EP1711487A1 - Dehydrophenylahistines et leurs analogues et la synthese des dehydrophenylahistines et de leurs analogues - Google Patents

Dehydrophenylahistines et leurs analogues et la synthese des dehydrophenylahistines et de leurs analogues

Info

Publication number
EP1711487A1
EP1711487A1 EP05712910A EP05712910A EP1711487A1 EP 1711487 A1 EP1711487 A1 EP 1711487A1 EP 05712910 A EP05712910 A EP 05712910A EP 05712910 A EP05712910 A EP 05712910A EP 1711487 A1 EP1711487 A1 EP 1711487A1
Authority
EP
European Patent Office
Prior art keywords
substituted
atom
heteroaryl
amino
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP05712910A
Other languages
German (de)
English (en)
Inventor
Michael A. Palladino
George Kenneth Lloyd
Yoshio Hayashi
Benjamin Nicholson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nereus Pharmaceuticals Inc
Original Assignee
Nereus Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nereus Pharmaceuticals Inc filed Critical Nereus Pharmaceuticals Inc
Publication of EP1711487A1 publication Critical patent/EP1711487A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • 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 procedures for making compounds useful in the treatment of cancer and the treatment of fungal infections.
  • Fungi especially pathogenic fungi and related infections, represent an increasing clinical challenge.
  • Existing antifungal agents are of limited efficacy and toxicity, and the development and/or discovery of strains of pathogenic fungi that are resistant to drags currently available or under development.
  • 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.
  • Blastomyces dermatitidis including Blastomyces dermatitidis; Pneumocystis carinii; Coccidioides immitis; Basidiobolus ranarum; Conidiobolus spp.; Histoplasma capsulatum; Rhizopus spp. including R. oryzae and R. microsporus; Cunninghamella spp.; Rhizomucor spp.; Paracoccidioides brasiliensis; Pseudallescheria boydii; Rhinosporidium seeberi; and Sporothrix schenckii (Kwon-Chung, KJ. & Bennett, J.E. 1992 Medical Mycology, Lea and Febiger, Malvern, PA).
  • tryprostatins A and B which are diketopiperazines consisting of proline and isoprenylated tryptophan residues
  • five other structurally-related diketopiperazines inhibited cell cycle progression 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 the microtubule assembly, see Usui, T. et al. 1998 Biochem J 333:543-48; Kondon, M. et al. 1998 J Antibiotics 51:801-04.
  • CLC-site colchicine binding-site
  • tubulin which is a macromolecule that consists of two 50 kDa subunits ( ⁇ - and ⁇ -tubulin) and is the major constituent of microtubules.
  • CLC-site colchicine binding-site
  • tubulin which is a macromolecule that consists of two 50 kDa subunits ( ⁇ - and ⁇ -tubulin) and is the major constituent of microtubules.
  • Microtubules are thought to be involved in several essential cell functions, such as axonal transport, cell motility and determination of cell morphology.
  • inhibitors of microtubule function may have broad biological activity, and be applicable to medicinal and agrochemical purposes.
  • colchicine (CLC)-site ligands such as CLC, steganacin, see Kupchan, S.M. et al, 1973 J Am Chem Soc 95:1335-36, podophyllotoxin, see Sackett, D.L., 1993 Pharmacol Ther 59:163-228, and combretastatins, 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.
  • CLC colchicine
  • diketopiperazine-type metabolites have been isolated from various fungi as mycotoxins, 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 Meth 25:5-17, little is known about the specific structure of the diketopiperazine-type metabolites or their derivatives and their antitumor activity, particularly in vivo.
  • Ri, R , and R 6 are each separately selected from the group consisting of a hydrogen atom, a halogen atom, and saturated C]-C 24 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 groups, hydroxy, carboxy, -CO-O-R 7 , cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R , wherein R is selected from a hydrogen atom, a halogen atom, and saturated C ⁇ -C 2 alkyl, uns
  • the methods comprise the steps of: [0020] reacting a diacyldiketopiperazine with a first aldehyde to produce an intermediate compound; and [0021] reacting said intermediate compound with a second aldehyde to produce said class of compounds with said generic structure, wherein [0022] said first aldehyde and said second aldehydes are selected from the group consisting of an oxazolecarboxaldeyhyde, imidazolecarboxaldehyde, a benzaldehyde, imidazolecarboxaldehyde derivatives, and benzaldehyde derivatives, thereby forming a compound wherein [0023]
  • the disclosed compounds have the structure of Formula (I) wherein: [0024] Ri, R 4 , and R 6 , are each separately selected from the group consisting of a hydrogen atom, a halogen atom, and saturated C ⁇ -C 2 alkyl, unsaturated C ⁇ -C 24 alkenyl,
  • the imidazolecarboxaldehyde is 5-(l,l-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 methylthiophen group, and methods of making such compounds, as well as the compounds described in Tables 2, 3 and 4, as well as methods of making such compounds.
  • Also disclosed are methods and materials for treating neoplastic tissue or preventing cancers or infection by a pathogenic fungus.
  • methods and materials are particularly well suited for treatment of mammalian subjects, more particularly humans, and involve administering to the subject a dehydrophenylahistin or its analog.
  • the method comprises administering to the subject a composition comprising an effective antitumor or antifungal amount of a dehydrophenylahistin or its analog.
  • Further embodiments relate to methods for treating a condition in an animal, which methods can 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 to reduce vascular density.
  • Exemplary conditions include neoplasms, such as cancers, as well as other conditions associated with or which rely upon vascularization, including for example, immune and non-immune inflammation, rheumatoid arthritis, chronic articular rheumatism, psoriasis, diabetic retinopathy, neovascular glaucoma, retinopathy of prematurity, macular degeneration, corneal graft rejection, retrolental fibroplasia, rubeosis, capillary proliferation in atherosclerotic plaques, osteoporosis, and the like, hi some embodiments, the disease is not cancer.
  • Other embodiments relate to methods of inducing vascular collapse in an animal.
  • the methods can include treating said animal with a therapeutically effective amount of a compound of the Formula (I) as described herein, for example.
  • the therapeutically effective amount of said compound can cause tubulin depolymerization in the vasculature.
  • the animal can be a human.
  • the disease can be a tumor, a diabetic retinopathy, an age-related macular degeneration, and the like, hi some aspects the disease is not cancer or cancer can be specifically excluded from the methods and uses.
  • the compound is KPU-02.
  • Still further embodiments relate to pharmaceutical compositions for treating or preventing vascular proliferation comprising a pharmaceutically effective amount of a compound disclosed herein together with a pharmaceutically acceptable carrier therefor.
  • the vascular proliferation can be a symptom of a disease, for example, cancer, age-related macular degeneration and diabetic retinopathy. .
  • Some embodiments relate to methods of preferentially targeting tumor vasculature over non-tumor tissue vasculature.
  • the methods can include the step of administering to an animal, preferably a human, a compound having the structure of Formula (I) as described herein.
  • the non-tumor tissue can be, for example, skin, muscle, brain, kidney, heart, spleen, gut, and the like.
  • the tumor vasculature can be preferentially targeted over non-tumor tissue vasculature, for example, by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% and 90%.
  • Other embodiments relate to methods of preferentially targeting tumor vasculature over non-tumor tissue vasculature, which methods can include administering to an animal an agent that preferentially targets tumor vasculature over non-tumor tissue vasculature.
  • Further embodiments relate 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 which relies upon vasculature.
  • the condition can be cancer, while in others, cancers particular types or all cancers are specifically excluded.
  • the condition can be any other that is associated with hypervascularization, associated with vasculature or which relies upon vasculature.
  • Figure 1 illustrates a reaction scheme for producing dehydrophenylahistins by reacting a diacyldiketopiperazine 1 with an imidazolecarboxaldeheyde 2 to yield an intermediate compound 3 which is reacted with a benzaldehyde 4 to produce a dehydrophenylahistin.
  • Figure 2 depicts the HPLC profile of the synthetic crude dehydrophenylahistin.
  • Figure 3 illustrates a reaction scheme for producing dehydrophenylahistins by reacting a diacyldiketopiperazine 1 with a benzaldehyde 4 to yield an intermediate compound 17 which is reacted with an imidazolecarboxaldeheyde 15 to produce a dehydrophenylahistin.
  • Figure 4 depicts the HPLC profiles of the crude synthetic tBu- dehyrophenylahistin produced from Route A and from Route B.
  • Figure 5 illustrates two modification strategies for dehydroPLH for potent cytotoxic activity.
  • Figure 6 depicts the putative active conformation of dehydroPLH at the phenyl moiety.
  • Figure 7 depicts Cytochrome P450 metabolism of phenylahistin.
  • Figure 8 illustrates the Z-E migration of tBu-dehydroPLH.
  • Figure 9 depicts the synthesis and prodrug image of acyl-E-tBu- dehydroPLH.
  • Figure 10 depicts the temperature gradient of 3-Z-Benzylidene-6-[5"-(l,l- dimethylallyl)-lH-imidazol-4"-Z-ylmethylene]-piperazine-2,5-dione.
  • Figure 11 depicts the temperature gradient of 3-Z-benzylidene-6-(5"-tert- butyl-lH-imidazol-4"-Z-ylmethylene)-piperazine-2,5-dione.
  • Figure 12 depicts the effect of KPU-2, KPU-35 and t-butyl-phenylahistin in comparison to colchicine and taxol on ⁇ uVEC monolayer permeability to FITC-Dextran.
  • Figure 13 depicts the effect of KPU-2 alone and in combination with CPT- 11 on estimated tumor growth in the ⁇ T-29 Human Colon Tumor Xenograft model.
  • Figure 14 depicts 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 HT-29 Human Colon Tumor Xenograft model.
  • Figure 15 depicts the effect of KPU-2 alone and in combination with CPT- 11 on estimated tumor growth in the HT-29 Human Colon Tumor Xenograft model.
  • Figure 16 depicts 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 HT-29 Human Colon Tumor Xenograft model.
  • Figure 17 depicts the effects of: A. KPU-2, B. KPU-35 and C.
  • Figure 18 depicts the effects of A. KPU-2, B. KPU-35 and C. t-butyl- phenylahistin alone and in combination with CPT-11 on the weight of tumors excised at autopsy in individual mice in the HT-29 Human Colon Tumor Xenograft model.
  • Figure 19 depicts the effects of KPU-2 alone and in combination with CPT-11 on tumor growth in the HT-29 human colon tumor xenograft model: comparison of three studies.
  • Figure 20 depicts the effects of KPU-2 alone and in combination with CPT-11 on final tumor weights in the HT-29 human colon tumor xenograft model: comparison of three studies.
  • Figure 21 depicts the effects of KPU-2 alone or in combination with Taxotere on estimated tumor growth in the DU-145 Human Prostate Tumor Xenograft Model.
  • Figure 22 depicts the effects of A. KPU-2, B. KPU-35 and C. t-butyl- phenylahistin alone and in combination with Taxotere on the estimated tumor growth based on observations made during the in-life portion of the DU-145 Human Prostate Tumor Xenograft Model.
  • Figure 23 depicts the effects of KPU-2 alone and in combination with Taxotere on the individual excised tumor weights at autopsy in the DU-145 Human Prostate Tumor Xenograft Model.
  • Figure 24 depicts the effects of KPU-35 alone and in combination with Taxotere on the individual excised tumor weights at autopsy in the DU-145 Human Prostate Tumor Xenograft Model.
  • Figure 25 depicts the effects of A. KPU-2, B. KPU-35 and C. t-butyl- phenylahistin alone and in combination with Taxotere in MCF-7 Human Breast Tumor Xenograft model.
  • Figure 26 depicts the effects of KPU-2 alone and in combination with Taxotere on estimated tumor growth in the A549 Human Lung Tumor Xenograft model.
  • Figure 27 depicts the effects of KPU-2 alone and in combination with Taxotere on the excised tumor weights at autopsy in the A549 Human Lung Tumor Xenograft model.
  • Figure 28 depicts the effects of KPU-2 alone and in combination with Paclitaxel on estimated tumor weight in the murine mammary fat pad implanted MDA-231 Human Breast Tumor model.
  • Figure 29 depicts effects of A. KPU-2, B. KPU-35 and C.
  • Figure 30 depicts effects of KPU-35 and KPU-02 on tumor vasculature in the dorsal skinfold chamber of Figure 30.
  • Figure 31 depicts effect of KPU-02 in combination with CPT-11 on the estimated tumor weight in the HT-29 human colon tumor xenograft model.
  • Figure 32 depicts effect of KPU-02 in combination with CPT-11 on the excised tumor weight in the HT-29 human colon tumor xenograft model.
  • Figure 33 depicts rapid tubulin depolymerization in HuVEC cells induced by KPU-02 and KPU-35.
  • Figure 34 depicts effect of KPU-02 on monolayer permeability in HuNEC cells.
  • Figure 35 depicts the effect of KPU-02 on tumor blood flow in the P22 rat sarcoma model using the 125I-IAP technique.
  • Figure 36 depicts the effect of KPU-02 15 mg/kg IP (expressed as % vehicle control) on blood flow in different tissues 1 and 24 hours post-dose.
  • Figure 37 depicts the tumor necrosis induced by KPU-02 7.5 and 15.0 mg/kg IP in the P22 rat sarcoma model
  • Figure 38 lists the activity of various fBu-dehydro-PLH derivatives at HT- 29 cells.
  • Figure 39 depicts 3D QSAR (CoMFA) analysis of fBu-dehydro-PLH derivatives.
  • Figure 40 depicts X-ray crystallographic analysis of fBu-dehydro-PLH derivatives.
  • Figure 41 depicts the biologically activity of various phenylahistin derivatives compared to colchicine.
  • Figure 42 depicts the effect on cell cycle progression of HeLa cells by tBu- dehydro-PLH (KPU-2) and KPU-35.
  • Figure 43 depicts the effect of dehydro-PLH and fBu-dehydro-PLH (KPU- 2) on drug-sensitive and drug-resistant tumor cell lines as compared to paclitaxel.
  • Figure 44A depicts turbidity spectra of microtubule protein polymerization in the presence of DMSO drag vehicle (0), 1.25 ⁇ M (D), 2.5 ⁇ M (— ), and 5 ⁇ M (o) KPU-02.
  • Figure 44B depicts turbidity spectra of microtubule protein polymerization in the presence of DMSO drag vehicle (0), 1.25 ⁇ M (D), 2.5 ⁇ M (— ), and 5 ⁇ M (o) CA4.
  • Figure 44C depicts turbidity spectra of microtubule protein polymerization in the presence of DMSO drag vehicle (0), 1.25 ⁇ M ( ⁇ ), 2.5 ⁇ M (— ), and 5 ⁇ M (o) CLC.
  • Figure 45 depicts inhibition of MT in the absence or presence of a range of KPU-02 (o),CA4 (G), and colchicine (0) concentrations.
  • Figure 46A depicts frequency histograms of mean microtubule lengths in vitro at steady state in the presence of KPU-02.
  • Figure 46B depicts frequency histograms of mean microtubule lengths in vitro at steady state in the presence of CA4.
  • Figure 46C depicts frequency histograms of mean microtubule lengths in vitro at steady state in the presence of CLC.
  • Figure 47A depicts electron micrographs of MAP-rich microtubules formed in vitro at steady state in the presence of KPU-02.
  • Figure 47B depicts electron micrographs of MAP-rich microtubules formed in vitro at steady state in the presence of CA4.
  • Figure 47C depicts electron micrographs of MAP-rich microtubules formed in vitro at steady state in the presence of CLC.
  • Figure 48 depicts a graphical summary of MT length decrease at steady state in the presence of KPU-02, CA4, and colchicine.
  • Figure 49A depicts fluorescence emission spectra of tubulin in the presence of increasing KPU-02.
  • Figure 49B depicts a fit to fluorescence emission maxima at 487 nm to obtain the Ka of tubulin for KPU-02. The inset depicts residuals.
  • Figure 49C depicts double reciprocal transformation of the binding data.
  • Figure 50 depicts the graphical results of a competitive inhibition assay of colchicine binding to tubulin with various concentrations of [ 3 H]CLC in the absence (0), or presence of 10 ⁇ M KPU-02 (o) or 10 ⁇ M CA4 ( ⁇ ).
