MXPA06008810A - Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof - Google Patents

Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof

Info

Publication number
MXPA06008810A
MXPA06008810A MXPA/A/2006/008810A MXPA06008810A MXPA06008810A MX PA06008810 A MXPA06008810 A MX PA06008810A MX PA06008810 A MXPA06008810 A MX PA06008810A MX PA06008810 A MXPA06008810 A MX PA06008810A
Authority
MX
Mexico
Prior art keywords
substituted
atom
nitro
heteroaryl
aryl
Prior art date
Application number
MXPA/A/2006/008810A
Other languages
Spanish (es)
Inventor
a palladino Michael
Kenneth Lloyd George
Hayashi Yoshio
Nicholson Benjampin
Original Assignee
Hayashi Yoshio
Kenneth Lloyd George
Nereus Pharmaceuticals Inc
Nicholson Benjamin
Palladino Michael A
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 Hayashi Yoshio, Kenneth Lloyd George, Nereus Pharmaceuticals Inc, Nicholson Benjamin, Palladino Michael A filed Critical Hayashi Yoshio
Publication of MXPA06008810A publication Critical patent/MXPA06008810A/en

Links

Abstract

Compounds represented by the following structure (I) are disclosed:as are methods for making such compounds, wherein said methods comprise reacting a diacyldiketopiperazine with a first aldehyde to produce an intermediate compound;and reacting the intermediate compound with a second aldehyde to produce the class of compounds with the generic structure, where the first aldehyde and the second aldehydes are selected from the group consisting of an oxazolecarboxaldeyhyde, imidazolecarboxaldehyde, a benzaldehyde, imidazolecarboxaldehyde derivatives, and benzaldehyde derivatives, thereby forming the above compound wherein R1, R1', R1'', R2, R3, R4, R5, and R6, X1 and X2, Y, Z, Z1, Z2, Z3, and Z4 may each be separately defined in a manner consistent with the accompanying description. Compositions and methods for treating vascular proliferation are also disclosed.

Description

DEHYDROPHENYAHISTINS AND THEIR ANALOGS AND THE SYNTHESIS OF DEHYDROPHENYAHISTINS AND THEIR ANALOGS Related Requests [0001] This application corresponds to a continuation-in-part of the patent application of the E.U.A. Serial Number 10 / 632,531 filed on August 1, 2003, and which claims priority of the application of the US patent. Serial number 60 / 542,073 filed on February 4, 2004 and the provisional patent application of the US. Serial Number 60 / 624,262 presented on November 1, 2004, all of which are titled DEHYDROPHENYLAHISTINS AND ANALOGS THEREOF AND THE SYNTHESIS OF DEHYDROPHENYLAHISTINS AND ANALOGS THEREOF; and each of which is hereby incorporated by reference in its entirety. BACKGROUND OF THE INVENTION Field of the Invention [0002] The present invention relates to compounds and methods of synthetic preparation in the fields of chemistry and medicine. More specifically, the present invention relates to compounds and methods for producing compounds useful in the treatment of cancer and in the treatment of fungal infections. Brief Description of the Related Art [0003] It is considered that a single universal cellular mechanism controls the regulation of the eukaryotic cell cycle process. See for example Hart ell, L. H. et al. , Science (1989), 246: 629-34. It is also known that when an abnormality arises in the cell cycle control mechanism, cancer or an immune disorder may occur. Accordingly, as is also known, antitumor agents and immune suppressors may be among the substances that regulate the cell cycle. Thus, new methods are required to produce eukaryotic cell cycle inhibitors such as antitumor and immune enhancement compounds, and should be useful in the treatment of human cancer as chemotherapeutic antitumor agents. See, Roberge, M. et al. , Cancer Res. (1994), 54, 6115-21. [0004] Fungi, especially pathogenic fungi and related infections, represent a growing clinical challenge. The existing antifungal agents are of limited efficacy and toxicity, and the development and / or discovery of strains of pathogenic fungi that are resistant to drugs currently available or under development. By way of example, fungi that are pathogenic in humans include among others Candida spp. including C. albicans, C. tropicalis, C. kefyr, C. krusei and C.galbrata; Aspergillus spp. including A. fumigatus and A. flavus; Cryptococcus neoformans; Blastomyces spp. including Blastomyces dermatitidis; Pneumocystis carinii; Coccidioides immitis; Basidiobolus ranaruni; Coi tidiobolus spp .; Histoplasma capsulatum; Rhizopus spp. including R. oryzae and R. microsporus; Cunninghamella spp .; Rhizomucor spp .; Paracoccidioides brasiliensis; Pseudallescheria boydii; Rhinosporidiu seeberi - and Sporotrix schenckii (Kwon-Chung, K.J. &Bennett, J.E. 1992 Medical Mycology, Lea and Febiger, Malvern, PA). [0005] Recently, it has been reported that tryptostats A and B (which are diketopiperazines consist of proline and tryptophan isoprenylate residues), and five other structurally related diketopiperazines, inhibit cell cycle advancement in the M phase, see Cui, C. et al. , 1996 J Antibiotics 49: 527-33; Cui, C. et al. 1996 J Antibiotics 49: 534-40, and that these compounds also affect microtubule structure, see Usui, T. et al. 1998 Biochem J 333: 543-48; Kondon, M. et al. 1998 J Antibiotics 51: 801-04. In addition, natural and synthetic compounds have been reported to inhibit mitosis, and thus inhibit the eukaryotic cell cycle, by binding to the colchicine or colchicine binding site (CLC site) in tubulin, which is a macromolecule consisting of two subunits of 50 kDa (alpha- and beta-tubulin) and is the main constituent of microtubules. See, e.g., Iwasaki, S., 1993 Med Res Rev 13: 183-198; Hamel, E. 1996 Med Res Rev 16: 207-31; Weisenberg, R. C. et al., 1969 Biochemistry 7 -.4466-79. It is considered that microtubules are involved in several essential cellular functions such as axonal transport, cell mobility and determination of cell morphology. Therefore, inhibitors of microtubule function may have broad biological activity, and be applicable for medicinal and agrochemical purposes. It is also possible that cochrane site ligands (CLC) such as CLC, steganacin, see Kupchan, S. M. et al. 1973 J Am Chem Soc 95: 1335-36, podophyllotoxin, see Sackett, D. L., 1993 Farmacol Elr 59: 163-228, and combrestatins, see Pettit, G. R. et al. , 1995 J Med Chem 38: 166-67, may prove to be valuable as eukaryotic cell cycle inhibitors and thus may be useful as chemotherapeutic agents. [0006] Although metabolites of the diketopiperazine type have been isolated from various fungi as mycotoxin, see Horak R. M. et al. , 1981 JCS Chem Comm 1265-67; Ali M. et al. , 1898 Toxicology Letters 48: 235-41, or as secondary metabolites, see Smedsgaard J. et al. , 1996 J Microbiol Met 25: 5-17, little is known about the specific structure of digotopiperazine-type metabolites or their derivatives and their antitumor activity, particularly in vivo. Not only have these compounds been isolated as mycotoxins, the chemical synthesis of a type of digotopiperazine-type metabolite, phenylahistine, has been described by Hayashi et al. in J. Org. Chem. (2000) 65, page 8402. In the art, this diketopiperazine-like metabolite derivative, dehydrophenylahistin, has been prepared by enzymatic hydrogenation of its parent phenylahistine. With incidences of growing cancer, there is a particular need to chemically produce a class of derivatives of substantially purified diketopiperazine-like metabolites that have cell proliferation-specific inhibitory activity of animals and high antitumor and selectivity activity. Therefore there is a particular need for an efficient method to synthetically produce diketopiperazine metabolite derivatives, substantially purified and structurally and biologically characterized. [0007] Also, PCT publication O / 0153290 (July 26,2001) describes a non-synthetic method for producing dehydrophenylahistin by exposing phenylahistine or a particular phenylahistin analog to a dehydrogenase obtained from Streptojmyces albulus.
SUMMARY OF THE INVENTION [0008] Compounds and methods for the synthetic manufacture of compounds are described for a class of compounds having the structure of Formula (I):
[0009] The disclosed compounds have the structure of formula (I) wherein: [0010] R <7> R <4>, and R <6>, each is selected separately from the group consisting of a hydrogen atom, a halogen atom and C! -C24 saturated alkyl, C? -C24 unsaturated alkenyl, alkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl groups hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? -C24 saturated alkyl, C, -C2 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl groups; [0011] Rx 'and Ri "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and C groups - C2 saturated alkyl, unsaturated alkenyl L-C2, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl , substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -C0-0-R, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl , and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? -C24 saturated alkyl, C? -C2 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl , heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; [0012] R2, R3, and R5 are each selected separately from the group consisting of a hydrogen atom, a halogen atom and C? -C12 alkyl groups, C1-C12 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, substituted sulfonyl and sulfonyl groups; [0013] Xi and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above; [0014] Y is selected from the group consisting of a nitrogen atom, a nitrogen atom substituted with a group R5 of the upper part, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; [0015] n is an integer equal to zero, one or two; [0016] Z, for each separate n, if not zero, and Z, Z2, Z3 and Z4 are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and [0017] dotted links can already be single or double bonds, - [0018] with the proviso that, in a particular compound, if Rx, Rx ', R2, R3, R4 and R5 are each a hydrogen atom , then it is not true that i and X2 each are an oxygen atom and R6 is already 3, 3-dimethylbutyl-1-ene or a hydrogen atom. [0019] The methods comprise the steps of: [0020] reacting a diacyldicetopiperazine with a first aldehyde, to produce an intermediate compound; and [0021] reacting the intermediate compound with a second aldehyde to produce the class of compounds with the generic structure, wherein [0022] the first aldehyde and the second aldehyde are selected from the group consisting of oxazolecarboxaldehyde, imidazolecarboxaldehyde, a benzaldehyde, derivatives of imidazolecarboxaldehyde, and benzaldehyde derivatives, in this manner forming a compound wherein [0023] The disclosed compounds have structure of the formula (I) wherein: [0024] R1; R4, and Rs, are each selected separately from the group consisting of a hydrogen atom, a halogen atom and C groups - C2 saturated alkyl, Cx-C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -C0-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C groups -C 2- saturated alkyl, unsubstituted C?-C 2 alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl , heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; [0025] Rx 'and Ri "are independently selected from the group consisting of a hydrogen atom, a halogen atom and L-C2 saturated alkyl groups, unsaturated alkenyl L-C2, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, aryl substituted, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C groups - C24 saturated alkyl, unsaturated C?-C 2 alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; [0026] R2, R3, and R5 each separately is selected from the group consisting of a hydrogen atom, an atom of halogen and groups C? -C? alq saturated uyl, C? -C12 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, sulfonyl and substituted sulfonyl group; [0027] Xx and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each already substituted or unsubstituted with a group Rs as defined above, - [0028] And it is chosen from the group consisting of a nitrogen atom, a nitrogen atom substituted with a group R? of the above, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; [0029] n is an integer equal to zero, one or two; [0030] Z, for each separate n, if it is not zero, and Zi, Z2, Z3 and Z4 are each chosen separately from a carbon atom, a sulfur atom, a nitrogen atom or an oxygen atom; and [0031] dotted links can already be single or double links. [0032] In preferred embodiments of the compound and method, the imidazolecarboxaldehyde is 5- (1, 1-dimethyl-2-ethyl) imidazole-4-carboxaldehyde and the benzaldehyde comprises a single methoxy group. Additional preferred embodiments of the compounds described herein include compounds having a t-butyl group, a dimethoxy group, a chloro group, and a methylthiophene group, and methods for producing these compounds as well as compounds described in Tables 2, 3 and 4 as well as as methods to produce these compounds. [0033] Methods and materials for treating neoplastic tissue and preventing cancers or infection by pathogenic fungi are also described. These methods and materials are particularly well suited for treatment of mammalian subjects, more particularly humans involve administering to the subject a dehydrophenylahistin or its analogue. The method comprises administering to the subject a composition comprising an antitumor or antifungal effective amount of a dehydrophenylahistin or its analog. [0034] Additional modalities refer to methods for treating a condition in an animal, these methods may include administering to the animal a compound as described herein, in an amount that is effective to reduce vascular proliferation or in an amount that is effective in reducing vascular density. Exemplary conditions include neoplasm, such as cancers, as well as other conditions associated with or based on vascularization, including for example immune and non-immune inflammation, rheumatoid arthritis, chronic joint rheumatism, psoriasis, diabetic retinopathy, neovascular glaucoma, premature retinopathy, degeneration macular, rejection. of corneal graft, retrolental fibroplasia, rubeosis, capillary proliferation in atherosclerotic plaques, osteoporosis, and the like. In some modalities, the disease is not cancer. [0035] Other embodiments refer to methods for inducing vascular collapse in an animal. The methods may include treating the animal with a therapeutically effective amount of a compound of the Formula (I) as described here, for example. The therapeutically effective amount of the compound can cause depolymerization of tubulin in the vasculature. [0036] Preferably the animal can be a human. Preferably, the disease can be a tumor, diabetic retinopathy, age-related mascular degeneration and the like. In some aspects the disease is not cancer or cancer can be specifically excluded from the methods and uses. Preferably, the compound is KPU-02. [0037] Still further embodiments refer to pharmaceutical compositions for treating or preventing vascular proliferation, comprising a pharmaceutically effective amount of a compound described herein together with a pharmaceutically acceptable carrier thereof. Vascular proliferation can be a symptom of a disease, for example cancer, age-related macular degeneration and diabetic retinopathy. [0038] Some embodiments refer to methods for preferential targetting in tumor vasculature on vasculature of non-tumor tissue. The methods may include the step of administering to an animal, preferably a human, a compound having structure of Formula (I) as described herein. Non-tumor tissue can be for example skin, muscle, brain, kidney, heart, spleen, intestine and the like. The tumor vasculature can be preferentially targeted on the musculature that is not tumor tissue, for example at about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% and 90%. [0039] Other embodiments relate to methods for preferential targeting of tumor vasculature versus vasculature of non-tumor tissue, these methods may include administering to an animal an agent that preferably targets or targets tumor vasculature over the tumor. the vasculature that is not tumor tissue. [0040] Additional modalities refer to uses of a compound having the structure of Formula (I) in the preparation of a medicament for the treatment of a condition associated with increased vasculature or that is based on vasculature. In some aspects, the condition may be cancer, while in other particular types of cancer or all cancers are specifically excluded. The condition can be any other associated with hypervascularization, associated with vasculature or based on vasculature. Examples include immune and non-immune inflammation, rheumatoid arthritis, chronic joint rheumatism, psoriasis, diabetic retinopathy, neovascular glaucoma, premature retinopathy, macular degeneration, corneal graft rejection, retrolental fibroplasia, rubeosis, capillary proliferation at atherosclerotic plaques, osteoporosis, and the like. BRIEF DESCRIPTION OF THE DRAWINGS [0041] The accompanying drawings which are incorporated in and form part of the specification simply illustrate certain preferred embodiments of the present invention. Along with the rest of the specification, it is intended that they serve to explain preferred modes for producing certain compounds of the invention to those skilled in the art. In the drawings: [0042] Figure 1 illustrates a reaction scheme to produce dehydrophenylahistin by reacting a diacyldiketopiperazine 1 with an imidazolecarboxaldehyde 2 to produce an intermediate compound 3 which is reacted with a benzaldehyde 4 to produce a dehydrophenylahistin. [0043] Figure 2 illustrates the HPLC profile of the synthetic crude dehydrophenylahistin. [0044] Figure 3 illustrates a reaction scheme for producing dehydrophenylahistin by reacting a diacyldiketopiperazine 1 with a benzaldehyde 4 to result in an intermediate 17 which is reacted with an imidazolecarboxaldehyde to produce a dehydrophenylahistin. [0045] Figure 4 illustrates the HPLC profile of the crude synthetic tBu-dehydrophenylahistin produced by Route A and Route B.
[0046] Figure 5 illustrates two modification strategies for dehydroPLH for potent cytotoxic activity. [0047] Figure 6 illustrates the putative active dehydroPLH conformation in the phenyl portion. [0048] Figure 7 illustrates cytochrome P450 metabolism of phenylahistine. [0049] Figure 8 illustrates the Z-E migration of tBu-dehydroPLH. [0050] Figure 9 illustrates the synthesis and pro-drug image of acyl-E-tBu-dehydroPLH. [0051] Figure 10 illustrates the temperature gradient of 3-Z-benzylidene-6- [5p- (1, 1-dimethylallyl) -1N-imidazol-4"-Z-ylmethylene] -piperazin-2, 5-dione. [0052] Figure 11 illustrates the temperature gradient of 3-Z-benzylidene-6- (5" - tert-butyl-lH-imidazole-4"-Z-ylmethylene) -piperazin-2,5-dione. [0053] Figure 12 illustrates the effect of KPU-2, KPU-35 and t-butyl-phenylahistine, in comparison with colchicine and taxol in permeability of HuVEC monolayer to FITC-Dextran. [0054] Figure 13 illustrates the effect of KPU-2 alone and in combination with CPT-11 on tumor growth estimated in the Human Colon Tumor Xenograft model. HT-29.
[0055] Figure 14 illustrates the effect of KPU-2 alone and in combination with CPT-11 on the weight of autopsy-cut tumors in individual mice in the Human Colon Tumor Xenograft model HT-29. [0056] Figure 15 illustrates the effect of KPU-2 alone and in combination with CPT-11 on tumor growth estimated in the Human Colon Tumor Xenograft model HT-29. [0057] Figure 16 illustrates the effect of KPU-2 alone and in combination with CPT-11 on the weight of tumors excised at autopsy in individual mice in the Human Colon Tumor Xenograft model HT-29. [0058] Figure 17 illustrates the effects of: A.
KPU-2, B. KPU-35 and C. t-butyl-phenylahistine alone and in combination with CPT-11 tumor growth estimated in the Human Colon Tumor Xenograft model HT-29. [0059] Figure 18 illustrates the effects of A.
KPU-2, B. KPU-35 and C. t-butyl-phenylahistine alone and in combination with CPT-11 in the weight of tumors cut at autopsy in individual mice in the model of Xenograft of Human Colon Tumor HT-29. [0060] Figure 19 illustrates the effects of KPU-2 alone and in combination with CPT-11 on tumor growth and on the Human Colon Tumor Xenograft model HT-29-. comparison of three studies. [0061] Figure 20 illustrates the effects of KPU-2 alone and in combination with CPT-11 on final tumor weights in the Human Colon Tumor Xenograft model HT-29: comparison of three studies. [0062] Figure 21 illustrates the effects of KPU-2 alone or in combination with Taxotere on tumor growth estimated in the Human Prostate Tumor Xenograft model DU-145. [0063] Figure 22 illustrates the effects of A. KPU-2, B. KPU-35 and C. t-butyl-phenylahistine alone and in combination with Taxotere on tumor growth estimated based on observations made during the portion in Life of the Human Prostate Tumor Xenograft Model DU-145. [0064] Figure 23 illustrates the effects of KPU-2 alone and in combination with Taxotere on individual cut tumor weights at autopsy in the Human Prostate Tumor Xenograft Model DU-145. [0065] Figure 24 illustrates the effects of KPU-35 alone and in combination with Taxotere on individual cut tumor weights at autopsy in the Human Prostate Tumor Xenograft Model DU-145.
[0066] Figure 25 illustrates the effects of A. KPU-2, B. KPU-35 and C. t-butyl-phenylahistine alone and in combination with Taxotere in model of Human Chest Tumor Xenograft MCF-7. [0067] Figure 26 illustrates the effects of KPU-2 alone and in combination with Taxotere on tumor growth estimated in the Human Pulmonary Tumor Xenograft model A549. [0068] Figure 27 illustrates the effects of KPU-2 alone and in combination with Taxotere on tumor weights cut at autopsy in the Human Lung Tumor Xenograft model A549. [0069] Figure 28 illustrates the effects of KPU-2 alone and in combination with Paclitaxel on the estimated tumor weight in the implanted murine mammary fat pad or panicle of the Human Breast Tumor model MDA-231. [0070] Figure 29 illustrates the effects of A. KPU-2, B. KPU-35 and C. t-butyl-phenylahistine alone and in combination with Paclitaxel model of Metastatic Tumor B16 FIO of Murine Melanoma. [0071] Figure 30 illustrates effects of KPU-35 and KPU-02 on tumor vasculature in the dorsal skin fold chamber of Figure 30.
[0072] Figure 31 illustrates the effect of KPU-02 in combination with CPT-11 on the tumor weight estimated in the Human Colon Tumor Xenograft model HT-29. [0073] Figure 32 illustrates the effect of KPU-02 in combination with CPT-11 on the tumor weight cut in the Human Colon Tumor Xenograft model HT-29. [0074] Figure 33 illustrates rapid depolymerization of tubulin in HuVEC cells induced by KPU-02 and KPU-35. [0075] Figure 34 illustrates the effect of KPU-02 on monolayer permeability in cells in HuVEC. [0076] Figure 35 illustrates the effect of KPU-02 on tumor blood flow in the P22 rat sarcoma model using the 125I-IAP technique. [0077] Figure 36 illustrates the effect of KPU-02 mg / kg IP (expressed as vehicle control%) in blood flow in different tissues 1 and 24 hours after the dose. [0078] Figure 37 illustrates the tumor necrosis induced by KPU-02 7.5 and 15.0 mg / kg IP in the rat sarcoma model P22. [0079] Figure 38 cites the activity of various tBu-dehydro-PLH derivatives in HT-29 cells. [0080] Figure 39 illustrates 3D QSAR analysis (CoMFA) of tBu-dehydro-PLH derivatives.
[0081] Figure 40 illustrates X-ray chromatographic analysis of tBu-dehydro-PLH derivatives. [0082] Figure 41 illustrates the biological activity of various phenylahistine derivatives compared to colchicine. [0083] Figure 42 illustrates the advancing effect of cell cycle of HeLa cells by tBu-dehydro-PLH (KPU-2) and KPU-35. [0084] Figure 43 illustrates the effect of dehydro-PLH and tBu-dehydro-PLH (KPU-2) on drug-sensitive and drug-resistant tumor cell lines compared to paclitaxel. [0085] Figure 44A illustrates turbidity spectra of microtubule protein polymerization in the presence of drug vehicle DMSO (0), 1.25 μM (p), 2.5 / μM (-), and 5 / iM (O) KPU-02. [0086] Figure 44B illustrates turbidity spectra of microtubule protein polymerization in the presence of drug vehicle DMSO (0), 1.25 μM (a), 2.5 / μM), and 5 / iM (O) CA4. [0087] Figure 44C illustrates turbidity spectra of microtubule protein polymerization in the presence of drug vehicle DMSO (0), 1.25 μM (a), 1. 5 μM (-), and 5 μM (O) CLC.
[0088] Figure 45 illustrates inhibition of MT in the absence or presence of a concentration range of KPU-02 (O), CA4 (p), and colchicine (0). [0089] Figure 46A illustrates frequency histograms of average microtubule lengths in vi tro in steady state in the presence of KPU-02. [0090] Figure 46B illustrates frequency histograms of average lengths of in vitro microtubules in steady state in the presence of CA4. [0091] Figure 46C illustrates frequency histograms of average microtubule lengths in vi tro in steady state in the presence of CLC. [0092] FIG. 47A illustrates photomicrographs of microtubule-electron electrons in MAP formed in vitro in steady state in the presence of KPU-02. [0093] Figure 47B illustrates photomicrographs of microtubule-rich electrons in MAP formed in vi tro at steady state in the presence of CA4. [0094] Figure 47C illustrates photomicrographs of microtubule-rich electron electrons in MAP formed in the stable state in the presence of CLC. [0095] Figure 48 illustrates a graphic summary of MT length decrease in steady state in the presence of KPU-02, CA4, and colchicine.
[0096] Figure 49A illustrates fluorescence emission spectra of tubulin in the presence of increasing KPU-02. [0097] Figure 49B illustrates a maximum setting of fluorescence emission at 487 nm to obtain Ka of tubulin for KPU-02. The insert illustrates waste. [0098] La, Figure 49C illustrates reciprocal double transformation of the link data. [0099] Figure 50 illustrates the graphical results of a competitive inhibition assay of colchicine to tubulin binding with various concentrations of [3H] CLC in the absence (0), or presence of 10 μM KPU-02 (O) or CA4 10 μM (o). [0100] Figure 51 illustrates log [compound] response curves for inhibition of mitotic advance by KPU-02, CA4, and CLC in MCF7 cells cultured in the presence of KPU-02 (o), CA4 (p), and colchicine (0) [0101] Figure 52 illustrates immunofluorescence microscopy images of MCF7 cells. a-d: Tubulin in control- (a) Tubulin in control, Cells treated with (b) KPU-02, (c) CA4, and (d) CLC; e-h: DNA in control- (e) DNA in control, cells treated with (f) KPU-02, (g) CA4, and (h) CLC.
[0102] Figure 53A illustrates immunofluorescence microscopy images of MCF7 cells treated with KPU-02. [0103] Figure 53B illustrates microscopy images of immunofluorescence of MCF7 cells treated with CA4. [0104] Figure 53C illustrates immunofluorescence microscopy images of MCF7 cells treated with CLC. [0105] Figure 54A illustrates immunofluorescence microscopy images of MCF7 cells treated with KPU-02. [0106] Figure 54B illustrates immunofluorescence microscopy images of MCF7 cells treated with CA4. [0107] Figure 54C illustrates immunofluorescence microscopy images of MCF7 cells treated with CLC. [0108] In certain Figures, compounds are identified using an alternate designation. A complete diagram to convert these alternate designations is as follows: Detailed Description of the Preferred Modality [0109] Each reference cited herein, including U.S. Pat. here cited, shall be considered incorporated by reference in this specification, to the fullest extent permitted by law. The patent application of the U.S.A. Serial Number 10 / 632,531, and the PCT patent application Serial Number PCTUS03 / 24232, both filed on August 1, 2003, and both with the title, "DEHYDROPHENYLAHISTINS AND ANALOGS THEREOF AND THE SYNTHESIS OF DEHYDROPHENYLAHISTINS AND ANALOGS THEREOF" are incorporated herein by reference totally. [0110] The disclosure provides methods for the synthetic preparation of compounds, including novel compounds, including dehydrophenylahistin and dehydrophenylahistin analogues, and provides methods for producing pharmaceutically acceptable cell cycle inhibitors, antitumor agents and antifungal agents in relatively high yield, wherein compounds and / or their derivatives are among the active ingredients in these cell cycle inhibitors, antitumor agents and antifungal agents. Other objects include providing novel compounds that are not obtained by currently available, non-synthetic methods. It is also an object to provide method for treating cancer, particularly human cancer, comprising the step of administering an effective tumor growth inhibitory amount of a member of a class of new antitumor compounds. This invention also provides a method for preventing or treating a pathogenic fungus in a subject which involves administering to the subject an effective antifungal amount of an Ina class member of novel antifungal compounds, for example administering a dehydrophenylahistin or its analogue in an amount and manner that provide the intended antifungal effect. In the preferred embodiment of the compounds and methods for producing and using these compounds described herein, but not necessarily in all embodiments of the present invention, these objectives are met. [0111] Also described herein are compounds and methods for producing a class of compounds wherein the compounds are represented by Formula (I):
[0112] wherein: [0113] Ri, R, and R6, each is separately selected from the group consisting of a hydrogen atom, a halogen atom, and C? -C24 saturated alkyl, unsaturated alkenyl L-C24, alkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano groups , alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? ~ C saturated alkyl, C? -C2 unsaturated alkenyl groups , cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl;
[0114] Ri 'and Ri "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and L-C24 saturated alkyl, C? -C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl groups , substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl , and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and Cx-C saturated alkyl, Cx-C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl , substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; [0115] R, R1 'and R1"are already covalently bound to each other or not; [0116] R2, R3, and Rs are each chosen separately from the group consisting of a hydrogen atom, a halogen atom and Ca-C? alkyl, C 1 -C 12 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, substituted sulfonyl and sulfonyl groups; [0117] Xi and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above, - [0118] ] And it is chosen from the group consisting of a nitrogen atom, a nitrogen atom substituted with a group R5 of the upper part, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a group substituted methylene; [0119] n is an integer equal to zero, one or two; [0120] Z, for each separate n, if not zero, and Zi, Z, Z3 and Z4 are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and [0121] the dotted links can already be single or double links. [0122] The method comprises the steps of: [0123] reacting a diacyldicetopiperazine with a first aldehyde to produce an intermediate compound; and [0124] reacting the intermediate compound with a second aldehyde to produce the class of compounds with the generic structure, wherein [0125] the first aldehyde and the second aldehyde are selected from the group consisting of an oxazolecarboxaldehyde, imidazolecarboxaldehyde, a benzaldehyde, imidazolecarboxaldehyde derivatives, and benzaldehyde derivatives, in this manner forming a compound of Formula (I) wherein: [0126] Ri, R, and Rs, each are selected separately from the group consisting of a hydrogen atom, a halogen atom and C groups -C 24 saturated alkyl, Ci-C 24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl , and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from an ato mo of hydrogen, a halogen atom and groups L-C24 saturated alkyl, Cx-C2 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, nitro substituted, phenyl, and substituted phenyl; [0127] R ly Ri "are independently selected from the group consisting of a hydrogen atom, a halogen atom and C groups - C2 saturated alkyl, unsaturated C2 -C2 alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -C0-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and L-C24 saturated alkyl groups, unsaturated alkenyl L-C2, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, - [0128] R, R3, and R5 each separately is selected from the group consisting of a hydrogen atom, an halogen and CI-L2 alkyl groups saturated, C? -C? 2 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, sulfonyl and substituted sulfonyl group, - [0129] Xi and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, and [0130] Y is selected from a group consisting of a nitrogen atom, a nitrogen atom substituted with a group R5 of the above, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; [0131] Z, for each separate n, if it is not zero, and Zi, Z2, Z3 and Z each is chosen separately from a carbon atom, a sulfur atom, a nitrogen atom or an oxygen atom; and [0132] dotted links can already be single or double links. [0133] Pharmaceutically acceptable salts and pro-drug esters of the compounds of Formulas (I) and (II) are also provided and methods are provided to simplify those compounds by the methods described herein. [0134] The term "pro-drug ester" especially with reference to a pro-drug ester of the compound of Formula (I) synthesized by the methods described herein, refers to a chemical derivative of the compound that is rapidly transformed in vivo to resulting in the compound, for example by hydrolysis in blood or internal tissues. The term "pro-drug ester" refers to derivatives of the compounds described herein formed by the addition of any of several ester-forming groups that are hydrolyzed under physiological conditions. Examples of pro-drug ester groups include pivaloyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, as well as other similar groups known in the art, including a group (5-R-2-oxo-1, 3-dioxolen-4-yl) methyl. Other examples of pro-drug ester groups can be found, for example, in T. Higuchi and V. Stella, in "Pro-drugs as Novel Delivery Systems", Vol. 14, A.C. S. Symposium Series, American Chemical Society (1975); and "Bioreversible Carriers in Drug Design: Theory and Application", edited by E. B. Roche, Pergamon Press: New York, 14-21 (1987) (provides examples of esters useful as pro-drugs for compounds containing carboxyl groups). [0135] The term "pro-drug ester" as used herein, also refers to a chemical derivative of the compound that rapidly transforms in vivo to result in the compound, for example by hydrolysis in the blood. The term "pro-drug ester" refers to derivatives of the compounds described herein formed by the addition of any of several ester-forming groups that are hydrolyzed under physiological conditions. Examples of pro-drug ester groups include pivoyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, as well as other of these groups known in the art, including a group (5-R-2-oxo-l, 3-dioxolen-4-yl). ) methyl. Other examples of pro-drug ester groups can be found, for example, in T. Higuchi and V. Stella, in "Pro-drugs as Novel Delivery Systems", Vol. 14, A.C. S. Symposium Series, American Chemical Society (1975); and "Bioreversible Carriers in Drug Design: Theory and Application", edited by E. B. Roche, Pergamon Press: New York, 14-21 (1987) (provide examples of esters useful as pro-drugs for compounds containing carboxyl groups). [0136] The term "pharmaceutically acceptable salt", especially when referring to a pharmaceutically acceptable salt of the compound of Formula (I) synthesized by the methods described herein, refers to any pharmaceutically acceptable salts of a compound, and preferably refers to an acid addition salt of a compound. Preferred examples of a pharmaceutically acceptable salt are the alkali metal salts (sodium or potassium), the alkaline earth metal salts (calcium or magnesium), or ammonium salts derived from ammonia or from pharmaceutically acceptable organic amines, for example L-C7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine or tris- (hydroxymethyl) -aminomethane. With respect to the compounds synthesized by the method which are basic amines, preferred examples of pharmaceutically acceptable salts are acid addition salts of pharmaceutically acceptable organic or inorganic acids, for example hydrohalic, sulfuric, phosphoric or aliphatic, or carboxylic or sulfonic acid or aromatic, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic acid, methanesulfonic, p-toluenesulfonic or naphthalenesulfonic. [0137] The term "pharmaceutically acceptable salt", as used herein, also refers to any pharmaceutically acceptable salts of a compound, and preferably refers to an acid addition salt of a compound. Preferred examples of the pharmaceutically acceptable salt are the alkali metal salts (sodium or potassium), the alkaline earth metal salts (calcium or magnesium), or ammonium salts derived from ammonia or from pharmaceutically acceptable organic amines, for example C? -C7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine or tris- (hydroxymethyl) -aminomethane. With respect to compounds which are basic amines, preferred examples of pharmaceutically acceptable salts are acid addition salts of pharmaceutically acceptable organic or inorganic acids, for example hydrohalic, sulfuric, phosphoric or aliphatic or aromatic carboxylic or sulfonic acid, for example acid acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, p-toluenesulfonic or naphthalenesulfonic. [0138] Preferred pharmaceutical compositions disclosed herein include pharmaceutically acceptable salts and pro-drug esters of the compound of Formula (I) synthesized by the method described herein. Accordingly, if the manufacture of pharmaceutical formulations involves intimate mixing of the pharmaceutical excipients and the active ingredient in its salt form, then it is preferred to use pharmaceutical excipients which are not basic, ie they are acidic or neutral excipients. [0139] In preferred embodiments of the methods of the compounds described herein, a pseudo three-ring, planar, relatively rigid structure can be formed. In order to stabilize this relatively rigid planar pseudo-three-ring structure, R3 can preferably be selected to be hydrogen.
