The compound BTO-964 [3,5-diiodo-4 (4'-methoxyphenoxy) benzaldehyde methyl] and the compound BTO-967 [3, 5 diiodo-4 (4 * -methoxyphenoxy-thiobenzoate) were tested in the CAM assay The endpoint of the assay was the quantitative determination of basal membrane biosynthesis by measuring the incorporation of 1 C-proline into the Type IV collagenous protein .
The CAM assay involves the development of live chicken embryos in Petri dishes under special sterile conditions. Therefore, only limited numbers of embryos can be used for the evaluation of the compounds in a single experiment. The compounds BTO-964 and BTO-967 were tested in separate tests. A conosido angiogenesis inhibitor, 2-methoxyestradiol (2-ME) was used as the positive control, and the human fibroblast sineing fastor (hFGF) was used to induce angiogenesis in the CAM. Fertilized eggs were supplied by Melody Ransh, Aptos, CA L-U-14C + -proline (specific astivity, 290 mCi / mmol) was purchased from New England Nuslear, Boston, MA. Collagenase and 2-SE were obtained from Sigma Chemical Co., St. Louis, MO. The silicone ring cups were obtained by cutting a silicone pipe (3 mm diameter) in 'small rings den "O" 1 mm thick. These silisone ring soups can be reused mushas veses if they are sterilized before testing. Plastis Petri disks (20 x 100 mm) were taken from Baxter Diagnostiss, 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 blanks for sterilization. The asetone-methanol mixture was then evaporated to dryness in a sterile sampana. The suspensions were dissolved in dimethylsulfoxide (DMSO) first and then diluted are a saline solution containing the methylcellulose. The final sonsentrasions were 2% DMSO and 0.5% methylsulose. All test solutions were added to CAM sada in 20 ml aliquots. The method of Folkman, et al. (Folkman et al (1974) Dev. Biol. 41: 391-394), are some modifisasiones, was used to sultivar chicken embryos somo follows: Fertile eggs strawberry were incubated for three days in a standard egg incubator. On day 3, the eggs were split under sterile condi tions and the embryos were placed in 20 x plastic Petri dishes 100 mm and cultured at 37ac in an embryo incubator with a water tank in the bottom shelf. Air was continuously bubbled into the water tank, using a 'small pump, so the humidity in the incubator remained constant. Observations were made daily to ensure that all embryos were healthy. The dead or unhealthy embryos of the insubator were removed immediately to avoid sontaminess. On day 9, a sterile silicone ring cup was placed in each CAM and 0.5 mCi of 1 C-proline with or without the test compound plus 2.5 ng of hFGF dissolved in a saline solution containing 0.5% methylsulose was delivered to Sada cup ring in a sterile bell. The 2-ME was tested in parallel to serve as a referensia compound. After the addition of the test materials, the embryos were returned to the incubator and the culture was sonicated. On day 12, all embryos were transferred to a cold room at 4-10ac. The antiangiogenic efesto of each test compound was determined using the collagenase assay to measure the incorporation of 14-proline into the solagenous protein.
B. Collagenase Assay for Measuring the Incorporation of 1C-Proline into the Collagenous Protein Using the outlined profiling in Maragoudakis et al., (1989), J. Pharm. Exp. Ther. 251: 679-682, the embryos were placed on ice and a 10 mm diameter CAM piece was cut under each ring cup and placed in a separate tube. To each tube was added 1.0 ml of a regulated saline solution are phosphate (PBS, pH 7.3) which are 0.11 mg of sisloheximide and 0.17 mg of dipyridyl. The tubes were soldered in a boiling water bath for 10 minutes and then cooled to room temperature. The PBS in each tube was discarded after centrifugation at 3000 x G, for 10 minutes. The CAM residue was washed with 3 ml of 15% TCA and then three times 3 ml of 5% TCA. The sentrifugate was taken to taste as it was previously dissed, among washed washing. At this point, the radioactivity bound to non-proteins and the CAM that are the 14C-solagenous protein were removed, resin synthesized was suspended in 0.9 ml of 0.1N NaOH and 1.1 ml of HEPES regulator, at a pH of 7.4. The pH of the sample was neutralized 0.8N HCl, using phenol red somo indicator. To digest the 14C-collagenous protein, 7.5 units of collagenase and 500 nmoles of calsium chloride in 40 ml of the HEPES regulator were added to the previous samples and the mixtures were incubated at 37ac for 4 hours. The reaction was stopped by adding 1.0 ml of 20% TCA containing 5 mg of tannic acid in each tube. After vortex mixing, the samples were centrifuged at 3000 x G for 10 minutes. An aliquot of the slane supernatant was taken by the scintillation count to quantify the radiolabeled tripeptides corresponding to the collagen of the basement membrane and other solagenous materials synthesized by CAM of 14C-prolna. The CAM pellets in each tube were solubilized in 0.5 ml of 1.0N NaOH by boiling them 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 Pier e Chemical Co. The radioactivity per milligram of CAM protein treated with a test compound in relation to that of control, gave the percent of inhibition of angiogenesis.