  • Figure 51 depicts log [compound] response curves for mitotic progression inhibition by KPU-02, CA4, and CLC in MCF7 cells cultured in the presence of KPU-02 (o), CA4 ( ⁇ ), and colchicine (0).
  • Figure 52 depicts immunofluorescence microscopy images of MCF7 cells, a-d: Tubulin in control— (a) Tubulin in control, (b) KPU-02, (c) CA4, and (d) CLC treated cells; e-h: DNA in control— (e) DNA in control, (f) KPU-02, (g) CA4, and (h) CLC treated cells.
  • Figure 53A depicts immunofluorescence microscopy images of MCF7 cells treated with KPU-02
  • Figure 53B depicts immunofluorescence microscopy images of MCF7 cells treated with CA4.
  • Figure 53C depicts immunofluorescence microscopy images of MCF7 cells treated with CLC.
  • Figure 54A depicts immunofluorescence microscopy images of MCF7 cells treated with KPU-02
  • Figure 54B depicts immunofluorescence microscopy images of MCF7 cells treated with CA4.
  • Figure 54C depicts immunofluorescence microscopy images of MCF7 cells treated with CLC.
  • compounds are identified using an alternative designation. A complete chart to convert these alternative designations is as follows:
  • the disclosure provides methods for the synthetic preparation of compounds, including novel compounds, including dehydrophenylahistin and dehydrophenylahistin analogs, and provides methods for producing pharmaceutically acceptable cell cycle inhibitors, antitumor agents and antifungal agents in relatively high yield, wherein said 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 not obtainable by currently available, non-synthetic methods. It is also an object to provide a method of treating cancer, particularly human cancer, comprising the step of administering an effective tumor-growth inhibiting amount of a member of a class of new anti-tumor 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 anti-fungal amount of a member of a class of new anti-fungal compounds, e.g., administering a dehydrophenylahistin or its analog in an amount and manner which provides the intended antifungal effect.
  • an effective anti-fungal amount of a member of a class of new anti-fungal compounds e.g., administering a dehydrophenylahistin or its analog in an amount and manner which provides the intended antifungal effect.
  • R ⁇ R 4 , and R 6 are each separately selected from the group consisting of a hydrogen atom, a halogen atom, and saturated C ⁇ -C 24 alkyl, unsaturated C ⁇ -C 24 alkenyl, cycloalkyl, cycloalkenyl, alkoxy, cycloalkoxy, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, substituted amino, nitro, azido, substituted nitro, phenyl, and substituted phenyl groups, hydroxy, carboxy, -CO-O-R , cyano, alkylthio, halogenated alkyl including polyhalogenated alkyl, halogenated carbonyl, and carbonyl -CCO-R 7 , wherein R 7 is selected from a hydrogen atom, a halogen atom, and saturated C ⁇ -C 24 alkyl, unsaturated C ⁇ -C 4 alkeny
  • the method comprises a method of producing compounds of Formula (I) by the steps of: [0123] reacting a diacyldiketopiperazine with a first aldehyde to produce an intermediate compound; and [0124] reacting said intermediate compound with a second aldehyde to produce said class of compounds with said generic structure, wherein [0125] said first aldehyde and said second aldehydes are selected from the group consisting of an oxazolecarboxaldeyhyde, imidazolecarboxaldehyde, a benzaldehyde, imidazolecarboxaldehyde derivatives, and benzaldehyde derivatives, thereby forming a compound of Formula (I) wherein [0126] Ri, R 4 , and R 6 , are each separately selected from the group consisting of a hydrogen atom, a halogen atom, and saturated C ⁇ -C 24 alkyl, unsaturated Cj-C 24 alkenyl, cycl
  • pro-drug ester especially when referring to a pro-drag ester of the compound of Formula (I) synthesized by the methods disclosed herein, refers to a chemical derivative of the compound that is rapidly transformed in vivo to yield the compound, for example, by hydrolysis in blood or inside tissues.
  • pro-drag ester refers to derivatives of the compounds disclosed herein formed by the addition of any of several ester-forming groups that are hydrolyzed under physiological conditions.
  • pro-drug ester groups examples include pivoyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, as well as other such groups known in the art, including a (5-R-2-oxo-l,3- dioxolen-4-yl)methyl group.
  • pro-drag ester groups can be found in, for example, T. Higuchi and N. Stella, in "Pro-drugs as Novel Delivery Systems", Vol. 14, A.C.S. Symposium Series, American Chemical Society (1975); and "Bioreversible Carriers in Drag Design: Theory and Application", edited by E. B.
  • pro-drag ester also refers to a chemical derivative of the compound that is rapidly transformed in vivo to yield the compound, for example, by hydrolysis in blood.
  • pro-drug ester refers to derivatives of the compounds disclosed herein formed by the addition of any of several ester-forming groups that are hydrolyzed under physiological conditions.
  • pro-drug ester groups examples include pivoyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, as well as other such groups known in the art, including a (5-R-2-oxo-l,3-dioxolen-4-yl)methyl group.
  • Other examples of pro-drug ester groups can be found in, for example, T. Higuchi and N. Stella, in "Pro-drugs as Novel Delivery Systems", Vol. 14, A.C.S. Symposium Series, American Chemical Society (1975); and "Bioreversible Carriers in Drag Design: Theory and Application", edited by E. B.
  • pharmaceutically acceptable salt especially when referring to a pharmaceutically acceptable salt of the compound of Formula (I) synthesized by the methods disclosed herein, refers to any pharmaceutically acceptable salts of a compound, and preferably refers to an acid addition salt of a compound.
  • Preferred examples of 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 ⁇ -C 7 alkylamine, cyclohexylamine, friethanolamine, ethylenediamine or tris-(hydroxymethyl)-aminomethane.
  • the preferred examples of pharmaceutically acceptable salts are acid addition salts of pharmaceutically acceptable inorganic or organic acids, for example, hydrohalic, sulfuric, phosphoric acid or aliphatic or aromatic carboxylic or sulfonic acid, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, p-toluensulfonic or naphthalenesulfonic acid.
  • pharmaceutically acceptable salt also refers to any pharmaceutically acceptable salts of a compound, and preferably refers to an acid addition salt of a compound.
  • Preferred examples of 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 ⁇ -C alkylamine, cyclohexylamine, triethanolamine, ethylenediamine or tris-(hydroxymethyl)-aminomethane.
  • the preferred examples of pharmaceutically acceptable salts are acid addition salts of pharmaceutically acceptable inorganic or organic acids, for example, hydrohalic, sulfuric, phosphoric acid or aliphatic or aromatic carboxylic or sulfonic acid, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, p- toluensulfonic or naphthalenesulfonic acid.
  • Preferred pharmaceutical compositions disclosed herein include pharmaceutically acceptable salts and pro-drug esters of the compound of Formula (I) synthesized by the method disclosed herein.
  • a relatively rigid, planar pseudo three-ring structure may be formed.
  • R may preferably be chosen to be hydrogen.
  • n is equal to zero or one, more preferable one, and Z 2 , Z , and Z , and each separately selected from an oxygen atom, a nitrogen atom, and a carbon atom, more preferable at one least one of Z 2 , Z 3 , and Z 4 being a carbon atom, and most preferable at least two of Z 2 , Z 3 , and Z 4 being a carbon atom. All Z's may simultaneous be carbon atoms.
  • Still other preferred embodiments of the methods and compositions disclosed herein involve compounds having the stractures of Formulae (la) and (lb), below:
  • variable groups are as defined herein.
  • halogen atom means any one 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.
  • alkyl means any unbranched or branched, substituted or unsubstituted, saturated hydrocarbon, with C ⁇ -C 6 unbranched, saturated, unsubstituted hydrocarbons being preferred, with methyl, ethyl, iosbutyl, and tert-butyl being most preferred.
  • substituted saturated hydrocarbons
  • C ⁇ -C 6 mono- and di- and per-halogen substituted saturated hydrocarbons and amino-substituted hydrocarbons are preferred, with perfluromethyl, perchloromethyl, perfluoro-tert-butyl, and perchloro-tert-butyl being the most preferred.
  • substituted has its ordinary meaning, as found in numerous contemporary patents from the related art. See, for example, U.S. Patent 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.
  • substituted is as broad as that provided in U.S. Patent No. 6,583,143, which defines the term substituted as any groups such as alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle and heterocyclealkyl, wherein at least one hydrogen atom is replaced with a substituent.
  • substituted is also as broad as the definition provided in U.S. Patent No.
  • substituted alkyl such that it refers to an alkyl group, preferably of from 1 to 10 carbon atoms, having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from 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, thioketo, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, --SO-alkyl, --SO
  • cycloalkyl refers to any non- aromatic hydrocarbon ring, preferably having five to twelve atoms comprising the ring.
  • acyl refers to alkyl or aryl groups derived from an oxoacid, with an acetyl group being preferred.
  • alkenyl means any unbranched or branched, substituted or unsubstituted, unsaturated hydrocarbon including polyunsaturated hydrocarbons, with Ci-C 6 unbranched, mono-unsaturated and di-unsaturated, unsubstituted hydrocarbons being preferred, and mono-unsaturated, di-halogen substituted hydrocarbons being most preferred.
  • a z- isoprenyl moiety is particularly preferred.
  • cycloalkenyl refers to any non- aromatic hydrocarbon ring, preferably having five to twelve atoms comprising the ring.
  • aryl refers to aromatic hydrocarbon rings, preferably having five, six, or seven atoms, and most preferably having six atoms comprising the ring.
  • heteroaryl refers to aromatic hydrocarbon rings in which at least one heteroatom, e.g., oxygen, sulfur, or nitrogen atom, is in the ring along with at least one carbon atom.
  • alkoxy refers to any unbranched, or branched, substituted or unsubstituted, saturated or unsaturated ether, with C ⁇ -C 6 unbranched, saturated, unsubstituted ethers being preferred, with methoxy being preferred, and also with dimethyl, diethyl, methyl-isobutyl, and methyl-tert-butyl ethers also being preferred.
  • cycloalkoxy refers to any non-aromatic hydrocarbon ring, preferably having five to twelve atoms comprising the ring.
  • the terms “purified,” “substantially purified,” and “isolated” as used herein refer to the compound being free of other, dissimilar compounds with which the compound is normally associated in its natural state, so that the compound of the invention comprises at least 0.5%>, 1%, 5%>, 10%, or 20%, and most preferably at least 50% or 75%> of the mass, by weight, of a given sample.
  • the compound of Formula (I) may be chemically synthesized or produced from reagents known and available in the art.
  • diacyldiketopiperazine modifications 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 diacyldiketopiperazine may be prepared, for example, by diacetylation of inexpensive 2,5-piperazinedione (TCI Cat. No. G0100, 25 g) with sodium acetate and sodium anhydride.
  • the diacetyl structure of the activated deketopiperazine can be replaced with other acyl groups, to include carbamates such as Boc (t-butoxycarbonyl), Z (benzoyloxycarbonyl).
  • the imidazolecarboxaldehyde may be prepared, for example, according the procedure disclosed in Hayashi et al, 2000 J Organic Chem 65: 8402 as depicted below: Tos-CI . V r. . DBU P fM ⁇ e, rt. " Y Q Ts O refluxCWC) ⁇ %
  • the synthetic method disclosed herein may be preferably performed in the presence of cesium carbonate as a base in DMF and in a deoxygenated atmosphere.
  • the inert atmosphere circumvents the probable oxidation of activated ⁇ -carbon atoms of the diketopiperazine ring during the treatment with cesium carbonate (see below) as reported, for example, by Watanabe et al, 18 th International Congress of Heterocyclic Chemistry in Yokohama, Japan (30 July 2001), Abstract, page 225.
  • Other embodiments of the synthetic method involve modifications to the compounds used in or otherwise involved in the synthesis of compounds represented by Formula (I).
  • Such 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 such modifications are discussed, for example, in Example 4.
  • the result of such modifications includes increased nitrogen content of the phenyl ring and/or the compound which may increase compound solubility.
  • Other modifications may incorporate derivatives of known tubulin inhibitors, thereby mimicking the activity of the tubulin inhibitors.
  • compositions [0154] The present invention also encompasses the compounds disclosed herein, optionally and preferably produced by the methods disclosed herein, in pharmaceutical compositions comprising a pharmaceutically acceptable carrier prepared for storage and subsequent adminisfration, which have a pharmaceutically effective amount of the products disclosed above in a pharmaceutically acceptable carrier or diluent.
  • a pharmaceutically acceptable carrier prepared for storage and subsequent adminisfration, which have a pharmaceutically effective amount of the products disclosed 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 edit. 1985).
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • sodium benzoate, ascorbic acid and esters of p- hydroxybenzoic acid may be added as preservatives.
  • antioxidants and suspending agents may be used.
  • the dehydrophenylahistin or dehydrophenylahistin analog compositions may be formulated and used as tablets, capsules, or elixirs for oral administration; suppositories for rectal administration; sterile solutions, suspensions for injectable adminisfration; patches for transdermal administration, and sub-dermal deposits and the like.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to 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.
  • the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like.
  • absorption enhancing preparations for example, liposomes, may be utilized.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may 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 soybean, grapefruit or almond oils, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions for oral use may be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PNP).
  • fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol
  • cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • concenfrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Such formulations can be made using methods known in the art (see, for example, U.S. Patent os.
  • compositions well known in the pharmaceutical art for uses that include intraocular, intranasal, and intraauricular delivery.
  • Pharmaceutical formulations include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eyedrops, or in gellan 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 microparticulates, drug- containing small polymeric particles that 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.
  • Such suitable pharmaceutical formulations are most often and preferably formulated to be sterile, isotonic and buffered for stability and comfort. Pharmaceutical compositions may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action. As disclosed in Remington's Pharmaceutical Sciences (Mack Publishing, 18 th Edition), and well-known to those skilled in the art, suitable formulations are most often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers. Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for such aural formulations include glycerin and water.
  • the compound of Formula (I) can be administered by either oral or a non-oral pathways.
  • it can be administered in capsule, tablet, granule, spray, syrup, or other such form.
  • it can be administered as an aqueous suspension, an oily preparation or the like or as a drip, suppository, salve, ointment or the like, when administered via injection or infusion, subcutaneously, mtreperitoneally, intravenously, intramuscularly, or the like.