[0140] In other preferred embodiments of the compounds and methods described herein, n is equal to zero or one, more preferably one, and Z2, Z3 and Z4 and each separately is chosen from an oxygen atom, a nitrogen atom , and a carbon atom, more preferably at least one of Z2, Z3 and Z4 is a carbon atom and more preferably at least two of Z2, Z3 and Z4 are a carbon atom. All Z can simultaneously be carbon atoms. [0141] Still other preferred embodiments of the methods and compositions disclosed herein involve compounds having the structures of the Formulas (la) and (Ib), following:
[0142] wherein the variable groups are as defined herein.
[0143] The term "halogen atom" as used herein, means any of the radio-stable atoms of column 7 of the Periodic Table of the Elements, i.e. fluorine, chlorine, bromine or iodine, with fluorine and chlorine being preferred. [0144] The term "alkyl", as used herein, means any unbranched or branched, substituted or unsubstituted, saturated hydrocarbon, with C? -C6 unbranched, saturated, unsubstituted hydrocarbons being preferred, with methyl, ethyl, isobutyl being preferred and tert-butyl. Among the substituted saturated hydrocarbons, Ci-Cg substituted mono- and di- and per-halogen substituted hydrocarbons and amino-substituted hydrocarbons are preferred, with preference being given to perfluromethyl, perchloromethyl, perfluoro-tert-butyl and perchloro-tert-butyl. The term "substituted" has its ordinary meaning, as found in numerous contemporary patents of the related art. See, for example, US patents. Nos. 6,583,143, 6,509,331; 6,506,787; 6,500,825; 5,922,683; 5,886,210; 5,874,443; and 6,350,759. Specifically, the definition of substitute is as broad as that set forth in US Pat. No. 6,583,143, which defines the substituted term as any groups such as alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle and heterocycloalkyl, wherein at least one hydrogen atom is replaced with a substituent. The term "substituted" is also as broad as the definition available from the US patent. No. 6,509,331, which defines the term "substituted alkyl" such that it refers to an alkyl group, preferably 1 to 10 carbon atoms, having 1 to 5 substituents and preferably 1 to 3 substituents, selected of the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyacylamino, cyano, halogen, hydroxyl, carboxyl, carboxylalkyl, keto, thioke thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocycloxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl , -S02-alkyl, -S0 -substituted alkyl, -S02-aryl and -S02-heteroaryl. The other patents cited above also provide standard definitions for the term "substituted" which are well understood by those skilled in the art. The term "cycloalkyl" refers to any non-aromatic hydrocarbon anhydride, preferably having five to twelve atoms comprising the ring. The term "acyl" refers to alkyl or aryl groups derived from an oxo acid, with a preferred acetyl group. [0145] The term "alkenyl", as used herein, means any unsaturated, unbranched or branched, substituted or unsubstituted hydrocarbon, including polyunsaturated hydrocarbons, with unbranched, mono-unsaturated and di-unsaturated C?-C hidrocar hydrocarbons, without substituting preferred, and mono-unsaturated hydrocarbons, with more preferred di-halogen substitution. In the Ri and R positions of the compound of structure (I), a z-isoprenyl moiety is particularly preferred. The term "cycloalkenyl" refers to any non-aromatic hydrocarbon ring, which preferably has five to twelve carbon atoms comprising the ring. [0146] The terms "aryl", "substituted aryl", "heteroaryl" and "substituted heteroaryl" as used herein, refer to aromatic hydrocarbon rings, preferably having five, six or seven atoms, and more preferably than They have six atoms that comprise the ring. "Heteroaryl" and "substituted heteroaryl" refer to aromatic hydrocarbon rings wherein at least one heteroatom, for example an oxygen, sulfur or nitrogen atom, is in the ring together with at least one carbon atom. [0147] The term "alkoxy" refers to any unbranched or branched ether, substituted or unsubstituted, saturated or unsaturated, with unsaturated C, -C, unbranched, unsubstituted ethers, with methoxy being preferred, and also with dimethyl, diethyl, methyl-isobutyl and methyl-tert-butyl are also preferred. The term "cycloalkoxy" refers to any non-aromatic hydrocarbon ring that preferably has five to twelve atoms comprising the ring. [0148] The terms "purified", "substantially purified", and "isolated" as used herein, refer to the compound that is free of other different compounds with which the compound is normally associated in its natural state, such as that the compound of the invention comprises at least 0.5%, 1%, 5%, 10%, or 20%, and more preferably at least 50% or 75% of the mass, by weight of a given sample. [0149] The compound of Formula (I) can be chemically synthesized or produced from reagents known and available in the art. For example, modifications of diacyldicetopiperazine (diacetyldicetopiperazine) have been described, for example, by Loughlin et al., 2000 Bioorg Med Chem Lett 10:91 or by Brocchini et al., In WO 95/21832. The diacyldicetopiperazine (diacetyldicetopiperazine) can be prepared, for example, by diacetylation of the economical 2,5-piperazinedione (TCI Cat. No. GO 100, 25 g) with sodium acetate and sodium anhydride. The diacetyl structure of the activated diketopiperazine can be replaced with other acyl groups, to include carbamates such as Boc (t-butoxycarbonyl), Z (benzoyloxycarbonyl). [0150] The imidazolecarboxaldehyde can be prepared, for example, according to the procedure described in Hayashi et al., 2000 J Organic Chem 65: 8402, as illustrated below: aa% 8% BSP & 92 & 5C% C! Z •. Ci HsNCBO, or o CHCÍEI, Reflux reflux? w 7?%? ß% S33 & OH 2 c
[0152] The synthetic method described herein can preferably be carried out in the presence of cesium carbonate with a base in DMF and in an atmosphere of deoxygenated. The inert atmosphere exceeds the probable oxidation of alpha-activated carbon atoms of the diketopiperazine ring during treatment with cesium carbonate (see below) as reported, for example by Watanabe et al., 18th International Congress of Heterocyclic Chemistry in Yokohama, Japan (July 30, 2001), Abstract, page 225.
Oxidation with air of Activated Carbonyl Compounds with Cesium Salts [0153] Other embodiments of the synthetic method involve modifications to the compounds employed in or otherwise involved in the synthesis of compounds represented by Formula (I). These derivatives may include modifications to the phenyl ring, introduction of other aromatic ring systems, position of the aromatic ring, alterations to the imidazole ring system and / or further modifications to the 5-position on the imidazole ring. Examples of these modifications are discussed, for example, in Example 4. The result of these modifications includes increasing the nitrogen content of the phenyl ring and / or the compound which can increase the solubility of the compound. Other modifications may incorporate derivatives of known tubulin inhibitors, thus mimicking the activity of tubulin inhibitors. Other modifications may simplify the synthesis of the beta-ketoester involved in the production of the imidazolecarboxaldehyde used in the methods described herein. Pharmaceutical Compositions [0154] The present invention also encompasses the compounds described herein, optionally and preferably produced by the methods described herein, in pharmaceutical compositions comprising a pharmaceutically acceptable carrier prepared for storage and subsequent administration, which has a pharmaceutically effective amount of the products described above in a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro, 1985). Preservatives, stabilizers, colorants and even flavoring agents can be provided in the pharmaceutical composition. For example, sodium benzoate, ascorbic acid and p-hydroxybenzoic acid esters can be added as preservatives. In addition, antioxidants and suspending agents may be employed. [0155] The dehydrophenylahistin or dehydrophenylahistin analogue compositions can be formulated and used as tablets, capsules or elixirs for oral administration; suppositories for rectal administration; sterile solutions, suspensions for injectable administration; patches for transdermal administration, and sub-dermal and similar deposits. The injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid before injection or infusion, or as emulsions. Suitable excipients are for example, water, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, human serum albumin and the like. In addition, if desired, the injectable pharmaceutical compositions may contain minor amounts of non-toxic auxiliary substances, such as wetting agents, pH buffering agents and the like. If desired, preparations that enhance absorption (e.g., liposomes), may be employed. [0156] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil or other organic oils such as soy, almond or grapefruit oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Suspensions for aqueous injection may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds, to allow the preparation of highly concentrated solutions. [0157] Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding a resulting mixture and processing the granule mixture, after adding suitable auxiliaries, if desired, to obtain dragee cores or tablets. Suitable excipients are in particular fillers such as sugars, including lactose, sucrose, mannitol or sorbitol; cellulose preparations such as, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth gum, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose and / or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as interlaced polyvinyl pyrrolidone, agar or alginic acid or its salt such as sodium alginate. Dragee cores are provided with convenient coatings. For this purpose, concentrated sugar solutions may be employed, which optionally may contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol and / or titanium dioxide, lacquer solutions and convenient 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 active compound doses. For this purpose, concentrated sugar solutions can be employed, which may optionally obtain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol and / or titanium dioxide, lacquer solutions and convenient organic solvents or solvent mixtures. Dyes or pigments may be added to the coatings of dragees or tablets for identification or to characterize different combinations of active compound doses. These formulations can be made using methods known in the art (see, for example, U.S. Patent Nos. 5,733,888 (Injectable compositions); 5,726,181 (compounds poorly soluble in water); 5,707,641; (Peptides or therapeutically active proteins); ,667,809 (lipophilic agent); 5,576,012 (Polymeric solubilizing agents); 5,707,615 (Anti-viral formulations); 5,683,676 (Medications in particles); 5,654,286 (Topical formulations); 5,688,529 (Oral suspensions); 5,445,829 (Prolonged-release formulations); 5,653,987 (Liquid Formulations); 5,641,515 (Controlled release formulations) and 5,601,845 (Spheroidal formulations). [0158] In addition, various pharmaceutically well known compositions are described herein in the pharmaceutical art for uses including intraocular, intranasal and intraauricular delivery. Pharmaceutical formulations include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eye drops, or gelan gum (Shedden et al., 2001 Clin Ther 23 (3).-440-50) or hydrogels (Mayer et al., 1996 Ophthalmologica 210: 101-3); ophthalmic ointments; ophthalmic suspensions, such as microparticles, small polymer particles containing drug, which are suspended in a liquid carrier medium (Joshi, A., 1994 J "Ocul Pharmacol 10: 29-45), lipid-soluble formulations (Aim et al., 1989 Prog Clin Biol Res 312.-447-58), and microspheres (Mordenti, 1999 Toxicol Sci 52: 101-6), and ocular inserts.These pharmaceutical formulations are most convenient and are preferably formulated to be sterile, isotonic and buffered for stability and comfort Pharmaceutical compositions may also include drops and sprays often prepared to simulate nasal secretions in many aspects to ensure maintenance of normal ciliary action, as described in Remington's Pharmaceutical Sciences (Mack Publishing, 18th Edition), and is well known to those skilled in the art, convenient formulations are more often and preferably isotonic, slightly cushioned to maintain a pH of 5.5 to 6.5 and more often and preferably include appropriate antimicrobial preservatives and drug stabilizers. Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for these aural formulations include glycerin and water. [0159] When used as a cell cycle inhibitor, a tumor growth inhibitor or a fungal growth inhibiting compound, the compound of Formula (I) can be administered by either oral or non-oral routes. When administered orally, it can be administered in the form of capsules, tablets, granules, sprays, syrups or other forms. When administered non-orally, it may be administered as an aqueous suspension, an oily preparation or the like or as a dropper, suppository, ointment or the like, when administered by injection or infusion, subcutaneously, intraperitoneally, intravenously, intramuscularly or the like . Similarly, it can be administered topically, rectally or vaginally as is considered appropriate by those skilled in the art to bring the compound into optimal contact with a tumor, thereby inhibiting tumor growth. Local administration at the tumor site is also contemplated, either before or after tumor resection, as controlled release formulations, reservoir formulations and infusion pump delivery. Methods of Administration [0160] The present invention also encompasses methods for producing and administering the described chemical compounds and the pharmaceutical compositions described. These disclosed methods include, among others, (a) administration via oral routes, this administration includes administration in the form of capsules, tablets, granules, sprays, syrups or the like; (b) administration through non-oral routes, this administration includes administration as an aqueous suspension, an oily preparation or the like or as a dropper, suppository, ointment or the like; administration by injection or infusion, subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally or the like; as well as (c) topical administration, (d) rectal administration or (e) vaginal administration, as deemed appropriate by those skilled in the art to bring the compound into contact with living tissue; and (f) administration by controlled release formulations, reservoir formulations, and infusion pump delivery. As further examples of these modes of administration and as further description of modes of administration, various methods for administration of the described chemical compounds and pharmaceutically compositions including modes of administration through intraocular, intranasal and intraauricular routes are described herein. [0161] The pharmaceutically effective amount of the dehydrophenylahistin or dehydrophenylahistin analogue composition, required as a dose, will depend on the route of administration, the type of animal, including human to be treated and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on factors such as weight, diet, concurrent medication, and other factors that those skilled in medical techniques will recognize.
[0162] In practicing the methods, the products or compositions can be used alone or in combination with each other, or in combination with other therapeutic or diagnostic agents. For example, as described herein, the compounds described herein are effective in the treatment of cancer, when used in combination with other active agents, specifically other chemotherapeutic agents, for example biological agents and the specific chemotherapeutics CPT-11, Taxotene (docataxel) and paclitaxel. The compounds described herein are also effective in the treatment of cancer when used in combination with other active agents, including anti-vascular agents, anti-angiogenic agents, such as Erbuitux (Imclone / bristol-Myers) and Iressa (AstraZeneca), other inhibitors. VEGF and biologics, more specifically at least one anti-VEGF antibody, especially monoclonal antibodies to the VEGF receptor, including DC101, a rat monoclonal antibody, which blocks the mouse VEGF receptor 2 (flk-1). These combinations can be used in vivo, ordinarily in a mammal, preferably in a human or animal. When used in vivo, the compounds described alone or in combination with other chemotherapeutic agents or other biological products can be administered to the mammal in a variety of forms, including parenterally, intravenously, by infusion or injection, subcutaneous, intramuscular, colonic, rectal, vaginal , nasal or intraperitoneal, using a variety of dosage forms. These methods can also be applied to chemical activity test in vivo. [0163] As will be readily apparent to a person skilled in the art, the useful in vivo dose to be administered and the particular mode of administration, will vary depending on the age, weight and species of mammals treated, the particular compounds employed and the use specific for which these compounds are used. The determination of effective dose levels, ie the dose levels necessary to achieve the desired result, can be achieved by a person skilled in the art, using routine pharmacological methods. Typically, clinical applications of human products are initiated at lower dose levels, with the dose level increasing until the desired effect is achieved. Alternatively, acceptable in vi tro studies may be employed to establish useful dosages and administration routes of the compositions identified by the present methods using established pharmacological methods.
[0164] In studies in non-human animals, applications of potential products are initiated at higher dose levels, decreasing the dose until the desired effect is no longer achieved or adverse side effects disappear. The dose may be in the range widely, depending on the desired effects and the therapeutic indication. Typically, doses may be between about 10 micrograms / kg and 100 mg / kg of body weight, preferably between about 100 micrograms / kg and 10 mg / kg of body weight. Alternate doses can be based and calculated on the surface area of the patient, as understood by those skilled in the art. The administration can be oral on a base every third day, every day sautéed, daily, twice a day or three times a day. [0165] The exact formulation, route of administration and dosage can be selected by the individual physician in view of the patient's condition. See, for example, Fingí et al., The Pharmacological Basis of Therapeutics, 1975. It should be noted that the attending physician must know how and when to terminate, interrupt or adjust the administration due to toxicity or organ dysfunctions. On the contrary, the attending physician will also know how to adjust the treatment to higher levels if the clinical response is not adequate (excluding toxicity). The magnitude of a dose of administration in the administration of a disorder of interest, it will vary with the severity of the condition to be treated and the route of administration. The severity of the condition can for example be evaluated in part, by standard prognostic evaluation methods. In addition, the dose and probably the dose frequency will also vary according to the age, body weight and response of the individual patient. A comparable program discussed above can be used in veterinary medicine. [0166] Depending on the specific conditions to be treated, these agents can be formulated and administered systemically or locally. A variety of techniques for formulation and administration can be found in Remington's Pharmaceutical Sciences, 18th Edition, Mack Publishing Co., Easton, PA (1990). Suitable routes of administration may include oral, rectal, transdermal, vaginal, transmucosal or intestinal; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, infusion, intraperitoneal, intranasal, or intraocular injections.
[0167] For injection or infusion, the agents can be formulated in aqueous solutions, for example in physiologically compatible buffers such as Hanks' solution, Ringer's solution or physiological saline buffer. For this transmucosal administration, penetrants appropriate to the barrier to permeate are used in the formulation. These penetrants are generally known in the art. The use of pharmaceutically acceptable carriers to formulate the compounds described herein for the practice of the invention in doses suitable for systemic administration is within the scope of the invention. With proper selection of carrier and suitable manufacturing practice, the compositions described herein, in particular those formulated as solutions, can be administered parenterally, such as by injection or intravenous infusion. The compounds can be easily formulated using pharmaceutically acceptable carriers well known in the art in doses suitable for oral administration. These carriers allow the compounds to be formulated as tablets, pills, capsules, liquids, gels, syrups, slimes, suspensions and the like, for oral ingestion by a patient to be treated.
[0168] Agents intended to be administered intracellularly, can be administered using techniques well known to those of ordinary skill in the art. For example, these agents can be encapsulated in liposomes, then administered as described above. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposomal contents are both protected from the external micro-environment and because the liposomes fuse with the cell membranes, they are efficiently delivered into the cell cytoplasm. Additionally, due to their hydrophobicity, small organic molecules can be administered directly intracellularly. [0169] The determination of the effective amounts is well within the capacity for those skilled in the art, especially in light of the detailed description provided herein. In addition to the active ingredients, these pharmaceutical compositions may contain convenient pharmaceutically acceptable carriers comprising excipients and auxiliaries that facilitate processing of the active compounds into preparations that can be employed pharmaceutically. Preparations formulated for oral administration may be in the form of tablets, dragees, capsules or solutions. The pharmaceutical compositions can be manufactured in a manner that is known per se, for example by conventional mixing, dissolving, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping or lyophilizing processes. [0170] Compounds described herein can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound or a subset of the compounds, which share certain chemical molecules, can be established by determining in vitro toxicity to a cell line, such as a mammalian cell line, and preferably a human cell line. . The results of these studies are often predictive of toxicity in animals, such as mammals or more specifically humans. Alternatively, the toxicity of particular compounds in an animal model, such as mice, rats, rabbits or monkeys, can be determined using known methods. The efficacy of a particular compound can be established using various methods recognized in the art, such as in. vitro, animal models, or clinical tests in humans. In vi tro models recognized in the specialty exist for almost every kind of condition, including the conditions brought down by the compounds described here, including cancer, cardiovascular disease and various fungal infections. Similarly, models in acceptable animals can be used to establish efficacy of chemicals to treat these conditions. When a model is selected to determine efficacy, the person with skill can be guided by the state of the art to select an appropriate model, dose and route of administration and regimen. Of course, clinical trials in humans can also be used to determine the efficacy of a compound in humans. [0171] When used as an anticancer agent, or a tumor growth inhibitor compound, the compounds described herein can be administered by either oral or non-oral routes. When administered orally, they can be administered in the form of capsules, tablets, granules, sprays, syrups or other such forms. When administered non-orally, they may be administered as an aqueous suspension, an oily preparation or the like or as a drip, suppository, ointment or the like, when administered by injection or infusion, subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally or similar. Similarly, they may be administered topically, rectally or vaginally, as deemed appropriate by those skilled in the art, to bring the compound into optimal contact with a tumor, thereby inhibiting tumor growth. Local administration at the site of the tumor or other disease condition is also contemplated, either before or after resection of the tumor, or as part of a treatment recognized in the specialty for the disease condition. Controlled release formulations, reservoir formulations, and infusion pump delivery are similarly contemplated. [0172] When used as an anticancer agent or an anti-tumor agent, it can be administered orally or non-orally to a human patient in the amount of about .0007 mg / day to about 7,000 mg / day of the active ingredient, and more preferably about 0.07 mg / day to about 70 mg / day of the active ingredient a, preferably, once per day or less, preferably, more than two to about ten times per day. Alternatively, and preferably also, the compound can preferably be administered in the amounts established continuously, for example by intravenous drip. Thus, for a patient weighing 70 kilograms, the preferred daily dose of the active tumor ingredient will be approximately 0.0007 mg / kg / day to approximately 35 mg / kg / day including 1.0 mg / kg / day and 0.5 mg / kg / day, and more preferably from 0.007 mg / kg / day to approximately 0.050 mg / kg / day, including 0.035 mg / kg / day. However, as will be understood by those skilled in the art, in certain situations it may be necessary to administer the anti-tumor compound, in amounts that exceed or exceed by far the preferred dose range set forth above, to treat effectively and efficiently. Aggressive tumors particularly advanced or lethal. [0173] When used as an antifungal agent, the preferable amount of dehydrophenylahistin or its effective analogue for treatment or prevention of a particular fungal pathogen, will depend in part on the characteristics of the fungus and the extent of the infection, and can be determined by standard clinical techniques. In vi tro or in vivo assays can optionally be employed to help identify optimal dose ranges. Effective doses can be extrapolated from dose-response curves derived from in vi tro analysis or preferably from animal models. The precise dose level should be determined by the attending physician or other health care provider and will depend on well-known factors, including the route of administration, and the individual's age, body weight, sex and general health.; the nature, severity and clinical stage of the infection; the use (or not) of concomitant therapies. [0174] The effective dose of dehydrophenylahistin or its analogue will typically be in the range of about 0.01 to about 50 mg / kg, preferably about 0.1 to about 10 mg / kg of mammalian body weight per day, administered in one or multiple doses. In general, the compound can be administered to patients who require this treatment in a range of daily doses of about 1 to about 2000 mg per patient. [0175] To formulate the dosage including the compounds described herein as a tumor growth inhibiting compound, known surfactants, excipients, smoothing agents, suspending agents and pharmaceutically acceptable film-forming substances and coating assistants and the like, may be employed . Preferably, alcohols, esters, sulphated aliphatic alcohols and the like can be used as surfactants; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium aluminate metasilicate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, calcium carboxymethyl cellulose and similar, can be used as excipients; magnesium stearate, talc, hardened oil and the like can be used as smoothing agents; Coconut oil, olive oil, sesame oil, peanut oil, soybean oil can be used as suspending agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or dimethyl acetate-methacrylate copolymer, as a polyvinyl derivative can be employed as suspending agents; and plasticizers such as phthalate ester and the like can be employed as suspending agents. In addition to the above preferred ingredients, sweeteners, fragrances, colorants, preservatives and the like may be added to the administered formulation of the compound, particularly when the compound is to be administered orally. [0176] The compositions described herein in pharmaceutical compositions may also comprise a pharmaceutically acceptable carrier. These compositions can be prepared for storage and subsequent administration. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art and are described for example in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro, ed., 1985). For example, these compositions can be formulated and used as tablets capsules or solutions for oral administration; suppositories for rectal or vaginal administration; Sterile solutions or suspensions for injectable administration. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid before injection or infusion or as emulsions. Suitable excipients include but are not limited to, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride and the like. In addition, if desired, the injectable pharmaceutical compositions may contain minor amounts of non-toxic auxiliary substances, such as wetting agents, pH buffering agents and the like. If desired, preparations for absorption enhancement (for example liposomes) can be employed. [0177] The pharmaceutically effective amount of the composition required as a dose will depend on the route of administration, the type of animal to be treated and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on factors such as weight, diet, concurrent medication and other factors that will be recognized by those skilled in the medical specialty. [0178] The products or compositions as described above can be used alone or in combination with each other or in combination with other therapeutic or diagnostic agents. Specifically, the products of the compounds described can be used alone or in combination with other chemotherapeutic or biological agents, include antibodies, for the treatment of cancer, or in combination with other anti-infectives for the treatment of fungal infection. These products or compositions can be used in vivo or in vi tro. The useful doses and the most useful modes of administration will vary depending on the age, weight and animal treated, the particular compounds employed and the specific use for which this composition or compositions are employed. The magnitude of a dose in the administration or treatment for a particular disorder will vary with the severity of the condition to be treated and the route of administration, and depending on the disease conditions and their severity, the compositions may be formulated and administered either systemically or locally A variety of techniques for formulation and administration can be found in Remington's Pharmaceutical Sciences, 18th ed. , Mack Publishing Co., Easton, PA (1990). t0179] To formulate the compounds of Formula (I), preferably synthetically produced according to the methods described herein, such as a cell cycle inhibitor, a tumor growth inhibitor or an antifungal compound, known excipient surfactants, smoothing agents, suspending agents and pharmaceutically acceptable film-forming substances and coating assistants and the like, may be employed. Preferably, alcohols, esters, sulfated aliphatic alcohols and the like can be used as surfactants; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium aluminate metasilicate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, carboxymethyl cellulose calcium and the like they can be used as excipients; magnesium stearate, talc, hardened oil and the like can be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soybean oil, can be used as suspension agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or dimethyl acetate-methacrylate copolymer, since a polyvinyl derivative can be used as a suspending agent; and plasticizers such as phthalate ester and the like can be used as suspending agents. In addition to the above preferred ingredients, sweeteners, fragrances, colorants, preservatives and the like can be added to the administered formulation of the compound, produced by the method, particularly when the compound is to be administered orally. [0180] Cell cycle inhibitors, antitumor agents and antifungal agents that can be produced by the method, can be administered orally or non-orally to a human patient in an amount of about 0.001 mg / kg / day to about 10,000 mg / kg / day of the active ingredient, and more preferably about 0.1 mg / kg / day to about 100 mg / kg / day of the active ingredient, preferably one every three days on a cyclic basis, one every sautéed day, once a day , twice per day or less preferably, more than two up to about ten times per day. Alternatively and preferably also, the compound produced by the method can preferably be administered in the amounts established in a continuous manner, for example by intravenous drip. Thus, for example in a patient weighing 70 kilograms, the preferred daily dose of the active anti-tumor ingredient would be from about 0.07 mg / day to about 700 grams / day, and more preferably, 7 mg / day to about 7 grams /day. However, as will be understood by those skilled in the art, in certain situations, it may be necessary to administer the anti-tumor compound produced by the method in amounts that exceed or even far exceed the preferred dose range set forth above, for Effectively and aggressively treat lethal or particularly advanced tumors. [0181] In the case of using the cell cycle inhibitor produced by the methods as a biochemical test reagent, the compound produced by the methods of the invention inhibits the advance of the cell cycle when dissolved in an organic solvent or hydrated organic solvent , and is directly applied to any of several systems of cultured cells. Usable organic solvents include for example methanol, methylsulfoxide and the like. The formulation may for example be a powder, granule or other solid inhibitor, or a liquid inhibitor prepared using an organic solvent or an organic hydrated solvent. While the preferred concentration of the compound produced by the method of the invention to be used as a cell cycle inhibitor is generally in the range of about 1 to about 100 μg / ml, the most appropriate use amount varies depending on the type of cell system. cultured cells and the purpose of use, as will be appreciated by persons with ordinary skill in the art. Also, in certain applications, it may be necessary or preferred for persons with ordinary skill in the art to use an amount outside the above range. [0182] From a pharmaceutical perspective, certain embodiments provide methods for avoiding or treating fungal infections and / or pathogenic fungi in a subject, involve administering to the subject a composition that includes a dehydrophenylahistin or its analog, for example administering dehydrophenylahistin or its analog in an amount and form that provide the intended antifungal effect. [0183] Other modalities include the treatment or prevention of infection in a patient by a pathogenic fungus such as those cited above or referred to below. [0184] Another embodiment refers to the treatment or prevention of infection in a patient by a pathogenic fungus that is resistant to one or more other antifungal agents, especially an agent other than dehydrophenylahistin or its analogue, including for example amphotericin B or its analogs or derivatives (including 14 (s) -hydroxyamfotericin B methyl ester, the hydrazide of amphotericin B with l-amino-4-methylpiperazine, and other derivatives) other polyrne macrolide antibiotics, including for example nystatin, cycidine, pimaricin and natamycin; flucytosine; griseofulvin; echinocandin or aureobasidin, including naturally occurring and semi-synthetic analogs, -dihydrobenzo [a] naptacenequinone; antifungals of nucleoside peptide including polyoxins and nico icins; allylamines such as naftifine and other squalene epoxidase inhibitors; and azoles, imidazoles and triazoles such as for example clotrimazole, miconazole, ketoconazole, econazole, butoconazole, oxiconazole, terconazole, itraconazole or fluconazole and the like. For additional conventional antifungal agents and new agents under development, see for example Turner and Rodriguez, 1996 Current Pharmaceutical Design, 2: 209-224.