Tables I and II summarize the results of two separate experiments. The results of these two experiments indicate that there is an "anti-angiogenic effect of the BTO-967 and BTO-964 complexes in the CAM assay.
Table I Biofuente compounds in the anqiogenesis induced by hFGF-B Table II Biofuente compounds in the anqiogenesis induced by hFGF-B? b Significantly less than the control, P < 0.01.
IV. TESTS USED TO CLASSIFY ANTIMITOLOGICAL PROPERTIES A. Microtubule Enzyme Inhibition Assays A cell-free assay to measure inhibition of the microtubule pool can be performed by first mixing tubulin and tubulin labeled rhodamine. a reversion of 4: 1 (Hyman, A. et al. (1990) Meth. Enzymol, 196m 478-485; Belmont, LD et al (1996) Cell 84, 623-631). This tubulin solution was then added on ice to a regulator (BRB 80, 80 mM potassium salt of PIPES (pH 7.5), 5 mM MgCl 2, 1 mM EGTA) containing 1 mM of GTP and 1 mM. from PTT up to a final consension of 15 μM Drugs are different sonsentrations (0.5 μl) were added to 50 μl samples of the regulated tubulin, and 10 μl of each solution was transferred to microfuge tubes Each tube resibid 0.4 μl Myrotrobule seeding (Belmont, LD supra) .The tubes were incubated at 37ac for 10 minutes, before adding 100 μl of RBR 80 which is 1% glutaraldehyde.Each reaction mixture (2.5 μl) was transferred to a slide of Microscopy for fluorescence microscopy, Microtubules in intact cells were visualized using a mouse anti-a-tubulin monoclonal antibody and a polyclonal anti-mouse mouse antibody, labeled with fluorescein. In short, the HeLa cells were placed in plates in cursors of the chamber of 2 cavities (Nunc, Napierville, IL) at 1.5 x 104 / ml and insubjected in 5% of CO2 at 37ac for 24 hours, before treatment are the drugs for 1 hour. After removing the medium, the sealed myrotubules were stabilized using the BRB-80 which is 4 mM EGTA and 0.5% Triton X 100. The cells were fixed for 3 minutes in methanol cooled to -20ac, the TBS regulator was washed (0.15 M NaCl, 0.02 M Tris-HCl, pH 7.4) and permeabilized are TBS / 0.5% Triton X-100. After several washes are TBS / 0.1% Triton X-100, the cells were blocked with a anti-body dilution regulator (TBS, 0.1% Triton X ^ -lOO, 2% BSA, 0.1% sodium azide) for 10 minutes. The cells were stained for obscurity with a primary antibody for 1.5 hours and then the secondary antibody was added to the antibody dilution buffer, which contains 1 μg / ml Hoechst 33342 and incubated in the dark for 45 minutes. The slides were mounted with n-propyl gallate (2% w / v in 30% 0.1M Tris / glycerol, pH 9.0) and sealed under glass cover caps. B. Competitive Tubulin Binding Assays Competitive binding of a compound to the tubulin colchysin binding site was performed using a rotary-tube method (Woods, JA et al (1995) British J. Cancer 71, 705- 711). The BTO-956 solution labeled with 14C (20 μM, 10 mCi) was mixed with the tubulin and sodium chloride at different sonsentrasions and was incubated at room temperature for 1.5 hours in a regulator, which is 0.1 M MES (pH of 6.8) 1 mM EGTA, 1 mM EDTA and 1 mM MgCl2- Each reassuring mix was hardened in a stand that is 1 ml of balanced G50 Sephadex are a regulator that are 40 mM MES (pH 7.5), 40 mM Tris and 1 mM of MgSO4"The soloms were centrifuged at 900 x G for 3 minutes and the eluents were mixed as one is 3 ml of CytoScint (ICN) for analysis by the liquid scintillation count.