  • the present invention also encompasses methods for making and for administering the disclosed chemical compounds and the disclosed pharmaceutical compositions.
  • Such disclosed methods include, among others, (a) administration though oral pathways, which administration includes administration in capsule, tablet, granule, spray, syrup, or other such forms; (b) administration through non-oral pathways, which administration includes administration as an aqueous suspension, an oily preparation or the like or as a drip, suppository, salve, ointment or the like; administration via injection or infusion, subcutaneously, intraperitoneally, intravenously, inframuscularly, infradermally, or the like; as well as (c) administration topically, (d) administration rectally, or (e) adminisfration vaginally, as deemed appropriate by those of skill in the art for bringing the compound into contact with living tissue; and (f) administration via controlled released formulations, depot formulations, and infusion pump delivery.
  • the disclosed herein are various methods for administration of the disclosed chemical compounds and pharmaceutical compositions including modes of adminisfration through intraocular, intranasal, and intraauricular pathways.
  • the pharmaceutically effective amount of the dehydrophenylahistin or dehydrophenylahistin analog composition required as a dose will depend on the route of administration, the type of animal, including human, being freated, and the physical characteristics of the specific animal under consideration.
  • the dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.
  • the products or compositions can be used alone or in combination with one another, or in combination with other therapeutic or diagnostic agents.
  • the compounds disclosed herein are effective in the treatment of cancer when used in combination with other actives, specifically other chemotherapeutics, for example biologies and the specific chemotherapeutics CPT-11, Taxotene (docataxel) and paclitaxel.
  • the compounds disclosed herein are also effective in the treatment of cancer when used in combination with other actives, including anti-vascular agents, anti-angiogenenic agents, such as Erbuitux (Imclone/bristol-Myers) and Iressa (AsfraZeneca), other NEGF inhibitors and biologies, more specifically, at least one anti- NEGF antibodies, especially monoclonal antibodies to the VEGF receptor, including DC 101, a rat monoclonal antibody, which blocks the mouse NEGF receptor 2 (flk-1).
  • actives including anti-vascular agents, anti-angiogenenic agents, such as Erbuitux (Imclone/bristol-Myers) and Iressa (AsfraZeneca), other NEGF inhibitors and biologies, more specifically, at least one anti- NEGF antibodies, especially monoclonal antibodies to the VEGF receptor, including DC 101, a rat monoclonal antibody, which blocks the mouse NEGF receptor 2
  • Such combinations may be utilized in vivo, ordinarily in a mammal, preferably in a human, or in vitro, h employing them in vivo, the disclosed compounds, alone or in combination with other chemotherapeutics or other biologic products, may be administered to the mammal in a variety of ways, including parenterally, intravenously, via infusion or injection, subcutaneously, intramuscularly, colonically, rectally, vaginally, nasally or intraperitoneally, employing a variety of dosage forms. Such methods may also be applied to testing chemical activity in vivo.
  • the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight and mammalian species freated, the particular compounds employed, and the specific use for which these compounds are employed.
  • the determination of effective dosage levels can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes of adminisfration of the compositions identified by the present methods using established pharmacological methods.
  • dosages may range broadly, depending upon the desired affects and the therapeutic indication. Typically, dosages may be between about 10 microgram/kg and 100 mg/kg body weight, preferably between about 100 micro gram/kg and 10 mg/kg body weight. Alternatively dosages may be based and calculated upon the surface area of the patient, as understood by those of skill in the art. Administration may be oral on an every third day, every other day, daily, twice daily, or thrice daily basis. [0165] The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.
  • the magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • Suitable administration routes may include oral, rectal, transdermal, vaginal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, via infusion, intraperitoneal, infranasal, or intraocular injections.
  • the agents may be formulated in aqueous solutions, for example, in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • peneflops appropriate to the barrier to be permeated are used in the formulation.
  • Such peneflops are generally known in the art.
  • Use of pharmaceutically acceptable carriers to formulate the compounds herein disclosed for the practice of the invention into dosages suitable for systemic administration is within the scope of the invention. With proper choice of carrier and suitable manufacturing practice, the compositions disclosed herein, in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection or infusion.
  • the compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral adminisfration. Such carriers enable the compounds to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Agents intended to be administered intracellularly may be administered using techniques well known to those of ordinary skill in the art. For example, such agents may be encapsulated into 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.
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
  • compositions may be manufactured in a manner that is itself known, for example, by means of conventional mixing, dissolving, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • Compounds disclosed herein can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties, may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • the toxicity of particular compounds in an animal model may be determined using known methods.
  • the efficacy of a particular compound may be established using several art recognized methods, such as in vitro methods, animal models, or human clinical trials. Art-recognized in vitro models exist for nearly every class of condition, including the conditions abated by the compounds disclosed herein, including cancer, cardiovascular disease and various fungal infections. Similarly, acceptable animal models may be used to establish efficacy of chemicals to treat such conditions. When selecting a model to determine efficacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, and route of administration, and regime.
  • the compounds disclosed herein may be administered by either oral or a non-oral pathways.
  • it can be administered in capsule, tablet, granule, spray, syrup, or other such form.
  • it can be administered as an aqueous suspension, an oily preparation or the like or as a drip, suppository, salve, ointment or the like, when administered via injection or infusion, subcutaneously, intreperitoneally, infravenously, inframuscularly, infradermally, or the like.
  • the compound may be administered topically, rectally, or vaginally, as deemed appropriate by those of skill in the art for bringing the compound into optimal contact with a tumor, thus inhibiting the growth of the tumor.
  • Local administration at the site of the tumor or other disease condition is also contemplated, either before or after tumor resection, or as part of an art-recognized treatment of the disease condition. Controlled release formulations, depot formulations, and infusion pump delivery are similarly contemplated.
  • an anti-cancer agent or an anti-tumor agent When used as an anti-cancer agent or an anti-tumor agent, may be orally or non-orally administered 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 at, preferably, one time per day or, less preferably, over two to about ten times per day.
  • the compound may preferably be administered in the stated amounts continuously by, for example, an intravenous drip.
  • the preferred daily dose of the active anti-tumor ingredient would be about 0.0007 mg/kg/day to about 35 mg/kg/day including 1.0 mg/kg/day and 0.5 mg/kg/day, and more preferable, from 0.007 mg/kg/day to about 0.050 mg/kg/day, including 0.035 mg/kg/day. Nonetheless, as will be understood by those of skill in the art, in certain situations it may be necessary to administer the anti-tumor compound in amounts that excess, or even far exceed, the above-stated, preferred dosage range to effectively and aggressively treat particularly advanced or lethal tumors.
  • the preferable amount of the dehydrophenylahistin or its analog effective in the freatment or prevention of a particular fungal pathogen will depend in part on the characteristics of the fungus and the extent of infection, and can be determined by standard clinical techniques. In vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. Effective doses may be extrapolated from dose-response curves derived from in vitro analysis or preferably from animal models.
  • the precise dosage level should be determined by the attending physician or other health care provider and will depend upon well known factors, including route of adminisfration, and the age, body weight, sex and general health of the individual; the nature, severity and clinical stage of the infection; the use (or not) of concomitant therapies.
  • the effective dose of the dehydrophenylahistin or its analog will typically be in the range of about 0.01 to about 50 mg/kgs, preferably about 0.1 to about 10 mg/kg of mammalian body weight per day, administered in single or multiple doses. Generally, the compound may be administered to patients in need of such treatment in a daily dose range of about 1 to about 2000 mg per patient.
  • ком ⁇ онентs to formulate the dosage including the compounds disclosed herein as a tumor-growth-inhibiting compound, known surface active agents, excipients, smoothing agents, suspension agents and pharmaceutically acceptable film-forming substances and coating assistants, and the like may be used.
  • alcohols, esters, sulfated aliphatic alcohols, and the like may be used as surface active agents; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium methasilicate aluminate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, calcium carboxymethyl cellulose, and the like may be used as excipients; magnesium stearate, talc, hardened oil and the like may be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soya may be used as suspension agents or lubricants; cellulose acetate phthalate as a derivative of a
  • compositions disclosed herein in a pharmaceutical compositions may also comprise a pharmaceutically acceptable carrier. Such compositions may be prepared for storage and for 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 edit. 1985).
  • compositions may be formulated and used as tablets, capsules or solutions for oral administration; suppositories for rectal or vaginal adminisfration; sterile solutions or suspensions for injectable administration, jectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to 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.
  • the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like. If desired, absorption enhancing preparations (for example, liposomes), may be utilized.
  • the pharmaceutically effective amount of the composition required as a dose will depend on the route of administration, the type of animal being 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 such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.
  • the products or compositions, as described above, may be used alone or in combination with one another, or in combination with other therapeutic or diagnostic agents.
  • the disclosed compounds products may be used alone or in combination with other chemotherapeutics or biologies, including antibodies, for the freatment of cancer, or in combination with other anti-infective for the treatment of fungal infection.
  • These products or compositions can be utilized in vivo or in vitro.
  • the useful dosages and the most useful modes of administration will vary depending upon the age, weight and animal freated, the particular compounds employed, and the specific use for which these composition or compositions are employed.
  • the magnitude of a dose in the management or freatment for a particular disorder will vary with the severity of the condition to be treated and to the route of adminisfration, and depending on the disease conditions and their severity, the compositions may be formulated and administered either systemically or locally.
  • alcohols, esters, sulfated aliphatic alcohols, and the like may be used as surface active agents; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium methasilicate aluminate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, calcium carboxymethyl cellulose, and the like may be used as excipients; magnesium stearate, talc, hardened oil and the like may be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soya may be used as suspension agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or methyiacetate-methacrylate copolymer as a derivative of polyvinyl may be used as suspension agents; and plasticizers such as ester phthalates and the like may be used as suspension agents.
  • the cell cycle inhibitors, the antitumor agents, and the antifungal agents that may be produced by the method may be orally of non-orally administered to a human patient in the 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 at, preferably, once every three days on a cyclic basis, once every other day, one time per day, twice per day, or less preferably, over two to about ten times per day.
  • the compound produced by the method may preferably be administered in the stated amounts continuously by, for example, an intravenous drip.
  • the preferred daily dose of the active anti-tumor ingredient would be about 0.07 mg/day to about 700 grams/day, and more preferable, 7 mg/day to about 7 grams/day. Nonetheless, as will be understood by those of skill in the art, in certain situations it may be necessary to administer the anti-tumor compound produced by the method in amounts that excess, or even far exceed, the above- stated, preferred dosage range to effectively and aggressively treat particularly advanced or lethal tumors.
  • the compound produced by methods of the invention inhibits the progression of the cell cycle when it is dissolved in an organic solvent or hydrous organic solvent and it is directly applied to any of various cultured cell systems.
  • Usable organic solvents include, for example, methanol, methylsulfoxide, and the like.
  • the formulation can, for example, be a powder, granular or other solid inhibitor, or a liquid inhibitor prepared using an organic solvent or a hydrous organic solvent.
  • a preferred concentration of the compound produced by the method of the invention for use as a cell cycle inhibitor is generally in the range of about 1 to about 100 ⁇ g/ml, the most appropriate use amount varies depending on the type of cultured cell system and the purpose of use, as will be appreciated by persons of ordinary skill in the art. Also, in certain applications it may be necessary or preferred to persons of ordinary skill in the art to use an amount outside the foregoing range.
  • certain embodiments provide methods for preventing or treating fungal infections and/or a pathogenic fungus in a subject, involve administering to the subject a composition including a dehydrophenylahistin or its analog, for example, administering the dehydrophenylahistin or its analog in an amount and manner which provides the intended antifungal effect.
  • Other embodiments include the treatment or prevention of infection in a patient by a pathogenic fungus such as those listed above or referred to below.
  • Another embodiment relates to the treatment or prevention of infection in a patient by a pathogenic fungus which is resistant to one or more other antifungal agents, especially an agent other than dehydrophenylahistin or its analog, including e.g.