Another embodiment involves the treatment or prevention of infection in a patient by a pathogenic fungus in cases where the patient is allergic to, otherwise intolerant of, unresponsive to one or more other antifungal agents or in which the use of other agents antifungals is otherwise contraindicated. Those other antifungal agents include, among others, the antifungal agents described above and elsewhere here. [0185] In the above methods for treatment or prevention, a dehydrophenylahistin or its analogue is administered to the subject in an effective antifungal amount. [0186] Other embodiments relate to the treatment or prevention of infection by a pathogenic fungus in a patient, by administration of a dehydrophenylahistin or its analog, in conjunction with the administration of one or more other antifungal agents, including for example any of the agents or type of previously mentioned agents (for example in combination with treatment with amphotericin B, preferably in a lipid or liposome formulation, an azole or triazole such as fluconazole, for example, an aureobasidin, dihydrobenzo [a] naptacenequinone, or an equinocardin ) as with a different dehydrophenylahistin or its analogue. [0187] Dehydrophenylahistin or its analogue can be administered before, after or at the same time as the other antifungal agent is administered. In certain embodiments, combination therapy will allow the use of reduced amounts of one or both antifungal components relative to the amount used, if used alone. [0188] Still other embodiments refer to the administration of a dehydrophenylahistin or its analogue to a subject for treatment or prevention of infection by a pathogenic fungus, wherein the subject is immunosuppressed or immunocompromised, for example as a result of genetic disorder or disease such as diabetes or HIV (HIV) or other infection, chemotherapy or radiation treatment for cancer or other disease, or immunosuppression induced by drug or otherwise, in connection with tissue or organ transplantation or the treatment of an autoimmune disorder. When the patient is or will be treated with an immunosuppressive agent, for example in connection with tissue or organ transplantation, a dehydrophenylahistin or its analog may be co-administered with the immunosuppressive agent (s) to try to avoid a pathogenic fungal infection.
[0189] Another aspect of this invention is the treatment or prevention of infection by a pathogenic fungus in an infected patient, or one suspected of being infected, with HIV, by administration of an antifungal dehydrophenylahistin or its analog, in conjunction with the administration of one or more anti-HIV therapeutics (including for example HIV protease inhibitors, reverse transcriptase inhibitors or antiviral agents). The dehydrophenylahistin or its analog may be administered before, after or at the same time as the administration of the anti-HIV agent (s). [0190] Another aspect of this invention is the treatment or prevention of infection by a pathogenic fungus in a patient by administration of an antifungal dehydrophenylahistin or its analog, in conjunction with the administration of one or more other antibiotic compounds, especially one or more antibacterial agents, preferably in an effective amount and regime to treat or prevent bacterial infection. Again, dehydrophenylahistin or its analog may be administered before, after or at the same time as administration of the other agent (s). [0191] Pathogenic fungal infections that can be treated or prevented by the methods described include, among others, Aspergillosis, including invasive pulmonary aspergillosis; Blastomycosis, including deep or rapidly progressive infections and blastomycosis in the central nervous system; Candidiasis, including retrograde candidiasis of the urinary tract, for example in patients with kidney stones, urinary tract obstruction, renal transplantation or poorly controlled diabetes mellitus; Coccidioidomycosis, including chronic disease that does not respond well to other chemotherapy; Cryptococcosis; Histopolasmosis; Mucormycosis, including for example craniofacial mucormycosis and pulmonary mucormycosis; Paracoccidioidomycosis; and Sporotrichosis. It should be noted that the administration of a composition comprising an antifungal amount of one or more of dehydrophenylahistin or its analogues, may be particularly useful to treat or prevent a pathogenic fungal infection in a mammalian subject when the fungus is resistant to one or more therapies. antifungal, or where the use of one or more other antifungal therapies is contraindicated, for example as mentioned above. [0192] Antifungal pharmaceutical compositions containing at least one antifungal dehydrophenylahistin or its analog, are also provided for use in practicing the described methods. Those pharmaceutical compositions can be packaged together with an appropriate packaging insert which contains, inter alia, instructions and information regarding its antifungal use. Pharmaceutical compositions are also provided which contain one or more dehydrophenylahistin or its analog together with a second antifungal agent. Methods of Treatment of Fungal Infections [0193] Certain embodiments described herein relate to methods to treat or prevent a pathogenic fungal infection including for example Aspergillosis, including invasive pulmonary aspergillosis; Blastomycosis, including deep or rapidly progressive infections and blastomycosis in the central nervous system; Candidiasis, including retrograde candidiasis of the urinary tract, for example in patients with kidney stones, obstruction of the urinary tract, renal transplantation or poorly controlled diabetes mellitus; Coccidioidomycosis, including chronic disease that does not respond well to other chemotherapy; Cryptococcosis; Histopolasmosis; Mucormycosis, including for example craniofacial mucormycosis and pulmonary mucormycosis; Paracoccidioidomycosis; and Sporotrichosis. The methods may involve administering at least one antifungal dehydrophenylahistin or its analogue, as described above, to a human subject such as to treat or prevent fungal infection. In certain embodiments, dehydrophenylahistin or its analog may be administered in conjunction with administration of one or more non-dehydrophenylahistin or its analogous antifungal agents such as amphotericin B, or an imidazole or triazole agent such as those previously mentioned. [0194] Pathogenic fungal infection can be topical, for example caused inter alia by species of Candida, Trichophyton, Microsporum or Epiderinophyton or mucosal, for example, caused by Candida albicans (for example oral and vaginal candidiasis). The infection can be systemic, for example caused by Candida albicans, Cryptococcus neoformans, Aspergillus fumigatus, Coccidiodes, Paracoccidiodes, Histoplasma or Blastomyces spp. The infection may also involve eutychotic mycetoma, chromoblastomycosis, cryptococcal meningitis, or phycomycosis. t0195] Additional modalities refer to methods to treat or prevent a pathogenic fungal infection selected from the group consisting of Candida spp. including C. albicans, C. tropicalis, C. kefyr, C krusei and C. galbrata; Aspergillus spp. including A. fumigatus and A. flavas; Cryptococcus neoibrrraans; Blastomyces spp. including Blastomyces dermati tidis;Pneumocvstis carinii; Coccidioides immitis; Basidiobolus ranarum; Conidiobolus spp .; Histoplasma capsulatum; Rhizopus spp. including R. oryzae and R. microsporus; Cunninghamella spp .; Rhizoniucor spp .; Paracoccidioides brasiliensis; Pseudallescheria boydii; Rhinosporidium see beri; and Sporothrix schenckii. Again, the method may involve administering a non-immunosuppressive antifungal dehydrophenylahistin or its analogue to a patient that requires it, such that the fungal infection is treated or prevented without inducing an adverse immunosuppressive effect. [0196] Additional modalities refer to methods to treat or prevent a pathogenic fungal infection that is resistant to another antifungal therapy, including pathogenic fungal infections that are resistant to one or more antifungal agents mentioned elsewhere herein such as amphotericin B, flucytosine, one of the imidazoles or triazoles (including for example fluconazole, ketoconazole, itraconazole and the other previously mentioned examples). The methods may involve administering to the patient one or more antifungal dehydrophenylahistin or its analogue, in an amount and dose regime such that fungal infection resistant to another antifungal therapy is treated or prevented in the subject.
[0197] Additional modalities refer to methods to treat or prevent a fungal pathogenic infection in a patient who is allergic to, intolerant of or who does not respond to another antifungal therapy or in whom the use of other antifungal agents is otherwise contraindicated, including one or more other antifungal agents mentioned elsewhere herein, such as amphotericin B, flucytosine, one of the imidazoles or triazoles (including for example fluconazole, ketoconazole, itraconazole and the other previously mentioned examples). The methods may involve administering to the patient one or more antifungal dehydrophenylahistin or its analog, in an amount such that the fungal infection is treated or prevented. Dehydrophenylahistin or its package analogs [0198] Certain embodiments refer to dehydrophenylahistin or its package analogues, preferably dehydrophenylahistin or its non-immunosuppressive immunosuppressant analogs in package, this term is intended to include at least one dehydrophenylahistin or its analogue, as described above, packaged with instructions for administering dehydrophenylahistin or its analogues as an antifungal agent, without causing an undesirable effect within a human subject. In some embodiments, the non-immunosuppressive antifungal dehydrophenylahistin or its analogue is a member of one of the preferred subsets of compounds described above. The dehydrophenylahistin or its analog can be packaged only with instructions or it can be packaged with another dehydrophenylahistin or its analogue, rapamycin or another ingredient or additive, for example one or more of the ingredients of the pharmaceutical compositions. The package may contain one or more containers filled with one or more of the ingredients of the pharmaceutical compositions. Associated as an option with this or these containers may be a notice in the form prescribed by a governmental regulatory agency for the manufacture, use or sale of pharmaceutical or biological products, this notice reflects approval by the agency of manufacture, use or sale for human administration. [0199] The following non-limiting examples are intended to describe the preferred methods using certain preferred embodiments. Variations in the details of the particular methods employed and in the precise chemical compositions obtained will undoubtedly be appreciated by those skilled in the art. EXAMPLE 1 A. Synthesis of Dehydrophenylahistin [0200] Dehydrophenylahistin is synthesized by condensation according to the following basic reaction scheme, as shown in Figure 1: N, N'-diacetyl-2, 5-piperazinedione [0201] 25.0 g of 2, Global 5-piperazinedione [2,5-piperazinedione (Aldrich G640-6), 25.0 g, 0.218 mol] in 100 mL acetic anhydride (Ac20) is mixed with sodium acetate (NaOAc) (17.96 g, 0.0218 mol) ). The mixture is heated to 110 degrees C for 8 hours using a double coil condenser under an Ar atmosphere. After Ac20 is removed by evaporation, the residue is dissolved in AcOEt, washed with 10% citric acid, 10% NaHCO3 and saturated NaCl (three times each), dried over Na2SO, and concentrated in vacuo. The residue is triturated with ether to form a solid. This solid was recrystallized from EtOAc with ether-hexane to give 26.4 g (61%) of N, N'-diacetyl-2,5-piperazinedione 1. l-Acetyl-3-. { (Z) -1- [5- (1, 1-dimethyl-2-propenyl) -1H-4-imidazolyl] methylidene} ] -2, 5- piperazinedione 2 [0202] To a solution of 5- (1, 1-dimethyl-2-propenyl) imidazole-4-carboxaldehyde (100 mg, 0.609 mmol) in DMF (2 mL) is added the compound 1 (241 mg, 1.22 mmol) and the solution is repeatedly evacuated in a short time to remove oxygen and flushed with Ar, followed by the addition of Cs2CO3 (198 mg, 0.609 mmol) and the evacuation-discharge or wash-through process. drag is repeated again. The resulting mixture is stirred for 5 hours at room temperature. After the solvent is removed by evaporation, the residue is dissolved in the mixture of EtOAc and 10% Na 2 CO 3, and the organic phase is washed with 10% Na 2 CO 3 again and NaCl saturated three times, dried over Na 2 SO 4 and concentrated to the vacuum The residual oil is purified by column chromatography on silica using CHCl3-MeOH (100: 0 to 50: 1) as an eluent to give 60 mg (33%) of a pale yellow solid 2. Dehydrophenylahistin [0203] To a solution of 2 (30 mg, 0.099 mmol) in DMF (0.8 mL) is added benzaldehyde (51 μL, 0.496 mmol, 5eq) and the solution is repeatedly evacuated in a short time to remove oxygen and flushed with Ar, followed by the addition of Cs2C03 (53 mg, 0.149 mmol, 1.5 eq) and the draining-washing process is repeated again. The resulting mixture is heated for 2.5 hours at 80 degrees C. (The temperature must be increased slowly, a rapid heating increases the production of E-isomer in the benzylidene portion.) After the solvent is removed by evaporation, the residue is dissolved in EtOAc, washed with water twice and in saturated NaCl three times, dried over Na2SO4 and concentrated in vacuo. On TLC using CHCl3-MeOH (10: 1), a spot can be observed with bright green-yellow luminescence at 365 nm UV. The purity of this crude product was more than 75% of the HPLC analysis. The resulting residue is dissolved in 90% aqueous MeOH and applied to a reverse phase HPLC column (YMC-Pack, ODS-AM, 20 x 250 mm) and eluted using a linear gradient of 70 to 74% MeOH in water for 16 minutes. at a flow rate of 12 mL / min, and the desired fraction is collected and concentrated by evaporation to give 19.7 mg (60%) of yellow dehydrophenylahistin. The HPLC profile of the synthetic crude dehydrophenylahistin is illustrated in Figure 2. [0204] In the purification of dehydrophenylahistin as shown in Figure 4, a major peak was the desired Z-form compound of dehydrophenylahistin. The formation of an E-isomer is observed as a minor component (approximately 10%), which elutes in a more polar peak than the Z-isomer. Like other minor peaks, the reduced Z and E compounds where the dimethylallyl part of dehydrophenylahistin is reduced, were also observed. The formation of these reduced compounds was due to aldehyde 2 with a reduced impurity, which is generated during the reduction with DIBAL-H and is not separated in the subsequent process. [0205] These minor compounds can be removed by preparative HPLC purification, resulting in dehydrophenylahistin with the Z-configuration in the benzylidene moiety in a yield of 60% (20% yield in two steps) with more than 95% purity. Compounds with E-configuration on the imidazole side of the diketopiperazine ring are not observed in this HPLC diagram, suggesting that the first reaction of compound 1 to 3 in Figure 1 is selective Z. B. Chemical Characteristics: [0206] The above dehydrophenylahistin compound is a pale yellow solid. Its structure is confirmed by standard NMR analysis. EXAMPLE 2 Synthesis and physical characterization of t-dehydrophenylahistin derivatives [0207] Dehydrophenylahistin structural derivatives are synthesized according to the following reaction schemes to produce tBu-dehydrophenylahistin. Synthesis by Route A (see Figure 1) is similar in certain aspects to the synthesis of dehydrophenylahistin synthesized as in Example 1. tBu-dehidroPLH Route A: [0208] N, N'-diacetyl-2, 5-piperazinedione 1 is prepared as in Example 1. 1) l-Acetyl-3 -. { (Z) -1- [5-tert-Butyl-lH-4-imidazolyl] methylidene} -2, 5-piperazinedione (16)
[0209] To a solution of 5-tert-butylimidazole-4-carboxaldehyde (3.02 g, 19.8 mmol) in DMF (30 mL) is added compound 1 (5.89 g, 29.72 mmol) and the solution is repeatedly evacuated in a short time to remove oxygen and cleaning by discharge with Ar, followed by addition of Cs2C03 (9.7 g, 29.72 mmol) and the evacuation-flushing process is repeated again. The resulting mixture is stirred for 5 hours at room temperature. After the solvent is removed by evaporation, the residue is dissolved in the mixture of EtOAc and 10% Na 2 CO 3, and the organic phase is washed with 10% Na 2 CO 3 again and NaCl saturated three times, dried over Na 2 SO and concentrated at empty. The residual oil is purified by column chromatography on silica using CHCl3-MeOH (100: 0 to 50: 1) as eluent to give 1.90 g (33%) of a pale yellow solid 16. a H NMR (270 MHz, CDC13) delta 12. 14 (d, br-s, ÍH), 9.22 (br-s, lH), 7.57 (s, lH), 7.18, (s, ÍH), 4.47 (s, 2H), 2.65 (s, 3H) , 1.47 (s, 9H). 2) t-Bu-dehydrophenylahistin
[0210] To a solution of l-acetyl-3-. { (Z) -1- [5-tert-Butyl-l, l-4-imidazolyl] methylidene} ] -2, 5-piperazinedione (16) (11 mg, 0.038 mmol) in DMF (1.0 mL), benzaldehyde (19 μL, 0.19 mmol, 5 eq) was added and the solution was evacuated repeatedly in a short time to remove the oxygen and flushing with Ar, followed by the addition of Cs2C03 (43 mg, 0.132 mmol, 3.5eq) and the flushing-evacuation process was repeated again. The resulting mixture was heated for 2.5 hours at 80 degrees C. After the solvent was removed by evaporation, the residue was dissolved in EtOAc, washed with water twice and NaCl saturated three times, dried over Na2SO4 and concentrated in vacuo. The resulting residue was dissolved in 90% aq MeOH and applied to a reverse phase HPLC column (YMC-Pack, ODS-AM, 20 x 250 mm) and eluted using a linear gradient from 70 to 74% MeOH in water for 16 minutes at a flow rate of 12 mL / min, and the desired fraction was collected and concentrated by evaporation to yield 6.4 mg (50%) of yellow tert-butyl-dehydrophenylahistin. XH NMR (270 MHz, CDC13) delta 12.34 br-s, HH), 9.18 (br-s, ÍH), 8.09 (s, lH), 7.59 (s, HH), 7.31-7. 49 (m, 5H), 7.01 s, 2H), 1.46 (s, 9H). [0211] The reaction of dehydrophenylahistin to produce tBu-dehydrophenylahistin is identical to Example 1.
[0212] The total yield of the recovered tBu-dehydrophenylahistin was 16.5%. Route B: [0213] N, N'-diacetyl-2,5-piperazinedione 1 was prepared as Example 1. 1) l-Acetyl-3- [(-benzylidene] -2,5-piperazinedione (17)
[0214] To a solution of benzaldehyde 4 (0.54 g, 5.05 mmol) in DMF (5 mL) is added compound 1 (2.0 g, 10.1 mmol) and the solution was repeatedly evacuated in a short time to remove the oxygen and flushed with Ar, followed by the addition of CsC03 (1.65 g, . 05 mmoles) and the process of evacuation-washing by dragging was repeated again. The resulting mixture is stirred for 3.5 h at room temperature. After the solvent is removed by evaporation, the residue is dissolved in the mixture of EtOAc and 10% Na 2 CO 3, and the organic phase is washed with 10% Na 2 CO 3 again and NaCl saturated three times, dried over Na 2 SO 4 and concentrated in vacuo. . The residual solid was recrystallized from MeOH-ether to obtain a whitish solid of 17; yield 1.95 g (79%). 2) t-Bu-dehydrophenylahistin [0215] To a solution of l-acetyl-3- [(Z) -benzylidene] -2,5-piperazinedione (17) (48 mg, 0.197 mmol) in DMF (1.0 mL) was add 5-tert-butylimidazole-4-carboxaldehyde 15 (30 mg, 0.197 mmol) and the solution was repeatedly evacuated in a short time to remove the oxygen and washed by stripping with Ar, followed by the addition of Cs2CO3 (96 mg, 0.296 mmol) and lava, the process of evacuation-washing by dragging was repeated again. The resulting mixture was heated for 14 h at 80 degrees C. After the solvent was removed by evaporation, the residue was dissolved in EtOAc, washed with water twice and saturated NaCl three times, dried over Na 2 SO and concentrated in vacuo. The resulting residue was dissolved in MeOH aq 90% and applied to a reverse phase HPLC column (YMC-Pack, ODS-AM, 20 x 250 mm) and eluted using a linear gradient of 70 to 74% MeOH in water for 16 minutes at a flow rate of 12 mL / min, and the desired fraction was collected and concentrated by evaporation to give 0.8 mg (1.2%) of yellow tert-butyl-dehydrophenylahistin.
[0216] The total yield of the recovered tBu-dehydrophenylahistin was 0.9%. [0217] The HPLC profile of the crude synthetic tBu-dehydrophenylahistin from Route A and from Route B is illustrated in Figure 4. [0218] Two other tBu-dehydrophenylahistin derivatives were synthesized according to the Route A method In the synthesis of the additional b-dehydrophenylahistin derivatives, modifications were made to the compound benzaldehyde 4. [0219] Figure 4 illustrates the similarities of the HPLC profiles (column: YMC- ODS-AM Pack (20 x 250mm); Gradient: 65% to 75% in a methanol-water system for 20 minutes, then 10 minutes in a 100% methanol system; Flow rate: 12 mL / min; OD 230 nm) of the dehydrophenylahistin synthesized from Example 1 (Fig. 2) and the above-exemplified tBu-dehydrophenyilahistine compound produced by Route A. [0220] The introduction sequence of the aldehydes is relevant to the field and therefore an aspect of the synthesis. An analogue of dehydrophenylahistin was synthesized as a control or model, wherein the dimethylallyl group was changed to the tert-butyl group with a similar steric hindrance at the 5-position of the imidazole ring.
[0221] The synthesis of this "tert-butyl (tBu) -deshydrophenylahistin" using "Route A" was shown above: particularly, the sequence of introduction of the aldehyde exactly follows the synthesis of dehydrophenylahistin and exhibits a total yield of 16.5% of tBu-dehydrofenilahistina. This yield was similar to that of dehydrophenylahistin (20%). Using "Route B" where the sequence of introduction of the aldehydes is opposite to that of Route "A" for the synthesis of dehydrophenylahistin, only a trace amount of the desired tBu-dehydroPLH was obtained with a total yield of 0.9% although the introduction of the first benzaldehyde 4 would yield a 76% yield of the intermediate 17. This result indicated that it can be difficult to introduce the highly bulky imidazole-4-carboxaldehydes with a substitution group having a quaternary carbon at the 5-position adjacent to it. in the imidazole ring, in intermediate 17, suggesting that the sequence for introducing aldehyde is an important aspect to obtain high yield of dehydrophenylahistin or a dehydrophenylahistin analog using the synthesis written here. [0222] From the HPLC analysis of the final crude products as shown in Figure 4, a very high content of tBu-dehydrophenylahistin and small amount of by-product formation was observed in the crude sample of Route A (left) . However, a relatively minor amount of the desired tBu-dehydrophenylahistin and several other by-products was observed in the sample obtained using Route B (right). EXAMPLE 3 Large Scale Alternate Synthesis of Dehydrophenylahistin and Analogs Synthesis of 3-Z-Benzylidene-6- [5"- (1,1-dimethylallyl) -lH-imidazole -4" -Z-ylmethylene] piperazine-2, 5- diona [Dehydrophenylamine] (1)
[0223] Reagents: a) LDA, CH3CH0; b) Cough-Cl, pyridine; c) DBU; d) NaOH; e) C2C1202; f) KOOCCH2COOEt, BuLi; g) S02C12; h) H2NCHO, H20; i) LIA1H4; j) Mn02; k) 1,4-diacetyl-piperazin-2, 5-dione, Cs2C03; 1) benzaldehyde, Cs2CO methyl ether of 3-hydroxy-2,2-dimethyl-butyric acid
[0224] A solution of LDA in heptane / THF / ethylbenzene (2 M, 196 ml, 0.39 mol) is added under argon to a solution of methyl isobutyrate (45 ml, 0.39 mol) in THF (270 ml) at -60 degrees. and the resulting mixture is stirred for 30 min. A solution of acetaldehyde (27 ml, 0.48 mol) in THF (45 ml) pre-cooled to -60 degrees, it is added slowly and the resulting solution is stirred for 30 more minutes. Saturated ammonium chloride (50 ml) is added and the solution allowed to warm to room temperature. The reaction mixture is extracted with ethyl acetate and the extracts are washed with HCl (2 M), sodium bicarbonate and then brine. The organic layer is dried over magnesium sulfate, filtered, then evaporated to give a clear oil (52.6 g). Distillation 76-82 degrees / 30 mm Hg gave methyl ester of pure 3-hydroxy-2,2-dimethyl butyl acid (42.3 g, 74%). (Burk et al., J. Am. Chem. Soc., 117: 4423-4424 (1995)). [0225]? I NMR (400 MHz, CDCl3) delta 1.15 (d, J = 6 .1 Hz, 3H); 1.17 (s, 6H); 2.66 (d, J = 6.2 Hz, ÍH, -OH); 3.71 (s, 3H, -OMe); 3.87 (app quintet, J = 6.4 Hz, 1H, H3). 2-, 2-Dimethyl-3- (toluene-4-sulfyloxy) -butyric acid methyl ester [0226] To a cooled solution (0 degrees) of 3-hydroxy-2, 2-dimethyl-butyl methyl ester ( 52.0 g, 0.36 mol) in pyridine (100 ml), p-toluene sulfonyl chloride (69.0 g, 0.36 mol) is gradually added. The mixture is cooled to warm to room temperature and stirred for 60 hours. The reaction is again cooled in ice and acidified by the addition of HCl (2 M). The resulting solution is extracted with ethyl acetate, the extracts are washed with HCl, then brine, dried and evaporated to give an oil which forms a white precipitator upon standing. This mixture is dissolved in the minimum amount of ethyl acetate and then light oil is added to produce a white precipitate that is collected and washed with more light oil. The filtrate was partially evaporated and a second crop of crystals was obtained and added to the first to result in 2,2-dimethyl-3 - (toluene-4-sulfyloxy) -butyric acid methyl ester (81.2 g, 76% ). [0227] X H NMR (400 NMz, CDCl 3) delta 1.12 (s, 3H); 1.13 (s, 3H); 1.24 (d, J = 6.4 Hz, 3H); 2.45 (s, 3H, -PhMe) 3. 58 (s, 3H, -OMe); 4.94 (quartet, J = 6.4 Hz, 1H, H3), 7.33 (d, J = 8.0 Hz, 2H), 7.78 (d, J = 8.0 Hz, 2H). [0228] Evaporation of the final filtrate resulted in additional crude methyl ester of 2,2-dimethyl-3- (toluene-4-sulfonyloxy) -butyric acid (19.0 g, 18%). 2, 2-Dimethyl-but-3-enoic acid methyl ester
[0229] A solution of methyl ester of acid 2, 2-dimethyl-3- (toluene-4-sulfonyloxy) -butyric acid (18.06 g, 0.06 mol) in DBU (15 ml) is heated 140-160 for 3.5 hours. Allow the mixture to cool to room temperature and then dilute with ether. The mixture is washed with HCl (1 M), sodium bicarbonate and then brine. The ether layer is dried and partially evaporated to give a concentrated solution of 2,2-dimethyl-but-3-enoic acid methyl ester (10 g). (Savu and Katzenellenbogen, J. Org Chem, 46: 239-250 (1981)). Higher evaporation is avoided due to product volatility (eg 102 degrees). (Tsaconas et al., Aust. J. Chem., 53: 435-437 (2000)). [0230] X H NMR (400 NMz, CDCl 3) delta 1.31 (s, 6H); 3.68 (s, 3H); 5.06 (d, J = 17.1 Hz, 1H, -CH = CH2); . 11 (d, J = 10, 7 Hz, ÍH, -CH = CH2); 6 03 (dd, J = 17.1, 7 Hz, ÍH, -CH = CH2). 2, 2 -Dimethyl-but-3-ene acid
[0231] The above ethereal solution of 2,2-dimethyl-but-3-enoic acid methyl ester (10 g) is diluted with ethanol (25 ml), sodium hydroxide (4 M, 22 ml) is added and the mixture is stirred overnight. The solution is partially evaporated to remove the methanol and the resulting mixture is added to HCl (1 M, 100 ml). The product is extracted with ethyl acetate and the extracts are dried and evaporated to give 2,2-dimethyl-but-3-enoic acid (6.01 g, 88% 2 steps). (Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000). [0232] XH NMR (400 MHz, CDC13) delta 1.33 (s, 6H); 5.11 (d, J = 10.8 Hz, ÍH, -CH = CH2), 5.15 (d, J = 17.2 Hz, 1H, -CH = CH2), 6.05 (dd, J = 17.2, 10.8 Hz, ÍH, -CH = CH2). [0233] Hydrogen malonate from monoethyl (Wierenga and Skulnick, "Aliphatic and Aromatic beta-keto Esters from Monoethyl Malonate: Ethyl 2-Butyrylacetate," Organic Syntheses Collective Volume 7, 213).