V. USE OF THE COMPOUNDS AS A RADIOTHERAPY A RADIOTHERAPY sensitizers A. In vitro radio-sensitization assay Radio-sensitization studies were carried out on crested cancer cells. The test formulation was added to the cells before, during, or after irradiation. The radiation doses are typically measured in units of Gy / min, are a Gy equal to 100 rad, while a rad is the amount of ionizing radiation that results in the absorption of 100 ergs of energy per gram of the irradiated material. The relations of increase of the sensitization (SER) were determined to a "Survival level of 10% The value of C? .6 (ie the consension of the test suite that supplies a SER of 1.6) was determined by projecting the values of the SER against the consension of the test set. S-3 were maintained and maintained in a modified medium of Eagles, which is 10% fetal bovine serum, in a CO2-controlled insubator, and humidity.Experimental studies were initiated in exponential cultures growing on tissue culture discs. 60 mm, at treatment densities of 3-7 x 10 < 5 > cells / disso The time of duplicating the HeLa S-3 sultives is approximately 18 hours A solution of the formula (I) was dissolved in distilled water, just before use and diluted 1: 100 by the addition of the appropriate volume to the sultivated cells.For ultraviolet irradiation studies, the compound was added only during the repair period following the irradiation. X irradiation studies, the drug was added to the sultivos 1 hour before the irradiation and remained in sontasto are the cells during irradiation.
For the ultraviolet irradiation (without ionization), all the medium was removed from the plants and. , the lid is removed, the disks were exposed to light of 1.4 J / M2 of UV254 nm, emitted from a germisid lamp of G.E. The half fresso, are or without the test set, then added to the sultivos during the subsequent repair period. In some cases, the cells were immediately suppressed in order to establish a T0 value for breaks in the DNA siren. The irradiation X (ionizing) of the cultures was carried out in a TFI Bigshot X-ray unit at 3 mA, 50 keV, filtered with 1.5 mm of Be and delivering 0.56 Gy / min to the cells (through the cap and 5 ml of the medium) as determined by the Victoeen ionization chamber, calibrated at a range of 10 to 50 keV. Following irradiation X, the sultives immediately resounded for trials of the saponity of forming solonias. The DQ values were derived from the survas surplus computer, projected by the linear regression analysis. The ability of cells to form solonias, after irradiation, was determined by standard methods. The cultures treated with the test compound were irradiated and immediately treated with trypsin, counted and re-collated into plasmas in 5 ml of medium which are the appropriate number of cells (500 for the untreated trees and the trees exposed to 2.8). Gy x-rays, 2,000 for crops exposed to 20 J / M2 UV, 5,000 for crops exposed to 5.6 Gy X-rays and 10,000 for crops exposed to 8.4 Gy X-rays). The crops grew "for 10 days, in their time the solonias of 1.0-2.0 mm (50-200 cells) were evaluated by the fixation of methanol and the dyeing." The untreated HeLa cells exhibited sloniation efisiensias in the range of 34 to 46% , using this protosolo.