  • amphotericin B or analogs or derivatives thereof including 14(s)-hydroxyamphotericin B methyl ester, the hydrazide of amphotericin B with l-amino-4-methylpiperazine, and other derivatives
  • other polyene macrolide antibiotics including, e.g., nystatin, candicidin, pimaricin and natamycin; flucytosine; griseofulvin; echinocandins or aureobasidins, including naturally occurring and semi-synthetic analogs; dihydrobenzo[a]napthacenequinones; nucleoside peptide antifungals including the polyoxins and nikkomycins; allylamines such as naftifine and other squalene epoxidase inhibitors; and azoles, imidazoles and triazoles such as, e.g., clotrimazole, miconazole, ketoconazole,
  • Another embodiment involves the treatment or prevention of infection in a patient by a pathogenic fungus in cases in which the patient is allergic to, otherwise intolerant of, or nonresponsive to one or more other antifungal agents or in whom the use of other antifungal agents is otherwise contra-indicated.
  • Those other antifungal agents include, among others, those antifungal agents disclosed above and elsewhere herein.
  • a dehydrophenylahistin or its analog is administered to the subject in an effective antifungal amount.
  • 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 previously mentioned agents or types of agents (e.g. 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[alnapthacenequinone; or an echinocardin) as well as with a different dehydrophenylahistin or its analog.
  • agents e.g. 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[alnapthacenequinone; or an echin
  • the dehydrophenylahistin or its analog may be administered before, after or at the same time the other antifungal agent is administered, hi certain embodiments, the combination therapy will permit the use of reduced amounts of one or both antifungal components, relative to the amount used if used alone.
  • Still other embodiments relate to adminisfration of a dehydrophenylahistin or its analog to a subject for the treatment or prevention of infection by a pathogenic fungus, where the subject is immunosuppressed or immunocompromised, e.g.
  • a dehydrophenylahistin or its analog may be co-administered with the immunosuppressive agent(s) to treat or prevent a pathogenic fungal infection.
  • Another aspect of this invention is the treatment or prevention of infection by a pathogenic fungus in a patient infected, or suspected of being infected, with HIV, by adminisfration of an antifungal dehydrophenylahistin or its analog, in conjunction with the adminisfration of one or more anti-HIV therapeutics (including e.g. HIV protease inhibitors, reverse transcriptase inhibitors or anti-viral agents).
  • an antifungal dehydrophenylahistin or its analog in conjunction with the adminisfration of one or more anti-HIV therapeutics (including e.g. HIV protease inhibitors, reverse transcriptase inhibitors or anti-viral agents).
  • the dehydrophenylahistin or its analog may be administered before, after or at the same time as administration of the anti-HIV agent(s).
  • 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 regiment to treat or prevent bacterial infection.
  • the dehydrophenylahistin or its analog may be administered before, after or at the same time as administration of the other agent(s).
  • Pathogenic fungal infections which may be treated or prevented by the disclosed methods include, among others, Aspergillosis, including invasive pulmonary aspergillosis; Blastomycosis, including profound or rapidly progressive infections and blastomycosis in the central nervous system; Candidiasis, including refrograde candidiasis of the urinary fract, e.g. in patients with kidney stones, urinary tract obstruction, renal transplantation or poorly controlled diabetes mellitus; Coccidioidomycosis, including chronic disease which does not respond well to other chemotherapy; Cryptococcosis; Histopolasmosis; Mucormycosis, including e.g.
  • compositions comprising an antifungal amount of one or more dehydrophenylahistin or its analogs may be particularly useful for treating or preventing a pathogenic fungal infection in a mammalian subject where the fungus is resistant to one or more other antifungal therapies, or where the use of one or more other antifungal therapies is contraindicated, e.g., as mentioned above.
  • Antifungal pharmaceutical compositions containing at least one antifungal dehydrophenylahistin or its analog are also provided for use in practicing the disclosed methods.
  • compositions may be packaged together with an appropriate package insert containing, ter alia, directions and information relating to their antifungal use.
  • Pharmaceutical compositions are also provided which contain one or more dehydrophenylahistin or its analog together with a second antifungal agent.
  • Methods of Treating Fungal Infections [0193] Certain embodiments disclosed herein relate to methods for treating or preventing a pathogenic fungal infection, including for example Aspergillosis, including invasive pulmonary aspergillosis; Blastomycosis, including profound or rapidly progressive infections and blastomycosis in the central nervous system; Candidiasis, including retrograde candidiasis of the urinary tract, e.g.
  • the methods may involve administering at least one antifungal dehydrophenylahistin or its analog, as described above, to a human subject such that the fungal infection is treated or prevented.
  • the dehydrophenylahistin or its analog may be administered in conjunction with administration of one or more non-dehydrophenylahistin or its analog antifungal agents such as amphotericin B, or an imidazole or triazole agent such as those mentioned previously.
  • the pathogenic fungal infection may be topical, e.g., caused by, among other organisms, species of Candida, Trichophyton, Microsporum or Epiderinophyton or mucosal, e.g., caused by Candida albicans (e.g. thrush and vaginal candidiasis).
  • the infection may be systemic, e.g., caused by Candida albicans, Cryptococcus neoformans, Aspergillus fumigatus, Coccidiodes, Paracocciciodes, Histoplasma or Blastomyces spp.
  • the infection may also involve eumycotic mycetoma, chromoblastomycosis, cryptococcal meningitits or phycomycosis.
  • Further embodiments relate to methods for treating or preventing 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.
  • the method may involve administering a non-immunosuppressive antifungal dehydrophenylahistin or its analog to a patient in need thereof such that the fungal infection is treated or prevented without inducing an untoward immunosuppressive effect.
  • a pathogenic fungal infection which is resistant to other antifungal therapy, including pathogenic fungal infections which are resistant to one or more antifungal agents mentioned elsewhere herein such as amphotericin B, flucytosine, one of the imidazoles or triazoles (including e.g. 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 and dosing regimen such that a fungal infection resistant to another antifungal therapy in the subject is treated or prevented.
  • Further embodiments relate to methods for treating or preventing a pathogenic fungal infection in a patient who is allergic to, intolerant of or not responsive to another antifungal therapy or in whom the use of other antifungal agents is otherwise contra- indicated, including one or more other antifungal agents mentioned elsewhere herein such as amphotericin B, flucytosine, one of the imidazoles or triazoles (including e.g. fluconazole, ketoconazole, itraconazole and the other previously mentioned examples).
  • the methods may involve administering to such patient one or more antifungal dehydrophenylahistin or its analog, in an amount such that a fungal infection is treated or prevented.
  • Packaged Dehydrophenylahistin or its analogs [0198] Certain embodiments relate to packaged dehydrophenylahistin or its analogs, preferably packaged nonimmunosuppressive antifungal dehydrophenylahistin or its analogs, which term is intended to include at least one dehydrophenylahistin or its analog, as described above, packaged with instructions for administering the dehydrophenylahistin or its analog(s) as an antifungal agent without causing a untoward immunosuppressive effects within a human subject, hi some embodiments, the non-immunosuppressive antifungal dehydrophenylahistin or its analog is a member of one of the preferred subsets of compounds described above.
  • the dehydrophenylahistin or its analog can be packaged alone with the instructions or can be packaged with another dehydrophenylahistin or its analog, raparnycin or another ingredient or additive, e.g., one or more of the ingredients of the pharmaceutical compositions.
  • the package can contain one or more containers filled with one or more of the ingredients of the phan-naceutical compositions.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • N,N'-diacetyl-2,5-piperazinedione [0201] 25.0 g of global 2,5-piperazinedione 1 [2,5-piperazinedione (Aldrich G640-6), 25.0 g, 0.218 mol] in 100 mL of acetic anhydride (Ac 2 O) was mixed with sodium acetate (NaOAc) (17.96 g, .0218 mol). The mixture was heated at 110°C for 8 h using a double coiled condenser under an Ar atmosphere.
  • Dehydrophenylahistin [0203] To a solution of 2 (30 mg, 0.099 mmol) in DMF (0.8 mL) was added benzaldehyde (51 ⁇ L, 0.496 mmol, 5 eq) and the solution was repeatedly evacuated in a short time to remove oxygen and flushed with Ar, followed by the addition of Cs 2 CO 3 (53 mg, 0.149 mmol, 1.5 eq) and the evacuation-flushing process was repeated again. The resultant mixture was heated for 2.5 h at 80°C. (The temperature must be increased slowly.
  • A (see Figure 1) is similar in certain respects to the synthesis of the dehydrophenylahistin synthesized as in Example 1.
  • N,N'-diacethyl-2,5-piperazinedione 1 was prepared as in Example 1. 1) 1 -Acetyl-3 - UZ)- 1 - 5-tert-butyl- lH-4-imidazolyl]methylidene ⁇ ] -2, 5-piperazinedione (16)
  • Figure 4 illustrates the similarities of the HPLC profiles (Column: YMC- Pack ODS-AM (20 x 250mm); Gradient: 65% to 75% in a methanol-water system for 20 min, then 10 min in a 100% methanol system; Flow rate: 12mL/min; O.D. 230 nm) from the synthesized dehydrophenylahistin of Example 1 (Fig 2) and the above exemplified tBu- dehydrophenylahistin compound produced by Route A.
  • the sequence of introduction of the aldehydes is a relevant to the yield and is therefore aspect of the synthesis.
  • Reagents a) IDA, CH 3 CHO; b) Tos-Cl, pyridine; c) DBU; d) NaOH; e) C 2 Cl 2 O 2 ; f) KOOCCH 2 COOEt, BuLi; g) SO 2 Cl 2 ; h) H 2 NCHO, H 2 O; i) LiAlH 4 ; j) MnO 2 ; k) l,4-diacetyl-piperazine-2,5-dione, Cs 2 CO 3 ; 1) benzaldehyde, Cs 2 CO 3
  • Ethyl potassium malonate (25.0 g, 0.15 mol) was suspended in water (15.6 ml) and cooled in an ice bath. Concentrated HCl (12.5 ml) was added dropwise over 30 min, then the mixture was stirred for a further 10 min. The precipitate was filtered, then washed twice with ether. The filtrate was separated and the aqueous phase was exfracted with ether. The combined ethereal solutions were dried (MgSO 4 ) and evaporated to afford, as an oil, monoethyl hydrogen malonate (19.2 g, 99%) which was dried under vacuum overnight (or 50°/ 1 mm for 1 h) prior to use.
  • Oxalyl chloride (3.83 ml, 43.9 mmol) was added dropwise to a cooled (0°) solution 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 was stirred for 1 h at 0°, then for 16 h at room temperature. Fractional distillation (121 760 mmHg) afforded 2,2-dimethyl-but-3-enoyl chloride (4. l g, 71%).
  • Reagents g) SO 2 Cl 2 ; h) H 2 NCHO, H 2 O; I)LiAlH 4 ; j) MnO 2 ; k) 1,4- diacetyl-piperazine-2,5-dione, Cs 2 C0 3 ; 1) benzaldehyde, Cs 2 CO 3
  • LC conditions [0335] Reverse 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 90% aqueous MeCN [0339] Gradient: 0-100% B over 11.0 min [0340] Flow rate: 1.5 mL/min [0341] Wavelength: 214 nm [0342] MS conditions: [0343] Ion Source: Ionspray [0344] Detection: Ion counting [0345] Flow rate to the mass spectrometer: 300 ⁇ L/min after split from column (1.5 mL/min).
  • ESMS was done on a Perkin Elmer/Sciex-API III LC/MS/MS using an electro spray inlet.
  • Solvent 0.1% AcOH in 60% aqueous MeCN
  • Flow rate 25 ⁇ L/min
  • Ionspray 5000 V
  • Orifice plate 55 V
  • Acquisition time 2.30 min
  • Scan range 100-1000 amu/z
  • Scan step size 0.2 amu/z
  • Preparative RP-HPLC Purification Conditions [0354] Reverse phase HPLC purification was carried out using Nebula with the Waters XterraMS column (19x50 mm, 5 ⁇ m, C18) using the following conditions: [0355] Solvent A: 0.1% aqueous TFA [0356] Solvent B: 0.1% TFA in 90% aqueous MeCN [0357] Gradient: 5-95% B over 4 min [0358] Flow rate: 20
  • Dehydrophenylahistin [0362] To a solution of 2 (30 mg, 0.099 mmol) in DMF (0.8 mL) was added benzaldehyde (51 ⁇ L, 0.496 mmol, 5 eq) and the solution was repeatedly evacuated in a short time to remove oxygen and flushed with Ar, followed by the addition of Cs CO 3 (53 mg, 0.149 mmol, 1.5 eq) and the evacuation-flushing process was repeated again. The resultant mixture was heated for 2.5 h at 80°C. (The temperature must be increased slowly.
  • HT-29 ATCC HTB-38
  • McCoy's complete medium McCoy's 5 A medium with L-glutamine and 25mM HEPES supplemented with 10% FBS, ImM Na pyruvate, IX NEAA, 2mM L- glutamine, and Pen/Strep at lOOIU/ml and lOO ⁇ g/ml, respectively.
  • PC-3 (ATCC CRL- 1435), a human prostate adenocarcinoma, was maintained in F12K complete medium (F12K medium supplemented with 10% FBS; 2mM Glutamine; 1% HEPES; and Pen/Strep at lOOIU/ml and lOO ⁇ g/ml, respectively).
  • F12K complete medium F12K medium supplemented with 10% FBS; 2mM Glutamine; 1% HEPES; and Pen/Strep at lOOIU/ml and lOO ⁇ g/ml, respectively.
  • Cell lines were cultured at 37 °C, 5% CO 2 in a 95% humidified incubator.
  • HT-29 or PC-3 cells were seeded at 5,000 cells/well in 90 ⁇ l complete media into a Corning 3904 black- walled, clear-bottom tissue culture plate and the plate were incubated overnight to allow cells to establish and enter log phase growth.
  • HT-29 a human colorectal adenocarcinoma was maintained in McCoy's complete medium (McCoy's 5A medium with L-glutamine and 25mM HEPES supplemented with 10% FBS, ImM Na pyruvate, IX NEAA, 2mM L- glutamine, and Pen/Strep at lOOIU/ml and lOO ⁇ g/ml, respectively).
  • PC-3 a human prostate adenocarcinoma, was maintained in F12K complete medium (F12K medium supplemented with 10% FBS; 2mM Glutamine; 1% HEPES; and Pen Strep at lOOIU/ml and lOO ⁇ g/ml, respectively).
  • Cell lines were cultured at 37 °C, 5%o CO 2 in a 95% humidified incubator.
  • HT-29 or PC-3 cells were seeded at 5,000 cells/well in 90 ⁇ l complete media into a Corning 3904 black- walled, clear-bottom tissue culture plates and the plates were incubated overnight to allow cells to establish and enter log phase growth.
  • Derivatives are synthesized that: 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 the 5-position on the imidazole ring.
  • a 1 Modification of the phenyl ring besed on the structure of known anti-tubulin compounds Alkyl, Halogen, Alkoxy, Acetyl, Sulfonamide, Amino, Hydroxyl, Nitro, etc.
  • derivatives of the compound may include the following substitutions at the phenyl ring (A): -CF 3 , -S0 2 NH 2 (-SO 2 NR ⁇ R 2 ), -SO 3 H, -CONH 2 (-CONR ⁇ R 2 ), -COOH, etc.
  • Other ring systems (C) may also include the following:
  • Methanol solution of the derivative obtained by the above-listed examples are added to the wells of the uppermost row, specimens are diluted by the half-log dilution method and added, and the plate is incubated in a carbon dioxide gas incubator at 37°C for 48 hours.
  • the result is added in lots of lO ⁇ l with MTT reagent (3-(4,5-dimethyl-2-thiazole)-2,5-diphenyl-2H-tetra bromide)(l mg/ml • PBS), followed by incubation in a carbon dioxide gas incubator at 37 °C for 6 hours.
  • the culture medium is discarded and the crystal of produced in the cells are dissolved in lOO ⁇ l/well of dimethylsulfoxide.
  • Cell strain A431 is derived from human lung cancer. EMEM culture medium containing 10% bovine fetal serum and 1% MEM nonessential amino acid solution (SIGMA M2025) is used to incubate A431 cells at 37 °C in an incubator saturated with 5% carbon dioxide gas and water vapor.
  • SIGMA M2025 MEM nonessential amino acid solution