[0234] Potassium malonate (25.0 g, 0.15 mol) is suspended in water (15.6 ml) and cooled in an ice bath. Concentrated HCl (12.5 ml) is added dropwise for 30 minutes, then the mixture is stirred for a further 10 minutes. The precipitate is filtered, then washed twice with ether. The filtrate is separated and the aqueous part is extracted with ether. The combined ether solutions are dried (MgSO) and evaporated to result, as an oil, monoethyl hydrogen malonate (19. 2 g, 99%) that is dried under vacuum overnight (or 50 degrees / l mm for 1 hour) before use. Ethyl 4-4 -Dimethyl-3 -oxo-hex-5-acid ester
[0235] Hexalyl Chloride (3.83 mL, 43.9 mmol) is added dropwise to a cooled solution (0 degrees) of 2,2-dimethyl-but-3-enoic acid (5.0 g, 43.9 mmol) and DMF (1 drop) ) in anhydrous dichloromethane (25 ml). The mixture is stirred one hour at 0 degrees, for 16 hours at room temperature. Fractional distillation (121 degrees / 760 mm Hg) resulted in 2,2-dimethyl-but-3-enoyl chloride (4.1 g, 71%). [0236] Monoethyl hydrogen malonate (7.2 g, 0.05 mol) and bipyridyl (few milligrams) were dissolved in THF (90 ml) and the system was flushed with nitrogen. The solution was cooled to -70 degrees, then BuLi (2.5 M in hexanes, 37 ml, 0.09 mol) is added. After the addition of only ~ 10 ml of BuLi, the solution was turned to pink and additional THF (15 ml) was required to activate magnetic stirring. The cooling bath was removed and the remaining BuLi was added, the temperature was allowed to reach -10 degrees, before which the solution turned colorless. The mixture was again cooled to -60 degrees and a solution of 2,2-dimethyl-but-3-enoyl chloride (4.1 g, 0.03 mol) in THF (12 ml) was added dropwise. After completing the addition, the mixture was allowed to warm to 0 degrees and stirred for 3 hours, was then added to a 1: 1 mixture of ether / HCl IM (260 ml) at 0 degrees and stirred for an additional 1.5 h. The organic layer was removed, washed with HCl (1 M), sodium bicarbonate solution, brine, then dried and evaporated to give 4,4-dimethyl-3-oxo-hex-5-enoic acid ester (5.6 g, 98%). (Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000)) Distillation with a Kugeirohr oven (160 degrees / 1 mm Hg) resulted in the pure material.
[0237] X H NMR (400 MHz, CDCl 3) delta 1.26 (s, 6H); 1.27 (t, J = 6.9 Hz, 3H, CH2CH3); 3.51 (s, 2H); 4.18 (q, J = 6.9 Hz, 2H, -CH2CH3); 5.20 (d, J = 17.7 Hz, ÍH, - CH = CH2); 5.21 (d, J = 9.6 Hz, lH, -CH = CH2); 5.89 (dd, J = 17.7, 9.6 Hz, ÍH, -CH = CH2). Ethyl 2-chloro-4,4-dimethyl-3-oxo-hex-5-enoic acid ester
[0238] Sulfuryl chloride (0.84 ml, 10.4 mmol) is added to a cooled solution (0 degrees) of 4,4-dimethyl-3-oxo-hex-5-enoic acid ethyl ester (1.83 g, 9.93 mmol) in chloroform (7 ml). The resulting mixture was allowed to warm to room temperature and stirred for 30 minutes, after which the reflux was heated for 2 hours. After cooling to room temperature, the reaction mixture was diluted with chloroform, then washed with sodium bicarbonate, water, then brine. The organic portion was dried and evaporated to give as a brown oil, 2-chloro-4,4-dimethyl-3-oxo-hex-5-enoic acid ethyl ester (2.01 g, 93%). (Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000). [0239] XH NMR (400 MHz, CDCl3) delta 1.28 (t, J = 7.0 Hz, 3H, -CH2CH3 ), 1.33 (s, 3H), 1.34 (s, 3H), 4.24 (q, J = 7.0 Hz, 2H, -CH2CH3), 5.19 (s, 1H, 5.28 (d, J = 16.9 Hz, ÍH, -CH = CH2); 5.29 (d, J = 10.9 Hz, 1H, -CH = CH2); 5.96 (dd, J = 16.9, 10.9 Hz, ÍH, -CH = CH2) [0240] LC / MS tR = 8.45 (219.3 [M (Cl37) + H] + min. [0241] This material is reacted without further purification 5- (1,1-dimethyl-allyl) -3H-imidazole-4-carboxylic acid ethyl ester
[0242] A suspension of 2-chloro-4,4-dimethyl-3-oxo-hex-5-enoic acid ethyl ester (19.4 g, 0.09 mol) and water (1.94 ml, 0.11 mol) in formamide (36.8 ml) ) was shaken briefly, then distributed in 15 x 18 ml ampoules. The ampoules were sealed and heated at 150 degrees for 5 hours in a row. After cooling to room temperature, the contents of the ampules were thoroughly combined and extracted with chloroform. The extracts were dried and evaporated to give a concentrated formamide solution (14.7 g). This was added to a silica column (diameter 7 cm, height 11 cm) packed with MeOH l% / Et3N 1% in chloroform. Elution of the column with 2 L of this mixture followed by 2 L of 2% MeOH / 1% Et3N in chloroform resulted in the first fractions, a compound that is suspected to be 5- (1,1-dimethyl) ethyl ester allyl) -oxazole-4-carboxylic acid (1.23 g, 7%). [0243] HPLC (214nm) tR = 8.68 (50.4%) min. [0244] aH NMR (400 MHz, CDC13) delta 1.40 (t, J = 1 .1 Hz, 3H, -CH2CH3); 1.54 (s, 6H); 4.38 (t, J = 1 .1 Hz, 2H, -CH2CH3); 5.03 (d, J = 17.4 Hz, ÍH, -CH = CH2); 5.02 (d, J = 10.4 Hz, 1H, -CH = CH2); 6.26 (dd, J = 17.4, 10.4 Hz, ÍH, -CH = CH2); 7.83 (s, 1H). [0245] LCMS tR = 8.00 (210.1 [M + H] +, 361.1 [2M + H +]) min. [0246] Recovered from further reactions was the desired ethyl ester of 5- (1, 1-dimethyl-allyl) -3H-imidazole-4-carboxylic acid (3.13 g, 17%). (Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000)). [0247] HPLC (214nm) tR = 5.52 (96.0%) min. [0248] X H NMR (400 MHz, CDCl 3) delta 1.38 (t, J = 7.0 Hz, 3H); 1.57 (s, 6H); 4.35 (q, J = 7.0 Hz, 2H); 5.04-5.14 (m, 2H, -CH = CH2); 6.28 (dd, J = 18.0, 10.4 Hz, 1H, -CH = CH2); 7.52 (s, ÍH). [0249] LC / MS t R = 5.30 (209.1 [M + H] +, 417.2 [2M + H] +) min. [0250] Additional ethyl ester of 5- (1,1-dimethyl-allyl) -3H-imidazole-4-carboxylic acid was also recovered from the column (3.59 g, 19%) which was of lower purity but still sufficient for greater reaction [0251] Another by-product isolated from a similar reaction (smaller scale) by further elution of the column with 5% real MeOH Et3N 1% in chloroform, was a compound that is suspected to be 5- (1,1-dimethyl-allyl) ) -3H-imidazole-4-carboxylic acid (0.27 g, 9%). [0252] HPLC (245nm) tR = 5.14 (68.9%) min. [0253] XH NMR (400 MHz, CD3OD) delta 1.45 (s, 6H); 4.97 (d, J = 10.6 Hz, ÍH, -CH = CH2); 5.01 (d, J = 17. 7 Hz, 1H, -CH = CH2); 6.28 (dd, J = 17.7, 10.6 Hz, 1H, -CH = CH2); 7.68 (s, 1H). [0254] LCMS tR = 4.72 (181.0 [M + H] +, 361.1 [2M + H] +) min. [5- (1,1-Dimethyl-allyl) -3H-imidazol-4-yl] -methanol
[0255] A solution of 5- (1,1-dimethyl-allyl) -3H-imidazole-4-carboxylic acid ethyl ester (3.13 g, 15.0 mmol) in THF (60 ml) is added dropwise to a suspension of aluminum lithium hydride (95% suspension, 1.00 g, 25.0 mmol) in THF (40 ml) and the mixture removed at room temperature for 4 h. Water is added until gas evolution ceased, the mixture was stirred for 10 minutes, then filtered through a sintered funnel. The precipitate was washed with THF, then with methanol, the filtrate and the washings were combined, evaporated, then freeze dried to give [5- (1,1-dimethylallyl) -3H-imidazol-4-yl] -methanol (2.56 g, 102%). Residual water is removed by forming the azeotrope with chloroform before further reaction (see Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000)). [0256] HPLC (240nm) tR = 3.94 (56.8%) min. [0257] X H NMR (400 MHz, CD3OD) delta 1.43 (s, 6H); 4.57 (s, 2H); 5.01 (d, J = 10.5 Hz, ÍH, -CH = CH2); 5.03 (d, J = 17.7 Hz, 1H, -CH = CH2); 6.10 (dd, J = 17.7, 10.5 Hz, 1H, -CH = CH2); 7.46 (s, ÍH). [0258] LC / MS t = 3.77 (167.3 [M + H] +) min. 5- (1, 1 -Dimethyl-allyl) -3H-imidazole-4-carbaldehyde
[0259] Manganese dioxide (20 g, 0.23 mol) is added to a solution of [5- (1,1-dimethyl-allyl) -3H-imidazol-4-yl] -methanol (2.56 g, 0.02 mol) in acetone (300 ml) and the resulting mixture was stirred at room temperature for 5 hours. The mixture was filtered through filter paper and the residue was washed with acetone. The filtrate and washings are combined and evaporated to give 5- (1,1-dimethyl-allyl) -3H-imidazole-4-carbaldehyde (1.82 g, 51%). (Hayashi et al., J. Org. Chem., 65: 8402-8405 (2000)). [0260] HPLC (240nm) tR = 4.08 (91.5%) min. [0261] X H NMR (400 MHz, CDC13) delta 1.56 (s, 6H); 5.16 (d, J = 10. 6 Hz, ÍH, -CH = CH2); 5.19 (d, J = 17.3 Hz, ÍH, -CH = CH2); 6.22 (dd, J = 17.3, 10.6 Hz, 1H, -CH = CH2); 7.75 (s, ÍH), 10.02 (s, 1H, HCO). [0262] LC / MS t R = 3.75 (165.2 [M + H] +) min. l-Acetyl-3- [5 '- (1,1-dimethyl-allyl) -1H-imidazol-4'-Z-ylmethylene] -piperazin-2,5-dione
[0263] To a solution of 5- (1,1-dimethyl-allyl) -3H-imidazole-4-carbaldehyde (1.78 g, 0.01 mol) in DMF (35 ml) is added 1,4-diacetyl-piperazin-2 , 5-dione (8.59 g, 0.04 mol) and the mixture is evacuated, then washed by stripping with argon. The process of evacuation-washing by dragging is repeated twice more then cesium carbonate (3.53 g, 0.01 mol) is added. The process of evacuation-washing by dragging is repeated three more times, then the resulting mixture is heated to 45 degrees for 5 hours. The reaction mixture is partially evaporated (heating with high vacuum) until a small volume remains and the resulting solution is added dropwise to ice-water (50 ml). The yellow precipitate is collected, washed with water, then dried by freezing to give 1-acetyl-3- [5 '- (1,1-dimethyl-allyl) -lH-imidazol-4'-methylmethylene] -piperazine- 2, 5-dione (1.18 g, 36%). (Hayashi, Personal Communication (2001)). [0264] HPLC (214nm) tR = 6.01 (72.6%) min. [0265] X H NMR (400 MHz, CDCl 3) delta 1.53 (s, 6H); 2.64 (s, 3H); 4.47 (s, 2H); 5.19 (d, J = 17.3 Hz, 1H, -CH = CH2); 5.23 (d, J = 10.7 Hz, 1H, -CH = CH2); 6.06 (dd, J = 17.3, 10.7 Hz, ÍH, -CH = CH2); 7.16 (s, ÍH), 7.59 (s, ÍH), 9.47 (bs, lH); 12.11 (bs, lH) [observed -2% 1,4-diacetyl-piperazin-2,5-dione delta contamination 2.59 (s, 6H); 4.60 (s, 4H).] [0266] LC / MS t R = 6.65 (303.3 [M + H] +, 605.5 [2M + H] +) min. (n.b. different system used). 3-Z-Benzylidene-6- [5"- (1,1-dimethylallyl) -lH-imidazol-4" -Z-ylmethylene] -piperazine-2,5-dione
[0267] To a solution of l-acetyl-3- [5 '- (1,1-dimethyl-allyl) -lH-imidazol-4' -ylmethylene] -piperazin-2,5-dione (2.91 g, 9.62 mmol ) in DMF (70 ml) is added benzaldehyde (4.89 ml, 48.1 mmol) and the solution is evacuated then washed by stripping with argon. The process of evacuating-washing by dragging is repeated twice more, then cesium carbonate (4.70 g, 14.4 mmol) is added. The draining-washing process is repeated three more times after the resulting mixture is heated under the temperature gradient shown below. [0268] After a total time of 5 hours, the reaction was allowed to cool to room temperature also the mixture was added to water cooled by ice (500 ml). The precipitate was collected, washed with water 0 (300 ml), then dried by freezing to result in a yellow solid (2.80 g). This material was dissolved in chloroform (250 ml) filtered through filter paper and evaporated to azeotrope with the remaining water. The residual yellow precipitate (2.70 g, HPLC (214 nm) tR = 7.26 (93.6%) min.) Was partially dissolved in chloroform (20 ml), the suspension was sonicated for 5 minutes, after the solid was collected and dried in air to give 3-Z-benzylidene-6- [5"- (1,1-dimethylallyl) -1H-imidazole- 4"-Z-Imethylene] -piperazine-2, 5-dione (1.82 g, 54%) (Hayashi, Personal Communication (2001)), mp. 239-240 degrees (dec.). [0269] HPLC (214nm) tR = 6.80 (1.92) min, 7.33 (95.01%). [0270] X H NMR (400 MHz, CDCl 3) delta 1.53 (s, 6H); 5.18 (d, J = 11.6 Hz, ÍH, -CH = CH2); 5.21 (d, J = 11. 0 Hz, ÍH, -CH = CH2); 6.06 (dd, J "= 17.6, 11.0 Hz, ÍH, - CH = CH2), 6.99 (s, ÍH, -C-C = CH), 7.00 (s, ÍH, -C-C = CH); 7. 30-7.50 (m, 5 x ArH); 7.60 (s, H2"), 8.07 (bs, NH), 9.31 (bs, NH), 12.30 (bs, NH). [0271] LC / MS ta = 6.22 (349.3 [M + H] +, E isomer) , 6.73 (349.5 [M + H] +, 697.4 [2M + H] +, Z-isomer) min. [0272] ESMS m / z 349.5 [M + H] +, 390.3 [M + CH4CN] +. [0273] Evaporation of the chloroform solution gave additional 3-Z-benzylidene-6- [5"- (1,1-dimethylallyl) -1H-imidazol-4" -Z-ylmethylene] -piperazin-2,5-dione (0.76 g , 29%). [0274] HPLC (214nm) tR = 7.29 (84.5%) min. 3-E-Benzylidene-6- [5"- (1,1-dimethylallyl) -lH-imidazole-4" -Z- ilmetilen] -piperazin-2, 5-dione
[0275] Purification of preparative HPLC gave raw sample of synthesized material as previously produced the geometric isomer 3-E-benzylidene-6- [5"- (1,1-dimethylallyl) -lH-imidazole-4" -Z-ilmethylene ] -piperazin-2, 5-dione (1.7 mg). [0276] HPLC (214nm) tR = 6.75 (87.79) min. [0277] X H NMR (400 MHz, CDCl 3) delta 1.52 (s, 6H); 5.19 (d, J = 20.8 Hz, ÍH, -CH = CH2); 5.22 (d, J = 14.0 Hz, ÍH, -CH = CH2); 6.05 (dd, J = 18.0, 10.4 Hz, ÍH, -CH = CH2); 6.33 (s, ÍH, C-C = CH); 6.90-7.65 (m, 7H). [0278] ESMS m / z 349.5 [M + H] "390.4 [M + CH4CN] +. Synthesis of 3-Z-benzylidene-6- (5" -tert-butyl-lH-imidazole-4"-Z-il-methylene) ) -piperazin-2, 5-dione (2) I §
[0279] Reagents: g) S02C12; h) H2NCHO, H20; i) LiAlH4; j) Mn02; k) 1, 4-diacetyl-piperazin-2, 5-dione, Cs2C03; 1) Benzaldehyde, Cs2C03 Ethyl ester of 2-chloro-4,4-dimethyl-3-oxo-pentanoic acid
[0280] Sulfuryl chloride (14.0 ml, 0.17 mol) is added to a cooled solution (0 degrees) of ethyl pivaloylacetate (27.17 g, 0.16 mol) in chloroform (100 ml). The resulting mixture is allowed to warm to room temperature and stir for 30 minutes, after which it is heated to reflux for 2.5 hours. After cooling to room temperature, the reaction mixture is diluted with chloroform, then washed with sodium bicarbonate, water and then brine. [0281] The organic phase was dried and evaporated to give as a clear oil ethyl ester of 2-chloro-4,4-dimethyl-3-oxo-pentanoic acid (33.1 g, 102%). (Durant et al., "Aminoalkylimidazols and Process for their Production." Patent No. GB1341375 (Great Britain, 1973)). [0282] HPLC (214nm) tR = 8.80 (92.9%) min. [0283] X H NMR (400 MHz, CDCl 3) delta 1.27 (s, 9H); 1.29 (t, J = 7.2 Hz, 3H); 4.27 (q, J = 7.2 Hz, 2H); 5.22 (s, ÍH). [0284] 13 C NMR (100 MHz, CDC13) delta 13.8, 26. 3, 45.1, 54.5, 62.9, 165.1, 203.6. 5-ester-Butyl-3H-imidazole-carboxylic acid ethyl ester
[0285] A solution of 2-chloro-4,4-dimethyl-3-oxo-pentanoic acid ethyl ester (25.0 g, 0.12 mol) in formamide (47.5 ml) and water (2.5 ml) was stirred, then dispensed with 15 x 8 ml ampoules. All the ampoules were sealed and then heated to 150 degrees for 3.5 hours. The ampules were allowed to cool to room temperature, then water (20 ml) was added and the mixture was thoroughly extracted with chloroform. The chloroform was removed to give a solution of concentrated form (22.2 g) which was added to a flash silica column (diameter 6 cm, height 12 cm) packed in 1% MeOH / 1% Et3N in chloroform. Elution of the column with 2.5 L of this mixture followed by an L of 2% MeOH / 1% Et3N in chloroform gave in its first reactions, a product that is suspected to be 5-tert-butyl-oxazole-4-ethyl ester. -carboxylic (6.3 g, 26%). [0286] HPLC (214nm) ta = 8.77 min. [0287] a H NMR (400 MHz, CDC13) delta 1.41 (t, J = 7.2 Hz, 3H); 1.43 (s, 9H); 4.40 (q, J = 7.2 Hz, 2H); 7.81 (S, ÍH). [0288] 13 C NMR (100 MHz, CDC13) delta 14.1, 28.8, 32.5, 61.3, 136.9, 149.9, 156.4, 158.3. [0289] ESMS m / z 198.3 [M + H] +, 239.3 [M + CH4CN] +. [0290] LC / MS t t = 7.97 (198.1 [M + H] +) min. [0291] From subsequent reactions was recovered ethyl ester of 5-tert-butyl-3H-imidazole-4-carboxylic acid (6.20 g, 26%). (Durant et al., "Aminoalkylimidazols and Process for their Production. "Patent No. GB 1341375 (Great Britain, 1973)).
[0292] HPLC (214nm) tR = 5.41 (93.7%) min. [0293] XH NMR (400 MHz, CDC13) delta 1.38 (t, J = 7.0 Hz, 3H); 1.47 (s, 9H); 4.36 (q, J = 1.1 Hz, 2H); 7.54 (s, ÍH). [0294] 13 C NMR (100 MHz, CDC13) delta 13.7, 28. 8, 32.0, 59.8, 124.2, 133.3, 149.2, 162.6. [0295] ESMS m / z 197.3 [M + H] +, 238.3 [M + CH4CN] +. [0296] Greater elution of the column with an L of 5% MeOH / Et3N 1% gave a compound that is suspected to be 5-tert-butyl-3H-imidazole-4-carboxylic acid (0.50 g, 2%). [0297] HPLC (245nm) tR = 4.68 (83.1%) min. [0298] ^? NMR (400 MHz, CD3OD) delta 1.36 (s, 9H), - 7.69 (s, 1H). [0299] X H NMR (400 MHz, CDC13) delta 1.37 (s, 9H); 7.74 (s, 1H). [0300] 2 H NMR (400 MHz, CD3SO) delta 1.28 (s, 9H); 7.68 (s, ÍH). [0301] ESMS m / z 169.2 [M + H] +, 210.4 [M + CH4CN] +. (5- tert -butyl-3H-imidazol-4-yl) -methanol
[0302] A solution of 5-tert-butyl-3-imidazole-4-carboxylic acid ethyl ester (3.30 g, 16.8 mmol) in THF (60 ml) is added dropwise to a suspension of lithium aluminum hydride. (suspension 95%, 0.89 g, 22.2 mmol) in THF (40 ml) and the mixture is stirred at room temperature for 3 hours. Water is added until gas evolution ceased, the mixture was stirred for 3 minutes, then filtered through a sintered funnel. The precipitate was washed with THF, then with methanol, the filtrate and the washing were combined and evaporated. The residue was dried by freezing overnight to give, as a white solid (5-tert-butyl-3H-imidazol-4-yl) -methanol (2.71 g, 105%). (Durant et al., "Aminoalkylimidazols and Process for their Production." Patent No. GB1341375 (Great Britain, 1973)). [0303] HPLC (240nm) a = 3.70 (67.4%) min. [0304] ^? NMR (400 MHz, CD3OD) delta 1.36 (s, 9H); 4.62 (s, IR); 7.43 (s, ÍH). [0305] 13 C NMR (100 MHz, CD3OD) delta 31.1, 33.0, 57.9, 131.4, 133.9, 140.8. [0306] LC / MS t H = 3.41 (155.2 [M + H] +) min. [0307] This material is used without further purification. 5 -ter -butyl-3H-imidazole-4 -carbaldehyde
[0308] Manganese dioxide (30 g, 0.35 mol) is added to a heterogeneous solution of (5-tert-butyl-3H-imidazol-4-yl) -methanol (4.97 g, 0.03 mol) in acetone (700 ml) and the resulting mixture is stirred at room temperature for 4 h. The mixture was filtered through a pad of celite and the pad was washed with acetone. The filtrate and washing were combined and evaporated. The residues were triturated to yield as a colorless solid, 5-tert-butyl-3H-imidazole-4-carbaldehyde (2.50 g, 51%). (Hayashi, Personal Communication (2000)). [0309] HPLC (240nm) t = 3.71 (89.3%) min. [0310] X H NMR (400 MHz, CDC13) delta 1.48 (s, 9H); 7.67 (s, ÍH); 10.06 (s, ÍH). [0311] LC / MS t R = 3.38 (153. 2 [M + H] +) min. [0312] Evaporation of the filtrate from the trituration gave additional 5-tert-butyl-3H-imidazole-4-carbaldehyde (1.88 g, 38%). l-Acetyl-3- (51-tert-butyl-IH-imdazole -4 '-Z-ylmethylene) piperazine-2, 5-dione
[0313] To a solution of 5-tert-butyl-3H-imidazole-4-carbaldehyde (2.50 g, 164.4 mmol) in DMF (50 ml) is added 1,4-diacetyl-piperazine-2,5-dione (6.50) g, 32.8 mmol) and the solution is evacuated, washed by stripping with argon. The draining-washing process is repeated twice more, then cesium carbonate (5.35 g, 16.4 mmol) is added. The draining-washing process is repeated three more times, then the resulting mixture is stirred at room temperature for 5 h. The mixture was partially evaporated (heat and high vacuum) until a small volume remained and the resulting solution was added dropwise to the water (100 ml). The yellow precipitate was collected, then dried by freezing to result in l-acetyl-3- (5'-tertbutyl-lH-imidazol-4 '-Z-ylmethylene) -piperazin-2,5-dione (2.24 g, 47 %). (Hayashi, Personal Communication (2000)). [0314] HPLC (214nm) tR = 5.54 (94.4%) min. [0315] XH NMR (400 MHz, CDC13) delta 1.47 (s, 9H); 2.65 (s, 3H), 4.47 (s, 2H); 7.19 (s, ÍH); 7.57 (s, ÍH), 9.26 (s, 1H), 12.14 (s, 1H).
[0316] XH NMR (100 MHz, CDC13 + CD30D) delta 27.3, 30. 8, 32.1, 46.5, 110.0, 123.2, 131.4, 133.2, 141.7, 160.7, 162.8, 173.0. [0317] LC / MS a = 5.16 (291.2 [M + H] +, 581.6 [2M + H] + min 3-Z-Benzylidene-β- (5"-tert-butyl-1H-imidazole-4" - Z-ilmethylene) -piperazine-2, 5-dione
[0318] To a solution of l-acetyl-3- (5'-tert-butyl-lH-imidazol-4 '-Z-ylmethylene) -piperazine-2, 5-dione (2.43 g, 8.37 mmol) in DMF (55 ml) is added benzaldehyde (4.26 ml, 41.9 mmol) and the solution is evacuated, then washed by stripping with nitrogen. Evacuation-washing process by dragging is repeated twice more, then cesium carbonate (4.09 g, 12.6 mmol) is added. The draining-washing process is repeated three more times, then the resulting mixture is heated under the temperature gradient as shown below. After a total time of 5 hours, the reaction is allowed to cool to room temperature and the mixture is added to water cooled by ice (400 ml). The precipitate is collected, washed with water, then dried by freezing to give a yellow solid (2.57 g, HPLC (214 nm) at = 6.83 (83.1%) min.). This material is dissolved in chloroform (100 ml) and evaporated to azeotrope with remaining water, resulting in a brown oil. This is dissolved in chloroform (20 ml) and cooled in ice. After 90 minutes, the yellow precipitate was collected and air dried to result in 3-Z-benzylidene-6- (5"-ter-butyl-lH-imidazol-4" -Zylmethylene) -piperazin-2, 5- Diona (1.59 g, 56%). (Hayashi, Personal Communication (2000)). [0319] HPLC (214nm) tH = 6.38 (2.1%), 6.80 (95.2) min. [0320] X H NMR (400 MHz, CDC13) delta 1.46 (s, 9H); 7.01 (s, ÍH, -C-C = CH); 7.03 (s, 1H, -C-C = CH); 7.30-7.50 (m, 5H, Ar); 7.60 (s, ÍH); 8.09 (bs, NH); 9.51 (bs, NH); 12.40 (bs, NH). [0321] LC / MS t = 5.84 (337.4 [M + H] +, E-isomer), 6.25 (337.4 [M + H] +, 673.4 [2M + H] +, Z-isomer) min. [0322] ESMS m / z 337.3 [M + H] +, 378.1 [M + CH4CN] +. [0323] Evaporation of the chloroform solution gave additional 3-Z-benzylidene-6- (5"-tert-butyl-lH-imidazol-4" -Z-ylmethylene) -piperazin-2,5-dione (0.82 g, 29%). HPLC (214nm) rt = 6.82 (70.6%) min. Experimental General [0324] Sodium bicarbonate refers to a 5% solution.