B. In vivo radio-sensitization assay This example describes a method by which one skilled in the art can test the effect of the compounds of the invention in the radiotherapy of malignant tumors. The model system used in this study is well established for the determination of radiation effects in tumor tissue. See, for example, Twentyman et al (1980), "Nat 'l nancer Inst. 64: 595-603.; Brown et al (1980) J. Nat 'l Cancer Inst. 64: 603-611; Bernstein et al (1982) Radiation Res. 91: 624-637. The model uses RIF-1 tumor cells, which are well suited for studies of the radiation response, which include in vitro cell survival and tumor studies in vivo, partly because of their rapid growth regimen, are a double time of 65 hours and a cell time of 12 hours. The RIF-1 tumor is minimally immunogenic and metastasizes only at a late stage of sresimiento. Tumors are produced by subcutaneous inosulation in the lower backs of mice. This inosulation The suspension was of 2 x 103 cells of RIF-1 from the sultivo in 0.25 ml of the minimum alpha-skeletal medium (MEM, Gibso) supplemented are 10% fetal bovine serum (Johns Ssentifis). Masho C3H / He mice (Harían Sprague Dawley Ins., Indianapolis, Ind.) That are 5 to 7 weeks old at the time of inosulation are added for this experiment. The animals were anesthetized for inosulation. The tumors were then allowed to grow to 1 cm in average diameter. The measurements were made using a caliper, taking the length and width of the tumor and calculating the average of these two. The diameter measurements of the tumor were taken every 2 to 3 days from the moment of implanting the tumor cells. The tumors were left to be approximately 1 cm in average diameter, without any intervention. When the tumors reached this average diameter, the subject animals were randomly collated into one of three groups. One group did not receive radiation and no test suite, a second group received radiation but no test suite, and one terser group received both radiation and the test compound. For the treatment of radiation, an animal anesthetic was respected. The animals were immobilized and solosaron in a radiotherapy apparatus by the same ' time frame. Radiation exposure was from a single radiation dose 3000 Gy of 150 KeVX (average time of 10 minutes and 40 seconds). Preferably, the radiotherapy equipment (Protea ionization chamber) was salted before and after the session to ensure an absolute uniform dose. The radiotherapy was administered with a cone on the tumor and the lower back of the animal, which, in any case, ensures a maximum uniform delivery dose to the tumor, while minimizing the delivery of doses to the sensitive structures of the abdomen and pelvis. higher. The animals randomly collated in the somatic treatment group were given an intravenous dose of a Formula I compound. The day on which the subject animal was treated was designated on Day Zero. At cool intervals, usually every day, the tumors were measured in the same way as previously dissorted, and an average of two diameters of salsula. These data were projected as a function of time. The endpoint of the study occurred when the tumor reached twice the diameter of the original treatment, or approximately 2 sm. The animals were sedated in a CO2 chamber and the tumors were surgically removed, post-mortem. Representative tumors are monitored, embedded in paraffin and the slides were stained using the H & E stain and examined to confirm the histological presensia of fibrosarcoma RIP-1. The animals were sasrified and the tumors were collected before they reached twice the original diameter in the following sites: premature death of the animal following the treatment; ulserasion of the tumor; or infection or inflammation of the injection site.
SAW. USE OF COMPOUNDS AS SENSITIZERS FOR CHEMOTHERAPY The assays to determine the appropriate doses of the compounds of the invention, for use as sensitizers for chemotherapy and immunotherapy, are similar to those described for radiotherapy. To examine the "previous insubassion effect" of the inversions, fatigued cells were exposed to a fixed concentration of a test sample for 2 hours, followed by exposure to variable sonsentrasions of a chemotherapeutic or immunotherapeutic agent for 1 hour, at 37ac, and then they were tested for solonia formations. In evaluating the efesto of the "previous insufflation time" in the chemosensitization, the sanser cells were exposed to fixed sonsentrasions of the 'test for 0 to 4 hours at 37ac, followed by exposure to a chemotherapeutic agent under aerobic sonication, for 1 hour at 37ac, and then they were tested in the formation of colonies. Dose-dependent potensilization was also examined by exposing cancer cells to several consents of the test portion for 2 hours at 37 ° C and then at a fixed dose of chemotherapeutic agent for 1 hour at 37 ° C under aerobic conditions. use a simultaneous addition of the sensitizing and chemotherapeutic agent for 1 hour at 37 ° C., under aerobic sounds, can also be performed.
VII. CHEMICAL PREVENTION The efisasia of the resorbed somo-positives, chemical preventative agents, can be demonstrated using the in vitro and in vivo models of carcinogenesis induced by 5AzadC. A suitable model system uses premalignant murine fibroblasts (cell lines 4C8 and PR4) that express a proto-oncogene s-Ha-ras, transcriptionally astivated. These cells are not free, the suals are highly susceptible to malignant conversion by pharmacological doses of 4AzadC, they are subslones of the mouse NIH 3T3 fibroblasts, PR4N and 4C8-A10 (designated here PR4 and 4C8) and have been previously dessrito (see , by 'example, Wilson et al (1986) Anal. Biochem. , 152: 275-284; Dugaiszk (1983) Biochem, 22: 1605-1613). Both cell lines are phenotypic inversions isolated from cells 373 transformed with LTR / c-Ha-rasl-, after treatment for long period are interferon alpha / beta of the murine. The sultives were maintained in an Eagle medium modified by Dulbesso (DMEM) supplemented with 10% fetal calf serum, inastivated by salor (Gibso) and antibiotics. The sodium salts of the phenylacetic and phenylbutyric acids (Elan Pharmaseutisal Corporation) are dissolved in distilled water. The 5AzadC (Sigma St. Louis, Mo.) was dissolved in a regulated saline solution is phosphate (PBS) and was alsuotated in aliquots at -20ac until use. The exposure of 5AzadC to direct light is avoided at all times to prevent hydrolysis of the drug. For in vitro tests, 1-2 x 105 cells were plated on 100 mm discs and the test compound was added to the crescent medium 20 and 48 hours later. The cells were subsugously subsumed and observed in the phenotypic alterations. While the 4C8 and PR4 form inhibited monolayers of the contaste, which are epithelial-type cells, the transient exposure of these cultures to 0.1 μM of 5 AZadC during the logarithmic phase of growth, results in the neoplastic, rapid and massive transformation. Within a week of Treatment with AzadC, the vast majority of the sellar population became refractory and spherical, and forms multiple layers with increased saturation densities, which is indicative of the loss of crescent stimulation inhibition. The treatment of the cells are the test substance reduces or prevents these phenotypic sambios. The test substance can be administered before the treatment of the cells are the 5AzadC, simultaneously with the treatment of the 5AzadC or after the treatment with the 5AzadC.