  • Colchicine recognizes the same binding site on ⁇ -tubulin as PLH. Colchicine has four characteristic methoxy groups on its A and B rings. A series of substitutions with the single or multiple methoxy groups was performed and the results of cytotoxic activity are shown in Table 6. Table 6. Effect of the methoxy group substitution on the proliferation of HT-29 cells
  • the compounds having effective functional groups which showed higher activity than tBu- dehydroPLH, may also be further modified. And since the migration of the stereochemistry from Z to E under the visible light irradiation was observed, substituents that decrease the electron density in the conjugated double bonds may contribute to the reduction of Z to E migration by the light, results in more physicochemically stable structures. Temperature can also effect this migration. [0392] Modification at two parts of the ring can be preferred for the development of potent but also biologically stable compounds.
  • the phenyl ring of phenylahistin is oxidized by cytochrome P-450. Double modification that reduces the electron density of the phenyl ring may therefore be effective to avoid P-450 oxidation.
  • the phenyl ring may also be replaced by heteroaryl groups.
  • the result of such replacements in terms of the cytotoxic activity is shown in Table 11. Since the arylic nitrogen atoms can form a hydrogen bonding with a NH group of the diketopiperazine ring and restrict the conformation of the molecule between pyridine and diketopiperazine rings to an uniplanar structure, the active conformation of dehydroPLH would be required a certain level of dihedral angle formed by the steric repulsion between an amide hydrogen atom of the diketopiperazine ring and an o-hydrogen atom of the phenyl ring ( Figure 6). Table 11. Effect of the replacement with the heteroaryl ring on proliferation of HT-29 cells
  • the E-form may also be used as a prodrug of dehydroPLH or of one or more of its analogs, including those analogs described herein.
  • One of the undesired properties of anti-tubulin drugs involves its low selectivity between tumor and intact tissues, although these drugs belong to one of the molecular target therapies. This causes undesired side effects. However, if the compounds functions selectively only in tumor tissues, negative side effects of anti-microtubule drugs can be reduced.
  • the dehydroPLH (Z-form) can be produced from its E-isomer by visible light irradiation, the E-form is administered and photo irradiation is performed only at the tumor site, then only, the tumor is damaged by photo-produced Z-form and the adverse effect to the intact tissues is reduced.
  • the E-form can be protected chemically 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.
  • the vials After being charged with nitrogen, helium or other inert gas, the vials are stored in a cool, dark place. Before use, the contents are dissolved in ethanol and added to 100 ml of a 0.85% ⁇ physiological salt water solution.
  • the resultant solution is administered as a method of inhibiting the growth of a cancerous tumor in a human diagnosed as having such a tumor at between approximately 10 ml/day to approximately 1000 ml/day, intravenously, by drip, or via a subcutaneous or intraperitoneal injection, as deemed appropriate by those of ordinary skill in the art.
  • the granules are thoroughly dried and sifted to obtain a granule preparation suitable for packaging in bottles or by heat sealing.
  • the resultant granule preparations are orally administered at between approximately lOOml/day to approximately 1000 ml/day, depending on the symptoms, as deemed appropriate by those of ordinary skill in the art of treating cancerous tumors in humans.
  • Formulation to be Administered Topically Administration to an individual of an effective amount of the compound can also be accomplished topically by administering the compound(s) directly to the affected area of the skin of the individual.
  • the compound administered or applied is in the form of a composition including a pharmacologically acceptable topical carrier, such as a gel, an ointment, a lotion, or a cream, which includes, without limitation, such carriers as water, glycerol, alcohol, propylene glycol, fatty alcohols, triglycerides, fatty acid esters, or mineral oils.
  • 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, hurnectants, viscosity stabilizers, and similar agents may be added as necessary.
  • Percutaneous penetration enhancers such as Azone may also be included.
  • the compound may be disposed within devices placed upon, in, or under the skin. Such devices include patches, implants, and injections which release the compound into the skin, by either passive or active release mechanisms.
  • EXAMPLE 8 In vitro Pharmacology of KPU-2, KPU-35 and t-butyl phenylahistin [0404]
  • KPU-35 and t-butyl phenylahistin 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), ONCAR-3 (human ovarian tumor; ATCC; HTB-161), B16-F10 (murine melanoma; ATCC; CRL-6475) and CCD-27sk (normal human fibroblast; ATCC; CRL- 1475).
  • 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;
  • KPU-2 and KPU-35 were most effective on the HT-29 tumor cell line, both in terms of potency (active in the low nanomolar range) and efficacy (most responsive in terms of the maximum cytotoxic effect); t-butyl- phenylahistin exhibited its greatest potency against the PC-3 tumor cell line, although the greatest efficacy was displayed against the HT-29 cell line; KPU-2 and KPU-35 were generally 10-40 fold more potent than t-butyl-phenylahistin whereas the efficacy was similar for all three compounds in the different tumor cell lines; the HT-29, PC-3, MDA-MB-231 and ⁇ CI-H292 tumor cell lines all responded similarly to the NPI compounds, whereas the B16-F10 appeared to be somewhat less sensitive, t-butyl-phenylahistin displayed a marked differential between normal fibroblasts and the tumor cell lines, with a ratio ranging from >20 - >100, except for the ONCAR-3
  • Cyclosporin A (CsA) pretreatment did not alter the potency of the selected compounds.
  • taxol was virtually inactive in the MES- SA DX5 resistant cell line, whereas this compound was very potent in the sensitive cell line.
  • CsA treatment restored the sensitivity to taxol of the MES-SA/DX5 cell line.
  • the MES- SA/DX5 cell line also showed reduced susceptibility to etoposide (60 fold), doxorubicin (34 fold) and mitoxantrone (20 fold).
  • KPU-35 t-butyl-phenylahistin and Taxol in MES-SA Taxol Sensitive and MES-SA/DX5 Taxol Resistant Human Uterine Sarcoma Tumor Cell Lines
  • KPU-2, KPU-35 and t-butyl-phenylahistin are not susceptible to the same resistance mechanisms as Mitoxantrone in this cell line, and there is no cross-resistance from Mitoxantrone to these selected novel compounds in this model.
  • Table 14 Activity of KPU-2, KPU-35. t-butyl-phenylahistin and Mitoxantrone in the HL-60 Human Acute Promvelocvtic Leukemia Tumor Sensitive and Resistant Cell Line Pair
  • Apoptosis and its dysregulation play an important role in oncology; the selective induction of the programmed cell death cycle in tumor cells is the goal of many chemotherapeutic drug discovery programs. This induction of apoptosis can be demonstrated by different methods including the characteristic cell membrane blebbing, DNA fragmentation, hyperphosphorylation of the antiapoptotic factor Bcl-2, activation of the caspase cascade and cleavage of poly (ADP ribose) polymerase (PARP).
  • PARP poly (ADP ribose) polymerase
  • the characteristic signs of apoptotic cell death include cell membrane blebbing, disruption of nuclei, cell shrinkage and condensation and finally cell death, very distinctive from necrotic cell death.
  • KPU-2 induced the typical morphological changes associated with early stages of apoptosis in human prostate tumor cells. A similar finding was also clear in the treatment of HuVEC cells with KPU-2.
  • a late stage characteristic of apoptosis is internucleosomal DNA cleavage 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 DNA laddering in Jurkat cells (human T cell leukemia line) in comparison to halimide and dehydrophenylahistin (KPU-1). KPU-2 induced DNA laddering at the 1 nM concentration whereas halimide and KPU-1 were much less potent.
  • Enhanced Vascular Permeability in HuNEC Cells Compounds that depolymerize microtubules (e.g. 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 vascular cell permeability initially to electrolytes and soon after to large molecules. The enhanced permeability of HuVEC cells to a fluorescent-labeled dextran is used as a proxy assay for vascular collapse.
  • combretastatin A-4- phosphate ZD6126
  • KPU-2, KPU-35 and t-butyl-phenylahistin all rapidly (within 1 hour) induced significant HuVEC monolayer permeability, to an extent similar to colchicine.
  • the microtubule stabilizing agent taxol was inactive in this assay ( Figure 12).
  • KPU-2 was initially screened at a concentration of 10 ⁇ M in a panel of 60 different kinases; the ATP concentration was 10 ⁇ M.
  • ATP concentration was 10 ⁇ M.
  • Four kinases were inhibited by greater than 50%o in the primary screen and the IC50's determined in secondary screening are presented in Table 17. All of the IC50 values are in the low micromolar range, which indicates that inhibition of these kinases is not related to the low nanomolar activities observed for tumor cell cytotoxicity. Table 17.
  • EXAMPLE 9 In vivo Pharmacology [0429] Preliminary studies with KPU-2 were performed using the MX-1 (breast) and HT-29 (colon) xenograft models and the P-388 murine leukemia tumor model, in the mouse. Other tumor models selected on the basis of activity in the in vitro tumor panel were the DU-145 (prostate), MCF-7 (breast), and the A549 (lung) cell lines. The human pancreatic tumor (MiaPaCa-2) was also included. The novel compounds were studied as monotherapy and in combination with a clinically-used chemotherapeutic agent.
  • the doses of the selected novel compounds were determined from the acute tolerability testing (Maximally Tolerated Dose, MTD) and were adjusted if necessary during each study.
  • the doses of the clinically-used chemotherapeutic agents were selected on the basis of historical studies.
  • KPU-2 was the first compound to be studied in these five tumor models. Following the initial results from this study, all three compounds were compared in the HT- 29 human colon tumor, the DU-145 human prostate and the MCF-7 human breast tumor xenograft models.
  • the above models all use the subcutaneous xenograft implantation technique and are potentially subject to selective effects of a compound on the subcutaneous vasculature producing a magnified (or apparent) antitumor activity.
  • Xenograft Models Animals used were (exceptions are indicated for individual studies): female nude mice (nu/nu) between 5 and 6 weeks of age ( ⁇ 20g, Harlan); group size was 9-10 mice per group unless otherwise indicated. [0433] Cell lines used for tumor implantation were: HT-29 human colon tumor; MCF-7 human breast tumor; A549 human non small cell lung tumor; MiaPaCa-2 human pancreas tumor; DU-145 human prostate tumor. [0434] Selected novel compounds were administered as monotherapy via the intraperitoneal (i.p.) route at the doses indicated for the individual study; for the combination studies the selected reference chemotherapy agents were injected 15-30 min prior to the compound.
  • i.p. intraperitoneal route
  • 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 CPT-11 each mL of solution contained 20 mg of frinotecan hydrochloride, 45 mg of sorbitol NF powder, and 0.9 mg of lactic acid, the pH being adjusted to 7.4 with NaOH or HCl. Saline dilutions are used to achieve the injection concentrations used for the reference compounds.
  • HT-29 Human Colon Tumor Model Animals were implanted subcutaneously (s.c.) by trocar with fragments of HT-29 tumors harvested from s.c. growing tumors in nude mice hosts. When the tumor size reached 5 mm x 5 mm (about 10-17 days) the animals were matched into treatment and control groups. Mice were weighed twice weekly and tumor measurements were obtained using calipers twice weekly, starting on Day 1. The tumor measurements were converted to estimated mg tumor weight using the formula (W 2 xL)/2. When the estimated tumor weight of the control group reached an average of 1000 mg the mice were weighed, sacrificed and the tumor removed.
  • TGI tumor growth inhibition
  • the animals were matched into treatment and control groups.
  • the rest of the study proceeded as described for the HT-29 model, using taxotere and CPT-11 as the standard chemotherapy agents.
  • the tested compounds were administered via the intraperitoneal route on a q3dx5 dose schedule for the CPT-11 combination or on a qdx5 dose regimen for the combination with taxotere; CPT-11 was administered via the intraperitoneal route on a qwx3 schedule; taxotere was administered intravenously on a qodx3 dose regimen.
  • MiaPaCa-2 Human Pancreas Tumor Model Animals were implanted s.c. by trocar with fragments of MiaPaCa-2 tumors harvested from s.c. growing tumors in nude mice hosts. When the tumor size reached 5 mm x 5 mm (about 10 -17 days) the animals were matched into treatment and control 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, test compounds were administered every third day via the intraperitoneal route on Days 1, 4, 7, 10 and 15 (q3dx5); gemcitabine was administered via the intraperitoneal route on Days 1, 4, 7 and 10 (q3dx4).
  • DU-145 Human Prostate Tumor Model Male mice were implanted s.c. by trocar with fragments of DU-145 tumors harvested from s.c. growing tumors in nude male mice hosts. When the tumors reached ⁇ 5 mm x 5 mm ( at about 13 -17 days) the animals were matched into treatment and control groups. The remainder of the study proceeded as for the HT-29 model, using taxotere as the standard chemotherapy agent. [0445] hi this model unless otherwise noted for the individual study, 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 Implantation Tumor Models [0446] The animals used were: female nude mice (nu/nu) (MDA-231 study) or B6D2F1 (B16-F10 studies) mice between 5 and 6 weeks of age ( ⁇ 20g, Harlan); group size was 10 mice per group unless otherwise indicated. [0447] The cell lines used were: MDA-MB-231 human breast tumor and B 16-F 10 murine melanoma cells. [0448] NPI compounds were administered as monotherapy via 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 prior to the NPI compound.
  • MDA-231 Human Breast Tumor Female nude mice were injected in the mammary fat pad with 2x10 MDA-231 cells harvested from in vitro cell culture. When the tumor size reached 5 mm x 5 mm (about 14-28 days) the animals were matched into treatment and control groups. The study then 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 via the intraperitoneal route on Days 1, 4, 8, 11 and 15 (q3dx5); paclitaxel was administered via the intraperitoneal route on Days 1-5 (qdx5).
  • B16-F10 Metastatic Murine Melanoma Model [0451] Mice received B16-F10 cells (prepared from an in vitro cell culture of B16-F10 cells) by the iv route on Day 0. On Day 1 mice were randomized into freatment and control groups and treatment commenced. Mice were weighed twice weekly, starting on Day 1. All mice are sacrificed on Day 16, the lungs removed, weighed and the surface colonies counted. Results are expressed as mean colonies of freated mice/mean colonies of confrol mice (T/C) x 100%>). The metastasis growth inhibition (MGI) is this number subtracted from 100%). Paclitaxel was the standard chemotherapy agent used in this study.
  • test compounds were administered via the intraperitoneal route on Days 1-5 (qdx5); paclitaxel was administered intravenously on Days l-5(qdx5).
  • results are presented as means ⁇ SEM.
  • Statistical analysis of studies with several groups was performed using ANOVA with Neuman-Keuls post test, unless otherwise indicated. A one-tailed t-test was also used based on the hypothesis that the compound or drug, or the combination, would reduce tumor growth. Results Studies in the HT-29 Human Colon Tumor Xenograft Model 1.
  • KPU-2 +/- CPT-11 in the HT-29 Human Colon Tumor Xenograft Model assessed changes in dosage strength and dosing regimen for KPU-2 alone and in combination with a relevant chemotherapeutic CPT-11 in the HT-29 model.
  • KPU-2 was administered at doses 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 TGI for the combined therapy group exceeds the NCI criterion of 58% for a positive result.
  • Study of KPU-2 +/- Standard Chemotherapy vs. Five Human Tumor Xenograft Models [0456] This study consists of five different arms, each with its own protocol, timing, dosing regimen and reference compound. Each arm will be considered within the presentation of the particular tumor model.
  • the aim of the HT-29 arm 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 as compared to those used in the study described above, in which a marked synergy was observed between KPU-2 (7.5 mg/kg ip q3dx5) and CPT-11 (100 mg/kg ip qwx3).
  • KPU-2 7.5 mg/kg ip q3dx5
  • CPT-11 100 mg/kg ip qwx3
  • the combined therapy maintained efficacy and the estimated tumor growth for this group was significantly lower than for either monotherapy group. Accordingly, adminisfration of KPU-2 and CPT-11 inhibited tumor growth and is an effective anti-tumor treatment.
  • the observations of the in-life portion of the study are supported by measurement of the weights of the tumors excised at autopsy ( Figure 16).
  • the tumor weights for the combination group was significantly less than the Controls (p ⁇ 0.01), as were the tumor weights for CPT-11 alone (p ⁇ 0.05).
  • the tumor size for the combination group was generally smaller than for the other treated or control groups.
  • the TGI of the combination group was 65.8%, indicating a positive effect by the NCI criterion, while monotherapy did not reach the NCI criterion of TGI > 58%.
  • the combination therapy groups all indicated a marked synergy between the novel compounds and CPT-11.
  • the individual tumor weights demonstrate the effectiveness of the cotherapy freatment (Figure 18). In each case the TGI for the combination group surpasses the NCI criterion for a positive effect, whereas the TGI for CPT-11 monotherapy did not reach this level.
  • KPU-2.KPU-35 and t-butyl-phenylahistin in Combination with CPT-11 in the HT-29 Human Colon Tumor Xenograft Model When combined with CPT-11, KPU-2 enhanced the effect of CPT-11, the standard chemotherapeutic agent, to a level well in excess of the NCI criterion of a TGI > 58%o for a positive effect.
  • the results generated in the three studies are very comparable for both the in-life observations ( Figure 19) and for the weights of the tumors excised at autopsy ( Figure 20).
  • KPU-2 KPU-35 and t-butyl-phenylahistin Alone or in Combination with Taxotere in the DU-145 Human Prostate Xenograft Model
  • KPU-2 KPU-35 and t-butyl-phenylahistin Alone or in Combination with Taxotere in the DU-145 Human Prostate Xenograft Model
  • the cotherapy group had a TGI of 74.4%) as compared to the control group well in excess of the NCI criterion for a positive effect (TGI > 58%). Taxotere alone had a TGI of 26.1 %. 7.
  • TGI human breast Tumor Orthotopic Xenograft Model
  • Antifungal activity can also be determined in whole-animal models of fungal infection. For instance, one may employ the steroid-treated mouse model of pulmonary mucormycosis (Goldaill, L.Z. & Sugar, A.M. 1994 J Antimicrob Chemother 33:369-372).
  • a number of animals are given no dehydrophenylahistin or its analog, various doses of dehydrophenylahistin or its analog (and/or combinations with one or more other antifungal agents), or a positive control (e.g. Amphotericin B), respectively, beginning before, at the time of, or subsequent to infection with the fungus.
  • a positive control e.g. Amphotericin B
  • Animals may be freated once every 24 hours with the selected dose of dehydrophenylahistin or its analog, positive control, or vehicle only. Treatment is continued for a predetermined number of days, e.g. up to ten days. Animals are observed for some time after the treatment period, e.g. for a total of three weeks, with mortality being assessed daily.
  • Models can involve systemic, pulmonary, vaginal and other models of infection with or without other treatments (e.g. treatment with steroids) designed to mimic a human subject susceptible to infection.
  • one method for determining the in vivo therapeutic efficacies e.g.
  • a mouse is infected with the fungal pathogen such as by intravenous infection with approximately 10 times the 50%o lethal dose of the pathogen (10 6 C. albicans cells /mouse).
  • dehydrophenylahistin compounds are given to the mouse at a predetermined dosed volume.
  • the ED 50 is calculated by the method of Van der Waerden (Arch Exp Pathol Pharmakol 195:389-412, 1940) from the survival rate recorded on 20th day post-infection.
  • untreated control animals die 7 to 1 3 days post-infection.
  • CF-1 mice are infected by injection 1 or 5 x 10 6 CFU into the tail vein.
  • Antifungal agents are administered intravenously or subcutaneously in ethanokwater (10:90), 4 h post infection and once daily thereafter for 3 or 4 more days. Survival is monitored daily.
  • the ED 50 can be defined as that dose which allows for 50%) survival of mice.
  • EXAMPLE 11 Evaluating Antimicotic Activity [0479] Benzimidazoles and griseofulvin are anti-tubulin agents capable of binding to fungal microtubules. Once bound, these compounds interfere with cell division and intracellular transport in sensitive organisms, resulting in cell death. Commercially, benzimidazoles are used as fungicidal agents in veterinary medicine and plant disease control. A wide variety of fungal species, including Botiytis cinerea, Beauveria bassiana, Helminthosporium solani, Saccharomyces cerevisiae and Aspergillus are susceptible to these molecules. Toxicity concerns and increasing drug resistance, however, have negatively impacted their usage.
  • Griseofulvin is used clinically to treat ringworm infections of the skin, hair and nails, caused by Trichophyton sp., Microsporum sp., and Epidermophyton floccosum. Its antifungal spectrum, however, is restricted to this class of fungal organisms. Genotoxicity is also a significant side effect. Terbinafine, while an alternative first-line treatment, is more costly. Further, clinical resistance recently has been observed in Trichophyton rubrum (the major causative agent for all dermatophyte infections). [0480] hi Candida albicans, microtubule/microfilament formation is affected where cells are exposed to the microtubule inhibitors nocodazole and chloropropham.
  • test compounds and controls used in this study were (-)-Phenylahistin, KPU-1, KPU-2, KPU-11 and KPU-17, KPU-35, t-butyl phenylahistin, Colchicine (commercial microtubulin inhibitor tested versus 3 Candida isolates), Benomyl (commercial microtubulin inhibitor tested versus 3 Candida isolates), Griseofulvin (commercial microtubulin inhibitor and antibiotic control for testing versus 6 dermatophyte isolates), Amphotericin B (antibiotic control for testing versus 3 Candida isolates), Itraconazole (antibiotic control for testing versus 2 Aspergillus isolates).
  • Candida albicans Candida glabrata
  • Aspergillus fumigatus Trichophyton rubrum
  • Trichophyton mentagrophytes Trichophyton mentagrophytes
  • Antifungal susceptibility testing was accomplished according to the methods outlined in the 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 testing in RPMI-1640 with glutamine and without bicarbonate, an inoculum size of 0.4 - 5 x 10 4 , and incubation at 30 or 35°C for 48 hours. The minimum inhibitory concentration (MIC) was defined as the lowest concenfration that resulted in an 80% reduction in turbidity as compared to a drug-free control tube.
  • MIC minimum inhibitory concentration
  • MIC minimum inhibitory concentration
  • MLC minimum lethal concentration
  • the MLC was called at the first concentration that exhibited five or fewer colonies of fungal growth representing a 99.95% kill.
  • a minimum fungicidal concenfration was determined to assess the fungistatic/fungicidal nature of the compound. This procedure entails diluting drug-treated cell samples (removed from test wells containing compound at and above the MIC) to compound concentrations significantly below the inhibitory concenfration and depositing them on agar plates. The compound is scored as fungistatic if the cells are able to resume growth and fungicidal if no regrowth is possible because the compound had killed the organisms.
  • T. rubrum is the principal causative agent for human dermatophytic infections, and would be the key organism to target in the development of a clinical agent.
  • Compounds KPU-2, KPU-11 and KPU-17, KPU-35 & t- butylphenylahistin were equivalent in potency or in some cases more potent than griseofulvin, a current, standard pharmaceutical agent used for treating dermatophytic infections.
  • Compounds (-)-Phenylahistin and KPU-1 were significantly less potent than the other compounds when tested versus T. rubrum and weaker but more comparable to the others versus the sensitive T. mentagrophytes isolate.
  • VTAs vascular targeting agents
  • the compounds disclosed herein, including NPU- 02 and KPU-35, for example, can be as VTAs.
  • Many VTAs exhibit their vascular effects by interacting at the colchicine-binding site on microtubules. This interaction induces a characteristic, rapid collapse and occlusion of established vasculature in the tumor and therefore compromises the integrity of existing vessels leading to necrosis.
  • Vascular collapse can occur, for example, within 30-60 minutes of exposure to the VTA and involves changing the shape of the immature and proliferating, but not the quiescent and mature, endothelial cells in the central portion of the tumor.
  • This differential effect on vascular cells provides a rationale for the selective effects on the tumor due to the higher percentage of proliferating immature endothelial cells in the tumor blood vessels versus normal blood vessels.
  • VTAs can be classified into three overlapping spectra of activity: (1) potent vascular and cytotoxic effects, (2) potent vascular with weak cytotoxic effects, and (3) potent cytotoxic with weak vascular effects.
  • H2B-GFP Histone-green fluorescent protein
  • KPU-02 and KPU-35 were shown to induce a rapid vascular collapse leading to central necrosis, and the regression of established tumors after a single i.v. adminisfration.
  • mice were treated i.v. with a 2-minute infusion of 5 mg/kg KPU-35, a 5 minute i.v. infusion of 10 mg/kg KPU-02, or bolus of vehicle (10% solutol (w/w) + 2%> DMSO in water).
  • 5-minute infusions of 10 mg/kg 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.
  • KPU-02 induces rapid tubulin depolymerization in HuVEC cells.
  • Human umbilical vein endothelial cells (HuVECs; Cambrex CC2519A) were maintained at subconfluent densities in EGM-2 (Cambrex) media. The cells were cultured in an incubator at 37°C in 5% CO 2 and 95%> humidified air. For tubulin staining assays, HuVEC cells were seeded at a density of 3x10 4 cells/ml in EGM-2 on tissue culture compatible coverslips (Fisher). The plates were returned to the incubator for 2 days. [0499] Stock (20 mM) solutions of the test compounds were prepared in 100%> DMSO.
  • 400X concenfrated dilutions of the compounds were prepared in 100% DMSO. 5 ⁇ l volumes of the dilutions were added to individual wells resulting in a final concenfration of 200 nM. The final concentration of DMSO was 0.25%> in all samples. The plates were returned to the incubator for 30 minutes. HuVEC cells were treated for 30 min with 200 nM KPU-02 or KPU-35. [0500] The cells were rinsed in dPBS before fixation in 10%>(v/v) neutral buffered formalin for 10 minutes at room temperature. Following fixation, ⁇ -tubulin was visualized by indirect immunofluorescence.
  • the cells were permeabilized in 0.2%>(v/v) friton X-100/dPBS for 10 minutes.
  • the cells were washed prior to transferring the coverslips to a humidified chamber, the coverslips were blocked for two hours in antibody buffer (2%(w/v) BSA/ 0.1%(v/v) Tween 20/ dPBS).
  • the coverslips were incubated with 50 ⁇ l of 0.1 ⁇ g/ml mouse ⁇ -tubulin (Molecular Probes) in antibody buffer for 1 hour before washing and incubation with 50 ⁇ l of 1 ⁇ g/ml goat anti-mouse FITC (Jackson hnmunoResearch Laboratories) for one hour in the dark.
  • Human umbilical vein endothelial cells (HuVECs; Cambrex CC2519A) were maintained at subconfluent densities in EGM-2 (Cambrex) media. The cells were cultured in an incubator at 37°C in 5%> CO 2 and 95% humidified air. For monolayer permeability assays, HuVEC cells were seeded at 1x10 s cells/ ml in EGM-2 media on Fibronectin-coated 3.0 ⁇ m Fluoroblok inserts (Becton Dickinson) in 24-well plates. The plates were returned to the incubator for 4 days to allow the cells to reach confluency. [0503] Stock solutions (20 mM) of the test compounds were prepared in 100% DMSO.
  • 10X concenfrated serial dilutions of the compounds were prepared in EGM-2. 10 ⁇ l volumes of the serial dilutions were added to the test inserts in duplicate resulting in final concentrations ranging from 2 ⁇ M to 2 nM. The final concentration of DMSO was 0.25% in all samples. The cells were treated with 2 nM-2 ⁇ M KPU-02 for 15 minutes. [0504] FITC-Dextran (50 mg/ml) in dPBS (38.2 kDa; Sigma) was diluted 2.5 fold in EGM-2, 10 ⁇ l of FITC-Dextran was added to each insert. The final concentration of FITC-Dextran was 1 mg/ml.
  • Figure 34 shows that KPU-02 is able to induce monolayer permeability in a dose dependent manner.
  • the results shown in Figure 34 represent the mean + S.D. of three independent experiments.
  • VTAs that have entered into the clinic show similar qualitative effects on tumor blood using the IAP methodology (or similar technology) to demonstrate reduced blood flow in the P22 rat sarcoma tumor and in humans using the dce-MRI technique.
  • VTAs selectively damage the vasculature in the cenfral part of the tumor versus the periphery, which recovers functionality, support using these agents in combination with chemotherapeutics (Taxol, Vinblastine and Cisplatin), radiation and angiogenesis inhibitors directed against VEGF and EGF.
  • chemotherapeutics Tuxol, Vinblastine and Cisplatin
  • the new VTAs will supplement rather than supplant these therapies and should provide for greater antitumor activities.
  • Conditions include other neoplasms, retinopathies, and any other condition or disease that relies upon blood supply, preferably blood supply from new vasculature in order to remain viable and/or proliferate.
  • Many conditions are associated with excessive or inappropriate vasculature.
  • conditions associated with excessive vasculature include inflammatory disorders such as immune and non-immune inflammation, rheumatoid arthritis, chronic articular rheumatism and psoriasis; disorders associated with inappropriate or inopportune invasion of vessels such as diabetic retinopathy, neovascular glaucoma, retinopathy of prematurity, macular degeneration, corneal graft rejection, refrolental fibroplasia, rubeosis, capillary proliferation in atherosclerotic plaques and osteoporosis; and cancer associated disorders, including for example, solid tumors, tumor metastases, blood born tumors such as leukemias, angiofibromas, Kaposi sarcoma, benign tumors such as hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, as well as other cancers which require vascularization to support tumor growth.
  • inflammatory disorders such as immune and non-immune
  • vasculature-dependent diseases include, for example, Osier- Webber Syndrome; myocardial angiogenesis; plaque neovascularization; telangiectasia; hemophiliac joints and wound granulation.
  • excessive vasculature is also associated with clinical problems as part of biological and mechanical implants (tissue/organ implants, stents, etc.).
  • the instant compounds and compositions can be used to target vasculature, in preferably to preferentially target disease vasculature over non disease tissue vasculature, and thus the compounds and compositions can be used in the treatment of such conditions.
  • Other diseases in which vascularization plays a role, and to which the instant compounds and compositions can be used, are known by those of skill in the art.
  • retinopathies include age-related macular degeneration (ARMD), diabetic retinopathy, and the like.
  • Pathological angiogenesis is a major contributing factor to a number of retinopathies that collectively are major 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.
  • retinal and disk 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.
  • neovascularization is a serious complication in ⁇ 10% of patients with macular degeneration.
  • Ferris et al Arch Ophthalmol (1984), 102, 1640-1642 which is incorporated herein by reference in its entirety.
  • Vascular targeting agents such as Combretastatin A-4 (CA-4) have been shown to cause the disruption of neovessels in non-neoplastic tissue. Griggs et al Br J Cancer (2001) 84, 832-835, which is incorporated herein by reference in its entirety.
  • CA-4P was shown to inhibit the retinal neovascularization that occurs during proliferative retinopathy.
  • X-ray crystal analysis indicates that the conformation of potent derivatives require a certain amount of dihedral angle between the phenyl ring and the pseudo-tricyclic cor template formed by DKP and imidazole rings. Thus, modification with the appropriate conformational restriction of the phenyl ring may elicit potent activity. While not being bound to any particular theory, it may be that the binding mode of PLH derivatives at the chochicine binding site of tubulin is different from that of colchicines and its known homologues.
  • Microtubule protein was prepared as previously described (Farrell KW and Wilson L. (1987) Tubulin-colchicine complexes differentially poison opposite microtubule ends. Biochemistry 23(16):3741-8, which is incorporated herein by reference in its entirety).
  • MTP preparations consisting of 70%> tubulin and 30%> microtubule-associated proteins (MAPs) were isolated from bovine brain by three cycles of warm polymerization and cold depolymerization in PEM100 (100 M 1-4 piperazinediethansulfonic acid (Pipes), 1 mM MgSO 4 , 1 mM EGTA, pH 6.8) and 1 mM GTP. MTP was drop-frozen in liquid nifrogen and stored at -70°C until use. Tubulin was purified from microtubule protein by phosphocellulose chromatography (PC-tubulin) and stored in PEM50 (50 mM Pipes, 1 mM MgSO 4 , 1 mM EGTA, pH 6.8). Protein concenfration was determined by a Bradford assay (Sigma Chemicals, St. Louis, MO) using bovine serum albumin as the standard (Bradford, 1976).
  • Test Agents [0518] Stock solutions of KPU-02 were prepared at a concentration of 20 mM in DMSO. Stock solutions of Combretastatin A4 (National Cancer Institute, Bethesda, MD) (CA4) was prepared at a concentration of 5 mM in DMSO. Colchicine (Sigma Chemicals, St. Louis, MO) (CLC) was prepared at a concenfration of 3 mM in water. All agents were shielded from ambient light with amber Eppendorf tubes. Serial dilutions were made in DMSO and/or PEM50 to the desired concentrations.
  • MTP Steady-State Microtubule Polymer Mass
  • microtubules were separated and sedimented from unpolymerized MTP by centrifugation (150,000 x g, 45 minutes, 37°C). The supernatant was removed, and the microtubule pellets were depolymerized in deionized H O (24 hours, 0°C) before protein determination by the Bradford assay. [0521] The percent inhibition was calculated in two ways and the values obtained from the two ways were compared. In one way, a ratio of the microtubule protein in the pellet, drug to no drug, was calculated. Another ratio of microtubule protein in the pellet to the supernatant, drug to no drug, was also calculated.
  • Microtubule Mean Length Distributions [0522] Transmission elecfron microscopy was used to determine the mean length distribution of microtubules in the absence or presence of tested agent. At 75 minutes and prior to sedimentation, 10 ⁇ l aliquots from the polymer mass experiments were fixed by dilution into 290 ⁇ l PEMlOO-buffered 0.2%> glutaraldehyde. Thirty microliters of fixed sample was settled onto formvar-coated 150 ICG mesh elecfron microscope grids for 90 seconds. Excess sample was wicked off with Whatman filter paper.
  • cytochrome C (1 mg/ml) was applied for 30 seconds to enhance protofilament resolution and facilitate negative staining.
  • Uranyl acetate (1.5%) was applied for 20 seconds and the excess was wicked off. Grids were viewed in a Jeol elecfron microscope- 1200 EXl 1 at 2000X and 30,000X magnification, The Zeiss MOPIJI was used to determine microtubule length distributions and mean lengths for at least 100 microtubules per sample.
  • PC-tubulin (0.2 mg/ml) was incubated in PEM50 with 1 mM GTP, 1% DMSO, 10 ⁇ M of tested agent and 7-25 ⁇ M [ 3 H] CLC for 120 minutes at 37°C. Measurement of [ 3 H] CLC binding was followed by DEAE-cellulose filter-binding assay as described previously (Wilson, 1970). This method depends on the adsorption of tubulin to filter paper impregnated with DEAE-cellulose. Whatman DE81 filter paper was pre- wet with PEM50 prior to sample application. The total 100 ⁇ l reaction volume was applied to 2.5 cm disks of filter paper, over parafilm, on ice.
  • the paper disks were washed by immersion in five successive 50 ml changes of PEM50, 5 min/wash, 4°C, to remove all unbound colchicine.
  • the paper disks with adhering tubulin-bound colchicine were then counted directly in a scintillation vial containing 2 ml of Beckman Coulter Ready Protein solution (Fullerton, CA). All of the disks were washed together. Negligible binding of unbound CLC to the paper disks occurred in controls, either in the presence of absence of tubulin.
  • the K; values were calculated by linear regression of a double reciprocal plot of the experimental data in Microsoft Excel.
  • K m value of tubulin for CLC under the experimental conditions was first determined, with x intercept equal to -1/K m .
  • K m app K m in the presence of drug, was determined experimentally.
  • Fluorescence Spectroscopy Fluorescence Spectroscopy [0525] Fluorescence measurements were performed using a Perkin-Elmer LS50B specfrofluorimeter. PC-tubulin (0.2 mg/ml) was incubated in PEM50, 2 mM GTP, 3% DMSO, with 0 - 30 ⁇ M KPU-02. The interaction of KPU-02 with tubulin was reported by 4,4'-dianilino- l,l'-binaphthyl- 5,5'-disulfonic acid, dipotassium salt (bis-ANS; Molecular Probes, Eugene, OR) fluorescence, with an excitation wavelength of 395 nm and an emission wavelength maximum of 487 nm. Excitation and emission band passes were 10 nm.
  • Inhibition of microtubule polymerization by KPU-02 [0529] KPU-02, CA4, and CLC were assayed for their ability to alter the polymerization of MAP-rich tubulin (MTP) (2 mg/ml) in a cell-free system in vitro.
  • MTP MAP-rich tubulin
  • KPU-02 was a more potent inhibitor towards MTs assembled with glycerol and DMSO seeds as compared to MTs assembled in the presence of MAPs that copurify with tubulin.
  • microtubule polymer in the absence of stabilizing MAPs did not reach steady state over a 2-hour period, these assays demonstrated that KPU-02 interacts directly with purified tubulin and that it does not exert its primary effect through a MAP.
  • KPU-02 and CA4 inhibited MT polymerization more powerfully than CLC as measured by light scattering (Figure 44) and sedimentation analysis (Figure 45).
  • MTP (2 mg/ml) was polymerized into microtubules in the presence of a range of drug concentrations and allowed to reach steady state as monitored by light scattering at 350 nm.
  • Figure 41 depicts turbidity specfra of microtubule protein polymerization in the presence of DMSO drug vehicle (0), 1.25 ⁇ M ( ⁇ ), 2.5 ⁇ M (— ), and 5 ⁇ M (o) NPI-2358 (a), CA4 (b) and CLC (c).KPU-02 and CA4 inhibited MT polymerization with comparable potencies.
  • Figure 45 depicts inhibition of microtubule polymerization in the absence or presence of a range of KPU-02 (o),CA4 (p), and colchicine (0) concentrations.
  • the total polymer mass after 75 minutes of assembly was determined by sedimentation. Error bars are standard deviation values from three experiments.
  • the concentration at which polymerization was inhibited 50%> (ICso), is 2.4 ⁇ 0.4 ⁇ M for KPU-02, 2.2 ⁇ 0.3 ⁇ M for CA4, and 7.6 ⁇ 2.4 ⁇ M for CLC (Table 22). (Variances obtained by statistical analysis are reported as standard deviation values unless stated otherwise).
  • MTP displays aggregation kinetics, suggesting that KPU-02 and CN4 sequester protein to prevent microtubule assembly.
  • Table 22 Microtubule polymerization inhibition concentrations.
  • Figure 47 depicts electron microscopy used to record microtubules in the absence or presence of tested compounds. At 75 minutes, samples from polymer mass experiments were fixed and stained and viewed in a Jeol electron microscope-1200 EX11 at 2000x magnification. Representative electron micrographs of MAP-rich microtubules formed in vitro at steady state in the presence of (A) KPU-02, (b) CA4, and (C) CLC. Scale bar, 10 ⁇ M.
  • Figure 48 depicts a graphical summary, of MT length decrease at steady state in the presence of KPU-02, CA4, and colchicine. Black bars, 1.25 ⁇ M, and shaded bars, 2.5 ⁇ M drug.
  • KPU-02, CA4 and CA4 did not affect MT nucleation.
  • the numerous, short microtubules formed in the polymerization reactions evidence that the presence of KPU-02, CA4, or CLC does not affect nucleation. If nucleation were affected, then fewer, longer microtubules, as opposed to numerous, shorter microtubules would have been observed in drug-treated versus control samples.
  • KPU-02 and CA4 were comparably potent in decreasing the average MT length. At 1.25 ⁇ M, the lowest drug concentration analyzed by elecfron microscopy, KPU-02 and CA4 decreased mean MT length by approximately 70%, and CLC by 40%> (Figure 48). [0536] At drug concentrations over the IC 50 for in vitro microtubule polymerization, microtubules are not observed by elecfron microscopy for KPU-02 and CA4. hi contrast, microtubules were observed by electron microscopy for all concentrations of CLC assayed.
  • microtubule protein in the presence of KPU- 02 and CA4 displays aggregation kinetics, characterized by a linear increase in light absorbance over time ( Figure 44A and 44B), whereas in the presence of CLC, light scattering polymer reaches steady state ( Figure 44C).
  • Figure 44A and 44B a linear increase in light absorbance over time
  • Figure 44C steady state
  • Figure 49A depicts fluorescence emission spectra of tubulin in the presence of increasing KPU-02. Drug binding results in quenching of bis-ANS fluorescence.
  • Figure 49B depicts fluorescence emission maxima at 487 nm fit to obtain the K d of tubulin for KPU-02, 10 ⁇ M, standard deviation 1.6 ⁇ M. set, residuals.
  • Figure 49C depicts the double reciprocal transformation of the binding data assuming a one mole drug/mole tubulin. dimer.
  • FIG. 50 depicts the results of an inhibition assay where phosphocellulose-purified tubulin (0.2 mg/ml) was incubated with various concentrations of [ 3 H]CLC in the absence (0), or presence of 10 ⁇ M KPU-02 (o) or 10 ⁇ M CA4 ( ⁇ ).
  • Tubulin-CLC Km was 11 ⁇ 4.4 ⁇ M and inhibition constants for KPU-02 and CA4 were 3.2 ⁇ 1.7 ⁇ M and 2.4 ⁇ 0.3 ⁇ M, respectively. Constants were calculated from three independent experiments.
  • the K m may be considered the overall K of tubulin for CLC, however, due to the time-dependence of CLC binding, the K m is greater than the reported values for the K*
  • the Kj for KPU-02 and CA4 was 3.2 ⁇ 1.7 ⁇ M and 2.4 ⁇ 0.3 ⁇ M, respectively.
  • the Kj is defined as the amount of drug required to inhibit CLC binding by 50%) and it is based on the amount of radioactive CLC bound to tubulin.
  • the Kj is a measure of the drugs' ability to compete with CLC; it is not a direct measurement of drug-tubulin binding dissociation because of the method in which binding affinity is reported.
  • CA4 a structural analog of CLC, also competitively inhibited CLC binding. Without being bound to any particular theory, it appears that despite sharing a tubulin binding region with CLC, KPU-02 and CA4 interact with tubulin and inhibit microtubules by a mechanism distinct from that of CLC.
  • EXAMPLE 15 In Vivo Action on Microtubules 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 Eagle's medium supplemented with 5% fetal bovine serum, 0.1% penicillin/streptomycin and nonessential amino acids (Sigma) in 250-ml tissue culture flasks or 35-mm six-well plates (doubling time, 29 hours) at 37°C in 5% CO 2 . 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 tested agent or DMSO vehicle, and incubation was continued at 37°C for 20 hours.
  • Mitotic Progression The fraction of cells in mitosis at a given drug concenfration (mitotic index) was determined in the breast cancer cell line MCF7. Cells were plated at a density of 3 x 10 cells/ml in six-well plates. After 24 hours, cells were incubated in the absence or presence of drug over a range of concentrations (1 nM to 1 ⁇ M) for 20 hours.
  • 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 FITC-conjugated secondary antibody and DAPI. Coverslips were mounted using Prolong antifade media (Molecular Probes, Eugene OR).
  • microtubule dynamics [0545] The positions of the plus ends of microtubules over time were tracked using the Metamorph Track Points application exported to Microsoft Excel and analyzed using Real Time Measurement software. The lengths of individual microtubules were graphed as a function of time. Individual growth and shortening rates were determined by linear regression. Changes of > 0.5 ⁇ m between two points were considered to be growth or shortening events, and changes of ⁇ 0.5 ⁇ m between two points were considered to be periods of attenuated dynamics or pause. At least 25 microtubules were analyzed for each condition. Results are the mean and standard deviation of at least three independent experiments.
  • 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 time-based rescue frequency per microtubule was calculated by dividing the total number of rescues per microtubule by the time spent shortening.
  • the distance-based catastrophe and rescue frequencies were calculated similarly by dividing the number of transitions by the length grown or shortened, respectively.
  • Microtubules that were visible for ⁇ 2 min were included in the frequency analysis.
  • Dynamicity per microtubule was calculated as the length grown and shortened divided by the total life span of the microtubule. Microtubules that were visible for > 0.3 min were included in the dynamicity analysis.
  • Figure 51 depicts log [Drug] response curves for mitotic progression inhibition by KPU-02, CA4, and CLC.
  • MCF7 cells were cultured in the presence of NPI-2358 (o), CA4 (o), and colchicine (0).
  • To evaluate mitotic indices MCF7 cells were plated at a density of 3 x 10 4 cells/ml in six-well plates. After 24 hours, cells were incubated in the absence or presence of drug over a range of concentrations (1 nM to 1 ⁇ M) for 20 hours. Cells were fixed and stained with DAPI to visualize nuclei.
  • MT-targeting agents block mitosis at the metaphase to anaphase transition. Mitotic block at the metaphase to anaphase transition is associated with suppression of MT dynamics. Without being bound to any particular theory, the earlier prometaphase block, together with the depletion of MT polymer, suggests a distinct mechanism of action for KPU-02 as compared with other MT depolymerizing drugs, e.g., vinblastine, which at low concentrations stabilize MT dynamics.
  • MT depolymerizing drugs e.g., vinblastine
  • Figure 53 A-C depicts immunofluorescence microscopy images of MCF7 cells treated with KPU-02 (A), CA4 (B), and CLC (C) for 20 hours. Mitotic spindle destruction with increasing drug concentration. 1-4, Alpha and beta tubulin in confrol (1), a concentration of IC 25 for mitotic block (2), the IC 5 o for mitotic block (3), and twice the IC50 for mitotic block (4); 5-8, corresponding images of DNA for the adjoining panels. There were no normal, bipolar spindles at the IC 25 for KPU-02 or CA4 ( Figures 53A and B). Compound-treated cells had monopolar or bipolar spindles with uncongressed chromosomes.
  • Figure 54A-C depicts immunofluorescence microscopy images of MCF7 cells freated with KPU-02 (a), CA4 (b), and CLC (c) for 20 hours.
  • Alpha and beta tubulin in confrol (1) a concenfration of IC 25 for mitotic block (2), the IC 50 for mitotic block (3), and 2XIC 5 o for mitotic block (4); 5-8, corresponding images of DNA for the adjoining panels.
  • tubulin is sequestered in these interphase cells, despite the presence of intracellular stabilizing MAPs, just as MAP-rich tubulin is sequestered in in vitro polymer mass assays.
  • KPU-02 did not have a measurable effect on MT dynamic instability at concentrations effecting 25%> (Table 24) or 50% (Table 25) of the maximal mitotic block in MCF7 cells.
  • Table 24 concentrations effecting 25%> (Table 24) or 50% (Table 25) of the maximal mitotic block in MCF7 cells.
  • Table 25 50% of the maximal mitotic block in MCF7 cells.

Abstract

L'invention concerne des composés représentés par la structure suivante (I) ainsi que des méthodes de fabrication de ces composés. Ces méthodes consistent à faire réagir un diacyldicétopipérazine avec un premier aldéhyde afin de produire un composé intermédiaire ; et à faire réagir un composé intermédiaire avec un second aldéhyde afin de produire la classe de composés présentant la structure générique, le premier aldéhyde et le second aldéhydes étant choisis dans le groupe constitué d'un oxazolecarboxaldéhyde, d'un imidazolecarboxaldéhyde, d'un benzaldéhyde, de dérivés d'imidazolecarboxaldéhyde et de dérivés de benzaldéhyde, ce qui permet d'obtenir le composé selon l'invention. Dans la formule (I), R1, R1 , R1 , R2, R3, R4, R5, et R6, X1 et X2, Y, Z, Z1, Z2, Z3 et Z4 peuvent chacun être séparément définis d'une manière conforme au descriptif. L'invention concerne également des compositions et des méthodes qui permettent de traiter la prolifération vasculaire.
EP05712910A 2004-02-04 2005-02-04 Dehydrophenylahistines et leurs analogues et la synthese des dehydrophenylahistines et de leurs analogues Withdrawn EP1711487A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US54207304P 2004-02-04 2004-02-04
US62426204P 2004-11-01 2004-11-01
PCT/US2005/003636 WO2005077940A1 (fr) 2004-02-04 2005-02-04 Dehydrophenylahistines et leurs analogues et la synthese des dehydrophenylahistines et de leurs analogues

Publications (1)

Publication Number Publication Date
EP1711487A1 true EP1711487A1 (fr) 2006-10-18

Family

ID=34864489

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05712910A Withdrawn EP1711487A1 (fr) 2004-02-04 2005-02-04 Dehydrophenylahistines et leurs analogues et la synthese des dehydrophenylahistines et de leurs analogues

Country Status (9)

Country Link
EP (1) EP1711487A1 (fr)
JP (1) JP2007520565A (fr)
KR (1) KR101228104B1 (fr)
AU (1) AU2005212399B2 (fr)
BR (1) BRPI0506655A (fr)
CA (1) CA2553630A1 (fr)
IL (1) IL176932A0 (fr)
NZ (1) NZ548659A (fr)
WO (1) WO2005077940A1 (fr)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL166628A0 (en) 2002-08-02 2006-01-15 Nereus Pharmaceuticals Inc Dehydrophenyl lahistins and analogs thereof and the synthesis of dehydrophenyllahistins and analogs thereof
US7919497B2 (en) 2002-08-02 2011-04-05 Nereus Pharmaceuticals, Inc. Analogs of dehydrophenylahistins and their therapeutic use
US7935704B2 (en) 2003-08-01 2011-05-03 Nereus Pharmaceuticals, Inc. Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
EP1926724A1 (fr) * 2005-09-21 2008-06-04 Nereus Pharmaceuticals, Inc. Analogues de deshydrophenylahistines et utilisation therapeutique de ceux-ci
US8129527B2 (en) 2006-11-03 2012-03-06 Nereus Pharmacuticals, Inc. Analogs of dehydrophenylahistins and their therapeutic use
MY185650A (en) * 2013-10-11 2021-05-27 Beyondspring Inc Cancer treatment with combinantion of plinabulin and taxane
SG11201706281YA (en) * 2015-02-12 2017-09-28 Beyondspring Pharmaceuticals Inc Use of plinabulin in combination with immune checkpoint inhibitors
BR112017018964A2 (pt) 2015-03-06 2018-05-22 Beyondspring Pharmaceuticals Inc uso de plinabulin e métodos para tratar tumor cerebral
AU2016229294B2 (en) 2015-03-06 2021-11-04 Beyondspring Pharmaceuticals, Inc. Method of treating cancer associated with a RAS mutation
DK3334726T3 (da) * 2015-07-13 2022-05-16 Beyondspring Pharmaceuticals Inc Plinabulinsammensætninger
CA3013467A1 (fr) 2016-02-08 2017-08-17 Beyondspring Pharmaceuticals, Inc. Compositions contenant du tucaresol ou ses analogues
EP3463337A4 (fr) 2016-06-06 2020-02-12 Beyondspring Pharmaceuticals, Inc. Composition et méthode permettant de réduire la neutropénie
WO2018028420A1 (fr) * 2016-08-12 2018-02-15 青岛海洋生物医药研究院股份有限公司 Forme polycristalline de composé de type déhydrophénylahistine, son procédé de fabrication et de purification et son application.
WO2018129381A1 (fr) 2017-01-06 2018-07-12 Beyondspring Pharmaceuticals, Inc. Composés se liant à la tubuline et leur usage thérapeutique
BR112019015974A2 (pt) 2017-02-01 2020-03-31 Beyondspring Pharmaceuticals, Inc. Método para reduzir neutropenia
NZ757213A (en) * 2017-03-13 2022-01-28 Beyondspring Pharmaceuticals Inc Compositions of plinabulin and use thereof
US11786523B2 (en) 2018-01-24 2023-10-17 Beyondspring Pharmaceuticals, Inc. Composition and method for reducing thrombocytopenia
CN108524442B (zh) * 2018-06-05 2022-01-28 深圳海王医药科技研究院有限公司 一种抗肿瘤药物的注射剂及其制备方法
JP2018199726A (ja) * 2018-09-26 2018-12-20 ビヨンドスプリング インコーポレイテッド プリナブリン及びタキサンの組合せがん治療
CA3234699A1 (fr) * 2021-10-07 2023-04-13 Beyondspring Pharmaceuticals, Inc. Methodes de traitement de cancers et de tumeurs

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3318789A1 (de) * 1983-05-24 1984-11-29 Eduard Gerlach GmbH Chemische Fabrik, 4990 Lübbecke Deodorant
GB9402807D0 (en) * 1994-02-14 1994-04-06 Xenova Ltd Pharmaceutical compounds
US6069146A (en) * 1998-03-25 2000-05-30 The Regents Of The University Of California Halimide, a cytotoxic marine natural product, and derivatives thereof
US6358957B1 (en) * 1998-11-12 2002-03-19 Nereus Pharmaceuticals, Inc. Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds
KR100831400B1 (ko) * 2000-01-18 2008-05-21 니리어스 파마슈티컬즈, 인크. 세포분열 저해제 및 그의 제조방법
HUP0302358A3 (en) * 2000-05-09 2007-09-28 Adpharma Piperazinedione compounds and use of them for producing pharmaceutical compositions
IL166628A0 (en) * 2002-08-02 2006-01-15 Nereus Pharmaceuticals Inc Dehydrophenyl lahistins and analogs thereof and the synthesis of dehydrophenyllahistins and analogs thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2005077940A1 *

Also Published As

Publication number Publication date
IL176932A0 (en) 2006-12-10
KR101228104B1 (ko) 2013-02-01
NZ548659A (en) 2011-01-28
AU2005212399B2 (en) 2011-09-22
AU2005212399A1 (en) 2005-08-25
JP2007520565A (ja) 2007-07-26
WO2005077940A1 (fr) 2005-08-25
KR20060124743A (ko) 2006-12-05
CA2553630A1 (fr) 2005-08-25
BRPI0506655A (pt) 2007-05-08

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
US8247552B2 (en) Analogs of dehydrophenylahistins and their therapeutic use
US8129527B2 (en) Analogs of dehydrophenylahistins and their therapeutic use
JP4616649B2 (ja) デヒドロフェニラヒスチンおよびそれらの類似体、ならびにデヒドロフェニラヒスチンおよびそれらの類似体の合成
WO2007035841A9 (fr) Analogues de deshydrophenylahistines et utilisation therapeutique de ceux-ci
ZA200607151B (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
WO2011084962A1 (fr) Analogues de déshydrophénylahistines
MXPA06008810A (en) Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
CN101633655A (zh) 脱氢苯基阿夕斯丁及其类似物以及脱氢苯基阿夕斯丁及其类似物的合成

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20060823

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: PALLADINO, MICHAEL, A.

Inventor name: LLOYD, GEORGE, KENNETH

Inventor name: NICHOLSON, BENJAMIN

Inventor name: HAYASHI, YOSHIO

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1097836

Country of ref document: HK

17Q First examination report despatched

Effective date: 20081230

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20121020

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1097836

Country of ref document: HK