[0325] Organic solvents were dried over sodium sulfate unless otherwise stated. Analytical Conditions NMR conditions [0326] XH NMR analysis (400 MHz) was performed on a Varian Inova Unity 400 MHz NMR machine. Samples were run in deuterated chloroform containing 0.1% TMS (unless otherwise specified) . Chemical shifts are reported with respect to TMS (0.00 ppm) or CH30H at 3.30 ppm for samples run in CD30D. Coupling constants are expressed in hertz (Hz). Analytical HPLC Conditions [0327] System Conditions 6: [0328] RP-HPLC is performed on a Rainin Microsorb-MV C18 column (5 m, 100 ALPHA) 50 x 4.6 mm. [0329] Shock absorber A: aqueous 0.1% TFA [0330] Shock absorber B: 0.1% TFA in 90% aqueous MeCN. [0331] Gradient: 0-100% of buffer B for 11 minutes. [0332] Flow Expense: 1.5 mL / min LCMS Conditions [0333] LCMS is run on a PerkinElmer Sciex API-100 instrument. [0334] LC Conditions: [0335] Reversed phase HPLC analysis [0336] Column: Monitor 5 m C18 50x4.6 mm [0337] Solvent A: 0.1% TFA in water [0338] Solvent B: 0.085% TFA in aqueous MeCN 90% [0339] Gradient: 0-100% B over 11.0 minutes [0340] Flow Rate: 1.5 mL / min [0341] Wavelength: 214 nm [0342] MS Conditions: [0343] ion source: spray Ions [0344] Detection: Ion count [0345] Flow rate to the mass spectrometer: 300 μL / min after separating from the column (1.5 mL / min). ESMS conditions [0346] ESMS is performed on a Perkin Elmer / Sciex-API III LC / MS / MS using an electro-dew or electro-fogging inlet. [0347] Solvent: AcOH 0.1% in aqueous MeCN in 60% [0348] Flow Expense: 25 μL / min [0349] Ion spray: 5000V [0350] Orifice plate: 55 V [0351] Acquisition time: 2.30 min [0352] Scan interval: 100-1000 amu / z [0353] Step size: 0.2 amu / z Preparative RP-HPLC Purification Conditions [0354] Reverse phase HPLC purification was carried out using Nebula with Waters column XterraMS (19x50 mm, 5 μm, C18) using the following conditions: [0355] Solvent A: Aqueous TFA up to 0.1% [0356] Solvent B: 0.1% TFA in 90% aqueous MeCN [0357] Gradient: about 5 to 95% B [0358] Flow Rate: 20 mL / min [0359] Wavelength: 214 nm [0360] The abbreviations are as follows: br s: broad broad singlet; BuLi: n-butyl lithium; d: doublet; DBU: 1,8-diazabicyclo [5.4.0] undec-7-ene; ESMS: electro dew mass spectrometry; HCl: hydrochloric acid; HPLC: high performance liquid chromatography; LCMS: liquid chromatography mass spectrometry; LD: lithium diisopropylamide, - M +: molecular ion; m: multiplet; MeCN: acetamicil; M: mass spectrometry; MW: molecular weight; NMR: nuclear magnetic resonance; q: quartet; s: singlet; : triplet; tB: retention time; TFA: trifloracetic acid; THF: tetrahydrofuran; Detailed procedure for the synthesis of Deshi drofeni lahistina l-Acetyl-3 -. { (-l- [5- (1, 1-dimethyl-2-propenyl) -1H-4-imidazolyl] methylidene].] -2,5-piperazinedione (2)
[0361] To a solution of 5- (1, l-dimethyl-2-propenyl) imidazole-4-carboxaldehyde (100 mg, 0.609 mmol) in DMF (2 mL) is added compound 1 (241 mg, 1.22 mmol) and the solution was repeatedly evacuated in a short time to remove the oxygen and flushed with Ar, followed by the addition of CsC03 (198 mg, 0.609 mmol) and the evacuation-flushing process is repeated again. The oxygen removal of pressure because this removal is achieved that decreases the oxidation of the alpha carbon in position 6 of the diketopiperazine ring. The resulting mixture is stirred for 5 hours at room temperature. After the solvent is removed by evaporation the acid is dissolved in a mixture of EtOAc and 10% Na 2 CO 3; and the organic phase is washed with 10% Na 2 CO 3 again and NaCl saturated three times, dried over Na 2 CO 3 and concentrated in vacuo. The residual oil is purified by column chromatography on silica using CHCl3-MeOH (100: 0 to 50: 1) as an eluent to give 60 mg (33%) as a pale yellow solid 2. Dehydrophenylahistin [0362] To a solution of 2 (30 mg, 0.099 mmol) in DMF (0.8 mL), benzaldehyde (51 μL, 0.496 mmol, 5 eq) was added and the solution was repeatedly evacuated in a short time to remove the oxygen and flushed with Ar, followed by by the addition of Cs2CO3 (53 mg, 0.149 mmol, 1.5eq) and the process of evacuation-washing by dragging was repeated again. The resulting mixture is heated for 2.5 h at 80 degrees C. (The temperature must be increased slowly, a rapid heating increases the production of E isomer in the benzylidene portion). After the solvent was removed by evaporation, the residue was dissolved in EtOAc, washed with water twice and saturated NaCl three times, dried over NaSO 4 and concentrated in vacuo. On TLC using CHCl3-MeOH (10: 1), a bright yellow-green luminescence spot can be observed at 365 nm UV. The purity of this crude product was greater than 75% from the HPLC analysis. The resulting residue was dissolved in 90% aq MeOH and applied to a reverse phase HPLC column (YMC-Pack, ODS-AM, 20 x 250 mm) and eluted using a linear gradient from 70 to 74% MeOH in water during 16 min at a flow rate of 12 mL / min, and the desired fraction was collected and concentrated by evaporation to give 19.7 mg (60%), although the yields are not optimized for each stage, of yellow dehydrophenylahistin. EXAMPLE 4 Biological Characteristics of Dehydrophenylahistin and Dehydrophenylahistin Analogs A. Biological Evaluation [0363] The biological characteristics of synthesized tBu-dehydrophenylahistin and dehydrophenylahistin are evaluated in both HT29 human colon cells and PC-3 prostatic adenocarcinoma cells. [0364] HT-29 (ATCC HTB-38) a human colorectal adenocarcinoma is maintained in McCoy's complete medium (McCoy 5A medium with L-glutamine and 25mM HEPES supplemented with 10% FBS, Na lmM pyruvate, NEAA IX, L -glutamine 2 mM, and Pen / Strep at lOOIU / ml and 100 μg / ml, respectively). PC-3 (ATCC CRL-1435), a human prostate adenocarcinoma, is maintained in complete F12K medium (F12K medium supplemented with 10% FBS, 2mM glutamine, 1% HEPES, and Pen / Strep at 100 IU / ml and 100 μg / ml, respectively). Cell lines were cultured at 37 degrees C, C02 at 5% in an incubator and humidified at 95%. [0365] For tumor cytotoxicity assays HT-29 or PC-3 cells were seeded at 5,000 cells / well in 90 μl of complete medium in a Corning 3904 black-bottomed clear-bottom tissue culture plate and the plate was incubated overnight to allow the cells to settle and enter log phase growth. Solutions of 20 mM material of dehydrophenylahistin and tBu-dehydrophenylahistin were prepared in 100% DMSO and stored at -20 degrees C. Concentrated 10X serial dilutions of the two compounds were prepared in appropriate culture medium for final concentrations in the range of 2.0 x 10"5 M to 2.0 x 10" xo M. Volumes of ten μl of the 10X serial dilutions were added to the triplicate test wells and the plates returned to the incubator for 48 hours. The final concentration of DMSO was 0.25% in all samples.
[0366] After 48 hours of exposure to the drug, 10 μl of 0.2 mg / ml resazurin (obtained from Sigma-Aldrich Chemical Co.) in Mg2 + free PBS, Ca2 + is added to each well and the plates are return to the incubator for 3 to 4 hours. Plates were removed and resazurin fluorescence was measured using 530 nm excitation and 590 nm emission filters in a fusion fluorometer (Packard Instruments). The resazurin dye without cells was used to determine the background, which was subtracted from the data for all the experimental wells. The data was analyzed using the Prism software (GraphPad Software). Data were normalized to the average of cells treated with medium alone (100% of cell growth) and EC50 values were determined using an algorithm for standard sigmoidal dose-response curve fitting. [0367] As indicated in Table 1 below, tBu-dehydrophenylahistin demonstrates approximately four times greater cytotoxic activity compared to Table 1. Cytotoxic effect of dehydrophenylahistin and derivative.
Dehydrofenilahistine tBu-dehidrofenilahistina EC50 (nM) Cell? PLH tBu-? PHL HT29 48 13 PC-3 5. 4 1. 0 [0368] See also Figure 41 for additional data of HT-29, PC-3, and P-388 cells. B. Structure and Activity Study of Dehydrophenylahistin Derivatives [0369] The cytotoxic effects of phenylahistine, dehydrophenylahistin and various dehydrophenylahistin derivatives were examined in murine P388 leukemia cells, HT-29 human colon cells and PC-3 prostatic adenocarcinoma cells. . [0370] As explained above, HT-29 a human colorectal adenocarcinoma is maintained in McCoy's complete medium (McCoy's 5A medium with L-glutamine and 25mM HEPES supplemented with 10% FBS, Na lmM pyruvate, NEAA IX, 2mM L-glutamine, and Pen / Strep at lOOIU / ml and 100g / ml, respectively). PC-3, a human prostate adenocarcinoma is maintained in complete medium F12K (F12K medium supplemented with 10% FBS, 2mM Glutamine, 1% HEPES, and Pen / Strep at 100μg / ml and 100μg / ml, respectively). Cell lines were cultured at 37 degrees C, 5% C02 in a 95% humidified incubator. [0371] For tumor cytotoxicity assays, HT-29 or PC-3 cells were seeded at 5,000 cells / well in 90 μl of complete medium in light-bottom tissue culture plates, with Corning 3904 black wall and plates they were incubated overnight, to allow the cells to settle and enter log phase growth. Solutions of 20 mM material of dehydrophenylahistin and tBu-dehydrophenylahistin were prepared in 100% DMSO and stored at -20 degrees C. Serum 10X concentrated dilutions of the two compounds were prepared in appropriate culture medium for final concentrations in the range of 2.0 x 10"5 M to 2.0 x 10" xo M. Volumes of 10 μl of the 10X serial dilutions were added to the triplicate test wells and the plates returned to the incubator for 48 hours. The final concentration of DMSO was 0.25% in all samples. [0372] After 48 hours of drug exposure 10 μl of 0.2 mg / ml resazurin (obtained from Sigma-Aldrich Chemical Co.) in Mg2 + -free PBS, Ca2 + is added to each well and the plates are returned to the incubator for 3-4 hours. Plates were removed and resazurin fluorescence was measured using 530 nm excitation filters and 590 nm emission filters in a Fusion fluorometer (Packard Instruments). Resazurin dye without cells was used to determine the background, which was subtracted from the data for all the experimental wells. The data was analyzed using the Prism program (GraphPad Software). The data were normalized to the average of cells treated with medium only (100% cell growth) and EC50 values were determined, using a standard sigmoidal dose-response curve fitting algorithm. [0373] EC50 and IC50 values of phenylahistine, dehydrophenylahistin and dehydrophenylahistin derivatives are summarized in Table 2 below. Table 2. SAR study of phenylarthiin or dehydrophenylahistin and dehydrophenylahistin derivatives
[0374] Modifications to the phenyl ring have a significant effect on cytotoxic activities. Compared to the activity of tBu-dehydrophenylahistin (# 6), the activity of the methoxy group in the meta position (KPU-9) exhibited the highest activity as the other derivatives with an IC50 of 20.8 + 3.3 nM in P388 cells. The KPU-9 derivative also exhibits cytotoxicity in HT-29 cells (EC50 31nM). Dehydrophenylahistin, tBu-dehydrophenylahistin (KPU-2) and the KPU-9 derivative all exhibit cytotoxicity in P388 cells. C. Structure and Activity Study of Additional Dehydrophenylathystein Derivatives [0375] The cytotoxic effects of phenylarthiin, dehydrophenylahistin, and various additional dehydrophenylahistin derivatives were examined in HT-29 human colon cells and PC-3 prostatic adenocarcinoma cells using the methodology described above.
Table 3. SAR SAR study of phenylarthin, dehydrophenylahistin and additional dehydrophenylahistin derivatives
[0376] Additional cytotoxicity assay were performed as described above under this example, using Resazurin fluorescence as an indicator of cell viability. The results are shown below in Table 3.1. Table 3.1. Study of phenylarthiin, dehydrophenylahistin and additional dehydrophenylahistin derivatives (Continuation) EXAMPLE 5 Other analogues of dehydrophenylahistin A. Modifications for the synthesis of dehydrophenylahistin derivatives [0377] Other derivatives of dehydrophenylahistin are synthesized using the above techniques alone or in conjunction with other well known organic synthesis techniques. [0378] Modifications to the diacyldicetopiperazine and the first and second aldehydes involved in the synthesis method, vary according to the desired derivative to be produced. The derivatives that are synthesized: A) modify the phenyl ring and / or introduce other aromatic ring systems, B) alter the position of the aromatic ring, C) alter the imidazole aromatic ring system, and / or D) modify position 5 on the imidazole ring. [0379] The following figure illustrates modified regions of the dehydrophenylahistin compound to produce dehydrophenylahistin derivatives. Non-limiting examples of modifications are described and based on this description, it will be understood by those skilled in the art.
A 1) Modification of the phenyl ring based on the structure of known anti-tubulin compounds, Alkyl, Halogen, Alkoxy, Acetyl, Sulfonamide, Amino, Hydroxyl, Nitro, etc.
Colchicine introduction of other aromatic ring systems B Position of the aromatic ring C Change to the other ring system D Greater modification of position 5 in the imidazole ring
[0297] The expansion in the above modifications to the dehydrophenylahistin compound, derivatives of the compound can include the following substitutions in the phenyl ring (A): -CF3, -S02NH2 (-S02NR2R2), -S03H, -CONH2 (-CONR? R2) , -COOH, etc. Other ring systems (C) may also include the following: B. Examples of synthesized dehydrophenylahistin derivatives [0380] Additional examples of synthesized dehydrophenylahistin derivatives are described in Table 4. Table 4. Additional synthesized derivatives of dehydrophenylahistin C. Evaluation of dehydrophenylahistin derivatives [0381] The evaluation of derivatives described above is estimated according to the methods described in Example 3. Additional evaluation of the derivatives, extends to specific activities such as determining the inhibitory effect on cell proliferation, the effects in a specific cellular mechanism (ie microtubular function), effects in advance of the cell cycle, evaluation of anti-tumor activity in vitro against cancer cell lines, etc. Some protocols of the evaluation method are given below. 1) Inhibitory Effect of Cellular Proliferation of Dehydrophenylahistin and its Analogs [0382] In each well of a 96-well microtiter plate, 100 μl of human lung cancer derived A-549 cells prepared at 10 5 cells / ml are placed in a culture medium obtained by adding 10% bovine fetus serum to EMEM culture medium (Nissui Seiyaku Co., Ltd. ) that has an antitumor effect against A-549 cells derived from human lung cancer. Methanol solution of the derivative obtained by the aforementioned examples are added to the wells of the uppermost row, the specimens are diluted by the semi-log dilution method and added, and the plate is incubated in a carbon dioxide gas incubator at 37 degrees C for 48 hours. The result is added in batches of 10 μl with MTT reagent (3- (4,5-dimethyl-2-thiazole) -2,5-diphenyl-2H-tetrabromide) (1 mg / ml • PBS), followed by incubation in an incubator with carbon oxide gas at 37 degrees C for 6 hours. The culture medium is discarded and the crystal produced in the cells is dissolved in 100 μl / well of dimethisulfoxide. Light absorption of 595 nm is measured with a microplate reader. By comparing the light absorptions of the untreated cells with those of the cells treated with a specimen of a known concentration, the specimen concentration that inhibits 50% cell proliferation (IC50) is calculated. 2) Cell Cycle Inhibitory Activity of Dehydrophenylahistin and Its Analogues [0383] Cell strain A431 is derived from human lung cancer. EMEM culture medium containing 10% fetal bovine serum and 1% MEM non-essential amino acid solution (SIGMA M2025), is used to incubate A431 cells at 37 degrees C in an incubator saturated with carbon dioxide gas. 5% and water vapor. The refined specimen of dehydrophenylahistine obtained by the above methods is added to the cells in the log growth phase and the advance of the cell cycle is analyzed by flow cytometer and microscope observations. [0384] The effect on cell cycle advancement of HeLa cells is illustrated in Figure 42. EXAMPLE 6 Structure-Activity Relationship of Synthesized Dehydrophenylahistin Derivatives (DehydroPLH) 1) Generality in synthesis of derivatives [0385] Many, although not all the dehydroPLH derivatives described herein include one, two or three modifications in the phenyl ring (Figure 5 below). The derivatives were synthesized by the methods described above. As illustrated in Table 5, certain compounds showed more potent cytotoxic activity than dehydroPLH and tBu-dehydroPLH. The most potent compound exhibiting an EC50 value of 3 nM was KPU-90. This value was 16-fold and 4-fold higher than that of dehydroPLH and tBu-dehydroPLH, respectively. These derivatives have mono-substitution at the -o or -m positions of the phenyl ring with the halogen atoms such as fluorine and chlorine atoms or the methyl, vinyl or methoxy group. Derivatives with substitutions to heteroaryl structures such as naphthalene, thiophene and furan rings also produce potent activity. KPU-35, 42, 69, 80 and 81 also showed superior activity than tBu-dehydroPLH. Table 5. Potent synthetic dehydroPLH derivatives 2) Introduction of methoxy groups to the phenyl ring [0386] Colchicine recognizes the same binding site in beta-tubulin as PLH. Colchicine has four characteristic methoxy groups in its rings A and B. A series of substitutions with single or multiple methoxy groups was performed and the results of cytotoxic activity are illustrated in Table 6. Table 6. Methoxy group substitution effect in the proliferation of HT-29 cells
[0387] The result showed that substitutions in the -m or -o positions increase the cytotoxic activity against HT-29 cells. PU-9 and 16 showed high activity. Methoxy derivatives with triple substitution (KPU-11, 17 and 45) also showed activity. The structure of KPU-24 was assigned by MASS analysis. 3) Modification with groups that withdraw electrons [0388] To study the most expanded structure activity relationship in the phenyl ring, a series of different functional groups is introduced, which include both groups that withdraw and donate electrons. The result of cytotoxicity against HT-29 cells is shown in Tables 7 and 8, respectively. [0389] Substitution in the -o or -m positions effectively increased the activity. These results were very consistent with the methoxy group case. Table 7. Effect of the group that retains electrons in HT-29 cell proliferation Table 8. Effect of the electron donor group on the proliferation of HT-29 cells
[0390] The present disclosure is not bound by or limited by any particular scientific theory. However, it is appreciated that people skilled in the art can interpret the results presented here, to suggest that a relatively smaller functional group, which affects less steric hindrance, may be preferred to produce more potent activity, and slightly larger groups such as the ethoxy group (when compared to the methoxy group) or the Br atom (when compared to the Cl atom) can affect unfavorable steric hindrance for interaction with, for example the tubulin binding site. Furthermore, because the electrical property of these substituents does not affect the activity, it is suggested that these relatively small substituents do not directly interact with the beta-tubulin binding site, but rather that it restricts the conformation of dehydroPLH suitable for the bond or Union. 0, as another possible hypothesis, the hydrophobic property may be a more important factor in the binding site for the -o-m positions in beta-tubulin, since the introduction of the hydrophilic hydroxyl group, which can form the hydrogen bond As a hydrogen donor, activity decreases drastically. [0391] As shown in Table 9, the effect of the substituents on the cytotoxic activity at the -o position can be ordered, as in the case of the -m position, as shown in Table 10. The compounds having Effective functional groups, which show superior activity than tBu-dehydroPLH, can also be further modified. Since the migration of the stereochemistry from Z to E under the irradiation of visible light is observed, substituents that decrease the electron density in the conjugated double bonds can contribute to the reduction of migration Z to E by light, resulting in more physicochemically stable structures. The temperature can also affect this migration. [0392] Modification in two parts of the ring may be preferred for the development of potent but also biologically stable compounds. The phenyl ring of phenylahistine is oxidized by cytochrome P-450. The double modification that reduces the electron density of the phenyl ring can therefore be effective to prevent P-450 oxidation. In this way, the combination of the small group that removes electrons such as the fluorine atom to the element that can increase activity such as -OMe, -Me, -Cl, -F and Br, can result in more potent drug compounds and biologically stable. Table 9. Summary of change in position -o Table 10 Modification summary in the m-position 4) Substitution of the phenyl ring in aryl heterocycles [0393] The phenyl ring can also be replaced by heteroaryl groups. The result of these replacements in terms of the cytotoxic activity is illustrated in Table 11. Since the aryl nitrogen atoms can form a hydrogen bond with an NH group of the diketopiperazine ring and restrict the conformation of the molecule between pyridine and diketopiperazine rings to a uniplanar structure, the active dehydroPLH conformation will require a certain level of dihedral angle formed by the steric repulsion between an amide hydrogen atom of the diketopiperazine ring and a hydrogen atom or phenyl ring (Figure 6). Table 11. Effect of replacement with the heteroaryl ring on HT-29 cell proliferation
[0394] Replacing the phenyl ring with a smaller furan or thiophene group, for example KPU-29 or -42, exhibits activity. The phenyl ring can be changed to another aromatic structure, while the potent activity is maintained. 5) Phenylarthidine metabolism [0395] In the recent study, (+) - phenylahistine was treated with hepatic microsome of rat or human liver P450s. In the human case, at least seven metabolites were detected, and two of them, namely Pl and P3, were major metabolites, representing more than 60% of the metabolites recovered.
[0396] Since there is no exo-olefin structure in tBu-dehydroPLH, synthesized derivatives present do not have oxidation such as Pl and P4. However, oxidations such as P3 and P5 are formed during hepatic metabolism. Several derivatives, which avoid this metabolism, are effective to prevent P450 oxidation in the phenyl ring. The imidazole ring can also be modified to avoid unfavorable oxidation. 6) Physical-chemical stability of dehydroPLH [0397] Physical-chemical stability is one of the unfavorable problems of dehydroPLH. In phenylahistine, since it does not structure additional olefin in the benzyl part, there is no such problem. However, in dehydroPLH, the benzylidene portion can be easily activated, probably with visible light, and migration Z to E frequently occurs due to the existence of longer conjugation of the double bond. The migration occurred even under normal room light. In the cytotoxic assay, some of the compounds migrate to E-form during incubation, although this migration is probably balanced in the 1: 1 ratio in the case of dehydroPLH. This migration can be controlled. Migration Z to E is also known in combretastatin A4, the same type of tubulin inhibitor, and a few studies to improve this problem were reported. 7) Prodrug Synthesis [0398] Form E can also be used as a dehydroPLH prodrug or one or more of these analogs, including those analogs described herein. One of the undesirable properties of anti-tubulin drugs involves their low selectivity between tumor and intact tissues, although these drugs belong to one of the molecular target or target therapies. This causes undesirable side effects. However, if the compound works selectively only in tumor tissues, negative side effects of anti-microtubule drugs can be reduced. Since the dehydroPLH (form Z) can be produced from its E isomer by irradiation with visible light, form E is administered and photo irradiation is performed only at the tumor site, then only the tumor is damaged by the Z-shape photo -produced and the adverse effect on intact tissues, is reduced. [0399] Form E can be chemically protected by the addition of a bulky but biodegradable acyl group, which is introduced into the diketopiperazine ring as a prodrug. This acyl group can be cleaved by the protease in the body. Therefore, the acylated compound E is maintained before administration, after administration it is changed to the real E form, which can migrate to the bioactive Z form by local irradiation photo. [0400] The synthetic scheme of this acyl-E form of tBu-dehydroPLH is summarized in Figure 9. EXAMPLE 7 Pharmaceutical Formulations of the Synthesized Dehydrophenylahines 1) Formulations Administered Intravenously, by Dripping, Injection, Infusion or Similar [0401] A Flasks containing 5 g of glucose powder, 10 mg of a compound synthesized by the method are added to each aseptically and sealed. After being charged with nitrogen, helium or other inert gas, the bottles are stored in a cold, dark place. Before use, the contents are dissolved in ethanol and added to 100 ml of a 0.85% physiological salt water solution. The resulting solution is administered as a method to inhibit the growth of a cancerous tumor in a human who is diagnosed to have this tumor between about 10 ml / day to about 1000 ml / day, intravenously, by drip or by a subcutaneous or intraperitoneal injection, as deemed appropriate by those of ordinary skill in the art. 2) Formulation for Orally Or Similar Administration [0402] A mixture obtained by complete mixing of 1 g of a compound synthesized by the method, 98 g of lactose and 1 g of hydroxypropyl cellulose, is formed into granules by any conventional method. The granules are dried and lowered completely to obtain a preparation of granules suitable for packaging in bottles or by heat sealing. The resultant granule preparations are administered orally between about 100 ml / day to about 1000 ml / day, depending on the symptoms, as judged appropriate by those of ordinary skill in the art to treat cancerous tumors in humans. 3) Formulation for Topical Administration [0403] Administration to an individual of an effective amount of the compound can also be achieved topically by administering the compound or compounds directly to the affected area of the individual's skin. For this purpose, the administered or applied compound is in the form of a composition that includes a pharmacologically acceptable topical carrier, such as a gel, an ointment, a lotion or a cream, including without limitation, carriers such as water, glycerol, alcohol, propylene glycol, fatty alcohols, triglycerides, fatty acid esters or mineral oils. Other topical carriers include liquid petroleum, isopropyl palmitate, polyethylene glycol, ethanol (95%), polyoxyethylene monolaurate (5%) in water, or sodium lauryl sulfate (5%) in water. Other materials such as anti-oxidants, humectants, viscosity stabilizers and similar agents can be added as needed. Percutaneous penetration enhancers such as Azone can also be included. In addition, in certain instances, it is expected that the compound can be placed within devices arranged in, on or under the skin. These devices include patches, implants and injections that release the compound in the skin, either by passive or active release mechanisms. EXAMPLE 8 Pharmacology In vi tro of KPU-2, KPU-35 and t-butyl phenylahistine [0404] In vitro efficacy studies performed with KPU-2, KPU-35 and t-butyl phenylahistine include: A) a panel of six tumor cell lines, B) studies on tumor cells resistant to multiple drugs, and C) studies to determine the mechanism of action.