For in vivo tests of the ability of the test suites to prevent the malignant state, untreated female 6'.9-week-old untreated mice were injected unsupervised (s.s.) are 0.5 x 10 cells. Twenty-four hours later, 400 μg of the SAzadC, prepared in 200 μl of PBS, was administered intraperitoneally (i.p.) in animal body (approximately 20 mg / kg). The test suite is also administered to the animal. The number, size and weight of the tumors was recorded after 3-4 weeks. For the histological examination, the tumors were cut, fixed in the Bouin's solution (very low písriso: 37% formaldehyde: rough glasial asystid, 15: 5: 1 vol / vol) and stained are H & E. A simple injection of the mouse i.p are 5AzadC (20 mg / kg) resulted typi- cally in the development of the tumor at the site of inosu- sion of the 4C8 cells in the sontroles. However, animals protected by an invertebrate host did not develop tumors or form slow-growing lesions at the site of 4C8 inoculation. An additional test of the ability of the invention bundles to prevent malignant stenosis involves the inhibition of the sealing cussion on matrigel, which is a base-membrane resonance (Collaborative Research). This test models the ability of cells to degrade and cross tissue barriers.
The cells were exposed for 48 hours, in tissue plastis disks of tissue cultures are 5AzadC alone or in symbiosis with the test compound. The treatment with the test compound was continued for 1-2 additional weeks, after which the cells were returned to solosa in plasmas on disks of 16 mm that had been previously re-coated are 250 μl of matrigel (10 mg / ml). The test set is added to the disks or omitted in order to determine if the test is reversible. In the absence of the test compound, the network-like formation, invasive cell features, occurred within 12 hours, and invasion in the matrigel is evident after 6 to 9 days.
VIII. ASSAY TO MEASURE THE CAPACITY OF THE COMPOUNDS OF THE FORMULA i IN REDUCING THE LEVELS OF THE NECROSIS FACTOR TUMOR (TNF-a) This example provides an assay that can be used to classify the compounds of Formula I in their ability to reduce the expression of cytosines (for example TNF-α) in a mammal.
A. Materials and Methods 1. Cell Line - ** • The PU5-1.8 line of the murine masrophage was purchased from the American Type Culture Collection (ATCC, Rosckville, MD). The cells in the supplemented DMEM medium were 100 mM sodium pyruvate, 0.1 mM non-essential amino acids, 2 mM glutamine and 5% fetal bovine serum (Life Technologies, Staten Island, NY). in a humid atmosphere of 5% CO 2 and 95% air at 37 ° C, the cells were passed twice a week, draining firmly the suction of the flask to dislodge the adherent cells, both non adherent and adherent cells were passed. grown exponentially were seeded at 5 x 105 / ml, 4 ml per 60 mm disso, 24 hours before the experiment.The test suites were delivered in 1 ml volumes of medium added to each disk, at the start of the experiment. the disks were incubated at 37 c in 5% C02 and 95% air for 3 hours 2. Reagents The sDNA of the Tumor Nesrosis Fastor (TNF-a) obtained from ATCC (Roskville, MD) [a-32P] -dCTP (250 μCi) and nylon membranes (hybond N) were obtained from Amersham (Arlington Heighs, IL). Solquisina (used as a sontrol) was sourced from Sigma Chemisal Company (St. Louis, MO.