TO) . Study of KPU-2, KPU-35 and t-butyl phenylahistine in a panel of six tumor cell lines [0405] The following cell lines (source in parentheses) were used: HT29 (human colon tumor; ATCC; HTB- 38), PC3 (human prostate tumor, ATCC, CRL-1435), MDA-MB-231 (human breast tumor, ATCC, HTB-26), NCI-H292 (human non-small cell lung tumor, -ATCC; CRL-1848), OVCAR-3 (human ovarian tumor; ATCC; HTB-161), B16-F10 (murine melanoma; ATCC; CRL-6475) and CCD-27sk (normal human fibroblasts; ATCC; CRL-1475) . Cells were maintained at sub-confluent densities in their respective culture media. [0406] Cytotoxicity assays were performed as described above in Example 4, using Resazurin fluorescence as an indicator of cell viability. [0407] The disclosed compounds are effective agents against a variety of different and distinct tumor cell lines. Specifically, for example, KPU-2 and KPU-35 were more effective in the HT-29 tumor cell line, both in terms of potency (active in the low nanomolar range) and efficacy (of greater response in terms of maximum cytotoxic effect).; t-butyl-phenylahistine exhibits its greatest potency against the PC-3 tumor cell line, although the greatest efficacy was exhibited against the HT-29 cell line; KPU-2 and KPU-35 in general were 10-40 times more potent than t-butyl-phenylahistine while the efficacy was similar for all three compounds in the different tumor cell lines; the tumor cell lines HT-29, PC-3, MDA-MB-231 and NCI-H292 all respond similarly to the NPI compounds, while B16-F10 appears to be somewhat less sensitive. T-butyl-phenylahistine exhibited a marked difference between normal fibroblasts and tumor cell lines, with a ratio in the range of > 20 - > 100, except for the OVCAR-3 cell line. Table 12 Activity of KPU-2, KPU-35 and t-butol phenylarthin in Tumor Panel Monitoring (CONTINUATION) B). Drug-resistant Cell Lines [0408] One of the main challenges in the use of chemotherapeutic agents in clinical oncology is the development of resistance to the effect of the drug by the tumor cells. There are several mechanisms for the development of resistance, each of which will have differential effects on chemotherapeutic drugs. These mechanisms include increased expression of ATP-dependent evacuation pumps such as the P-glycoprotein encoded by MDR1 or the multi-drug resistance associated with protein 1 encoded by MRP1. Reduced drug absorption, alteration of the drug target, increased repair of drug-induced DNA damage, alteration of the apoptotic pathway and activation of cytochrome P450 enzymes are other examples of mechanisms by which cancer cells become resistant to anti-cancer drugs. The selected compounds were studied in three different cell lines that exhibit two different resistance mechanisms; the over-expression of P-glycoprotein and altered topoisomerase II activity. 1) Pair of Tumor Cell Lines of Human Uterine Sarcoma: MES-SA (Taxol Sensitive) and MES-SA / Dx5 (Taxol Resistant). [0409] This cell line expresses elevated mdr-lmRNA and P-glycoprotein (an extrusion pump mechanism). Pretreatment with cyclosporin-A (CsA) blocks P-glycoprotein and reinstates activity in the resistant cell line by those compounds for which resistance is due to elevated P-glycoprotein. [0410] As can be seen in Table 13, KPU-2, and KPU-35 have the same potency in the resistant cell line as in the sensitive line and the potency of t-butl-phenylahistine was only slightly reduced. Pretreatment with cyclosporin A (CsA) does not alter the potency of the selected compounds. In contrast, taxol was virtually inactive in the cell line resistant to MES-SA / DX5, whereas this compound was very potent in the line of sensitive cells. Treatment with CsA restored the sensitivity to taxol of the MES-SA / DX5 cell line. The MESSA / DX5 cell line also showed reduced susceptibility to etoposide (60 times), doxorubicin (34 times) and mitoxantrone (20 times). [0411] These data indicate that the effects of KPU-2, KPU-35 and t-butyl-phenylahistine are not susceptible to the mechanism of resistance related to taxol (p-glycoprotein) in this cell line and there is no cross-resistance of taxol to these select compounds in this model. Table 13 Activity of KPU-2 KPU-35, t-butyl-phenylahistine and Taxol in Tumor Cell Lines of Human Uterine Sarcoma Resistant to Taxol MES-SA / DX5 Taxol and Sensitive to Taxol MES-SA
[0412] See also the additional data presented in Figure 43. 2) Pair of Cell Lines of Human Acute Promyelocytic Leukemia: HL-60 (Mitoxantrone-sensitive) and HL-60 / MX-2 (Mitoxantrone-Resistant) [0413] This cell line is considered to have atypical drug resistance properties, with altered topoisomerase II catalytic activity without over-expression of P-glycoprotein. [0414] As can be seen in Table 14, these results indicate that the potencies of the novel novel compounds are very similar in sensitive and resistant HL-60 cell lines. In contrast, Mitoxantrone loses efficacy by a factor of 24-fold in the cell line resistant to HL-60 / MX-2. [0415] Thus, KPU-2, KPU-35 and t-butyl-phenylahistine are not susceptible to the same resistance mechanisms as Mitoxantrone in this cell line, and there is no cross-resistance of Mitoxantrone to these select novel compounds in this model. Table 14. Activity of KPU-2, _ KPTJ-35 ^ t-butyl-phenylahistine and Mitoxantrone in the Pair of Lines Resistant and Sensitive to Leukemia Tumors Promyelocytic Human Aging HL-60 Compound HL-60 HL-60 Resistant Sensitive 3) . Pair of Human Chest Carcinoma Cell Line: MCF-7 (Taxol Sensitive) and MCF-7 / ADR (Taxol Resistant) [0416] This study involves KPU-2 compared to taxol. KPU-2 demonstrated similar potencies in both sensitive and resistant members of this pair of cell lines. In contrast, taxol was virtually inactive in the resistant cell line while there was low nanomolar potency in the sensitive cell line (Table 15). [0417] These studies confirm in a different human tumor cell line that resistance to taxol is not transferred to KPU-2. Table 15. Activity of KPU-2 and Taxol in the Pair of Cell Lines Resistant and Sensitive to Human Breast Carcinoma MCF-7 Composite MCF-7 Sensitive MCF-7 / ADR Resistant C) Studies of the Mechanism of Action 1). Action on Microtubule Function [0418] Human umbilical vein endothelial cells (HuVEC of Cambrex) were used in this study to evaluate the effects of KPU-2 and t-butyl-phenylahistine compared to colchicine and taxol on tubulin by staining by alpha-tubulin. [0419] Thirty minutes of exposure to KPU-2, t-butl-phenylahistine or colchicine (all at 2 μM) induce microtubule depolymerization as indicated by the lack of intact microtubule structure in contrast to that observed in the DMSO control and formation of cell membrane blebs (a clear indication of apoptosis) in HuVEC cells, while taxol does not induce microtubule depolymerization under these conditions. Colchicine is a known microtubule depolymerizing agent while taxol is a tubulin stabilizing agent. Similar results are obtained when CCD-27sk cells are exposed to KPU-2 or colchicine. 2) . Induction of Apoptosis [0420] Apoptosis and its de-regulation play an important role in oncology; The selective induction of the programmed cell death cycle in tumor cells is the goal of many programs for the discovery of chemotherapeutic drugs. This induction of apoptosis can be demonstrated by different methods, including blistering on characteristic cell membrane, DNA fragmentation, hyperphosphorylation of the antiapoptotic factor Bel-2, activation of the caspase cascade and cleavage of poly (ADP ribose) polymerase (PARP). [0421] Characteristic signs of apoptotic cell death include cell membrane blister formation, rupture of nuclei, cell shrinkage and condensation and finally cell death, very distinctive of necrotic cell death. KPU-2 induces the typical morphological changes associated with the early stages of apoptosis in human prostate tumor cells. A similar finding was also clear in the treatment of HuVEC cells with KPU-2. 3) . DNA Fragmentation [0422] A later stage characteristic of apoptosis is excision of internucleosomal DNA that results in a distinctive ladder pattern that can be visualized by gel electrophoresis. This approach was used to study the effect of KPU-2 on scale formation of DNA (DNA) in Jurkat cells (human T cell leukemia line) compared to halimide and dehydrophenylahistin (KPU-1). Formulation of DNA ladder induced by KPU-2 at the 1 nM concentration while halimide and KPU-1 were much less potent. 4) . Activation of the Caspasa Cascade [0423] Several enzymes in the caspase cascade are activated during apoptosis, including Caspase-3, -8 and -9. The activity of Caspase-3 is monitored in Jurkat cells after treatment with KPU-2, KPU-35 and t-butyl-phenylahistine. [0424] The results indicate that caspase-3 is activated in a dose-dependent manner by treatment with all three compounds in a manner similar to halimide. Activation of caspase-3 occurred over a similar concentration range for IC50s of cytotoxicity in the Jurkat cell line (Table 16). Table 16 Cytotoxicity of KUP-2, KUP-35 and t-butyl-phenylahistine in Jurkat Cells ) . Cleavage of Poly (ADP-ribose) (PARP) Polymerase in Jurkat Cells [0425] To estimate the ability of these compounds to induce apoptosis in Jurkat cells, the cleavage of poly (ADP-ribose) polymerase was monitored (PARP). PARP is a 116 kDa nuclear protein that is one of the main targets or intracellular targets of Caspase-3. The cleavage of PARP generates a stable product of 89 kDa, and this process can be easily monitored by western technique. The cleavage of PARP by caspases is one of the hallmarks of apoptosis, and as such it serves as an excellent marker for this process. KPU-2 at 100 nM induces cleavage of PARP in Jurkat cells, 10 hours after exposure of the cells to the compound. KPU-2 seems to be more active than either halimide or KPU-1. 6). Enhanced Vascular Permeability in HuVEC Cells [0426] Compounds that depolymerize microtubules (ie combretastatin A-4-phosphate, ZD6126) have been shown to induce vascular collapse in tumors in vivo. This vascular collapse is preceded by a rapid induction of permeability of vascular cells initially to electrolytes and shortly thereafter to large molecules. The improved permeability of HuVEC cells to a fluorescent labeled dextran is used as a substitute assay for vascular collapse. [0427] KPU-2, KPU-35 and t-butyl-phenylahistine all rapidly (within 1 hour) induce significant permeability of tHuVEC monolayer, in a ratio similar to colchicine. The taxol microtubule stabilizing agent was inactive in this assay (Figure 12). 7). Profile on a Large Kinase Screen [0428] KPU-2 was initially monitored at a concentration of 10 μM in a panel of 60 different kinases; the concentration of ATP was 10 μM. Four kinases were inhibited by more than 50% in the primary screening and the IC50 were determined in the secondary screening are presented in Table 17. All IC50 values are in the low micromolar range, indicating that the inhibition of these kinases is not relate to the low nanomolar activities observed by cytotoxicity of tumor cells. Table 17. Activity of KPU-2 against Select Kinases EXAMPLE 9 In Vivo Pharmacology [0429] Preliminary studies with KPU-2 were performed using the semi-graft models of MX-1 (breast) and HT-29 (colon) and the murine leukemia tumor model P-388, in the mouse. Other tumor models selected based on activity in the tumor panel in vi tro were the cell lines DU-145 (prostate), MCF-7 (breast), and A549 (lung). Human pancreatic tumor (MiaPaCa-2) was also included. The novel compounds were studied as monotherapy and in combination with a clinically employed chemotherapeutic agent. The dose of the selected novel compounds was determined from the acute tolerability test (Maximum Tolerated Dose (MTD)) and adjusted as necessary during each study. The doses of chemotherapeutic agents used clinically were selected based on historical studies. [0430] KPU-2 was the first compound to be studied in these five tumor models. Following the initial results of this study, all three compounds were compared in xenograft models HT-29 of human colon tumor, DU-145 of human prostate and MCF-7 of human breast tumor. [0431] The above models all use the subcutaneous xenograft implant technique and all are potentially subject to selective effects of a compound in the subcutaneous vasculature that produces an amplified (or apparent) anti-tumor activity. In order to overcome this possibility, two other tumor models have been incorporated into the investigation. One of these is the observation of lung metastasis after intravenous injection of B16-F10 mouse melanoma tumor cells. The other model is the implant of human breast tumor cells MDA-231 in the mouse mammary fat panniculus. While this latter model is a xenograft model, the subcutaneous vasculature does not play a role. Methods 1). Xenograft models [0432] The animals used were of (except as indicated for individual studies): nude female mice (nu / nu) between 5 and 6 weeks of age (~ 20g, Harian); Group size was 9-10 mice per group, unless indicated otherwise. [0433] The cell lines used for tumor implantation were: HT-29 human colon tumor; human breast tumor MCF-7; human non-small cell lung tumor A549; human pancreas tumor MiaPaCa-2; human prostate tumor DU-145. [0434] Selected novel compounds were administered as monotherapy by the intraperitoneal route (i.p.) at the doses indicated for the individual study; for the studies in combination, the selected reference chemotherapy agents were injected 15-30 min before the compound. [0435] The vehicles used in these studies were: 12.5% DMSO, 5% Cremaphor and 82.5% peanut oil for the selected novel compounds; (1: 3) Polysorbate 80: 13% ethanol for taxotere; (1: 1) Cremaphor: ethanol for paclitaxel; for irinotecan hydrochloride each ml of solution contains 20 mg of irinotecan hydrochloride, 45 mg of NF sorbitol powder, and 0.9 mg of lactic acid, the pH is adjusted to 7.4 with NaOH or HCl. Saline dilutions are used to achieve the injection concentrations used for the reference compounds. Human colon tumor model HT-29 [0436] Animals were subcutaneously implanted (s.c.) by trocaring with fragments of HT-29 tumors collected from growth tumors. c. in hosts nude mice. When the tumor size reached 5 mm x 5 mm (approximately 10-17 days) the animals were formed into treatment and control groups. Mice were weighed twice weekly and tumor measurements were obtained using gauge compasses weekly twice, starting from Day 1. Tumor measurements were converted to tumor weight in mg estimated using the formula (W2xL) / 2. When the estimated tumor weight of the control group reached an average of 1000 mg, the mice were weighed, sacrificed and the tumor was removed. The tumors were weighed and the average tumor weight per group was calculated and the inhibition of tumor growth (TGI = tumor growth inhibition) was determined for each group (100% minus the change in the average weight of the tumor treated / the change in the average control tumor weight x 100. [0437] In this model, unless otherwise noted for the individual study, the selected novel compounds were injected intraperitoneally every third day for 15 days [1, 4, 8, 11 and 15 (q3dx5)], - CPT-11 was administered intraperitoneally on days 1, 8 and 15 (qwx3) .Child breast tumor model MCF-7 [0438] Female naked mice (-20 g) were implanted sc with 21-day release estrogen granules (0.25 mg), 24 hours before sc implantation with MCF-7 tumor fragments (collected from sc tumors in nude mouse hosts) The study proceeded as described in model HT-29 , using taxotere as the standard chemotherapy agent. [0439] In this model, unless otherwise noted for individual study, the novel compounds were injected intraperitoneally daily, Days 1-5, inclusive (qdx5); Taxotere was administered intravenously on Days 1, 3 and 5 (qodx3). Human lung tumor model A549 [0440] In s.c. were implanted by trocar with A549 tumor fragments collected from s.c. in hosts of nude mice. When the tumor size reaches 5 mm x 5 mm (approximately 10-17 days) the animals are mated in control treatment groups. The rest of the study proceeded as described for the HT-29 model, using taxotere and CPT-11 as the standard chemotherapy agents. [0441] In this model, unless otherwise noted for the individual study, the tested compounds were administered by the intraperitoneal route in a dose schedule aq3dx5 for the CPT-11 combination or in a qdx5 dose regimen for the combination with taxotere; CPT-11 was administered by the intraperitoneal route in a qwx3 program, - taxotere was administered intravenously in a qodx3 dose regimen. Human pancreatic tumor model MiaPaCa-2 [0442] Animals were implanted s.c. by trocar fragments of MiaPaCa-2 tumors collected from s.c. in hosts of nude mice. When the tumor size reaches 5 mm x 5 mm (approximately 10-17 days) the animals were mated in control treatment groups. The rest of the study proceeded as described for the HT-29 model, using gemcitabine as the standard chemotherapy agent. [0443] In this model, unless otherwise noted for the individual study, the test compounds administered every third day by the intraperitoneal route on Days 1, 4, 7, 10 and 15 (q3dx5), - gemcitabine by the intraperitoneal route on Days 1, 4, 7 and 10 (q3dx4). Human prostate tumor model DU-145 [0444] Male mice were implanted s.c. by trocar with fragments of DU-145 tumors collected from s.c. in hosts of male nude mice. When the tumors reached ~ 5 mm x 5 mm (at about 13-17 days) the animals were mated in control treatment groups. The rest of the study proceeded as for the HT-29 model, using taxotere as the standard chemotherapy agent. [0445] In this model, unless otherwise noted for the individual study, the test compounds were administered via the intraperitoneal route on Days 1, 3, 5, 8 and 11 (q3dx5); Taxotere was administered intravenously on Days 1, 3 and 5 (q2dx3). 2) . Non-subcutaneous implant tumor models [0446] The animals used were: female nude mice (nu / nu) (study MDA-231) or mice B6D2F1 (studies B16-F10) between 5 and 6 weeks of age (~ 20g, Harían); the group size was 10 mice per group unless otherwise indicated.
[0447] The cell lines used were: human breast tumor MDA-MB-231 and murine melanoma cells B16-F10. [0448] NPI compounds were administered as monotherapy by the intraperitoneal route at the doses indicated for the individual study; for the combination studies, the selected reference chemotherapy agents were injected 15-30 min before the NPI compound. Human breast tumor MDA-231 [0449] Female nude mice were injected into the panicle or mammary fat pad with 2xl06 MDA-231 cells harvested from cell culture in vi tro. When the tumor size reached 5 mm x 5 mm (approximately 14-28 days), the animals were mated in control treatment groups. The study proceeded as described for the HT-29 model, using paclitaxel as the standard chemotherapy agent. [0450] In this model, unless otherwise noted for the individual study, the test compounds were administered by the intraperitoneal route on Days 1, 4, 8, 11 and 15 (q3dx5), -paclitaxel was administered by the intraperitoneal route on Days l-5 (qdx5). Metastatic Murine Melanoma Model B16-F10 [0451] Mice received B16-F10 cells (prepared from a cell culture of B16-F10 cells) by the iv route on Day 0. On Day 1, the mice were randomly distributed in control treatment groups and treatment was started. Mice were weighed twice weekly, starting on Day 1. All mice are sacrificed on Day 16, the lungs are removed, weighed and the surface colonies counted. The results are expressed as average treated mice / average colonies of control (T / C) x 100% mice. Inhibition of metastasis growth (MGI = metastasis growth inhibition) is the number subtracted from 100%. Paclitaxel was the standard chemotherapy agent used in this study. [0452] In this model, unless otherwise noted for the individual study, the test compounds are administered by the intraperitoneal route on Days 1-5 (qdx5), - paclitaxel was administered intravenously on Days 1-5 ( qdx5). [0453] When appropriate (n> 3), results are presented as averages ± SEM. Analysis of statistical studies with several groups are performed using ANOVA with the Neuman-Keuls post test, unless otherwise indicated. A test-t of a tail also it is used based on the hypothesis that the compound or drug or combination will reduce the growth of the tumor. Results Studies in the human colon tumor xenograft model HT-29 1. In Vivo Evaluation of KPU-2 +/- CPT-11 in the HT-29 human colon tumor xenograft model [0454] This study estimates changes in dose concentration and dose regimen for KPU-2 alone and in combination with a relevant chemotherapeutic CPT-11 in the HT-29 model. [0455] KPU-2 is administered at a dose of 7.5 mg / kg ip daily for five days (qdx5), 3.75 mg / kg ip bid for five days, 7.5 mg / kg ip every second day for 10 days (qodx5) and 7.5 mg / kg ip every third day for 15 days (q3dx5). The combination of CTP-11 with NPI-2358 at a dose of 7.5 mg / kg ip q3dx5 results in a significantly greater effect than for any compound alone that withstood the duration of the study (Figure 13). Those observations during the lifetime portion of the study duration were confirmed by the final tumor weights of the average group at autopsy, for which only the group in combination exhibited a statistically significantly lower tumor weight than the control ones. In addition, the difference between the average tumor weights of the combination therapy and the monotherapy groups CPT-11 was statistically significant (Figure 14). When the individual final tumor weights at autopsy are examined, the greatest effect of co-therapy is clear (Figure 14). TGl of co-therapy was 78% compared to 38.9% for CPT-11 alone. TGl for the combination therapy group exceeds the NCI criteria of 58% for a positive result. 2. Standard chemotherapy studies KPU-2 +/- against five models of human tumor xenoinj [0456] This study consists of five different branches, each with its own protocol, timing, dose regimen and reference compound. Each branch will be considered within the presentation of the particular tumor model. [0457] The objective of the HT-29 branch of the study was to investigate a slightly higher dose of KPU-2 (10 mg / kg ip q3dx5) in the HT-29 human colon tumor xenograft model compared to those employed in the study described above, where marked synergy was observed between KPU-2 (7.5 mg / kg ipq3dx5) and CPT-11 (100 mg / kg ipqwx3). [0458] As can be seen in Figure 15, the combination of KPU-2 and CPT-11 in this model results in a marked synergy in the inhibition of tumor growth, with tumor growth being almost completely inhibited until the day of treatment 29 in the combination therapy group. The combination therapy maintains efficacy and the estimated tumor growth for this group was significantly lower than for any monotherapy group. Accordingly, the administration of KPU-2 and CPT-11 inhibits tumor growth and is effective antitumor treatment. [0459] Observations of the portion in life of the study (estimated tumor growth, Figure 15) are supported by the measurement of the weights of the tumors cut at autopsy (Figure 16). The tumor weights for the combination group were significantly lower than the controls (p <; 0.01), as the tumor weights for CPT-11 alone (p <0.05). [0460] When the individual final tumor weights are considered (Fig 16), the tumor size for the combination group was generally smaller than for the other treatment or control groups. TGl of the combination group was 65.8%, indicating a positive effect by the NCI criterion, while monotherapy does not meet the NCI criterion of > 58% 3. Activity study of KPU-2, KPU-35 and t-butyl-phenylahistine in the HT-29 human colon tumor xenograft study [0461] The results of this study are presented in Figure 17 and Table 18 The combination therapy groups all indicate a marked synergy between the novel compounds and CPT-11. Individual tumor weights demonstrated the effectiveness of the co-therapy treatment (Figure 18). In each case, TGl for the combined group exceeds the NCI criterion for a positive effect, while TGl for the CPT-11 monotherapy does not reach this level. Table 18. Compendium of studies performed in the HT-29 human colon tumor model.
(CONTINUATION) * p < 0.05 vs Control; ** p < 0.01 vs Control; # p < 0.05 vs CPT-11 Solo; ## p < 0.01 vs CPT-11 Solo; + = Number of deaths 4. Compendium of the effects of KPU-2, KPU-35 and t-butyl-phenylahistine in combination with CPT-11 in the human colon tumor xenograft model HT-29 [0462] When combined with CPT-11, KPU-2 improves the effect of CPT-11, the standard chemotherapeutic agent, at a level that far exceeds the NCI criterion of TGl > 58% for a positive effect. The results generated in the three studies are very comparable both for life observations (Figure 19) and for the weights of tumors cut at autopsy (Figure 20). Studies in the human prostate tumor xenograft model DU-145 [0463] Two studies have been completed with this model: the first study involves copying alone and in combination with taxotere; The second study compared KPU-2, KPU-35 and t-butyl-phenylahistine alone and in combination with taxotere. 1. Effect of KPU-2 in Combination with Taxotere in the Human Prostate Tumor Xenograft Model DU-145 [0464] As can be seen from the data obtained during the life portion of this study (Fig. 21), the treatment The most effective human prostate tumor DU-145 was a combination therapy of KPU-2 plus taxotere. The effect of the treatment was more pronounced at the beginning of the study and seems to be reduced as the study progresses. Of the 20-27 treatment days, the combination therapy provides an apparent TGI that exceeds the NCI criterion (TGI> 58%), and the estimated tumor weight of the combination therapy was significantly lower than for any mono-therapy. 2. Activity in KPU-2 KPU-35 and t-butyl-phenylahistine alone in Combination with Taxotere in the Human Prostate Xenograft Model DU-145 [0465] Based on the data obtained with KPU-2 in combination with taxotere in the study described above, a second study comparing KPU-2 with KPU-35 and t-butyl-phenylahistine alone and in combination with taxotere was initiated. [0466] Observations made during the live portion of this study indicate that the combination of either KPU-2 or KPU-35 with taxotere has greater reduction in tumor growth than for taxotere alone (Figure 22). Tumor growth was almost completely blocked by KPU-35 in combination with taxotere. [0467] The weights of tumors cut at autopsy confirmed the observations made during the life segment of the study. The combination of either KPU-2 (Figure 23) or KPU-35 (Figure 24) with taxotere was significantly more effective than taxotere alone in blocking tumor growth. In the case of KPU-35, three of ten mice showed evidence for tumor shrinkage. The rates of inhibition of tumor growth indicate a marked inhibition of tumor growth for KPU-2 (average group = 74.1%) and an almost total blockage for KPU-35 (average group = 92.5%). Taxotere alone does not meet the criteria established by NCI for a positive effect (TGA> 58%). 5. Studies in the Human Breast Tumor Xenograft Model MCF-7 [0468] This study compares the effects of KPU-2, KPU-35 and t-butyl-phenylahistine in the human breast tumor xenograft model MCF- 7 Doses of the compounds were administered on days 1, 2, 3, 4, and 7; Taxotere is administered on days 1, 3 and 7. [0469] The selected novel compounds have early activation, statistically significant effects when used in combination with taxotere in that model, apparently almost completely blocks the estimated tumor growth.
(Figure 25). Of the three compounds, KPU-2 seems to be the most effective, with t-butyl-phenylahistine which also exhibits a significant potentiation of taxotere. 6. Studies in Lung Tumor Xenograft Model of Non-Human Human Cells A549 [0470] The observations in life during their study (Figure 26) indicate that the combination of KPU-2 (7.5 mg / kg ip, qdx5) with taxotere, resulted in a marked inhibition of tumor growth compared to the control of any group of monotherapies. This was confirmed by the autopsy tumor weights, since the average of the co-therapy group was significantly lower than that of taxotere alone or the control group (Figure 27). The tumor weights of the co-therapy group formed a swarm of low tumor weights, indicating the consistency of effect. [0471] When the tumor growth rate is calculated, the co-therapy group had a TGI of 74.4% compared to the excessively controlled control group of the NCI criterion for a positive effect (TGl 58%). Taxotere only had TGI of 26.1%. 7. Studies in Xenoinj Model Orthotopic Human Breast Tumor MDA-231 [0472] This model involves the placement of human tumor tissue in the panicle or mouse mammary fat pad, a substitute for the natural environment. In this way, the possibility of a positive effect due to a specific action in the subcutaneous vascular bed is avoided. This study compared the effect of KPU-2 (7.5 mg / kg ip, q3dx5) alone and in combination with paclitaxel (16 mg / kg ip, qdx5). [0473] At three weeks in the study there was a significant inhibition of tumor growth in the combination therapy group, a highly significant effect. This effect seems to be more marked than for taxotere alone (Figure 28). 8. Studies in the Metastatic Tumor Model B16 FIO of Murine Melanoma [0474] This study examined the effect of KPU-2, KPU-35 and t-butyl-phenylahistine alone and in combination with paclitaxel in the number of metastases that appear on the surface of the lung 16 days after the intravenous injection of B16 FIO melanoma cells into the mouse. This model is not a model of xenograft; however, it does not involve a high degree of vacularization in the tumor mass. [0475] In this model, the most effective supplement was KPU-2 alone (Figure 29), which has an average metastasis count of approximately 10% lower than for paclitaxel (MGIs of 41.6% and 35.0%, respectively). While that study does not by itself establish that combination therapy is more effective than mono-therapy, it indicates that KPU-2, KPU-35 and t-butyl-phenylahistine are more effective in highly vascularized tumors. EXAMPLE 10 Assays for Activity Against Pathogenic Fungi [0476] The comparative activity of a dehydrophenylahistin or its analogue against pathogenic fungi, with respect to known antifungal compounds described above for use in determining dehydrophenylahistin or its analogue AF / IS value, is measured directly against the fungal organism, for example by micro-titration plate adaptation of the NCCLS broth macro-dilution method described in Diagn Micro and Infect Diseases 21: 129-133 (1995). Antifungal activity can also be determined in whole animal models of fungal infection. For example, the pattern model plotted with pulmonary mucormycosis steroids (Goldaill, L. Z. &Sugar, A.M. 1994 J Antimicrob Chemother 33: 369-372) can be used. By way of illustration, in these studies, a number of animals are not given dehydrophenylahistin or its analogue, various doses of dehydrophenylahistin or its analogue (and / or) combinations with one or more other antifungal agents (or a positive control) for example Amphotericin B), respectively starting earlier, at or after the infection with the fungus. Animals can be treated once every 24 hours with the selected dose of dehydrophenylahistine or its analog, positive control or vehicle alone. The treatment is continued for a predetermined number of days, for example up to ten days. The animals were observed for a certain time after the treatment period, for example for a total of three weeks, with mortality estimated daily. The models may involve models of systemic, pulmonary, vaginal and other infections with or without other treatments (eg, steroid treatment) designed to mimic a human subject susceptible to infection. [0477] To further illustrate, a method for determining therapeutic efficacies in vivo (ED50, for example, expressed in mg dehydrophenylahistine or its analog / kg subject) is a rodent model system. For example, a mouse is infected with the fungal pathogen such as by intravenous infection with approximately ten times the lethal dose 50% of the pathogen (106 cells of C. albicans / mouse). Immediately after the fungal infection, the dehydrophenylahistin compounds are delivered to the mouse at a predetermined dose volume. ED50 is calculated by the Van der Waerden method (Arch Exp Pathol Pharmakol 195: 389-412, 1940) from the survival rate recorded on the 20th day post-infection. In general, untreated control animals die 17 to 13 days after infection. [0478] In other illustrative embodiments, C. albicans Wisconsin (C43) and C. tropicalis (C112), grown on Sabouraud dextrose agar (SDA) tilted plates for 48 h at 28 degrees C, are suspended in saline and adjusted to 46% of transmission at 550 nm in a spectrophotometer. The inoculum is additionally adjusted by hematocytometer and confirmed by plate count from approximately 1 or 5 x 107 CFU / ml. CF-1 mice are infected by injection of 1 or 5 x 10 CFU into the tail vein. Antifungal agents are administered intravenously or subcutaneously in ethanol: water (10:90), 4 h after infection and once daily afterwards for 3 or 4 more days. Survival is monitored daily. ED50 can be defined as that dose that allows 50% survival of mice. EXAMPLE 11 Evaluation of Antifungal Activity [0479] Benzimidazoles and griseofulvin are anti-tubulin agents capable of binding to fungal microtubules. Once bound, these compounds interfere with cell division and intercellular transport in sensitive organisms, resulting in cell death. Commercially, benzimidazoles are used as fungicidal agents in veterinary medicine and control of plant diseases. A wide variety of fungal species include Botrytis cinerea, Beauveria bassiana, Helminthosporium solani, Saccharomyces cerevisiae and Aspergillus are susceptible to these molecules. Considerations regarding toxicity and increasing drug resistance have nevertheless negatively impacted its use. Griseofulvin is used chemically to treat tub infections in the skin, hair and nails, caused by Tricofi ton sp., Microsporum sp., And Epidermofi ton, floccosum. Its antifungal spectrum however restricts this class of fungal organisms. Genotoxicity is also a significant side effect. Terbinafine, while it is an alternative first line treatment is more expensive. In addition, clinical resistance has recently been observed in Trichophyton rubrum (the major causative agent for all dermatophyte infections). [0480] In Candida albicans, microtubule / micro-filament formation is affected when the cells are exposed to the microtubule inhibitors nucodazole and chloropropane. These results further validate the screening of skeletal site inhibitors as effective anti-fungal agents. Accordingly, several of the compounds described herein were evaluated for anti-fungal activity. [0481] Specifically, the described compounds were evaluated in parallel with commercially available microtobulin inhibitors as well as recognized antifungal agents. The test compounds and controls used in this study: (-) -Fenilahistine, KPU-1, KPU-2, KPU-11 and KPU-17, KPU-35, t-butyl phenylahistine, Colchicine (microtubulin inhibitor, fray approved against 3 Candida isolates) Benomyl (microtubulin inhibitor, if approved against 3 Candida isolates) Griseofulvin (microtubulin inhibitor, commercial and antibiotic control for test against 6 dermatophyte isolates), Amphotericin B (controls antibiotic for test against 3 isolates of Candida), Itraconazole (controls antibiotic for test against two Aspergillus isolates). [0482] Microorganisms with which these compounds were tested, include: Candida albicans, Candida glabrata, Aspergillus fumigatus, Trichofiton rubrum, Trichofiton mentagrofites, Epidermofiton floccosum. Except for Candida glabrata (an isolate) two isolates of such a species were tested. [0483] The antifungal susceptibility test was achieved according to the methods established in National Committee for Clinical Laboratory Standards, M38-A "Reference Method for Broth Dilution Antifungal Susceptibility Testing of Conidium-Forming Filamentous Fungi; Approved Standard. "This includes RPMI-1640 test with glutamine and without bicarbonate, an inoculum size of 0.4-5 x 104, and incubation at 30 or 35 degrees C for 48 hours.The minimum inhibitory concentration (MIC = minimum inhibitory concentration ) is defined as the lowest concentration that results in an 80% reduction in turbidity compared to a drug-free control tube.The drug concentrations were 0.03-16 μg / ml for the compounds investigated, 0.015-8 μg / ml. ml for itraconazole and griseofulvin. [0484] The minimum inhibitory concentration (MIC) in which a compound prevents growth of the target microorganism was estimated according to the modified version of the NCCLS protocol.Minimum inhibitory concentrations (MIC) were determined in the first 24-hour interval where growth could be determinant in the drug-free control tube The MIC defined was the lowest concentration exhibiting an 80% reduction in turbidity in the Comparison with growth control. The minimum lethal concentration (MLC = minimum lethal concentration) was determined by applying a 0.1 μl layer of the concentration of MIC concentration on MIC. MLC was declared at the first concentration exhibiting 5 or fewer fungal growth colonies representing an extermination of 99.95%. When a MIC is obtained, a minimum fungicidal constitution (MFC = minimum fungicidal concentration) is determined to estimate the fungistatic / fungicidal nature of the compound. This procedure involves diluting samples of cells treated with drugs (withdrawal of test wells containing compound in and on MIC) at compound concentrations that are significantly below the inhibitory concentration and deposited on agar plates. The compound is classified as fungistatic if the cells are able to resume growth and fungicide if re-growth is not possible because the compound kills the organisms. [0485] Compounds described herein are effective against two species of Trichophyton. T. rubrum is the main causative agent for human dermatophyte infections and will be the key organism to target the development of the clinical agent. [0486] Compounds KPU-2, KPU-11 and KPU-17, KPU-35 and t-butyl-phenylahistine were potent or in some cases more potent equivalents than griseofulin, a current standard pharmaceutical agent, used to treat dermatophytic infections. [0487] The compounds (-) -Fenilahistine and KPU-1 were significantly less potent than the other compounds with which they can find T. rubrum weaker but more comparable with the others against the sensitive T. mentagropli tes isolate. [0488] In those cases when a CFM can be determined, the results indicate that these compounds are fungistatic in nature (see tables 19 and 20). Table 19. Antifungal Activity of Dehydrophenylahistin and its Analogs.
(CONTINUATION) Table 20. Antifungal Activity of Dehydrofenlahistin and its Analogs EXAMPLE 12 Evaluating Vascular Diana Activity [0489] Tumors and neoplastic conditions can be treated using the compounds described herein. Occlusion of the blood supply of tumors with target or vascular targeting agents (VTAs) induces regression of the tumors. The compounds described herein include NPU-02 and KPU-35, for example they can be as VTAs. Many VTAs exhibit their vascular defects by interacting at the site of the colchicine link in microtubules. This interaction induces a characteristic, rapid collapse and occlusion of the vasculature established in the tumor and therefore compromises the stability of existing vessels that lead to necrosis. [0490] Vascular collapse may occur for example within 30 or 60 minutes of exposure to VTA and involves changing the shape of the invaded and proliferating cells, but not the immature static cells, in the central portion of the tumor. This differential effect in vascular terms provides a rationale for selective effects in the tumor due to the higher percentage of immature endothelial cells proliferating in the tumor blood vessels against normal blood vessels. VTAs can be classified into three superimposed spectra of activity: (1) potent vascular and cytotoxic effects, (2) potent vascular effects with weak cytotoxicity and (3) potent cytotoxic effects with weak vascular. Activity of Invasive Diana Vascular of KPU-02 and KPU-35 [0491] Models in animals are essential to investigate new therapies that inhibit tumor-induced angiogenesis, target or target the established tumor vasculature and inhibit tumor growth . [0492] A models of murine "pseudo-orthotopic" breast cancer were used to address these aspects. Torres Filho et al., Microvascular Research (1995) 49, 212-226, which is incorporated herein by reference in its entirety. To create the "pseudo-orthotopic medium", the cover of a dorsal skin flap chamber is removed and small pieces of fatty mammary panicle from donor mice were implanted in the chamber. At the top of the fatty panniculus graft, tumor steroids containing N202 breast tumor cells transduced with green fluorescent protein Histone (H2B) -green fluorescent protein (GFP), were applied. The use of H2B-GFP transduced cells allows visualization of tumor growth and monitoring of mitosis and apotosis. [0493] Video fluorescence microscopy allows relatively non-invasive study of tumor microcirculation in conscious mice. This model can provide data regarding the effect of compounds on tumor vasculature, tumor growth, mitosis and apotosis and is useful for examining the activity of compounds either alone or in combination with other therapeutic agents. Using this model, KPU-02 and KPU-35 were shown to induce rapid vascular collapse leading to central necrosis, and regression of established tumors after a single i.v administration. [0494] On day 12 of tumor growth, i.v. mice were treated. with a 2-minute infusion of 5 mg / kg of KPU-35, an i.v. of 5 minutes of 10 mg / kg of KPU-02, or vehicle bolus (10% solutol (W / W) + 2% DMSO in water). On day 13, 15-minute infusions of 10 mg / kg of KPU-02, KPU-35 or vehicle were administered. Treatments with KPU-02 or KPU-35 were well tolerated. Mice were observed for two additional days. The area of tumor, expenditure of blood flow and vascular density within and surrounding the tumor, were visualized. Real-time observations were recorded at various points in time using still photos and video microscopy. [0495] This study demonstrates the rapid collapse of central vasculature after i.v. simple with any of KPU-02 or KPU-35. Changes in vascular functions resulted in significant central tumor necrosis, without an observed effect on the vasculature of the surrounding fatty panniculus or skin (Figure 30). These observations support the selectivity and specificity of KPU-02 and KPU-35, both of which can individually interrupt or break the established tumor vasculature. In Vivo Activity of KPU-02 in human tumor xenograft [0496] When KPU-02 is administered with CPT-11 (Irinotecan), Taxotere or Paclitaxel, marked anti-tumor activity was seen in human colon (HT-29), breast (MCF-7; MDA-MB231) and lung lung xenograft models (A549) (Table 21). The effect of KPU-02 in the HT-29 model was robust, reproducible in three studies and showed a dose-dependent effect, ie 7.5 mg / kg was statistically higher than 2.5 mg / kg (Figures 32, 33). In Vitro Activity of KPU-02 and KPU-35 in HuVEC cells [0497] The in vivo effects described above of KPU-02 and KPU-35 in the tumor vasculature were supported by the in vitro effects of the same compounds on HuVEC cells . Human umbilical vein endothelial cells are considered a good model in tumor endothelium, which is considered "immature". The tumor endothelium lacks supportive wall-vascular cells and is increasingly dependent on the microtubule network for integrity of the tumor vasculature. Therefore, the disruption of the tumor microtubule network causes vascular collapse.
KPEJ-02 induces rapid depolymerization of tubulin in HuVEC cells. [0498] Human umbilical endothelial vein cells (HuVECs; Cambrex CC2519A) were maintained at subconfluent densities in EGM-2 medium (Cambrex). The cells were cultured in a 37 degree C incubator at 5% C02 and 95% humidified air. For tubulin extinction assays, HuVEC cells were seeded at a density of 3 × 10 04 cells / ml in EGM-2 on slides or covers compatible with tissue culture (Fisher). The plates were returned to the incubator for 2 days. [0499] Standard solutions (20 mM) of the test compounds were prepared in 100% DMSO. Concentrated 400X dilutions of the compounds were prepared in 100% DMSO. Volumes of 5 μl of the dilutions were added to individual wells, resulting in a final concentration of 200 nM. The final agreement of DMSO was 0.25% in all the samples. The plates were returned to the incubator for 30 minutes. HuVEC cells were treated for 30 min with KPU-02 or 200 nM KPU-35. [0500] The cells were rinsed in dPBS before fixation in 10% (v / v) neutral buffered formalin for 10 minutes at room temperature. After alpha-tubulin fixation is visualized by indirect immuno-fluorescent. Specifically, the cells were waterproofed in triton X-100 / dPBS 0.2% (v / v) by minutes. The cells were washed before transferring the covers to a humidified chamber, the covers were blocked for two hours in antibody buffer (BSA 2% (v / v) / Tween20 0.1% (v / v) / dPBS). The covers were incubated with 50 μl of mouse alpha-tubulin 0.1 μg / ml (Molecular Probes) in antibody buffer for one hour before washing and incubation with 50 μl of 1 μg / ml FITC goat anti-mouse (Jackson ImmunoResearch Laboratories) for one hour in the dark. Finally the cells were washed and treated with 2 μg / ml of DAPI (Molecular Probes) for 10 minutes before rinsing in H20 and assemble with Vectashield assembly measurement (Vector Labs). The cells were imaged using a 60x oil immersion objective in a vertical microscope (Olympus BX51). The images were captured digitally using a CCD camera and software or Magnafire 2.0 program (Olympus). Post-image processing was done in Photoshop Elements 2.0 (Adobe) and in Microsoft Powerpoint. [0501] Figure 33 shows that KPU-02 and KPU-35 rapidly induce depullarization of tubulin in HuVEC cells. KPU-02 induces dose-dependent mono-layer permeability in HuVEC cells. [0502] Human umbilical vein endothelial cells (HuVECs; Cambrex CC2519A) were maintained at subconfluent densities in EGM-2 medium (Cambrex). The cells were cultured in a 37 incubator in 5% C02 and 95% humidified air. For monolayer permeability assays, HuVEC cells were seeded at IxlO5 cells / ml in EGM-2 medium in 3.0 μm Fluoroblock grafts coated with Fibronectin (Becton Dickinson) in 24-well plates. The plates were returned to the incubator for 4 days to allow the cells to reach confluence. [0503] Standard solutions (20 mM) of the test compounds were prepared in 100% DMSO. Serial 10X concentrated dilutions of the compounds were prepared in EGM-2. Volumes 10 μl of serial dilutions were added to the duplicate test grafts resulting in final concentrations in the range of 2 μM to 2 nM. The final agreement of DMSO was 0.25% in all the samples. The cells were treated with 2nM-2μM KPU-02 for 15 minutes. [0504] FITC-Dextran (50 mg / ml) in dPBS of (38.2 kDa; Sigma) is diluted 2.5 times in EGM-2, 10 μl of FITC-Dextran is added to each graft. The final concentration of FITC-Dextran was 1 mg / ml. The plates were returned to the incubator and 30 minutes later the fluorescence of the lower chambers of the wells of 24 plates was read using a Fusion fluorometer (Packard Bioscience) with lambdaex filters = 485 nm and lambdaem = 530 nm. [0505] Figure 34 shows that KPU-02 is capable of inducing mono-layer permeability in a dose-dependent manner. The results illustrated in Figure 34 represent the mean ± S.D. of three independent experiments. Blood Flow in the Rat Sarcoma Model P22 with 125 I-IAP [0506] Blood flow in tumor is estimated in a model using a quantitative technique of 125 I-iodine-antipyrine (IAP) in rats having a sarcoma number of rat P22. KPU-02 (15 mg / kg, IP) markedly and selectively reduced tumor blood flow to 23% vehicle at 1 hour after administration; blood flow remained markedly reduced 24 hours later (59% of the vehicle). In contrast, the blood flow in non-tumor tissues concentrated in a much smaller proportion than 1 hour (see Figure 35). [0507] Reduction of blood flow at 24 hours post-dose was more variable between tissues for KPU-02 compared to the vehicle as illustrated in Figure 36. Blood flow to the tumor was most affected. Other tissues exhibited a small reduction in blood flow. Skeletal muscle blood flow appeared to increase 24 hours after the dose. [0508] The effects of KPU-02 observed at 1 hour appear to be longer lasting and more selective for tumor blood flow than previously reported for CA4P using the same technique. [0509] In an experiment with the rat sacroma model P22, it was demonstrated that KPU-02 7.5 and 15 mg / kg IP (n = 2 per dose) produces a dose-dependent tumor necrosis for 24 hours post-dose , with the highest dose resulting in almost total tumor necrosis as illustrated in Figure 37. All tumors in rats treated as KPU-02 showed evidence of necrosis, while tumors in vehicle-treated rats did not. . VTAs that have entered a clinic (eg, CA4P, ZD6126, AVE8062) show similar qualitative effects on tumor blood using the IAP methodology (or similar technology) to demonstrate reduced blood flow in the rat sarcoma tumor P22 and in humans using the dce-MRI technique. See Stevenson JP, Rosen M, Sun W, Gallagher M, Haller DG, Vaughn D, et al., "Phase I trial of the antivascular agent combretastatin A4 phosphate on a 5-day schedule to patients with cancer: magnetic resonance imaging evidence for altered tumor blood flow, "J Clin Oncol 2003; 21 (23): 4428-38; Evelhoch JL, LoRusso PM, He Z, DelProposto Z, Polin L, Corbett TH, et al., "Magnetic resonanceimaging measurements of the response of murine and human tumors to the vascular-targeting agent ZD6126," Clin Cancer Res 2004; 10 (11): 3650-7; and Gadgeel SM, LoRusso PM, Wozniak AJ, Wheeler C. "A dose-escalation study of the novel vascular-targeting agent, ZD6126, in patients with solid tumors," Proc Am Soc Clin Oncol 2002; 21: abstract 438; each of which is hereby incorporated by reference in its entirety. Combination Therapy with Microtubule Directed Agents. [0510] The findings that VTAs selectively damage the vasculature in the central part of the tumor against the periphery, which recover functionality, support using these agents in combination with chemotherapeutic agents (Taxol, Vinblastine and Cisplatin), radiation and angiogenesis inhibitors directed against VEGF and EGF. The new VTAs will supplement rather than supplant these therapies and should provide greater anti-tumor activities. Treatment of other conditions [0511] In addition to cancer, other diseases can be treated using the VTAs described here. Conditions that include other neoplasms, retinopathies and any other condition or disease that is based on blood supply, preferably blood supply of new vasculature in order to remain viable and / or proliferate. [0512] Many conditions are associated with excessive or inappropriate vasculature. Examples of conditions associated with excessive vasculature include inflammatory disorders such as immune and non-immune inflammation, rheumatoid arthritis, chronic joint rheumatism and psoriasis; disorders associated with inappropriate or untimely invasion of vessels such as diabetic retinopathy, neovascular glaucoma, retinopathy of prematurity, macular degeneration, rejection of corneal graft, retrolental fibroplasia, rubeosis, capillary proliferation in arteriosclerotic plaques and osteoporosis; and disorders associated with cancer, including for example solid tumors, tumor metastases, blood-borne tumors such as leukemia, angiofibromas, kaposi's sarcoma, benign tumors such as hemangiomas, acoustic neuromas, neurofibromas, trachoma, and pyogenic granulomas, as well as other cancers that require vascularization to support tumor growth. Traditional examples of vasculatory dependent diseases include for example the Osler-Webber syndrome; myocardial angiogenesis, plaque neovascularization; telangiectasia; Hemophilic joints and wound granulation. In addition, excessive vasculature is also associated with clinical problems such as parts of biological and mechanical implants (tissue / organ implants, endoprostheses or intraluminal cannulas, etc.). The present compounds and compositions can be used to target the vasculature, preferably to target diseased vasculature on non-diseased tissue vasculature and thus the compounds and compositions can be used in the treatment of these conditions. Other diseases wherein vascularization plays a role, and in which the present compounds and compositions may be employed, are known to those skilled in the art. [0513] Examples of retinopathies include age-related macular degeneration (ARMD = age-related macular degeneration), diabetic retinopathy, and the like. Pathological angiogenesis is a major contributor to a number of retinopathies that are collectively the leading cause of blindness in the developed world. Kahn and Hiller Am J Ophthalmol (1974) 78.58-67, which is incorporated herein by reference in its entirety. For example, retinal and disc neovascularization occurs in 30-50% of patients with diabetic retinopathy for more than 20 years. Yanko et al., Retina (2003) 23, 518-522, which is incorporated herein by reference in its entirety. In addition, neovascularization is a serious complication in 10% of patients with macular degeneration. Ferris et al Arch Ophthalmol (1984), 102, 1640-1642, which is hereby incorporated by reference. [0514] Vascular target agents such as Combretastatin A-4 (CA-4) have been shown to cause the rupture of new vessels in neovatic tissue. Griggs et al Br J Cancer (2001) 84, 832-835, which is hereby incorporated by reference in its entirety. Additionally, CA-4P is shown to inhibit retinal neovascularization that occurs during proliferative retinopathy. Griggs et al Am J Path (2002) 160, 1097-1103, which is incorporated herein by reference in its entirety. Finally, CA-4P phosphate is shown to suppress the development of retinal neovascularization induced by VEGF and inhibits development and / or causes partial regression of choroidal neovascularization. Nambu et al-, Invest Ophthalmology & Visual Sci (2003) 44.3650-3655, which is incorporated herein by reference in its entirety. The compounds described herein can be used to treat retinopathy. For example, the methodologies of Griggs (2001 and 2002) and Nambu are used to treat retinopathies. In addition, the compounds and compositions described herein can be used to treat these retinopathies by applying the compounds and / or compositions to the target area in an amount effective to reduce vascular density and / or vascular proliferation. Table 21. Effect of KPU-02 in combination with chemotherapy in human tumor xenograft models Tumor Dose Reference Model KPU-02 Chemotherapeutics (# Studies) (mg / kg ip) Colon 7.5 CPT-11 HT29 Days Days 1,8,15 (3) 1,4,8,11,15 Chest Taxotere MCF-7 7.5 Days (2) qdx5 1,3, 5 Chest 7.5 Paclitaxel MDA-231 Days qdx5 Tumor Dose Reference Model KPU-02 Chemotherapeutics (# Studies) (mg / kg ip) (1) 1,4,8,11,15 Lung Taxotere A549 7.5 Days (1) qdx5 1,3,5 Table 21 (Continued.) Tumor Inhibition of tumor regression growth of tumor (# / total) (%) Model Reference KPU-02 + NPI agent + reference Reference (# Agent Reference Studies) Colon HT29 37 + 3 79 + 8 0/30 4/30 (3) Chest MCF-7 12; 58 26; 81 0/20 3/20 (2) Chest MDA-231 53 71 1/10 0/10 (1) Tumor Inhibition of tumor regression tumor growth (# / total) (%) Model Reference PU-02 + Agent NPI + reference Reference (# Agent Reference Studies) Lung A549 26 74 0/10 0/10 (1) EXAMPLE 13 Relationship of Structure-Activity [0515] The activity effect of various modifications on the phenyl ring of tBu-dehydroPLH is illustrated by the data in Figure 38. It is apparent that substitution with relatively hydrophobic and functional minor groups in the position -mu -o, increases or maintains the cytotoxic activity in the HT-29 cells while substitutions in the -p position, decreases the activity. While not bound by any particular theory, these data suggest a rigorous recognition of the phenyl ring by tubulin. [0516] 3D-QSAR analysis (CoMFA) (see Figure 39) also supports the existence of sterically favorable fields at positions -m and -o and sterically unfavorable fields exist at position -p. Analysis of X-ray crystals (see Figure 40) indicates that the conformation of potent derivatives requires a certain amount of dihedral angle between the phenyl ring and the pseudo-tricyclic cor template formed by DKP and imidazole rings. In this way, modification with the appropriate conformation restriction of the phenyl ring, can produce potent activity. While not bound by any particular theory, it may be that the mode of linking PLH derivatives at the tubulin colchicine binding site is different from colchicine and its known counterparts. EXAMPLE 14 In Vitro Action in Microtubules Microtubule Protein and Tubulin Purification [0517] Microtubule protein (MTP) is prepared as previously described (Farrell KW and Wilson L. (1987) Tubulin-colchicine complexes differentially poison opposite ends of microtubules, Biochemistry 23 (16): 3741-8, which is incorporated herein by reference in its entirety). Preparations of MTP consisting of 70% tubulin and 30% microtubule-associated proteins (MAPs = microtubule-associated proteins) were isolated from bovine brain by three cycles of hot polymerization and cold depolymerization in PEM100 (1-4 piperazindiethansulfonic acid 100 mM (Pipes), 1 mM MgSO4, 1 mM EGTA, pH 6.8) and 1 mM GTP. MTP was frozen by dripping in liquid nitrogen and stored at -70 degrees C until use. Tubulin is purified from microtubule protein by phosphocellulose chromatography (PC-tubulin) and stored in PEM50 (50 mM Pipes, 1 mM MgSO4, 1 mM EGTA, pH 6.8). Protein concentration is determined by a Bradpord assay (Sigma Chemicals, St. Louis, MO) using bovine serum albumin as the norm (Bradpord, 1976). Test agents [0518] Standard solutions of KPU-02 were prepared at a concentration of 20 mM in DMSO. Standard solutions of Combretastatin A4 (National Cancer Institute, Bethesda, MD) (CA4) are prepared at a concentration of 5 mM in DMSO. Colchicine (Sigma Chemicals, St. Louis, MO) (CLC) is prepared at a concentration of 3 mM in water. All agents were protected from ambient light with Eppendorf amber tubes. Serial dilutions were performed in DMSO and / or PEM50 at the desired concentrations.
Determination of Microtubule Polymer Mass in Stable State [0519] MTP (2 mg / ml) was polymerized in microtubules in the presence of a range of drug concentrations in PEM100 containing 1 mM GTP and a final DMSO concentration of 0.5% Samples were monitored by light scattering at 350 nm at 37 degrees C for 75 minutes. [0520] Polymerization reactions were centrifuged and microtubule protein concentrations in the supernatant, a measure of soluble tubulin in steady state, and the precipitate, a measure of microtubule polymer, is used to calculate the inhibition of polymerization. After incubation, the polymerized microtubules were separated and pelleted from MTP without polymerization by centrifugation (150,000 x g, 45 minutes, 37 degrees C). The supernatant was removed, and the microtubule precipitates were depolymerized in deionized H20 (24 hours, 0 degrees C) before protein determination by the Bradford assay. [0521] Percent inhibition was calculated in two ways and the values obtained from the two forms were compared. In one form, a proportion of the microtubule protein in the precipitate, drug to no drug, was calculated. Another proportion of microtubule protein in the precipitate to the supernatant, drug to no drug was also calculated. The numbers were in close agreement and the previous values were used because they underwent less variance and experimental perturbation. Average Microtubule Length Distributions [0522] Transmission electron microscopy is used to determine the average microtubule length distribution in the absence or presence of the tested agent. At 75 minutes and before sedimentation, 10 μl aliquots of the polymer mass experiments were fixed by dilution in 290 μl of 0.2% glutaraldehyde buffered with PEMIOO. Thirty microliters of fixed sample are sedimented in electron microscope grids of 150 ICG mesh coated with formvar for 90 seconds. The excess sample was sucked by capillarity with Whatman filter paper. Thirty microliters of cytochrome C (1 mg / ml) are applied for 30 seconds to improve the resolution of protofilaments and facilitate negative staining. Uranil acetate (1.5%) is applied for 20 seconds and the excess is absorbed by capillarity. Grids were seen on a Jeol-1200EX11 electronic microscope at 2000X and 30,000X magnification. The Zeiss MOPHI was used to determine microtubule length distributions and average lengths for at least 100 microtubules per sample. Competition assays CLC [0523] PC-tubulin (0.2 mg / ml) is incubated in PEM50 with 1 mM GTP, 1% DMSO, 10 μM tested agent and 7-25FM [3H] CLC for 120 minutes at 37 degrees C Linkage measurement and [3H] CLC was followed by cellulose filter-DEAE binding assay as previously described (Wilson, 1970). This method depends on the adsorption of tubulin to the filter paper impregnated with DEAE-cellulose. Whatman DE81 filter paper was pre-moistened with PEM50 before sample application. The total reaction volume of 100 μl is applied to 2.5 cm discs of filter paper, on parafilm, on ice. The paper discs were washed by immersion in five successive 50 ml changes of PEM50, 5 minutes / wash, 4 degrees C, to remove all unlinked colchicine. The paper discs with colchicine bound to adherent tubulin were counted directly in a flask containing 2 ml of Beckman protein solution Coulter Ready Protein Solution (Fullerton, CA). All discs were washed together.
Unreliable link of CLC without binding to paper discs occurs in controls, either in the presence or absence of tubulin.
[0524] The Ki values were calculated by linear regression of a reciprocal double trace of the experimental data in Microsoft Excel. The Km value of tubulin for CLC under the experimental conditions was determined first, with x intersecting equal to -I / Km in the presence of drug, was determined experimentally. Ki is determined using the ratio K ^ app = alpha Km, and for competitive inhibition alpha = Km (l + [I] / Ki). Fluorescence Spectroscopy [0525] Fluorescence measurements were made using a Perkin-Elmer LS50B spectrofluorimeter. PC-tubulin (0.2 mg / ml) is incubated in PEM50, 2 mM GTP, 3% DMSO, with 0-30 μM KPU-02. The interaction of KPU-02 with tubulin is reported by fluorescence of 4,4 '-dianilino-1,1' -bubfthyl-5,5'-disulfonic acid, dipotassium salt (bis-ANS; Molecular Probes, Eugene, OR), with an excitation wavelength of 395 nm and a maximum emission wavelength of 487 nm. The excitation and emission band passages were 10 nm. This experiment was performed twice. [0526] The bis-ANS fluorophore probes the hydrophobic surface of proteins and a change in intensity of the bís-ANS fluorescence signals is a result of a change in the solvent accessible surface area of a protein. If there is a change in conformation that modifies the tubulin-bis-ANS interaction before ligand binding, then bis-ANS can be used to report links. [0527] PC-tubulin (0.2 mg / ml) is incubated with KPU-02 0-30 μM at 25 degrees C for 20 minutes. Bis-ANS (25 μM) was then added and relative fluorescence intensities of samples were measured at 25 degrees C in 15 minutes. White absorber spectra were collected and showed that KPU-02 plus bis-ANS produced negligible fluorescence in the experimental wavelength range. [0528] The Ka is determined by fitting experimental data in Sigmaplot and Microsoft Excel using the equation F = ((-Fmax xL) / (a + L)) + F0 where F is the fluorescence intensity of bis-ANS-tubulin in the presence of total concentration of ligand L, Fmax is the intensity of fluorescence bis-ANS of tubulin totally with ligand, and Fo is fluorescence bis-ANS in the absence of drug. Fma? it is determined by plotting 1 / (F0-F) against l / L and extrapolating to l / L = 0. The fraction of binding sites B occupied by KPU-02 is determined using the following relationship: B = (F0-F) / (F0-Fma?). The concentration of free ligand is determined with Lfree = L-B [C] where [C] is the molar concentration of ligand binding sites considering a single binding site per tubulin dimer. Inhibition of microtubule polymerization by KPU-02 [0529] KPU-02, CA4, and CLC were tested for their ability to alter the polymerization of MAP-rich tubulin (MTP) (2 mg / ml) in a cell-free system. vitro. Initially, the polymerization inhibition was assayed using protein free tubulin associated with microtubules, purified with phosphocellulose (data not shown). KPU-02 was a more potent inhibitor towards MTs assembled with glycerol and DMSO seeds compared to MTs assembled in the presence of MAPs that co-purify with tubulin. Although the microtubule polymer in the absence of stabilizing MAPs is not stable over a period of two hours, these tests demonstrate that KPU-02 interacts directly with purified tubulin and that it does not exert its primary effect through a MAP. [0530] KPU-02 and CA4 inhibit MT polymerization more powerfully than CLC as measured by light scattering analysis (Figure 44) and sedimentation (Figure 45). MTP (2 mg / ml) is polymerized in microtubules in the presence of a range of drug concentrations and allows to reach steady state as verified by light scattering at 350 nm. Figure 41 illustrates turbidity spectra of microtubule protein polymerization in the presence of the drug vehicle DMSO (0), 1.25 μM (D), 2.5 μM (-), and 5 μM (O) NPI-2358 (a), CA4 (b) and CLC (c). KPU-02 and polymerization inhibited by CA4 with comparable potencies. Figure 45 illustrates inhibition of microtubule polymerization in the absence or presence of a range of concentrations of KPU-02 (o), CA4 (ü), and colchicine (0). The total polymer mass after 75 minutes of assembly was determined by sedimentation. Error bars are standard deviation values of three experiments. The concentration at which the polymerization was inhibited by 50% (IC50), is 2.4 + 0. 4 μM for KPU-02, 2.2 ± 0.3 μM for CA4, and 7.6 + 2.4 μM for CLC (Table 22). (Variances obtained by statistical analysis are reported as standard deviation values unless otherwise stated). At concentrations higher than IC50 for in vitro polymerization of MAP-rich tubulin, MTP exhibits aggregation kinetics, suggesting that KPU-02 and CA4 sequester protein to prevent microtubule assembly. Table 22. Concentrations for inhibition of microtubule polymerization.