The lipopolysaccharide (LPS) of Escherichia coli was sourced from DIFCO Laboratories (Detroit, MI). All plastid supplies were purchased from VWR Scientific Products (San Francisco, CA). 3. Northern blot Total RNA was isolated by the method of guanidinium chloride-cesium, as dessribe in N. S. Waleh, J. Gallo, T.D. Grant, B.J. Murphy, R.H. Kramer and R.M. Southerland (1994), "Disminating Selective Regulation of Integrin Resectors in Squamous Cell Carcinoma Steroids," C ncer Res. , 54: 838-843. Five to 10 μg of total RNA were subjected to electrophoresis in 1% agarose gels, containing 6% formaldehyde. Following the electrophoresis, the gels were stained with ethidium bromide to visualize the positions of 28S and 18S in the RNA. The - RNA were then transferred to nylon membranes (Ambersham Hybond N) by capillary staining and fixed to the filter by exposure to UV light. The spots were tested with sequelae of the 32P-labeled sDNA of human TNFa, obtained by Amerisan Type Culture Collestion (ATCC). TNF-α sDNA is a 1.1 kb PstI fragment of plasmid pE4 in E. coli MM294 (ATCC 39894). Hybridization was carried out at 42ac in 50% formamide, 5x SSC, 5% Denhardt solder, 0.1% SDS and 0.3 mg / ml salmon sperm DNA. The filters were washed by 1 x SSC, 0.1% SDS, two times at room temperature for 15 minutes and once at 55ac in 0.1 x SSC, 0.1% SDS for 1 hour. The filters were exposed to a x-ray film at a temperature of -70 ° C, using an intensifying screen (Coronex Hi-Plus). The hybridized bands were quantified by analyzing the images obtained using a video densitometer (Applied Imaging Corporation, Santa Clara, CA). The densities of the film were salified using a density of optimal density. The treatment of murine PU5- 1.8 masrophages are LPS (100 ng / ml) for 3 hours resulting in a significant increase (>; 7 times) at the level of TNF-a mRNA, as determined by Northern blot analysis. The treatment of cells with colchicine at 10 μM concentration had no effect on the expression of TNF-a mRNA. However, the addition of the 10 μM colchysin to the treated sultives is LPS resulting in a substantial redussion of the removal of TNF-a mRNA. To establish a relationship of consension effect, the masrophages were exposed to several concentrations of the formula I formulations, in the presence of the LPS stimulus, for 3 hours. The sanctities of TNF-a mRNA are delineated from the formulations of Formula I. The formulations of Formula I that were tested were statistically as effective as the solquisin in the down-regulation of TNF-a. Thus, the above test can be used to show that the compounds of Formula I have the ability to reduce the level of TNF-α produced by a cell. It will be understood that the previous hesitation is intended to be illustrative and not restrictive. Mushas modalities will be apparent to experts in the subject of the lestura of the previous dessripsión. The alsanse of the invention, therefore,. it will be determined not with reference to this previous dissension, but it must, instead, be determined are reference to the annexed claims, together they are the absolute alsanse of the equivalents to the suals such claims are entitled. The expositions of all the articles and references, which include the patent applications and publications, will be insorporated here as a reference for all purposes.

Claims (30)

1. A method to inhibit the growth of a tumor cell, this method comprises putting in check the tumor cell with a compound, which has the estrustura: or its aseptable salts pharmaceutically. in which: X, if present, is a selected member of the group that is of: A - CH2 - and - C - A, together the sarbones atoms to the suals are united, form a carbocyclic or heterosylic ring of 3, 4, 5, or 6 members; R9 and R10 are independently from the group consisting of: hydrogen, alkyl and halogen. And, it is a selected member of the group consisting of: H, alkyl and alkoxy; Z is a member selected from the group consisting of: -CEbOQ Q, is a member selected from the group consisting of: H, alkyl and S-alkyl; R, R, R3 and R4 are independently members of the group consisting of H, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alkoxy, halogen nitro and amino; and R, R, R7 and R8 are independently selected members of the group consisting of H, alkyl, alkenyl, alkynyl, hydroxyl, alkoxy and halogen with the proviso that if or Z is - Cx, \ OQ and Q is methyl, and Y is different from methoxy and ethoxy.
2. The method of agreement is the vindication 1, in which: X is - O -; And it's methoxy; Q is methyl; R and R are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
3. The method of agreement is the vindication 1, in which: X is - O -; - And it's hydrogen; Z is \ OQ Q is methyl; R1 and R4 are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
4. The method of agreement with claim 1, in which: X is - O -; And it's alkyl; Q is methyl; R and R are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
5. The method of agreement is the vindication 1, in which: X is - O -; And it's methoxy; O Z is - C "H R 1 and R 4 are both hydrogen, R 2 and R 3 if both n iodine, and R 5, R 6, R 7 and R 8 are all hydrogen.
6. The method according to claim 1, wherein: X is -O-; And it's methoxy; Z is - CH20Q; Q is hydrogen; R1 and R4 are both hydrogen; R2 and R3 are both iodo; and _, R5, R6, R7 and R8 are all hydrogen.
7. The method of agreement with claim 1, wherein: X is O; And it's methoxy; Z is CHOQ; Q is methyl; R1 and R4 are both hydrogen; R and R without both iodine; and R5, R6, R7 and R8 are all hydrogen.
8. The method, of agreement is the reivindisasión 1, -in which the sompuesto is selected from the group consisting of: BTO-969 Y
9. The method, according to claim 1, wherein the tumor cell is selected from the group of cells of the lung, colon, breast, ovary, prostate and liver.
10. The method, according to claim 1, wherein the tumor cell is in a mammalian subject.
11. The method, according to claim 1, wherein the tumor cell is a squamous cell carcinoma.
12. The method, according to claim 1, wherein the compound is formulated in a pharmaceutically acceptable form, is an exipient or carrier.
13. The method, according to I agree with claim 1, in which the compound is formulated in a liposome.
14. The method, I agree is the reivindisasión 1, in which the sompuesto is administered orally.
15. The method, according to claim 1, which also suffers the stage of observing the redussion in the growth of the tumor cell.
16. A method for inhibiting the growth of a tumor cell in a mammalian subject, this method comprises administering to the subject a therapeutically effective amount of a compound having the structure: or its pharmaceutically acceptable salts. in the sual: X, if present, is a member selected from the group consisting of: A CH2 - C - -. / A, together with the sarbono atoms to which it joins, forms a carbosisyllic or heterocyclic ring of 3, 4, 5, or 6 members; R9 and R10 are independently selected from the group consisting of: hydrogen, alkyl and halogen. And, it is a selected member of the group consisting of: H, alkyl and alkoxy; Z is a member of the group that is made up of: f ss -CHaQQ Q, is a member selected from the group consisting of: H, alkyl and S-alkyl; R, R, R and R are members selected, independently, from the group consisting of H, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alsoxi, halogen nitro and amino; and R, R, R7 and R8 are independently members of the group consisting of H, alkyl, alkenyl, alkynyl, hydroxyl, alkoxy and halogen are the sonsion that if OZ is \ OQ and Q is methyl, then Y is different from methoxy and ethoxy.
17. A method for inhibiting the sresence of a tumor cell, according to claim 16, wherein the administration is carried out in combination with immunotherapy.
18. A method for inhibiting the cresting of a tumor cell, according to claim 17, further comprising the step of administering to the mammal a tumor vasuna.
19. A farmasuccesis somposision, comprising a pharmaceutically acceptable carrier and a host that has the stress: or its aseptable salts pharmaceutically. in the sual: X, if present, is a member selected from the group consisting of: - CH2 - and - c - A, together with the carbon atoms to which it is attached, forms a carbocyclic or heterocyclic ring of 3, 4, 5, or 6 members; R9 and R10 are independently selected from the group consisting of hydrogen, alkyl and halogen. And, it is a member selected from the group consisting of: H, alkyl and alkoxy; Z is a selessioned member of the group that consists of Q, is a member selected from the group consisting of: H, alkyl and S-alkyl; R1, R2, R3 and R4 are independently members of the group consisting of H, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alsoxi, halogen nitro and amino; and R.sup.5, R.sup.6, R.sup.7 and R.sup.8 are independently linked members of the group consisting of H, alkyl, alkenyl, alkynyl, hydroxyl, alsoxi and halogen are the sonde and Q is methyl, and Y is different from methoxy and ethoxy.
20. The farmaséutisa somposision, of agreement are the reivindisasión 19, in which: X is - O -; And it's methoxy; Q is methyl; R1 and R4 are both hydroquinone; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
21. The pharmaceutical composition, according to claim 19, wherein: X is - O -; And it's hydrogen; O Z is OQ Q is methyl; R1 and R4 are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
22. The farmasuccease somposision, of agreement, is claim 19, in which: X is - 0 -; And it's alkyl; Z is S / \ OQ Q is methyl; R1 and R4 are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
23. The somposisióh farmaséutisa, of agreement are the reivindisasión 19, in which: X is - O -; 'And it's methoxy; R1 and R4 are both hydrogen; R2 and R3 without both iodine; and R5, R6, R7 and R + 8 are all hydrogen.
24. The farmaséutisa somposision, according to claim 19, in which: X is - O -; And it's methoxy; Z is - CH20Q; Q is hydrogen; R1 and R4 are both hydrogen; R2 and R3 are both iodo; and R5, R6, R7 and R8 are all hydrogen.
25. The farmaséutisa somposision, of agreement are the reivindisasión 19, in which: X is - O -; And it's methoxy; Z is CH2OQ; Q is methyl; -? R1 and R4 are both hydrogen; R2 and R3 without both iodine; and R5, R6, R7 and R8 are all hydrogen.
26. The pharmaceutical composition, according to claim 19, wherein the compound is selected from the group consisting of: ;
27. A method to treat a patient who suffers from a neoplastic disease state, this method offers to administer to the patient a sanctity anti-neoplastic effector of ionizing or non-ionizing radiation, in a set they are the therapy of an anti-neoplastic efffective sanctity of a body that has the estrustura: or its pharmaceutically acceptable salts, wherein: X, if present, is a member selected from the group consisting of: A - O - - S -, CH2 - C - A, together with the carbon atoms to which it is attached, forms a carbosylic or heterocyclic ring of 3, 4, 5, 6 or 6 members; R9 and R10 are independently selected from the group consisting of: hydrogen, alkyl and halogen. And, it is a member selected from the group consisting of: H, alkyl and alsoxi; Z is a member of the group that consists of: -OfcOQ. Q, is a member of the group consisting of: H, alkyl and S-alkyl; R1, R2, R3 and R4 are independently members of the group consisting of H, alkyl, alkenyl, alkynyl, aryl, hydroxyl, alsoxi, halogen nitro and amino; and with the convention that if O Z is \ OQ and Q is methyl, and Y is different from methoxy and ethoxy.
28. The method of agreement, with claim 27, in which the neoplastic disease state is a leusemia.
29. The method of agreement is the vindication 27, in which the state of neoplastic disease is a carcinoma.
30. A method for treating a patient suffering from a neoplastic disease state, this method provides the patient with an effective anti-neoplastic amount of ionizing or non-ionizing radiation in conjunction with the therapy of an effective sensitizing amount of a compound having the structure : or its salts are acceptable, preferably. in the sual:
MXPA/A/1999/008753A 1997-03-26 1999-09-24 Di-aryl ethers and their derivatives as anti-cancer agents MXPA99008753A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US041679 1997-03-26
US60/041679 1997-03-26

Publications (1)

Publication Number Publication Date
MXPA99008753A true MXPA99008753A (en) 2000-07-01

Family

ID=

Similar Documents

Publication Publication Date Title
US6169104B1 (en) Di-aryl ethers and their derivatives as anti-cancer agents
US6433012B1 (en) Method for inhibiting inflammatory disease
US6150407A (en) Methods for inhibiting angiogenesis
JP6234340B2 (en) How to treat a tumor
US6632798B2 (en) Methods for inhibiting angiogenesis
CA1331460C (en) Antiestrogenic hydrazones
TWI862414B (en) Pharmaceutical composition for preventing or treating treatment-resistant cancer
US12285406B1 (en) Method of treating cancer with a selenide
KR20100051837A (en) Treatment of pediatric tumors
MXPA99008753A (en) Di-aryl ethers and their derivatives as anti-cancer agents
US20040048808A1 (en) Methods for inhibiting angiogenesis
US6326405B1 (en) Anticancer agent
US5525626A (en) Treating susceptible human malignant tumors with 7-hydroxy-1,2-benzopyrone
MXPA00009345A (en) Benzoates derivatives for inhibiting angiogenesis
CA2280181A1 (en) Enhanced anti-angiogenic activity of permanently charged derivatives of steroid hormones
HK40043269A (en) Activators of the unfolded protein response
JPS6041058B2 (en) Method for producing acetic acid derivatives