[0531] As illustrated in Figure 44, all three tested agents produce a concentration-dependent inhibition of the microtubule polymerization extension from 1.25-5 μM. There are two important differences to notice between the spectra. First, the initial rate of increase in absorbance over time decreases with increasing drug concentration (Figure 44A and 44B). The spectra indicate that there is a delay period for formation of MT in the presence of KPU-02 and CA4. Drugs that significantly and rapidly reduce the tubulin-soluble assembly assembly will decrease the initial rate of polymerization. In contrast, the initial polymerization rate is unchanged at all CLC concentrations (Figure 44C). Second, MTP in the presence of KPU-02 or CA4 does not reach stable state at high drug concentrations (above 5 μM), as shown by the absorbance values that increase linearly with time (Figures 44A and 44B). In contrast, MTP in the presence of CLC reaches steady state at high drug concentrations (Figure 44C). [0532] The amount of drug required to inhibit 50% polymerization (IC50), is determined from the analysis of the linear relationship between the decrease in microtubule polymer sedimented by centrifugation with the increase in drug concentration (Figure 45). The error bars in Figure 45 represent standard deviation values of at least three independent experiments. Decrease in microtubule average length measured by transmission electron microscopy [0533] Transmission electron microscopy was performed in microtubule-agent polymerization reactions, to describe the polymer formed in steady state and evaluate conclusions taken from the light scattering spectrum. KPU-02, CA4, and CLC all decrease the lengths of the microtubules formed in stable state. MTs were progressively shorter with increasing concentration of the drug (Figures 46, 47 and 48). Figure 46 illustrates frequency histograms of average lengths of microtubules in vitro in steady state in the presence of (A) KPU-02, (B) CA4, and (C) CLC. The Zeiss MOPIII is used to determine microtubule length distributions and average lengths. At least 100 microtubules per drug concentration were counted. Figure 47 illustrates electron microscopy used to record microtubules in the absence or presence of the tested compounds. At 75 minutes, samples of the polymer mass experiments were fixed and stained and viewed in a Jeol-1200EX11 electron microscope at a magnification of 2000x. Representative electron microphotographs of MAP-rich microtubules formed in vitro in steady state in the presence of (A) KPU-02, (b) CA4, and (C) CLC. The scale bar, 10 μM. Figure 48 illustrates a graphic summary of MV length decrease in steady state in the presence of KPU-02, CA4, and colchicine. The black bars, 1.25 μM, and shaded bars, 2.5 μM of drug. In the presence of KPU-02, and CA4, MTs are progressively shorter with increased drug concentration, until the concentration of the drug in which MTP exhibits aggregation kinetics as detected by turbidity, and no MTs are observed. Error bars are values of standard deviation of the measurement of at least 100 microtubules. [0534] KPU-02, CA4 and CLC do not affect MT nucleation. The numerous short microtubules formed in the polymerization reactions show that the presence of KPU-02, CA4, or CLC does not affect nucleation. If nucleation were to be affected, then fewer and longer microtubules, as opposed to numerous and shorter microtubules, would have been observed in the control samples versus those treated with the drug. [0535] KPU-02 and CA4 were comparatively potent to decrease the average MT length. At 1.25μM, the lowest concentration of drug analyzed by electron microscopy, KPU-02 and CA4 decrease the average MT length by approximately 70%, and CLC by 40% (Figure 48). [0536] At drug concentrations on IC50 for microtubule polymerization in vi tro microtubules are not observed by electron microscopy for KPU-02 and CA4. In contrast, microtubules were observed by electron microscopy for all CLC concentrations tested. At concentrations over IC50, the microtubule protein in the presence of KPU-02 and CA4 exhibits aggregation kinetics, characterized by a linear increase in light absorbance over time (Figures 44A and 44B), whereas in the presence of CLC , the polymer light scattering reaches stable state (Figure 44C). Despite the observation that MTP with KPU-02 or CA4 increased absorbance at 350nm over time, no drug-specific protein aggregates were observed. Fluorescence spectroscopy [0537] Tubulin (0.2 mg / ml) was incubated with a range of concentrations of KPU-02 for 20 minutes at 25 C in PEM50 and 2 mM GTP. The fluorescence of bis-ANS neutralized by KPU-02 in a concentration-dependent manner (Figure 49A). For KPU-02 and tubulin as measured by non-linear regression analysis of bis-ANS fluorescence intensity at maximum emission, K ^ = 10 + 1.6 μM (standard error) (Figure 49B). The reciprocal double line of link data, considering a single link site for KPU-02 per tubulin dimer, produces a dissociation constant of 6.4 μM (Figure 49C). The two different Kd values obtained by linear and non-linear regression analysis methods were close enough and the values were considered approximately equivalent. Figure 49A illustrates fluorescence emission spectra of tubulin in the presence of KPU-02 increase. The drug binding results in neutralization of its bis-ANS fluorescence. Figure 49B illustrates fluorescence emission maxima at 487 nm adjusted to obtain the tubulin for KPU-02, 10 μM, 1.6 μM standard deviation. Insert, residual. Figure 49C illustrates the reciprocal double transformation of binding data considering one mole of drug / mol of tubulin dimer. Competitive inhibition of CLC binding [0538] KPU-02 and CA4 competitively inhibited CLC binding with tubulin (Figure 50). Figure 50 illustrates the results of an inhibition assay wherein tubulin purified with phosphocellulose (0.2 mg / ml) is incubated with various concentrations of [3 H] CLC in the absence (0), or presence of 10 μM KPU-02 (or ) or CA4 10 μM (o). K ^ of Tubulin-CLC was 11 + 4.4 μM and the inhibition constants for KPU-02 and CA4 were 3.2 ± 1.7 μM and 2.4 + 0.3 μM, respectively. The constants were calculated for three independent experiments. The colchicine-tubulin binding reaction depends on time and temperature and the bond dissociation constant is Ka = 0.1-1 μM, depending on the test conditions (Wilson L and Meza. (1973) The mechanism of action of colchicine. Colchicine bindinf properties of sea urchin sperm tail outer doublet tubulin, Journal of Cell Biology 58 (3): 709-19, which is hereby incorporated by reference in its entirety). Under the test conditions, the Kp of tubulin for CLC is 11 + 4.4 μM. The K ^ can be considered the total a tubulin for CLC, however, due to the dependence of the CLC link time, the Km is greater than the values reported for K¿. The Ki for KPU-02 and CA4 was 3.2 ± 1.7 μM and 2.4 ± 0.3 μM, respectively. Ki is defined as the amount of drug required to inhibit CLC binding by 50% and is based on the amount of radioactive CLC that is bound to tubulin. The i is a measure of the drug's ability to compete with CLC; it is not a direct measure of the dissociation of drug-tubulin binding due to the method in which the binding affinity is reported. Results At all CLC concentrations tested, tubulin rich in MAP reached steady state. In contrast, at higher concentrations of KPU-02 or CA4 drug, tubulin-rich MAP does not polymerize at steady state and microtubules were not observed by electron microscopy. KPU-02 and CA4 effectively decrease the concentration of available tubulin. This decrease in the soluble tubulin assembly increases the critical concentration of MT and prevents polymerization. The stoichiometric amounts of KPU-02 and CA4 required to decrease the polymer mass in vitro to match with the data that the microtubule protein does not reach a stable state over those concentrations, suggesting that KPU-02 and CA4 act by a mechanism of sequestration where the soluble tubulin binds and prevents it from polymerizing. Observations by electron microscopy in microtubules rich in stable state MAP, formed in the presence of the tested agents, were consistent with the proposed mechanism in which KPU-02 and CA4 sequester tubulin. There was a concentration-dependent decrease in average microtubule length in the presence of KPU-02, CA4, and CLC. In the presence of KPU-02 and CA4, there was a decrease dependent on the drug concentration in the initial state of polymerization, indicating that these drugs reduce the tubulin available for elongation. This decrease in the initial polymerization rate was not seen with CLC due to its slow association with tubulin. further, microtubules were formed at CLC concentrations on their IC50 for polymerization, but no microtubules were formed at concentrations of KPU-02 or CA4 on their IC50 for polymerization. While not bound by any particular theory, the concentration of soluble tubulin bound by KPU-02 or CA4 must be below the critical concentration required to proceed to the polymerization of tubulin. [0539] Linkage studies indicate that tubulin has a lower affinity for KPU-02 than for CLC. Inhibition of CLC binding with tubulin by KPU-02 and CA4 occurs within an incubation period of 20 minutes, indicating that the association of KPU-02 and CA4 with tubulin approaches equilibrium relatively faster than for CLC (data not shown ). KPU-02 competitively inhibits the binding of CLC with tubulin in a site superimposed with the CLC binding site, consistent with studies characterizing phenylahistine (halimide) (Kanoh K, Kohno S, Kataka J, Takahashi J and Uno I. (1999 ) (-) - Phenylahistine slows cells in mitosis by inhibiting tubulin polymerization The Journal of Antibiotics 52 (2): 134-141, which is incorporated herein by reference in its entirety). CA4, a structural analog of CLC, also competitively inhibits the CLC linkage. Without being bound by any particular theory, it seems that although they share a tubulin binding region with CLC, KPU-02 and CA4, they interact with tubulin and inhibit microtubules by a mechanism other than CLC. EXAMPLE 15. Microtubule in vivo action Cell culture studies [0540] MCF7 human breast carcinoma cells (American Type Culture Collection, Manassas, VA) stably transfected with GFP-alpha-tubulin (Clontech, Palo Alto, CA) were cultured in Dulbecco's modified Tagle medium supplemented with 5% fetal bovine serum, 0.1% penicillin / streptomycin and non-essential amino acids (Sigma) in 250 ml tissue culture flasks or plates. six 35-mm wells (doubling time, 29 hours) at 37 degrees C in C02 at %. Cells were incubated with KPU-02, CA4, or CLC, prepared as described in Example 14, by replacing the original medium with an equal volume of medium containing the required concentration of the tested agent or DMSO vehicle, and incubation is continued. at 37 degrees C for 20 hours. Mitotic Advance [0541] The fraction of cells in mitosis now given drug concentration (mitotic index), is determined in the MCF7 breast cancer cell line. The cells were coated at a density of 3 × 10 04 cells / ml in six-well plates. After 24 hours, the cells were incubated in the absence or presence of drug in a range of concentrations (1 nM to 1 μM) for 20 hours. Media was collected and cells were rinsed with versene (137 mM NaCl, 2.7 M KCl, 1.5 mM KH2P04, 8.1 mM Na2P04 and 0.5 mM EDTA), detached with trypsin and added back to the medium to ensure that the floating and poorly connected mitotic cells will be included in the analysis. The cells were fixed with 10% formalin in PBS overnight at 37 degrees C, permeabilized with methanol for 10 minutes and stained with 4,6-diamino-phenylindole (DAPI) to visualize nuclei. The stained cells were dispersed on coverslips in the middle of Vectashield assemblies (Burlingame, CA) and sealed the slide with nail varnish. Fluorescence microscopy was used to determine mitotic indices. The results were average standard deviation of 4-7 experiments where 300 cells were counted for each concentration. The Ic50 was the concentration of drug that experimentally induced 50% of the maximum mitotic accumulation at 20 hours. Immunofluorescence microscopy. [0542] Cells were prepared for mitotic advance, except that the cells were seeded on coverslips treated with L-lysine (50 μg / ml, sigma) on the day of staining, cells were rinsed in PBS and fixed in 10% formalin overnight at 37 degrees C. the cells were rinsed in PBS, permeabilized in methanol at -20 degrees C and hydrated with PBS. The coverslips were treated with 20% normal goat serum in PBS / BSA (1%) for one hour at room temperature. The cells were incubated in a mouse monoclonal cocktail of anti-alpha- and beta-tubulin, DM1A / DM1B diluted in PBS / BSA for 1 hour at room temperature, then stained with secondary antibodies and conjugated with FITC and DAPI. The coverslips were mounted using Prolong anti-fading medium (Molecular Probes, Eugene OR). Preparation of Cells for Microtubule Analysis and Dynamics. [0543] Cells were prepared for mitotic advance, except that to promote cell dispersion, the cells were seeded onto glass coverslips that had been pretreated with poly-L-licina (50 μg / ml, sigma) for 2 hours, followed by by laminin and fibronectin (10 μg / ml, sigma) for 1 hour at 37 degrees C. The cells were incubated chondroma or DMSO for 20 hours and deprived of serum. Prior to analysis, the coverslips were transferred to recording or recording medium (culture medium lacking red-phenol and sodium bicarbonate buffered with 25mM HEPES and supplemented with 3.5 g / L sucrose). To avoid photobleaching, Oxyrase (30 μl / ml, Oxyrase Inc., Mansfield, OH) is added to the recording medium immediately before sealing the cells in a chamber circumscribed with double coverslips. Interval Microscopy and Image Acquisition [0544] Microtubules were observed using Nikon Eclipse E800 fluorescence microscopy with a plan objective lens apochromat 1. 4N. A. x 100. The stage was circumscribed in a Pyrex box and maintained at 36 ± 1 degree C by a forced air heating system. 30 images of each cell were acquired at 4-s intervals using a Photometrics Cool SNAP HQ digital camera (Tucson, AZ) directed by the Metamorph program (Universal Imaging, Media, PA) at 10 MHz, with an exposure time of 300 ms, a gain of 2, and 2 x 2 combination to improve the brilliance. Microtubule Dynamics Analysis [0545] The locations of the microtubule plus ends over time were followed using the Metamorph Track Points application exported to Microsoft Excel and analyzed using the Real Time Measurement program. The lengths of individual microtubules were plotted as a function of time. Individual growth and cut-off speeds were terminated by linear regression. Changes of >; 0.5 μm between two points were considered growth or shortening events, and changes of < 0.5 μM between two points, were considered periods of dynamics or pauses attenuated. At least 25 microtubules were analyzed for each condition. The results are the average and standard deviation of at least three independent experiments. [0546] The time-based catastrophe frequency for each microtubule was calculated by dividing the number of catastrophes per microtubule by the time spent in growth or attenuation. The microtubule time-based rescue frequency is calculated by dividing the total number of rescues per microtubule by the time spent in the shortening. Rescue and catastrophe frequencies based on distance were generally calculated by dividing the number of transitions by the developed or shortened length, respectively. Microtubules that were visible by < 2 min were included in the frequency analysis. The structural dynamics (dynamicity) by microtubules is calculated as the length of growth and cut out divided by the total life span of the microtubule. Microtubules that were visible for 0.3 min were included in the dynamics analysis. Advance of Cell Cycle Blocked in prometa-phase [0547] The concentration interval for KPU-02, CA4 and CLC on which cells accumulate in mitosis was determined. After 20 hours, 60-70% of the cells were inhibited in the prometa-phase, compared to 30-40% of meta-phase cells in studies in MT depolymerizers such as vinca alkaloids and 2-methoxysodiol and MT stabilizers such as taxol, epothilone B and discodermolide (Jordan MA (2002) Current Medicinal Chemistry- Anti-cancer Agents: 1-11, (which is hereby incorporated by reference in its entirety). The drug concentration needed for 50% maximum mitotic blocking (IC50) is evaluated between 1 nM and 1 μM of drug (Figure 51). Figure 51 illustrates response curves [Drug] for inhibition of mitotic advancement by KPU-02, CA4, and CLC. MCF7 cells were cultured in the presence of NPI-2358 (o), CA4 (a), and colchicine (0). To evaluate mitotic indices, MCF7 cells were coated at a density of 3 x 10 4 cells / ml in 6-well plates. After 24 hours, the cells were incubated in the absence or presence of drug over a range of concentrations (1 nM to 1 μM) for 20 hours. The cells were fixed and stained with DAPI to visualize the nuclei. Fluorescence microscopy is used to determine mitotic indices. The results are the average and standard deviation of three or four experiments where 300 cells were counted for each drug concentration. The mitotic block ICS0 for KPU-02 was 17.4 ± 1.2 nM, CA4 was 5.4 ± 0.7 nM, and CLC was 23.8 ± 3.1 nM (Table 23). Table 23. Inhibition of mitotic advance.
[0548] Most agents directed to MT block mitosis in the transition from meta-phase to ana-phase. The mitotic block in the transition from metaphase to anaphase is associated with suppression of MT dynamics. Without being bound by any particular theory, the previous phase promise block, together with MT MT depletion, suggests a different mechanism of action for KPU-02 compared to other MT depolymerizing drugs, for example vinblastine, which at low concentrations stabilize the dynamics of MT. Depolymerization of the achromatic spindle and the interface arrangement MTs [0549] KPU-02, CA4, and CLC were observed as potent microtubule depolymerizers in MCF7 cells. Although achromatic spindle microtubules are more susceptible to depolymerization and / or polymerization inhibition than microtubules of interphase array, both populations of microtubules were affected (Figure 52). Figure 52 illustrates immuno-fluorescence microscopy images of MCF7 cells. Interphase arrays are relatively more stable to polymerization by KPU-02, CA4 CLC than the achromatic spindles. Cells were prepared and plated for mitotic advance and treated with the IC50 mitotic block for each drug for 20 hours. Cells were incubated in a mouse monoclonal cocktail of anti-alpha-and-beta-tubulin, DM1A / DM1B then stained with conjugated secondary antibody FITC and DAPI. ad, Tubulin in control (a), KPU-02 (b), CA4 (c), and CLC (d) treated cells, and e ~ h, DNA in control (e) cells treated with KPU-02 (f), CA4 (g), and CLC (h). Narrow arrows indicate achromatic spindle polymer and mitotic chromosomes and thicker arrows indicate interphase and nuclei arrangement. [0550] To IC25 for mitotic block, KPU-02 dramatically alters the spindle morphology. Figure 53A-C illustrates immuno-fluorescence microscopy images of MCF7 cells treated with KPU-02 (A), CA4 (B), and CLC (C) for 20 hours. Destruction of an achromatic spindle with increasing concentration of drugs. 1-4, Alpha and beta tubulin in control (1), a concentration of IC25 for mitotic block (2), the ICS0 for mitotic block (3), and twice the IC50 for the mitotic block (4); 5-8, corresponding to DNA images for the attached panels. There were no normal, bipolar spindles in IC25 for KPU-02 or CA4 (Figures 53A and B). Cells treated with compound had monopolar or bipolar spindles with non-congregating chromosomes. By contrast, normal bipolar spindles persist in IC25 for CLC (Figure 53C). In IC50 for KPU-02, 75% of the mitotic cells contain foci or tubulin esters, and the remaining cells had no detectable mitotic polymer. In the presence of CLC, half of the cells were bipolar with non-clustered chromosomes and the remaining half were monopolar. At double IC50 concentrations for mitotic block, there was little detectable MT polymer in mitotic cells treated as KPU-02, CA4, or CLC. [0551] The microtubule interface sets were more resistant to depolymerization than the achromatic spindles for all compounds examined (Figure 52). However, a qualitative decrease in polymers is observed in a dose-dependent manner for all three compounds (Figure 54 A-C). Figure 54A-C illustrates immunofluorescence microscopy images of MCF7 cells treated with KPU-02 (a), CA4 (b), and CLC (c) for 20 hours.
Depolymerization of MT interface with increasing concentration of drugs. 1-4, Alpha and beta tubulin in control (1), a concentration IC25 for mitotic block (2), IC50 for mitotic block (3), 2X IC50 for mitotic block (4), - 5-8 corresponding DNA images for the attached panels. Supposedly, tubulin is sequestered in these interphase cells despite the presence of intracellular stabilizing MAPs, such as tubulin rich in MAP are sequestered in in vitro polymer mass assays. Missing or modulating MT dynamic instability in live MCF7 cells [0552] KPU-02, as well as CA4 does not have a measurable effect on the dynamic instability of MT at concentrations affecting 25% (Table 24) or 50% (Table 25) of the maximum mitotic block in MCF7 cells. Without being bound by any particular theory these data suggest that the anti-proliferative mechanism of action KPU-02 (and CA4) is primarily due to inhibition of MT polymerization rather than suppression of microtubule dynamics. Table 24. Dynamic instability in the mitotic block Table 24 (continued) Table 25. MV instability in the mitotic block Table 25 (continued)
[0553] The examples described above are set forth only to assist in the understanding of the invention. In this way, a person skilled in the art will appreciate that the methods and compounds described encompass and otherwise may provide additional dehydrophenylahhistine derivatives. [0554] A person skilled in the art will readily appreciate that the present invention is well adapted to obtain, for example, the purposes and advantages mentioned, as well as other inherent ones. The methods and methods described herein are representative of preferred embodiments and are exemplary and are not intended as limitations on the scope of the invention. Changes there and other uses will occur to those with skill in the art that are encompassed within the spirit of the invention. [0555] It will be readily apparent to a person skilled in the art that variants substitutions and modifications can be made in the invention described herein, without departing from the scope and spirit of the invention. [0556] As shown above, all patents and publications mentioned in this specification are indicative of the levels of those skilled in the art to which the invention relates. All patents and publications herein are incorporated by reference to the extent permitted by law, such that each patent and individual publication may be treated as specifically and individually indicated to be incorporated by reference. [0557] The invention described in illustrative form herein, conveniently may be practiced in the absence of any element or elements, limitation or limitations that are not specifically described herein. The terms and expressions that have been used, are used as terms of description and not limitation, and there is no intention that the use of these terms and expressions indicate the exclusion of equivalents of the characteristics described and described or their portions. It is recognized that various modifications are possible within the scope of the invention. Thus, it will be understood that although the present invention has been specifically described by modalities, preferences and characteristics, modifications and optional variations of the concepts described herein may be contemplated by those skilled in the art and that said modifications and variations are considered which fit within the scope of the invention.

Claims (41)

  1. CLAIMS 1. A compound that has the structure of the formula (I) s wherein: Rl7 R4, and R6, each is selected separately from the group consisting of a hydrogen atom, a halogen atom and Cx-C2 saturated alkyl, C? -C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy , cycloalkoxy, aryl, substituted aryl, heteroaryl, or substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl groups which includes polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7 / wherein R7 is selected from a hydrogen atom, a halogen atom and saturated L-C24 alkyl groups, C? -C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy , cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; Rx 'and Ri "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and L-C24 saturated alkyl, unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl groups. , heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? -C2 saturated alkyl, C? ~ C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl: R, Rx 'and Ri "are already covalently bound to each other or not; R2, R3, and R5 are each chosen separately from the group consisting of a hydrogen atom, a halogen atom and C groups -C12 alkyl, C2C2 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy , aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, substituted sulfonyl and sulfonyl groups; Xx and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above; Y is selected from the group consisting of a nitrogen atom, a nitrogen atom substituted with R 5 of the upper part, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; n is an integer equal to zero, one or two; Z, for each separate n, if not zero, and Zx, Z2, Z3 and Z4 are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and dotted links can already be single or double links; and where the compound induces vascular collapse.
  2. 2. The compound according to claim 1, characterized in that the first aldehyde is an imidazolecarboxaldehyde.
  3. 3. The compound according to claim 1, characterized in that the second aldehyde is a benzaldehyde. .
  4. The compound according to claim 1, characterized in that each of R2, R3, R5 and R6 is a hydrogen atom.
  5. 5. The compound according to claim 1, characterized by each of Xx and X2 is an oxygen atom.
  6. 6. The compound according to claim 1, characterized in that R4 is a saturated C? -C1 alkyl.
  7. 7. The compound according to claim 6, characterized in that the saturated Cx-C12 alkyl is a tertiary butyl group.
  8. 8. The compound according to claim 1, characterized in that Rx comprises a substituted phenyl.
  9. 9. The compound according to claim 8, characterized in that the substituted phenyl group is methoxybenzene.
  10. 10. The compound according to claim 1, characterized in that the first aldehyde is a benzaldehyde.
  11. 11. The compound according to claim 1, characterized in that the second aldehyde is an imidazolecarboxaldehyde.
  12. 12. The compound according to claim 1, characterized in that n is equal to zero or one.
  13. 13. The compound according to claim 1, characterized in that n is equal to one.
  14. 14. The compound according to claim 1, characterized in that n is equal to one and Z, Zi, Z2, Z3 and Z4 are each a carbon atom.
  15. A method for treating a condition in an animal, comprising administering to the animal the compound of claim 1, in an amount that is effective to reduce vascular proliferation or in an amount effective to reduce vascular density.
  16. 16. The method according to claim 15, characterized in that the condition is selected from the group consisting of immune and non-immune inflammation, rheumatoid arthritis, chronic articular rheumatism, psoriasis, diabetic retinopathy, neovascular glaucoma, premature retinopathy, macular degeneration, rejection of corneal graft, retrolental fibroplasia, rubeosis, capillary proliferation in atherosclerotic plaques and osteoporosis.
  17. 17. The method according to claim 15, characterized by the condition is a neoplastic condition.
  18. 18. The method according to claim 17, characterized in that the neoplastic condition is cancer.
  19. 19. The method according to claim 15, characterized in that the condition is not cancer.
  20. 20. The method according to claim 15 or 16, characterized in that the condition is a retinopathy.
  21. 21. The method according to claim 20, characterized in that the retinopathy is diabetic retinopathy.
  22. 22. The method according to claim 20, characterized in that retinopathy is an age-related macular degeneration.
  23. 23. The method according to claim 15, characterized in that the animal is a human.
  24. 24. The method according to claim 15, characterized in that the compound is KPU-02.
  25. 25. The method according to claim 15, characterized in that the condition is a condition associated with hypervascularization.
  26. 26. A method for inducing vascular collapse in an animal, comprising treating the animal with a therapeutically effective amount of a compound of Formula (I), 0) characterized in that the therapeutically effective amount of compound causes depolymerization of tubulin in the vasculature, and wherein the compound has the following structure: wherein Rx, R4, and R6, each is separately chosen the group consisting of one atom of hydrogen, a halogen atom and C? -C24 saturated alkyl, C? -C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl groups including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7 / wherein R7 is selected from an atom of hydrogen, a halogen atom and C groups - C24 saturated alkyl, Cx-C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitr or, azido, substituted nitro, phenyl, and substituted phenyl; i 'and Ra "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and C groups - C24 saturated alkyl, Cx-C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl , heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? -C24 saturated alkyl, unsaturated alkenyl Ca-C24, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, heteroaryl substituted, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; R, Ri 'and Ri "are already covalently bound to each other or not; R2, R3, and R5 are each selected separately from the group consisting of a hydrogen atom, a halogen atom, and Cx-C12 alkyl, unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, aryl groups substituted, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, sulfonyl and substituted sulfonyl groups; i and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above; Y is selected from the group consisting of a nitrogen atom, a nitrogen atom substituted with R 5, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; n is an integer equal to zero, one or two; Z, for each separate n, if not zero, and Zx, Z2, Z3 and Z are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and dotted links can already be single or double links.
  27. 27. The method according to claim 26, characterized in that the animal is a human.
  28. 28. The method according to claim 26, characterized in that the human has a disease selected from the group consisting of tumor, diabetic retinopathy, and macular degeneration related to age.
  29. 29. The method according to claim 26, characterized in that the disease is not cancer.
  30. 30. The method according to claim 26, characterized in that the compound is KPU-02.
  31. 31. A pharmaceutical composition for treatment or prevention of vascular proliferation comprising a pharmaceutically effective amount of a compound according to claim 1 together with a pharmaceutically acceptable carrier.
  32. 32. The composition according to claim 31, characterized in that the vascular proliferation is a symptom of a disease selected from cancer, an age-related macular degeneration and diabetic retinopathy.
  33. 33. A method for preferential targetting in tumor vasculature against tissue vasculature without tumor, characterized in that it comprises: administering to an animal a compound having the structure of Formula (I), wherein: Rx, R4, and R6, each is separately chosen the group consisting of a hydrogen atom, a halogen atom and C groups -C 24 saturated alkyl, unsaturated C 2 -C 2 alkenyl, alkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl , substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl- CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and saturated Cx-C24 alkyl, unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl groups do, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; Ri 'and Rx "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and C groups ~ C24 saturated alkyl, Cx-C2 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl , heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -C0-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated algayl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and saturated L-C2 alkyl, Cx-C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl , amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl: R, Rx 'and Ri "are already covalently bound to each other or not; R2, R3, and R5 are each independently selected from the group consisting of a hydrogen atom, a halogen atom, and L-CI2 alkyl, C? -C12 unsaturated alkenyl, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl groups, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, substituted sulfonyl and sulfonyl groups; i and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above; Y is selected from the group consisting of a nitrogen atom, a nitrogen atom substituted with R 5, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a substituted methylene group; n is an integer equal to zero, one or two; Z, for each separate n, if not zero, and Zx, Z2, Z3 and Z4 are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and dotted links can already be single or double links.
  34. 34. The method according to claim 33, characterized in that the non-tumor tissue is selected from the group consisting of skin, muscle, brain, kidney, heart, spleen and intestines.
  35. 35. The method according to claim 33, characterized in that the tumor vasculature is preferentially directed on vasculature of non-tumor tissue by approximately 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% and 90%.
  36. 36. The method according to claim 33, characterized in that the animal is a human.
  37. 37. A method for preferential targetting in tumor vasculature against tissue-free vasculature, characterized in that it comprises administering to an animal an agent that preferentially targets tumor vasculature against tissue vasculature without tumor.
  38. 38. Use of a compound having the structure of Formula (I) in the preparation of a medicament for the treatment of a condition associated with increased vasculature or based on vasculature, wherein: Rx, R4, and Re, each one is selected separately from the group consisting of a hydrogen atom, a halogen atom and saturated L-C24 alkyl, C? -C24 unsaturated alkenyl, alkyl alkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO -R7, wherein R7 is selected from a hydrogen atom, a halogen atom and C? -C24 saturated alkyl, unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl groups , amino, a substituted mino, nitro, azido, substituted nitro, phenyl, and substituted phenyl; i 'and Rx "is independently selected from the group consisting of a hydrogen atom, a halogen atom, and C groups - C24 saturated alkyl, C? -C24 unsaturated alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, aryl substituted, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl, hydroxy, carboxy, -CO-0-R7, cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R7, wherein R7 is selected from a hydrogen atom, a halogen atom and Cx-C24 saturated alkyl, unsaturated alkenyl L-C2, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, heteroaryl substituted, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl: R, Rx 'and Ri "are already covalently bound to each other or not; R2, R3, and Rs are each independently selected from the group consisting of a hydrogen atom, a halogen atom and C groups - C2 alkyl, unsaturated alkenyl L-CI2, acyl, cycloalkyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, and substituted nitro, substituted sulfonyl and sulfonyl groups; Xi and X2 are selected separately from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, each either unsubstituted or substituted with a group R5 as defined above; And it is chosen from the group consisting of a nitrogen atom, a nitrogen atom substituted with a group R5 of the upper part, an oxygen atom, a sulfur atom, an oxidized sulfur atom, a methylene group and a methylene group replaced; n is an integer equal to zero, one or two; Z, for each separate n, if not zero, and Zi, Z2, Z3 and Z4 are each chosen separately from a carbon atom, sulfur atom, nitrogen atom or oxygen atom; and dotted links can already be single or double links.
  39. 39. The use according to claim 38, characterized in that the condition is cancer.
  40. 40. The use according to claim 38, characterized in that the condition is not cancer.
  41. 41. The use according to claim 38, characterized in that the condition is chosen from the group consisting of immune and non-immune inflammation, rheumatoid arthritis, chronic articular rheumatism, psoriasis, diabetic retinopathy, neovascular glaucoma, premature retinopathy, macular degeneration, graft rejection. corneal, retrolental fibroplasia, rubeosis, capillary proliferation in atherosclerotic plaques and osteoporosis.
MXPA/A/2006/008810A 2004-02-04 2006-08-03 Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof MXPA06008810A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60/542,073 2004-02-04
US60/624,262 2004-11-01

Publications (1)

Publication Number Publication Date
MXPA06008810A true MXPA06008810A (en) 2006-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
AU2005212399B2 (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
US7935704B2 (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
JP4616649B2 (en) Synthesis of dehydrophenylahistine and their analogs, and dehydrophenylahistine and their analogs
US8247552B2 (en) Analogs of dehydrophenylahistins and their therapeutic use
US8129527B2 (en) Analogs of dehydrophenylahistins and their therapeutic use
WO2007035841A9 (en) Analogs of dehydrophenylahistins and their therapeutic use
ZA200607151B (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
WO2011084962A1 (en) Analogs of dehydrophenylahistins
MXPA06008810A (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof