US20050065217A1 - Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents - Google Patents

Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents Download PDF

Info

Publication number
US20050065217A1
US20050065217A1 US10/861,186 US86118604A US2005065217A1 US 20050065217 A1 US20050065217 A1 US 20050065217A1 US 86118604 A US86118604 A US 86118604A US 2005065217 A1 US2005065217 A1 US 2005065217A1
Authority
US
United States
Prior art keywords
compound
alkyl
hydrogen
aryl
carbon atoms
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.)
Abandoned
Application number
US10/861,186
Inventor
Kevin Pinney
Feng Wang
Maria Mejia
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.)
Baylor University
Original Assignee
Baylor University
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 Baylor University filed Critical Baylor University
Priority to US10/861,186 priority Critical patent/US20050065217A1/en
Publication of US20050065217A1 publication Critical patent/US20050065217A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/22Amides of acids of phosphorus
    • C07F9/24Esteramides
    • C07F9/2454Esteramides the amide moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/247Esteramides the amide moiety containing a substituent or a structure which is considered as characteristic of aromatic amines (N-C aromatic linkage)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • C07F9/5728Five-membered rings condensed with carbocyclic rings or carbocyclic ring systems

Definitions

  • Tubulin is currently among the most attractive therapeutic targets in new drug design for the treatment of solid tumors. 1c
  • CA-4 combretastatin A-4
  • the nuclear membrane is broken down and the cytoskeletal protein tubulin is able to form centrosomes (also called microtubule organizing centers) and through polymerization and depolymerization of tubulin the dividing chromosomes are separated.
  • centrosomes also called microtubule organizing centers
  • the most recognized and clinically useful members of this class of antimitotic, antitumor agents are vinblastine and vincristine 3 along with taxol.
  • FIG. 1 illustrates 3-(3′, 4′, 5′-trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxybenzo[b]thiophene.
  • FIG. 2 illustrates 2-(3′, 4′, 5′-trimethoxybenzoyl)-3-(4′-methoxyphenyl)-6-methoxybenzo[b]furan.
  • FIG. 3 illustrates benzo[b]thiophene Phenol (BBT-OH).
  • FIG. 4 illustrates benzo[b]thiophene prodrug (BBT-P).
  • FIG. 5 illustrates in vivo biological data for benzo[b]thiophene prodrug (BBT-P).
  • FIG. 6 illustrates a synthetic route for preparation of phenylindole derivatives.
  • FIG. 7 illustrates a COSY NMR for 2-phenyl indole (aromatic region) a compound 31.
  • FIG. 8 illustrates a cyclized isomer without aryl migration (no evidence for its formation).
  • FIG. 9 illustrates a preparation of 2-phenylindole 31 in a one-pot reaction.
  • FIG. 10 illustrates a designed synthetic route for preparation of indole-based analog.
  • FIG. 11 illustrates a preparation of indole-based analog.
  • FIG. 12 illustrates a synthesis of indole-based disodium prodrug salt.
  • FIG. 13 illustrates another synthesis of indole-based disodium prodrug.
  • FIG. 14 illustrates another synthesis of indole-based disodium prodrug.
  • FIG. 15 illustrates a synthesis of indole based phosphoramidate prodrug.
  • FIG. 16 illustrates another synthesis of indole-based disodium prodrug salt.
  • FIG. 17A illustrates a combretastatin A-4 pro-drug.
  • FIG. 17B illustrates a phosphoramidate analog 10.
  • FIG. 18 illustrates a synthesis of phosphoramidate 10.
  • FIG. 19 illustrates a model system used for phosphoramidate synthesis.
  • FIG. 20 illustrates a synthesis of phosphoramidate 10 from from (Z)-3′-nitro combrestastatin analog 7B.
  • FIG. 21 illustrates substituted 4-methoxyindole amines and/or phenols.
  • FIG. 22 illustrates substituted 4-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 23 illustrates further substituted 4-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 24 illustrates substituted 6-methoxyindole amines and/or phenols.
  • FIG. 25 illustrates substituted 6-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 26 illustrates substituted 6-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 27 illustrates substituted 4-methoxy-3-arylindole amines and/or phenols.
  • FIG. 28 illustrates substituted 4-methoxy-3-arylindole phospate -moieties and phosphoramidates.
  • FIG. 29 illustrates further substituted 4-methoxy-3-arylindole phospate moieties and phosphoramidates.
  • FIG. 30 illustrates 2-(4′-Methoxyphenyl)-3-(3′′, 4′′, 5′′-trimethoxybenzoyl)-4- methoxyindole.
  • FIG. 31 illustrates 2-(3′, 4′, 5′-Trimethoxybenzoyl)-3-(4′′-methoxyphenyl)-6- methoxyindole.
  • FIG. 32 illustrates 2-(3′, 4′, 5′-Trimethoxybenzoyl)-3-(4′′-methoxyphenyl)-4- methoxyindole.
  • FIG. 33 illustrates Disodium 2-(3′-phosphoramidate4′-methoxyphenyl)-3-(3′′, 4′′, 5′′- trimethoxybenzoyl)-6-methoxyindole.
  • FIG. 34 illustrates 2-(3′-Hydroxy-4′-methoxyphenyl)-3-(3′′, 4′′, 5′′-trimethoxybenzoyl)- 4-methoxyindole.
  • FIG. 35 illustrates 2-(3′-Amino-4′-methoxyphenyl)-3-(3′′, 4′′, 5′′-trimethoxybenzoyl)- 4-methoxyindole.
  • FIG. 36 illustrates Disodium 2-[(4′-methoxyphenyl)-3′-O-phosphate]-3-(3′′, 4′′, 5′′trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 37 illustrates 2-(3′-Diethylphosphoramidate-4′-methoxyphenyl)-3-(3′′, 4′′, 5′′- trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 38 illustrates Disodium 2-(3′-phosphoramidate-4′-methoxyphenyl)-3-(3′′, 4′′, 5′′- trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 39 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3′′-hydroxy-4′′-methoxyphenyl)-6- methoxyindole.
  • FIG. 40 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3′′-amino-4′′-methoxyphenyl)-6- 10 methoxyindole.
  • FIG. 41 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3 ′′- O-phosphate)]-6-methoxyindole.
  • FIG. 42 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3′′- diethylphosphoramidate)]-6-methoxyindole.
  • FIG. 43 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3′′- phosphoramidate)]-6-methoxyindole.
  • FIG. 44 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3′′-hydroxy-4′′-methoxyphenyl)-4- methoxyindole.
  • FIG. 45 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3′′-amino4′′-methoxyphenyl)-4- 20 methoxyindole.
  • FIG. 46 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3′′- O-phosphate)] -4-methoxyindole.
  • FIG. 47 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3′′- diethylphosphoramidate)]-4-methoxyindole.
  • FIG. 48 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4′′-methoxyphenyl-3 ′′- phosphoramidate)]-4-methoxyindole.
  • FIG. 49 illustrates substituted 3-phosphoramidate derivatives of combretastatin A-4.
  • FIG. 50 illustrates Disodium (Z)- I -[(4′-methoxyphenyl)-3′-phosphoramidate]-2- (3′′,4′′,5′′-trimethoxyphenyl)ethene
  • FIG. 51 illustrates substituted 3-phosphoramidate salts of combretastatin A-4.
  • the phenolic derivative of the 3,4-5-trimethoxybenzo[b]thiophene compound ( FIG. 3 ) has pronounced cytotoxicity and demonstrates outstanding inhibition of tubulin polymerization 36 and the pro-drug disodium phosphate salt form of this compound ( FIG. 4 ) demonstrates in vitro and in vivo cytotoxicity as a vascular targeting and destruction agent (which includes a component of tubulin binding (phenolic form of drug) 36,37 and subsequent inhibition of tubulin polymerization).
  • mice Female scid mice were single dose ip administered with CA-4P, and benzo[b]thiophene phosphate prodrug at 400 mg/kg (i.e. MDT of CA-4P) after one week of MHEC inoculation (1 ⁇ 10 ⁇ 6 /mouse). Studies were carried out through a collaboration with Professors Ronald W. Pero and Klaus Edvardsen, University of Lund, Sweden (Note: PbT Prodrug 20 is the same compound that is referred to as BBT-P).
  • FIGS. 6, 9 and 11 A typical synthesis of indole-based ligand 33 is shown in FIGS. 6, 9 and 11 .
  • Secondary amine 30 was prepared by treatment of m-anisidine and 2-bromo-4methoxyacetophenone under basic condition (ethanolic potassium hydroxide) at 0° C. Treatment of amine 30 with PPA resulted in the formation of two regioisomers. These isomers have poor solubility in EtOAc, CH 2 Cl 2 and EtOH. Indole 31 was purified (from indole 32) by trituration in acetone. The structure of this isomer was confirmed by NMR analysis. COSY NMR was taken in order to study, in detail, the coupling relationship between the protons.
  • the enlarged COSY spectrum for the aromatic region of ligand 31 is shown in FIG. 5 .
  • This COSY NMR spectrum shows a strong coupling between H a and H b which each appear as a doublet.
  • H c is coupled by the proton attached to the nitrogen into a small doublet.
  • H d is coupled only by H e into a corresponding doublet, while H e is coupled both by an ortho coupling (H d ) and by a meta coupling (H f ) into a doublet of doublet pattern.
  • H f is coupled by H e into a doublet.
  • Further evidence of the formation of 2-phenyl indole 31 is the chemical shift of the proton H c on the ring which contains nitrogen.
  • indole 31 was mixed with trimethoxybenzoyl chloride. Since both reagents are solid, a solvent with a high boiling point was needed. 1,2-dichlorobenzene was chosen in this case since it has a boiling point of 180° C. Under these condition, indole 33 was obtained in moderate yield following purification by flash column chromatography and recrystallization. NMR spectroscopy suggests that the structure of indole 33 is that indicated in FIG. 11 .
  • phosphorous based prodrug derivatives of the nitrogen analog of combretastatin A-4 may have therapeutic advantages as selective tumor vasculature destruction agents.
  • These compounds are primarily phosphoramidate derivatives and related phosphate dianions that are assembled on the 3 amino substituent of the nitrogen analog of CA-4.
  • Combretastatin CA-4P prodrug ( FIG. 17A ) is one of the leading new candidates from among a relatively small collection of known world compounds which display this vaxcular targeting. Discovered by Professor George R. Pettit (Arizona State University) from a willow tree (combretum caffrum) in South Africa in the 1970s, this compound is currently undergoing phase I clinical evaluation sponsored and licensed by OXiGENE, Inc.
  • CA-4 Combretastatin A-4
  • CA-4 is a potent inhibitor of tubulin polymerization which binds to the colchicine site on ⁇ -tubulin.
  • CA-4 itself does not demonstrate destruction of tumor vasculature, while CA-4 prodrug is very active in terms of tumor vasculature destruction. It is very likely that the phosphate ester portion of the prodrug undergoes dephosphorylation (perhaps through the action of endothelial alkaline phosphatases) selectively at sites of enhanced vascularization to reveal the potent CA-4 itself which destroys the tumor cell through an inhibition of tubulin polymerization.
  • the dephosphorylation event takes place selectively at tumor cells since tumor cells represent sites of prolific vascularization and alkaline phosphatases appear to be present at elevated concentrations in the endothelial cells lining tumor vasculature. This need for enhanced vascularization is not necessary for healthy cells. Hence, this dual-mode reactivity profile is clearly important in order to target tumor cells selectively over healthy cells. This is a proposal which has been advanced by Professor Ronald Pero (OXiGENE, Inc., University of Lund) for which a variety of strong evidence has been obtained.
  • Phosphoramidate 10 below was obtained following the procedure reported by Taylor and coworkers for unrelated aryl amines. 28 Treatment of arylamine 7B with diethylchiorophosphite in anhydrous ether followed by oxidation with m-CPBA produced the phosphoramidate 10 in moderate yield ( FIG. 18 ).
  • the IC 50 values for inhibition of tubulin polymerization are 1.2 ⁇ 0.1 ⁇ M for CA-4,>80 ⁇ M for CA-4 prodrug, 1.0 ⁇ 0.2 ⁇ M for phenstatin and 21 ⁇ 3 ⁇ M for phenstatin prodrug; similar results are expected for the amino-CA-4 8 and the phosphoramidate 10.
  • the IC 50 for the amino-CA-4 8 is 1.2 ⁇ 0.02 ⁇ M, and the phosphoramidate 10 has little if any activity.
  • 32 TABLE III Comparative GI 50 Values against Human Cancer Cell Lines for Amine-CA-4 8, Amine-CA-4 Prodrug 10. Phenstatin. Phenstatin Prodrug and Combretastatin A-4 Prodrug.
  • Amine-CA-4 Phenstatin Combretastatin Cell Type Cell-Line Amine-CA-4 8 a Prodrug 10 a Phenstatin Prodrug b A-4 Prodrug b Ovarian OVCAR-3 ND 1.9 ⁇ 10 ⁇ 1 2.3 ⁇ 10 ⁇ 3 2.5 ⁇ 10 ⁇ 3 2.3 ⁇ 10 ⁇ 2 CNS SF-295 ND 2.4 ⁇ 10 ⁇ 1 5.2 ⁇ 10 ⁇ 2 1.2 ⁇ 10 ⁇ 2 3.6 ⁇ 10 ⁇ 2 Lung-NSC NCI-H460 6.8 ⁇ 10 ⁇ 4 3.5 ⁇ 10 ⁇ 1 5.7 ⁇ 10 ⁇ 3 3.5 ⁇ 10 ⁇ 2 2.9 ⁇ 10 ⁇ 2 Colon KM20L2 ND 2.8 ⁇ 10 ⁇ 1
  • phosphoramidate analog 10 is able to provide a more soluble compound than the amine 8, thereby incrementing its bioavailability.
  • the P-N bond can be broken by serum phosphatases releasing the amine which can inhibit tubulin polymerization in a manner analogous to combretastatin
  • the growth of a tumor depends on the generation of blood vessels which will provide all the metabolites required during cell division.
  • the development of anti-angiogenic compounds is especially useful in the treatment of solid tumors, since these compounds have the potential capability of selectively disrupting the vasculature of tumor cells while leaving healthy cells in a viable situation.
  • the combretastatin A-4 prodrug has demonstrated anti-angiogenic activity since small doses of the drug are toxic to tumor vasculature. 34
  • Enhanced cytotoxic activity was observed against endothelial cells associated with the tumor vasculature of cancerous cells, while at the same time it was reported to have no effect against other endothelial cells which are located distant from the tumor itself.
  • Diethylchlorophosphite (0.103 g, 0.66 mmol) was dissolved in anhydrous diethyl ether (2.5 ml) and cooled to ⁇ 78° C.
  • Diisopropylethyl amine (0.187 g. 1.45 mmol) was dissolved in Et 2 O (1.0 ml) and added slowly over a period of 2 mm to the reaction mixture by syringe.
  • Amino-stilbene 8 was dissolved in Et 2 O (1.0 mL) and added slowly to the reaction mixture by syringe.
  • the reaction mixture was stirred under nitrogen at ⁇ 78° C. for 2 hours, followed by stirring for 1 hour at room temperature. The mixture was filtered, and the solvent was removed under reduced pressure.
  • IC 50 values for tubulin polymerization were determined according to the procedure described in Bai et al. Purified tubulin is obtained from bovine brain cells as described in Hamel and Lin. Various amounts of inhibitor were preincubated for 15 minutes at 37° C. with purified tubulin. After the incubation period, the reaction was cooled and GTP was added to induce tubulin polymerization. Polymerization was then monitored in a Gilford spectrophotometer at 350 nm.
  • the final reaction mixtures (0.25 ml) contained 1.5 mg/ml tubulin, 0.6 mg/ml microtubule-associated proteins (MAPs), 0.5 mM GTP, 0.5 mlM MgCl 2 , 4% DMSO and 0.1M 4-morpholineethanesulfonate buffer (MES, pH 6.4).
  • IC 50 is the amount of inhibitor needed to inhibit tubulin polymerization 50% with respect to the amount of inhibition that occurs in the absence of inhibitor.
  • the IC 50 value determined for 3 -(3′,4′,5′-trimethoxybenzoyl)-2-( 4′-methoxyphenyl)- 6-methoxyindole was 0.5-1.5 ⁇ M.
  • the ED50 value (defined as the effective dosage required to inhibit 50% of cell growth) of 3-( 3′,4′,5′ trimethoxybenzoyl)- 2-(4′-methoxyphenyl)-6-methoxyindole was found to be 0.0133 ⁇ g/mL.
  • 3-(3′,4′,5′-Trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxyindole was evaluated in terms of growth inhibitory activity against several human cancer cell lines, including pancreas, ovarian, CNS, lung-NSC, colon, and prostate lines.
  • the assay used is described in Monks et al. Briefly, the cell suspensions, diluted according to the particular cell type and the expected target cell density (5,000-40,000 cells per well based on cell growth characteristics), were added by pipet (100 ⁇ l) to 96-well microtiter plates. Inoculates were allowed a preincubation time of 24-28 hours at 37° C. for stabilization.
  • Indole and indole containing compounds, of therapeutic efficacy have been known for many, many years. What is truly unique about the indole compounds described in this application is the fact that these compounds are the first (to the best of our knowledge) indole-based ligands to incorporate the 3,4,5-trimethoxyaryl motif reminiscent of colchicine and combretastatin A-4 arranged in an appropriate molecular conformation such that a pseudo aryl-aryl pi stacking interaction can take place. It is our contention that such an aryl-aryl interaction of the appropriate centroid-to-centroid distance (approximately 4.7 ⁇ ) is imperative for enhanced binding affinity to the colchicine site on ⁇ -tubulin.
  • trimethoxyaryl motif seems optimal for enhanced tubulin binding, it is also very possible that another combination of alkoxy substituents (such as ethoxy, propoxy, isopropoxy, allyloxy, etc.) either as a trisubstituted pattern or as disubstituted (with one type of alkoxy moiety) and monosubstituted (with a different alkoxy moiety), or with three distinct types of alkoxy moieties may also have good tubulin binding characteristics. It is also conceivable that instead of having aryl alkoxy groups, it may be possible to substitute simply aryl-alkyl and aryl-alkenyl moieties and still maintain the enhanced cytotoxicity profile.
  • alkoxy substituents such as ethoxy, propoxy, isopropoxy, allyloxy, etc.
  • Phenolic groups may also have activity on these described indole ligands.
  • the synthesis of any of these modified indole-ligands will be very straight-forward for anyone skilled in the art, and often will only involve a different choice of initial starting materials.
  • To prepare these alternative ligands the same synthetic schemes ( FIGS. 6, 9 , 11 , 12 - 16 ), or similar schemes with only slight modifications may be employed.
  • FIGS. 6, 9 , 11 , 12 - 16 the same synthetic schemes with only slight modifications may be employed.
  • the carbonyl group can be replaced with an oxygen to generate a new compound which maintains the same or similar biological efficacy with tubulin.
  • the replacement of the carbonyl group in the described indole ligand may be replaced with an oxygen atom (ether linkage) to generate a new derivative which would be predicted to have good activity with tubulin.
  • This compound may be prepared by an addition elimination reaction utilizing the trimethoxyphenolic anion as a nucleophile as described by us for the benzo[b]thiophene compounds.
  • Other linkage atoms between the aryl aryl rings are conceivable as well.
  • compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Abstract

Trimethoxyphenyl substituted indole ligands have been discovered which demonstrate impressive cytotoxicity as well as a remarkable ability to inhibit tubulin polymerization. Such compounds as well as related derivatives are excellent clinical candidates for the treatment of cancer in humans. In addition, certain of these ligands, as pro-drugs, may well prove to be tumor selective vascular targeting and destruction chemotherapeutic agents or to have anti-angiogenesis activity resulting in the selective prevention and/or destruction of tumor cell vasculature.

Description

    BACKGROUND OF THE INVENTION
  • Tubulin is currently among the most attractive therapeutic targets in new drug design for the treatment of solid tumors.1c The heralded success of vincristine and taxol along with the promise of combretastatin A-4 (CA-4) prodrug and dolastatin 10, to name just a few, have firmly established the clinical efficacy of these antimitotic agents for cancer treatment.
  • An aggressive chemotherapeutic strategy toward the treatment of solid-tumor cancers continues to rely on the development of architecturally new and biologically more potent anti-tumor, anti-mitotic agents which mediate their effect through a direct binding interaction with tubulin. A variety of clinically-promising compounds which demonstrate potent cytotoxicity and antitumor activity are known to effect their primary mode of action through an efficient inhibition of tubulin polymerization.1 This class of compounds undergoes an initial interaction (binding) to the ubiquitous protein tubulin which in turn arrests the ability of tubulin to polymerize into microtubules which are essential components for cell maintenance and division.2 During metaphase of the cell cycle, the nuclear membrane is broken down and the cytoskeletal protein tubulin is able to form centrosomes (also called microtubule organizing centers) and through polymerization and depolymerization of tubulin the dividing chromosomes are separated. Currently, the most recognized and clinically useful members of this class of antimitotic, antitumor agents are vinblastine and vincristine3 along with taxol.4 Additionally, the natural products rhizoxin,5 combretastatin A-4 and A-2,6 curacin A,1 podophyllotoxin,7 epothilones A and B,8 dolastatin 109 and welwistatin10 (to name just a few) as well as certain synthetic analogues including phenstatin,11 the 2-styrylquinazolin-4(3H)-ones (SQO),12 and highly oxygenated derivatives of cis- and trans-stilbene13 and dihydrostilbene are all known to mediate their cytotoxic activity through a binding interaction with tubulin. The exact nature of this binding site interaction remains largely unknown , and definitely varies between the series of compounds. Photoaffinity labeling and other binding site elucidation techniques have identified several key binding sites on tubulin: colchicine site, vinca alkaloid site, and a site on the polymerized microtubule to which taxol binds.la.14
  • SUMMARY OF THE INVENTION
  • An important basic and essential aspect of this work requires a detailed understanding, on the molecular level, of the “small molecule” binding domain of both the α and β subunits of tubulin. The tertiary structure of the α, β tubulin heterodimer was reported earlier this year by Downing and co-workers at a resolution of 3.7 Å using a technique known as electron crystallography.15 This brilliant accomplishment culminates decades of work directed toward the elucidation of this structure and should facilitate the identification of small molecule binding sites, such as the colchicine site, through techniques such as photoaffinity and chemical affinity labeling.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates 3-(3′, 4′, 5′-trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxybenzo[b]thiophene.
  • FIG. 2 illustrates 2-(3′, 4′, 5′-trimethoxybenzoyl)-3-(4′-methoxyphenyl)-6-methoxybenzo[b]furan.
  • FIG. 3 illustrates benzo[b]thiophene Phenol (BBT-OH).
  • FIG. 4 illustrates benzo[b]thiophene prodrug (BBT-P).
  • FIG. 5 illustrates in vivo biological data for benzo[b]thiophene prodrug (BBT-P).
  • FIG. 6 illustrates a synthetic route for preparation of phenylindole derivatives.
  • FIG. 7 illustrates a COSY NMR for 2-phenyl indole (aromatic region) a compound 31.
  • FIG. 8 illustrates a cyclized isomer without aryl migration (no evidence for its formation).
  • FIG. 9 illustrates a preparation of 2-phenylindole 31 in a one-pot reaction.
  • FIG. 10 illustrates a designed synthetic route for preparation of indole-based analog.
  • FIG. 11 illustrates a preparation of indole-based analog.
  • FIG. 12 illustrates a synthesis of indole-based disodium prodrug salt.
  • FIG. 13 illustrates another synthesis of indole-based disodium prodrug.
  • FIG. 14 illustrates another synthesis of indole-based disodium prodrug.
  • FIG. 15 illustrates a synthesis of indole based phosphoramidate prodrug.
  • FIG. 16 illustrates another synthesis of indole-based disodium prodrug salt.
  • FIG. 17A illustrates a combretastatin A-4 pro-drug.
  • FIG. 17B illustrates a phosphoramidate analog 10.
  • FIG. 18 illustrates a synthesis of phosphoramidate 10.
  • FIG. 19 illustrates a model system used for phosphoramidate synthesis.
  • FIG. 20 illustrates a synthesis of phosphoramidate 10 from from (Z)-3′-nitro combrestastatin analog 7B.
  • FIG. 21 illustrates substituted 4-methoxyindole amines and/or phenols.
  • FIG. 22 illustrates substituted 4-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 23 illustrates further substituted 4-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 24 illustrates substituted 6-methoxyindole amines and/or phenols.
  • FIG. 25 illustrates substituted 6-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 26 illustrates substituted 6-methoxyindole phosphate ester moieties and phosphoramidates.
  • FIG. 27 illustrates substituted 4-methoxy-3-arylindole amines and/or phenols.
  • FIG. 28 illustrates substituted 4-methoxy-3-arylindole phospate -moieties and phosphoramidates.
  • FIG. 29 illustrates further substituted 4-methoxy-3-arylindole phospate moieties and phosphoramidates.
  • FIG. 30 illustrates 2-(4′-Methoxyphenyl)-3-(3″, 4″, 5″-trimethoxybenzoyl)-4- methoxyindole.
  • FIG. 31 illustrates 2-(3′, 4′, 5′-Trimethoxybenzoyl)-3-(4″-methoxyphenyl)-6- methoxyindole.
  • FIG. 32 illustrates 2-(3′, 4′, 5′-Trimethoxybenzoyl)-3-(4″-methoxyphenyl)-4- methoxyindole.
  • FIG. 33 illustrates Disodium 2-(3′-phosphoramidate4′-methoxyphenyl)-3-(3″, 4″, 5″- trimethoxybenzoyl)-6-methoxyindole.
  • FIG. 34 illustrates 2-(3′-Hydroxy-4′-methoxyphenyl)-3-(3″, 4″, 5″-trimethoxybenzoyl)- 4-methoxyindole.
  • FIG. 35 illustrates 2-(3′-Amino-4′-methoxyphenyl)-3-(3″, 4″, 5″-trimethoxybenzoyl)- 4-methoxyindole.
  • FIG. 36 illustrates Disodium 2-[(4′-methoxyphenyl)-3′-O-phosphate]-3-(3″, 4″, 5″trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 37 illustrates 2-(3′-Diethylphosphoramidate-4′-methoxyphenyl)-3-(3″, 4″, 5″- trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 38 illustrates Disodium 2-(3′-phosphoramidate-4′-methoxyphenyl)-3-(3″, 4″, 5″- trimethoxybenzoyl)-4-methoxyindole.
  • FIG. 39 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3″-hydroxy-4″-methoxyphenyl)-6- methoxyindole.
  • FIG. 40 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3″-amino-4″-methoxyphenyl)-6- 10 methoxyindole.
  • FIG. 41 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3 ″- O-phosphate)]-6-methoxyindole.
  • FIG. 42 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3″- diethylphosphoramidate)]-6-methoxyindole.
  • FIG. 43 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3″- phosphoramidate)]-6-methoxyindole.
  • FIG. 44 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3″-hydroxy-4″-methoxyphenyl)-4- methoxyindole.
  • FIG. 45 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-(3″-amino4″-methoxyphenyl)-4- 20 methoxyindole.
  • FIG. 46 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3″- O-phosphate)] -4-methoxyindole.
  • FIG. 47 illustrates 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3″- diethylphosphoramidate)]-4-methoxyindole.
  • FIG. 48 illustrates Disodium 2-(3′,4′,5′-trimethoxybenzoyl)-3-[(4″-methoxyphenyl-3 ″- phosphoramidate)]-4-methoxyindole.
  • FIG. 49 illustrates substituted 3-phosphoramidate derivatives of combretastatin A-4.
  • FIG. 50 illustrates Disodium (Z)- I -[(4′-methoxyphenyl)-3′-phosphoramidate]-2- (3″,4″,5″-trimethoxyphenyl)ethene
  • FIG. 51 illustrates substituted 3-phosphoramidate salts of combretastatin A-4.
  • DETAILED DESCRIPTION OF THE INVENTION
  • We have developed a working hypothesis suggesting that the discovery of new antimitotic agents may result from the judicious combination of a molecular template (scaffold) which in appropriately substituted form (ie. phenolic moieties, etc.) interacts with estrogen receptor (ER), suitably modified with structural features deemed imperative for tubulin binding (arylalkoxy groups, certain halogen substitutions, etc.). The methoxy aryl functionality seems especially important for increased interaction at the colchicine binding site in certain analogs.16 Upon formulation of this hypothesis concerning ER molecular templates, our initial design and synthesis efforts centered on benzo [b]thiophene ligands modeled after raloxifene, the selective estrogen receptor modulator (SERM) developed by Eli Lilly and Co.17 Our initial studies resulted in the preparation of a very active benzo[b]thiophene-based antitubulin agent. 18-21 In further support of our hypothesis, recent studies have shown that certain estrogen receptor (ER) binding compounds as structurally modified estradiol congeners (2-methoxyestradiol, for example) interact with tubulin and inhibit tubulin polymerization.22 Estradiol is, of course, perhaps the most important estrogen in humans, and it is intriguing and instructive that the addition of the methoxy aryl motif to this compound makes it interactive with tubulin. It is also noteworthy that 2-methoxyestradiol is a natural mammalian metabolite of estradiol and may play a cell growth regulatory role especially prominent during pregnancy.
  • The design premise that molecular skeletons of traditional estrogen receptor (ER) binding compounds can be modified with structural motifs reminiscent of colchicine and combretastatin A-4 to produce inhibitors of tubulin polymerization has been validated by the benzo[b]thiophene and benzol[b]furan classes of new antimitotic agents. 18-21 The lead compounds in each series (FIGS. 1 and 2), demonstrate remarkable biological activity against a variety of human cancer cell lines. For example, the 3,4,5-trimethoxybenzo[b]thiophene (FIG. 1) demonstrates potent cytotoxicity and inhibition of tubulin polymerization. In the NCI 60 cell line panel,23 this compound produces a mean panel G150=2.63×10−7 M (see Table I).
  • Inhibition of tubulin polymerization by 3-(3′, 4′, 5′-trimethoxybenzoyl)-2-(4′- methoxyphenyl)-6-methoxybenzo[b]thiophene. 50% inhibition of the maximum tubulin assembly rate with 1.1 μM drug same assay with conbretastatin A4 gives a value of 0.73 μM.
  • Human cancer cell line studies (in vitro) by 3-(3′, 4′, 5′-trimethoxybenzoyl)-2-(4′- methoxyphenyl)-6-methoxybenzo[b]thiophene.
    TABLE I
    Inhibition of tubulin polymerization by 2-(3′, 4′, 5′-
    trimethoxybenzoyl)-3-(4′-methoxyphenyl)-6-methoxy-
    benzo[b]furan. IC50 = 2.1 pM (totally flat at 4 pM).
    Human cancer cell line studies (in vitro) by 2-(3′, 4′, 5′-
    trimethoxybenzoyl)-3-(4′-methoxyphenyl)-6-
    methoxybenzo[b]furan.
    Type of Cancer Cell Line Cancer Cell Line GI50 (uglmL)
    Pancreas - adn BXPC-3 0.038
    Neuroblast SK-N-SH 0.025
    Thyroid ca SW1736 0.047
    Lung-NSC NCI-H460 0.041
    Pharynx-sqam FADU 0.035
    Prostate DU-145 0.062
  • In addition, the phenolic derivative of the 3,4-5-trimethoxybenzo[b]thiophene compound (FIG. 3) has pronounced cytotoxicity and demonstrates outstanding inhibition of tubulin polymerization36 and the pro-drug disodium phosphate salt form of this compound (FIG. 4) demonstrates in vitro and in vivo cytotoxicity as a vascular targeting and destruction agent (which includes a component of tubulin binding (phenolic form of drug) 36,37 and subsequent inhibition of tubulin polymerization).
  • Initial in vivo studies are very encouraging (see FIG. 5). Female scid mice were single dose ip administered with CA-4P, and benzo[b]thiophene phosphate prodrug at 400 mg/kg (i.e. MDT of CA-4P) after one week of MHEC inoculation (1×10−6/mouse). Studies were carried out through a collaboration with Professors Ronald W. Pero and Klaus Edvardsen, University of Lund, Sweden (Note: PbT Prodrug 20 is the same compound that is referred to as BBT-P).
  • Based on these promising research results, our interest in designing an indole based antimitotic agent was initiated, and a synthetic route (Schemes 1-4, see FIGS. 3 A-D) was designed according to the synthesis of the benzo[b]thiophene derivatives.
  • The possibility clearly exists that some of the new indole-based ligands described herein, which are structurally related to combretastatin A-4, may also function through additional biological mechanisms involving anti-angiogenic activity. Clearly the ability to selectively disrupt the blood-flow to developing tumor cells is a potential breakthrough in the ever up-hill battle against cancer. Certain phenylindoles have been noted for inhibiting tubulin polymerization.27
  • A typical synthesis of indole-based ligand 33 is shown in FIGS. 6, 9 and 11. Secondary amine 30 was prepared by treatment of m-anisidine and 2-bromo-4methoxyacetophenone under basic condition (ethanolic potassium hydroxide) at 0° C. Treatment of amine 30 with PPA resulted in the formation of two regioisomers. These isomers have poor solubility in EtOAc, CH2Cl2 and EtOH. Indole 31 was purified (from indole 32) by trituration in acetone. The structure of this isomer was confirmed by NMR analysis. COSY NMR was taken in order to study, in detail, the coupling relationship between the protons. The enlarged COSY spectrum for the aromatic region of ligand 31 is shown in FIG. 5. This COSY NMR spectrum, shows a strong coupling between Ha and Hb which each appear as a doublet. Hc is coupled by the proton attached to the nitrogen into a small doublet. Hd is coupled only by He into a corresponding doublet, while He is coupled both by an ortho coupling (Hd) and by a meta coupling (Hf) into a doublet of doublet pattern. Hf is coupled by He into a doublet. Further evidence of the formation of 2-phenyl indole 31 is the chemical shift of the proton Hc on the ring which contains nitrogen. Though computer modeling (ChemDraw Ultra 4.5), the theoretical chemical shift value of 6.4 ppm is predicted for proton Hc (at the 3 position), which matches the peak shown in the actual NMR spectrum at 6.6 ppm. For the case where the proton is at the 2 position (FIG. 8), the chemical shift is predicted to be 7.03 ppm, which does not match any peak in the spectrum that was obtained. Based collectively on these studies, the formation of isomer 31 is confirmed, and the migration of the methoxyphenyl system is evidenced. The other isomer (indole 32) is soluble in acetone and is much more difficult to obtain in pure form (see FIG. 6).
  • Alternatively, another synthetic methodology can also be applied to the preparation of the desired 2-phenylindole. In 1984, Angerer and co-workers reported the synthesis of 2-phenylindoles in a one-pot reaction sequence (FIG. 9) as a route toward the development of new therapeutic agents for the treatment of endocrine disorders.25
  • Following this procedure (FIG. 9), two arylindole regioisomers were obtained in good yield. Recrystallization in EtOH afforded the desired isomer, 2-phenylindole 31, as a white crystalline material.
  • In order to synthesize the indole-based analog 33, Friedel-Crafts acylation was carried out by treating indole 31 with 3,4,5-trimethoxybenzoyl chloride in the presence of the Lewis-Acid AlCl3 ( FIG. 10). The reaction did not work under the regular conditions and only starting material was obtained following work-up. Attempts to modify the reaction conditions by increasing the reaction temperature or using other Lewis Acids, such as TiCl4, proved futile as well. Starting material was recovered in all cases. One possible explanation for this result is the fact that the nitrogen atom (containing a lone pair of electrons and an acidic proton) may disrupt the acylation process. According to this analysis, a Grignard reagent (ethylmagnesium bromide) was used to protect this nitrogen prior to the Friedel-Crafis acylation step. Still, only starting material was obtained following the reaction. Therefore, a new synthetic approach was brought into this study.
  • In 1977, Inion and co-workers reported the synthesis of a variety of aminoalkoxy4-benzoyl-3-indoles.26 The benzoate indole product was prepared by treatment of indole with the appropriate benzoyl chloride with heating (130-150° C.). HCl is generated under these conditions. A similar synthetic approach was used in the synthesis of the desired trimethoxybenzoate indole ligand 33 (FIG. 11).
  • The precursor, indole 31, was mixed with trimethoxybenzoyl chloride. Since both reagents are solid, a solvent with a high boiling point was needed. 1,2-dichlorobenzene was chosen in this case since it has a boiling point of 180° C. Under these condition, indole 33 was obtained in moderate yield following purification by flash column chromatography and recrystallization. NMR spectroscopy suggests that the structure of indole 33 is that indicated in FIG. 11.
  • Based on promising results obtained with benzo[b]thiophene and benzofuran analogs, the preparation of phosphate salts is detailed in FIGS. 12-14, the preparation of analogs is detailed in FIGS. 15-16 and the preparation of similar indole-based phosphate prodrug salts and phosphoramidate derivatives is detailed in FIGS. 21-51.
  • In addition to the phosphate ester prodrugs that are described in this application for indole-based anti-mitotic agents, we have also discovered that phosphorous based prodrug derivatives of the nitrogen analog of combretastatin A-4 (CA-4) may have therapeutic advantages as selective tumor vasculature destruction agents. These compounds are primarily phosphoramidate derivatives and related phosphate dianions that are assembled on the 3
    Figure US20050065217A1-20050324-P00900
    amino substituent of the nitrogen analog of CA-4. Although we describe two specific compounds and several obvious analogs, it should be apparent to anyone skilled in the art, that there are numerous other nitrogen phosphorous bond designs that might be assembled from the 3-amino-combretastatin A-4 structure and that would display similar functionality as prodrugs for the selective destruction of tumor vasculature.
  • Further significance is given to new drugs that bind to the colchicine site since it has recently been shown that combretastatin CA-4 also demonstrates anti-angiogenesis activity.24 An emerging area of cancer chemotherapy centers on the development of both anti-angiogenesis drugs which disrupt the new microvessel formation of developing tumors and vascular targeting and destruction agents which selectively target the vasculature of tumor cells while leaving healthy cells intact. Combretastatin CA-4P prodrug (FIG. 17A) is one of the leading new candidates from among a relatively small collection of known world compounds which display this vaxcular targeting. Discovered by Professor George R. Pettit (Arizona State University) from a willow tree (combretum caffrum) in South Africa in the 1970s, this compound is currently undergoing phase I clinical evaluation sponsored and licensed by OXiGENE, Inc.
  • Combretastatin A-4 (CA-4) is a potent inhibitor of tubulin polymerization which binds to the colchicine site on β-tubulin. Interestingly, CA-4 itself does not demonstrate destruction of tumor vasculature, while CA-4 prodrug is very active in terms of tumor vasculature destruction. It is very likely that the phosphate ester portion of the prodrug undergoes dephosphorylation (perhaps through the action of endothelial alkaline phosphatases) selectively at sites of enhanced vascularization to reveal the potent CA-4 itself which destroys the tumor cell through an inhibition of tubulin polymerization. The dephosphorylation event takes place selectively at tumor cells since tumor cells represent sites of prolific vascularization and alkaline phosphatases appear to be present at elevated concentrations in the endothelial cells lining tumor vasculature. This need for enhanced vascularization is not necessary for healthy cells. Hence, this dual-mode reactivity profile is clearly important in order to target tumor cells selectively over healthy cells. This is a proposal which has been advanced by Professor Ronald Pero (OXiGENE, Inc., University of Lund) for which a variety of strong evidence has been obtained.
  • Based in part on the good and promising biological results obtained for the 3′-nitrogen analogs of combretastatin A-4, a phosphoramidate analog has been prepared as a new combretastatin A-4 nitrogen prodrug (FIG. 17B).
  • Phosphoramidate 10 below was obtained following the procedure reported by Taylor and coworkers for unrelated aryl amines.28 Treatment of arylamine 7B with diethylchiorophosphite in anhydrous ether followed by oxidation with m-CPBA produced the phosphoramidate 10 in moderate yield (FIG. 18).
  • A previous attempt in the synthesis of the phosphoramidate analog 10 utilized the methodology reported by Bilha Fisher and Larisa Sheihet.29 This methodology presents a phosphoramidate intermediate, which can be isolated from the reduction of nitro aryl compounds to the corresponding aryl amines using diethylchlorophosphite as a biphilic reagent. The (Z)-nitro combretastatin analog 7B was considered a viable starting material for the synthesis of the phosphoramidate prodrug 10. This reaction was also tried using (Z)-1-(3′,4′,5′-trimethoxyphenyl)-2-(4″-nitrophenyl)ethene (synthesized in a similar manner as the other combretastatin containing analogs reported previously) as a model system (FIG. 19). In neither case was the phosphoramidate product observed. It is thought that the presence of methoxy groups as strong electron donating substituents on the stilbene system disfavors the reaction (FIG. 20).
  • It should be obvious to anyone skilled in the art of phosphate of phosporamidate chemistry that there are numerous other synthetic methods which can be employed to prepare phosphoramidates (such as 10) and their related salts (-NHPO3 −22Na+).
    TABLE II30
    In vitro Human Cancer Cell Line Study of Phosphoramidate
    Analog 10. GI50, TGI, and LC50 are reported as concentrations
    in μg/mL ND = Not determined
    Cell Type Cell Line GI50 TGI LC50
    Pancreas-a BXPC-3 1.5 × 10−1 5.7 × 10−1 >10
    Ovarian OVCAR-3 1.9 × 10−1 8.6 × 10−1 >10
    CNS SF-295 2.4 × 10−1 >10 >10
    Lung-NSC NCI-H460 3.5 × 10−1 >10 >10
    Colon KM20L2 2.8 × 10−1 6.1 × 10−1 >10
    Prostate −DU-145 2.6 × 10−1 2.6 × 10−1 >10
    Leukemia P388 3.1 × 10−1 ND ND
  • Biological evaluation (in vitro) suggests that the phosphoramidate prodrug 10 is less effective than the corresponding amine 8 (Table II). Pettit and co-workers reported a similar loss in biological activity in vitro for the phosphate prodrugs of combretastatin A-4 and phenstatin compared to the original compounds (Table III).31 These results might be explained by the bulkiness of the phosphorous group and its steric hindrance toward binding site recognition. In fact, Pettit and co-workers reported no inhibition of tubulin polymerization with the combretastatin prodrug while only a 40% activity is present for the phenstatin prodrug compared to phenstatin. The IC50 values for inhibition of tubulin polymerization are 1.2±0.1 μM for CA-4,>80 μM for CA-4 prodrug, 1.0±0.2 μM for phenstatin and 21±3 μM for phenstatin prodrug; similar results are expected for the amino-CA-4 8 and the phosphoramidate 10.31 The IC50 for the amino-CA-4 8 is 1.2±0.02 μM, and the phosphoramidate 10 has little if any activity.32
    TABLE III
    Comparative GI50 Values Against Human Cancer Cell Lines for Amine-CA-4 8,
    Amine-CA-4 Prodrug 10. Phenstatin. Phenstatin Prodrug and Combretastatin A-4
    Prodrug. GI50, values are reported as concentrations in μg/mL ND = Not
    determined,a Data obtained in collaboration with Dr. George R. Pettit.30b
    Data obtained from synthesis of phenstatin phosphate.
    Amine-CA-4 Phenstatin Combretastatin
    Cell Type Cell-Line Amine-CA-4 8a Prodrug 10a Phenstatin Prodrugb A-4 Prodrugb
    Ovarian OVCAR-3 ND 1.9 × 10−1 2.3 × 10−3 2.5 × 10−3 2.3 × 10−2
    CNS SF-295 ND 2.4 × 10−1 5.2 × 10−2 1.2 × 10−2 3.6 × 10−2
    Lung-NSC NCI-H460 6.8 × 10−4 3.5 × 10−1 5.7 × 10−3 3.5 × 10−2 2.9 × 10−2
    Colon KM20L2 ND 2.8 × 10−1 4.0 × 10−4 2.7 × 10−1 3.4 × 10−1
  • In terms of in vivo systems, phosphoramidate analog 10 is able to provide a more soluble compound than the amine 8, thereby incrementing its bioavailability. Under, in vivo biological conditions, the P-N bond can be broken by serum phosphatases releasing the amine which can inhibit tubulin polymerization in a manner analogous to combretastatin
  • Anti-Angiogenesis
  • The growth of a tumor depends on the generation of blood vessels which will provide all the metabolites required during cell division. The development of anti-angiogenic compounds is especially useful in the treatment of solid tumors, since these compounds have the potential capability of selectively disrupting the vasculature of tumor cells while leaving healthy cells in a viable situation. The combretastatin A-4 prodrug has demonstrated anti-angiogenic activity since small doses of the drug are toxic to tumor vasculature.34 Enhanced cytotoxic activity was observed against endothelial cells associated with the tumor vasculature of cancerous cells, while at the same time it was reported to have no effect against other endothelial cells which are located distant from the tumor itself.34, 35 The mechanism of action of combretastatin A-4 prodrug, as an anti-angiogenic drug for cancer treatment, is under investigation because the development of blood vessels is crucial for the survival and growth of solid tumors. One proposed mechanism for anti-angiogenesis involves induction of apoptosis (cell suicide) of the cells instead of necrosis. An evaluation of the ability of the new phosphoramidate 10, along with structurally similar compounds, to induce apoptosis of endothelial cells will be undertaken in the near future.
  • Synthesis of the Phosphoramidate Analog
  • (Z)- 1 -(3′-Diethylphosphoramidate-4′-methoxyphenyl)-2-(3″, 4″, 5″-trimethoxyphenyl)ethene 10.
  • Diethylchlorophosphite (0.103 g, 0.66 mmol) was dissolved in anhydrous diethyl ether (2.5 ml) and cooled to −78° C. Diisopropylethyl amine (0.187 g. 1.45 mmol) was dissolved in Et2O (1.0 ml) and added slowly over a period of 2 mm to the reaction mixture by syringe. Amino-stilbene 8 was dissolved in Et2O (1.0 mL) and added slowly to the reaction mixture by syringe. The reaction mixture was stirred under nitrogen at −78° C. for 2 hours, followed by stirring for 1 hour at room temperature. The mixture was filtered, and the solvent was removed under reduced pressure. A yellow oil was obtained which was dissolved in dry CH2Cl2 (5 mL). The oil was cooled to 40° C. and a solution of m-CPBA (0.193 g, 1.12 mmol) in CH2Cl2 (5 mL) was added. It was stirred over one hour at room temperature. After this time, the reaction mixture was cooled to −40° C. and filtered through a sintered glass funnel. The liquid was collected with vigorous stirring over sodium sulfite (5%) (20 ml) in order to quench the reaction. The product was isolated by extraction with CH2Cl2, and washed with a saturated solution of NaHCO3. The yellow oil which was obtained was dried over MgSO4. Purification by flash chromatography (70/30, hexanes/EtOAc) afforded the phosphoramidate 10 as a yellow oil (0.130 g, 0.29 mmol, 44%).
  • 'H-NMR (CDCl3, 360 MHz) δ7.12 (d, J=1.9 Hz, IH, ArH), 6.88 (dd, J=8.4 Hz, 2.0 Hz, IH, ArH), 6.72 (dd, J=8.4 Hz, 1.7, IH, ArH), 6.49 (s, 2H, ArH), 6.51 (d, J=12.1 Hz, IH, vinyl CH), 6.41 (d, J=12.1 Hz, IH, vinyl CH), 5.67 (d, J=10.0 Hz, NH), 4.02 (m, 4H, CH2), 3.83 (s, 3H, OCH3), 3.83 (s, 3H, OC3), 3.68 (s, 6H, OCH3), 1.25 (t, 6H, J=7.1 Hz, CH3).
  • 13C-NMR (CDCl3, 90 MHz) δ152.7, 146.7, 146.6, 137.0, 132.7, 130.3, 129.7, 129.1, 129.0, 122.0, 117.0, 109.9, 106.0, 62.8, 60.7, 55.7, 16.1.
  • 31P-NMR (CDCl3, 145 MHz) δ0.84.
  • HRMS (EI) M+calcd for C22H30N07P 451.1760, found 451.1765.
  • EXAMPLE I Synthesis of the Indole-Based Anti-Tubulin Agents
  • Preparation of 2-Phenyl Indole 31
  • Method I (2 steps);
  • To a well-stirred solution of KOH (0.926 g, 16.5 mmol) in EtOH (18 ml) and H2O (9 ml) at rt was added m-anisidine (2.192 g, 17.80 mmol) by syringe. The solution was then stirred at 0° C. After 10 min, the solution of 2-bromo-4-methoxyacetophenone (4.09 g, 17.80 mmol) was added dropwise with an addition funnel over a 40 minute period. After 24 h, 0° C. to rt, water was added. The product was isolated by extraction (I H HCI, NaHCO3, brine, MgSO4). The product was purified by recrystallization (50:50 EtOAc:hexanes) to afford secondary amine 30 (2.46 g, 9.07 mmol, 52%) as yellow solid.
  • 'H NMR (CDCI3): δ7.98 (2H, D, J=8.9 Hz), 7.12 (IH, t, J 8.1 Hz), 6.97 (2H, d, J 8.9 Hz), 6.30 (3H, m), 4.54 (2H, s), 3.88 (3H, s), 3.79 (3H, s).
  • Polyphosphoric acid (PPA) was charged to around-bottom flask and the temperature was raised to 80° C. with vigirous stirring. To this flask was added the foregoing amine 30 (4.0 g, 14.74 mmol) in 6 portions over a 30 minute period. After 2 h, 80° C. to 90° C., water was added. The product was isolated by extraction ( EtOAc, NaHCO3, brine, MgSO4). Purification by recrystallization (acetone) afforded indole 31(0.544 g, 2.15 mmol, 15%) as a pale yellow solid.
  • 'H NMR (CDCl3): δ11.24 (IH, br, s), 7.72 (2H, d, J 8.82 Hz), 7.36 (IH, d, J=8.57 Hz), 7.00 (2H, d, J=8.84 Hz), 6.85 (IH, d, J=2.07 Hz), 6.66 (IH, d, J=1.66 Hz), 6.63 (IH, dd, J 8.59, 2.28 Hz), 3.78 (3H, s), 3.77 (3H, s).
  • 13C NMR (CDCl3): δ158.15, 155.22, 137.44, 136.33, 125.60, 124.93, 122.82, 120.04, 114.07, 109.00, 96.97, 94.01, 54.93, 54.88.
  • Method 2(1 step):
  • To a boiling mixture of in-anisidine (1.56 ml, 20.0 minol) and N,N-dimethylaniline (3.5 ml) was added 2-bromo-4-methoxyacetophenone (1.37 g in EtOAc, 6.00 mmol) slowly by syringe. After addition, the mixture was kept at 170° C. for 1 hour. The reaction mixture was cooled to room temperature and a dark colored solid was formed. EtOAc was added along with HCI (2 N). The aqueous layer was extracted with EtOAc several times. The combined organic layers were washed with brine, and dried over MgSO4. Solvent was removed under the reduced pressure to afford a dark brown colored solid. Purification by recrystallization in EtOH afforded indole 31 as a white crystalline material.
  • 'H NMR(CDCl3): δ11.24 (IH,br,s),7.72(2H,d, J 8.82 Hz), 7.36 (IH,d, J 8.57 Hz),7.00 (2H,d, J=8.84 Hz), 6.85 (IH, d, J=2.07 Hz), 6.66 (IH, d, J=1.66 Hz), 6.63 (IH, dd, J8.59, 2.28 Hz),3.78 (3H, s), 3.77 (3H, s).
  • 13C NMR (CDCl3): δ158.15, 155.22, 137.44, 136.33, 125.60, 124.93, 122.82, 120.04, 114.07, 109.00, 96.97, 94.01, 54.93, 54.88.
  • Melting Point: 208-229.5° C.
  • HRMS (El) M+calcd for CH16N02 253.3035, found 253.1060.
  • Preparation of Trimethoxybenzoate 2-Phenylindole 33
  • To a well stirred solution of indole 31(0.502 g, 1.98 mmol) in o.dichlorobenzene (10 ml) was added trimethoxybenzoylchloride (0.692 g, 3.00 mmol). The reaction mixture was heated to reflux for 12 hours. Solvent was removed by distillation under reduced pressure. After cooling down to room temperature, a dark solid formed which was dissolved in chloroform and purified by silica gel column chromatography with chloroform as the eluent. The collected mixture was again purified by column chromatography (50:50 hexanes:EtOAc) affording trimethoxybenzyl indole 33 (0.744 g, 1.66 mmol, 84%) as a yellow oily gel. Pale yellow-green crystals were obtained by recrystallization from a mixture of ethanol and hexanes.
  • 'H NMR (CDCl3): δ8.63 (IH, br, s), 7.88 (IH, d, J=9.39 Hz), 7.24 (2H, d, J=8.78 Hz), 6.95(2H, s), 6.90 (2H, m), 6.71 (2H, d, J=8.79 Hz), 3.86 (3H, s), 3.80 (3H, s), 3.73 (3H, s), 3.68 (6H, s);
  • 13C NMR (CDCl3): δ192.23, 159.73, 157.06, 152.42, 142.85, 141.01, 136.41, 134.65, 130.16, 124.28, 122.94, 122.17, 113.67, 112.46, 111.52, 107.24, 94.54, 60.78, 55.92, 55.54, 55.14.
  • Melting Point: 153-155° C.
  • Anal. Calcd for C26H25N06: C, 69.79; H, 5.63; H, 3.13. Found: C, 69.61; H, 5.63; N, 3.01.
  • EXAMPLE 2 Inhibition of Tubulin Polymerization Assay
  • IC50 values for tubulin polymerization were determined according to the procedure described in Bai et al. Purified tubulin is obtained from bovine brain cells as described in Hamel and Lin. Various amounts of inhibitor were preincubated for 15 minutes at 37° C. with purified tubulin. After the incubation period, the reaction was cooled and GTP was added to induce tubulin polymerization. Polymerization was then monitored in a Gilford spectrophotometer at 350 nm. The final reaction mixtures (0.25 ml) contained 1.5 mg/ml tubulin, 0.6 mg/ml microtubule-associated proteins (MAPs), 0.5 mM GTP, 0.5 mlM MgCl2, 4% DMSO and 0.1M 4-morpholineethanesulfonate buffer (MES, pH 6.4). IC50 is the amount of inhibitor needed to inhibit tubulin polymerization 50% with respect to the amount of inhibition that occurs in the absence of inhibitor. The IC50 value determined for 3 -(3′,4′,5′-trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxyindole was 0.5-1.5 μM.
  • EXAMPLE 3 Cytotoxic Assay with P388 Leukemia Cells
  • One of the newly prepared compounds was evaluated for cytotoxic activity against P388 leukemia cells using an assay system similar to the National Cancer Institute procedure described below and in Monks et al. The ED50 value (defined as the effective dosage required to inhibit 50% of cell growth) of 3-(3′,4′,5′ trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxyindole was found to be 0.0133 μg/mL.
  • EXAMPLE 4 Growth Inhibitory Activity Against Other Cancer Cell Lines
  • 3-(3′,4′,5′-Trimethoxybenzoyl)-2-(4′-methoxyphenyl)-6-methoxyindole was evaluated in terms of growth inhibitory activity against several human cancer cell lines, including pancreas, ovarian, CNS, lung-NSC, colon, and prostate lines. The assay used is described in Monks et al. Briefly, the cell suspensions, diluted according to the particular cell type and the expected target cell density (5,000-40,000 cells per well based on cell growth characteristics), were added by pipet (100 μl) to 96-well microtiter plates. Inoculates were allowed a preincubation time of 24-28 hours at 37° C. for stabilization. Incubation with the inhibitor compounds lasted for 48 hours in 5% CO2 atmosphere and 100% humidity. Determination of cell growth was done by in situ fixation of cells, followed by staining with a protein-binding dye, sulforhodamine B (SRB), which binds to the basic amino acids of cellular macromolecules. The solubilized stain was measured spectrophotometrically. The results of these assays are shown in Table 1. Gl50 is defined as the dosage required to inhibit tumor cell growth by 50%.
    TABLE IV
    Activity of Indole Ligand Against Selected Human
    Cancer Cell lines (In Vitro).
    Indole-based Ligand 33
    CELL TYPE CELL LINE GI50 (μG/mL)
    Pancreas-a BXPC-3 2.0 × 10−3
    Ovarian OVCAR-3 2.4 × 10−3
    CNS SF-295 2.4 × 10−3
    Lung-NSC NCI-H460 2.6 × 10−3
    Colon KM20L2 1.7 × 10−3
    Prostate DU-145 2.3 × 10−3
  • Indole and indole containing compounds, of therapeutic efficacy have been known for many, many years. What is truly unique about the indole compounds described in this application is the fact that these compounds are the first (to the best of our knowledge) indole-based ligands to incorporate the 3,4,5-trimethoxyaryl motif reminiscent of colchicine and combretastatin A-4 arranged in an appropriate molecular conformation such that a pseudo aryl-aryl pi stacking interaction can take place. It is our contention that such an aryl-aryl interaction of the appropriate centroid-to-centroid distance (approximately 4.7Å) is imperative for enhanced binding affinity to the colchicine site on β-tubulin. It is this binding that ultimately leads to an inhibition of tubulin polymerization which manifests itself as a cytotoxic event. It should be readily apparent to any practitioner skilled in the art that there are various ways of appending trimethoxyaryl and trimethoxyaroyl groups around an indole molecular scaffold in a manner which will result in a similar molecular conformation capable of undergoing pseudo pi-pi stacking. In addition, although the trimethoxyaryl motif seems optimal for enhanced tubulin binding, it is also very possible that another combination of alkoxy substituents (such as ethoxy, propoxy, isopropoxy, allyloxy, etc.) either as a trisubstituted pattern or as disubstituted (with one type of alkoxy moiety) and monosubstituted (with a different alkoxy moiety), or with three distinct types of alkoxy moieties may also have good tubulin binding characteristics. It is also conceivable that instead of having aryl alkoxy groups, it may be possible to substitute simply aryl-alkyl and aryl-alkenyl moieties and still maintain the enhanced cytotoxicity profile. Phenolic groups may also have activity on these described indole ligands. The synthesis of any of these modified indole-ligands will be very straight-forward for anyone skilled in the art, and often will only involve a different choice of initial starting materials. To prepare these alternative ligands, the same synthetic schemes (FIGS. 6, 9, 11, 12-16), or similar schemes with only slight modifications may be employed. In previous studies with the benzo[b]thiophene ligands, we have demonstrated that the carbonyl group can be replaced with an oxygen to generate a new compound which maintains the same or similar biological efficacy with tubulin. Similarly, the replacement of the carbonyl group in the described indole ligand may be replaced with an oxygen atom (ether linkage) to generate a new derivative which would be predicted to have good activity with tubulin. This compound may be prepared by an addition elimination reaction utilizing the trimethoxyphenolic anion as a nucleophile as described by us for the benzo[b]thiophene compounds. Other linkage atoms between the aryl aryl rings are conceivable as well.
  • All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • Literature Cited
  • 1) (a) For a recent review of numerous antitubulin and antimitotic agents see: Hamel, E., Antimitotic Natural Products and Their Interactions with Tubulin, Medicinal Research Reviews, 1996, 16, 207.
  • (b) Gerwick, W. H.; Proteau, P. J.; Nagle, D. G.; Hamel, B.; Blokhin, A.; Slate, D. L., Structure of Curacin A, a Novel Antimitotic, Antiproliferative, and Brine Shrimp Toxic Natural Product from the Marine Cyanobacterium Lyngbya majuscula, J. Org. Chem. 1994, 59, 1243.
  • (c) Gianna Kakou, P.; Sackett, D.; Fojo, T.; Tubulin/Microtubes: Still a promising Target for New Chemotherapeutic Agents, J. Natl Cancer Inst., 2000, 92, 182.
  • 2) Owellen, R. J.; Hartke, C. A.; Kickerson, R. M.; Hams, F. 0., Inhibition of Tubulin-Microtubule Polymerization by Drugs of the Vinca Alkaloid Class, Cancer Res. 1976, 36, 1499.
  • 3) Lavielle, G.; Havtefaye, P.; Schaeffer, C.; Boutin, J. A.; Cudennec, C. A.; Pierre, A., New α-Amino Phosphonic Acid Derivatives of Vinblastine: Chemistry and Antitumor Activity, J Med Chem. 1991, 34, 1998.
  • 4) (a) Kingston, D. G. I.; Samaranayake, G.; Ivey, C. A., The Chemistry of Taxol, a Clinically Useful Anticancer Agent, J Nat. Prod. 1990, 53, 1.
  • (b) Schiff, P. B.; Fant, J.; Horwitz, S. B., Promotion of Microtubule Assembly In Vitro by Taxol, Nature, 1979, 277, 665.
  • (c) Swindell, C. S.; Krauss, N. B.; Horwitz, S. B.; Ringel, I., Biologically Active Taxol Analogs with Deleted A-ring Side Chain Substituents and Variable C-2
    Figure US20050065217A1-20050324-P00900
    Configurations, J Med Chem. 1991, 34, 1176.
  • (d) Pamess, J.; Horwitz, S. B., Taxol Binds to Polymerized Tubulin In Vitro, J. Cell Biol. 1981, 91, 479.
  • 5) (a) Nakada, M.; Kobayashi, S.; Iwasaki, S.; Ohno, M., The First Total Synthesis of the Antitumor Macrolide Rhizoxin: Synthesis of the Key Building Blocks, Tetrahedron Lett. 1993, 34, 1035.
  • (b) Nakada, M.; Kobayashi, S.; Iwasaki, S.; Ohno, M., The First Total Synthesis of the Antitumor Macrolide Rhizoxin,Tetrahedron Lett. 1993, 34, 1039.
  • (c) Boger, D. L.; Curran, T. T., Synthesis of the Lower Subunit of Rhizoxin, J. Org. Chem. 1992, 57, 2235.
  • (d) Rao, A. V. R.; Sharma, G. V. M.; Bhanu, M. N., Radical Mediated Enantioselective Construction of C-1 to C-9 Segment of Rhizoxin, Tetrahedron Lett. 1992, 33, 3907.
  • (e) Kobayashi, S.; Nakada, M.; Ohno, M., Synthetic Study on an Antitumor Antibiotic Rhizoxin by Using an Enzymatic Process on Prochiral betaSubstituted Glutarates, Pure AppL. Chem. 1992, 64, 1121.
  • (f) Kobayashi, S.; Nakada, M.; Ohno, M., Synthetic Study on an Antitumor Antibiotic Rhizoxin by Using an Enzymatic Process on Prochiral betaSubstituted Glutarates Indian J. Chem., Sect. B. 1993, 32B, 159.
  • (g) Rao, A. V. R.; Bhanu, M. N.; Sharma, G. V. M., Studies Directed Towards the Total Synthesis of Rhizoxin: Stereoselective Synthesis of C-12 to C-18 Segment, Tetrahedron Lett. 1993, 34, 707.
  • 6) (a) Lin, C. M.; Ho, H. H.; Pettit, G. R.; Hamel, E., Antimitotic Natural Products Combretastatin A-4 and Combretastatin A-2: Studies on the Mechanism of Their Inhibition of the Binding of Colchicine to Tubulin, Biochemistry 1989, 28, 6984.
  • (b) Pettit, G. R.; Cragg, G. M.; Singh, S. B., Antineoplastic agents, 122. Constituents of Combretum caffrum, J Nat. Prod 1987, 50, 386.
  • (c) Pettit, G. R.; Singh, S. B.; Cragg, G. M., Synthesis of Natural (-)Combretastatin, J. Org. Chem. 1985, 50, 3404.
  • (d) Pettit, G. R.; Cragg, G. M.; Herald, D. L.; Schmidt, J. M.; Lohavanijaya, P., Isolation and Structure of combretastatin, Can. J Chem. 1982, 60, 1374.
  • (e) Dorr, R. T.; Dvorakova, K.; Snead, K.; Alberts, D. S.; Salmon, S. E.; Pettit, G. R., Antitumor Activity of Combretastatin A4 Phosphate, a Natural Product Tubulin Inhibitor, Invest. New Drugs, 1996, 14, 131.
  • 7) (a) Harmrnonds, T. R.; Denyer, S. P.; Jackson, D. B.; Irving, W. L., Studies To Show That With Podophyllotoxin the Early Replicative Stages of Herpes Simplex Virus Type 1 Depend Upon Functional Cytoplasmic Microtubules, J Med. Microbiol., 1996, 45, 167.
  • (b) Cortese, F.; Bhattacharyya, B.; Wolff, J., Podophyllotoxin as a Probe for the Colchicine Binding Site of Tubulin, J. Biol. Chem., 1977, 252, 1134.
  • 8) Nicolaou, K. C., Winssinger, N., Pastor, J., Ninkovic, S., Sarabia, F., He, Y., Vourloumis, D., Yang, Z., Oi, T., Giannakakou, P., Hamel, B., Sythesis of Epothilones A and B in Solid and Solution Phase, Nature, 1997, 387, 268-272.
  • 9) (a) Pettit, G. R., Kamano, Y., Herald, C. L., Tuinrman, A. A., Boettner, F. E., Kizu, H., Schmidt, J. M., Baczynskyj, L., Tomer, K. B., Bontems, R. J., The Isolation and Structure of a Remarkable Marine Animal Antineoplastic Contituent: Dolastatin 10, J Am. Chem. Soc., 1987,109, 6883-6885.
  • (b) Pettit, G. R., Srirangam, J. K., Barkoczy, J., Williams, M. D., Boyd, M. R., Hamel, E., Pettit, R. K., Hogan F., Bai, R., Chapuis, J. C., McAllister, S. C., Schmidt, J. M., Antineoplastic Agents 365: Dolastatin 10 SAR Probes, Anti-Cancer Drug Des., 1998, 13, 243-277.
  • 10) Zhang, X.; Smith, C. D., Microtubule Effects of Welwistatin, a Cyanobacterial Indolinone that Circumvents Multiple Drug Resistance, Molecular Pharmacology, 1996, 49,288.
  • 11) Pettit, G. R., Told, B., Herald, D. L., Verdier-Pinard, P., Boyd, M. R., Hamel, E., Pettit, R. K., Antineoplastic Agents 379. Synthesis of Phenstatin Phosphate, J Med Chem., 1998, 41, 1688-1695.
  • 12) Jiang, J. B.; Hesson, D. P.; Dusak, B. A.; Dexter, D. L.; Kang, G. J.; Hamel, B., Synthesis and Biological Evaluation of 2-Styrylquinazolin4(3H)-ones, a New Class of Antimitotic Anticancer Agents Which Inhibit Tubulin Polymerization, J Med. Chem. 1990, 33, 1721.
  • 13) Cushman, M.; Nagarathnam, D.; Gopal, D.; Chakraborti, A. K.; Lin, C. M.; Hamel, E. Synthesis and Evaluation of Stilbene and Dihydrostilbene Derivatives as Potential Anticancer Agents That Inhibit Tubulin Polymerization, J Med Chem. 1991, 34, 2579.
  • 14) (a) Sawada, T.; Kato, Y.; Kobayashi, H.; Hashimoto, Y.; Watanabe, T.; Sugiyama, Y.; Iwasaki, S., A Fluorescent Probe and a Photoaffinity Labeling Reagent to Study the Binding Site of Maytansine and Rhizoxin on Tubulin, Bioconjugate Chem., 1993, 4, 284.
  • (b) Rao, S.; Horwitz, S. B.; Ringel, I., Direct Photoaffinity Labeling of Tubulin with Taxol, J Natl Cancer Inst., 1992, 84, 785.
  • (c) Chavan, A. J.; Richardson, S. K.; Kim, H.; Haley, B. E.; Watt, D. S., Forskolin Photoaffinity Probes for the Evaluation of Tubulin Binding Sites, Bioconjugate Chem. 1993, 4, 268.
  • (d) Sawada, T.; Kobayashi, H.; Hashimoto, Y.; Iwasaki, S., Identification of the Fragment Photoaffinity-labeled with Azidodansyl-rhizoxin as Met-363-Lys-379 on beta-Tubulin, Biochem. Pharmacol. 1993, 45, 1387.
  • (e) Staretz, M. E.; Hastie, S. B., Synthesis, Photochemical Reactions, and Tubulin Binding of Novel Photoaffinity Labeling Derivatives of Coichicine, J Org. Chem. 1993, 58, 1589.
  • (f) Hahn, K. M.; Hastie, S. B.; Sundberg, R. J., Synthesis and Evaluation of 2-Diazo-3 ,3 ,3 -trifluoropropanoyl Derivatives of Colchicine and Podophyllotoxin as Photoaffinity Labels: Reactivity, Photochemistry, and Tubulin Binding, Photochem. Photobiol. 1992, 55, 17.
  • (g) Sawada, T.; Hashimoto, Y.; Li, Y.; Kobayashi, H.; Iwasaki, S., Fluorescent and Photoaffinity Labeling Derivatives of Rhizoxin, Biochem. Biophys. Res. Commun. 1991, 178, 558.
  • (h) Wolff, J.; j(nipling. L.; Cahnmann, H. J.; Palumbo, G., Direct Photoaffinity Labeling of Tubulin with Colchicine, Proc. Natl. Acad Sci. US.A. 1991, 88, 2820.
  • (i) Floyd. L. J.; Baines, L. D.; Williams, R. F., Photoaffinity Labeling of Tubulin with (2-Nitro-4-azidophenyl)deacetylcolchicine: Direct Evidence for Two Colchicine Binding Sites, Biochemistry, 1989, 28, 8515.
  • (j) Safa, A. R.; Hamel, E.; Felsted, R. L., Photoaffinity Labeling of Tubulin Subunits with a Photoactive Analog of Vinblastine, Biochemistry 1987, 26, 97.
  • (k) Williams, R. F.; Mumford, C. L.; Williams, G. A.; Floyd, L. J.; Aivaliotis, M. J.; Martinez, R. A.; Robinson, A. K.; Bames, L. D., A Photoaffinity Derivative of Colchicine: 6-(4′-Azido-2′-nitrophenylamnino)hexanoyldeacetylcolchicine. Photolabeling and Location of the Colchicine-binding Site on the alpha-subunit of Tubulin, J Biol. Chem. 1985, 260, 13794.
  • 15) Nogales, E., Wolf, S. G., and Downing, K. H., Structure of the ct,˜Tubulin Dimer by Electron Crystallography, Nature, 1998, 391, 199-203.
  • 16) Shirai, R.; Tokjida, K.; Koiso, Y.; Iwasaki, S., Synthesis and AntiTubulin Activity of Aza-Combretastatins, Biomedical Chem. Lett. 1994, 699.
  • 17) (a) Jones, C. K.; Jevnikar, M. G.; Pike, A. J.; Peters, M. K.; Black, L. J.; Thompson, A. R.; Falcone, J. F.; Clemens, J. A., Antiestrogens. 2. Structure-Activity Studies in a Series of 3-Aroyl-2-arylbenzo[b]thiophene Derivatives Leading to [6- Hydroxy-2-(4-hydroxyphenyl)benzo[b]thien-3-yl][4-[2-(1-piperidinyl) ethoxy] phenylimethanone Hydrochioride (LY156758), a Remarkably Effective Estrogen Antagonist with Only Minimal Intrinsic Estrogenicity, J. Med Chem. 1984, 27, 1057.
  • (b) Grese, T. A.; Cho, S.; Finley, D. R.; Godfrey, A. G.; Jones, C. D.; Lugar III, C. W.; Martin, M. J.; Matsumoto, K.; Pennington, L. D.; Winter, M. A.; Adrian, M. D.; Cole, H. W.; Magee, D. E.; Phillips, D. L.; Rowley, E. R.; Short, L.; Glasebrook, A. L.; Bryant, H. R., Structure-Activity Relationships of Selective Estrogen Receptor Modulators: Modifications to the 2-Arylbenzothiophene Core of Raloxifene, J Med Chem., 1997, 40, 146.
  • (c) Palkowitz, A. D.; Glasebrook, A. L.; Thrasher, K. J.; Hauser, K. L.; Short, L. L.; Phillips, D. L.; Muehl, B. S.; Sato, M.; Shetler, P. K.; Cullinan, G. J.; Pell, T. R.; Bryant, H. U., Discovery and Synthesis of [6-Hydroxy-3-[4-[2-(1- piperidinyl)ethoxy]phenoxy]-2-(4-hydroxyphenyl)]benzo[b]thiophene: A Novel, Highly Potent, Selective Estrogen Receptor Modulator, J. Med Chem., 1997, 40, 1407.
  • 18) Pinney, K. G., Anti-Mitotic Agents Which Inhibit Tubulin Polymerization, Baylor University, Application for United States Letters Patent, Filed, Mar. 6, 1997. Pat. No.: 5,886,025. Issued, Mar. 23, 1999.
  • 19) Pinney, K. G.; Mejia, P.; Mocharla, V. P.; Shirlai, A.; Pettit, G. R., Anti-Mitotic Agents Which Inhibit Tubulin Polymerization, PCT Application Pending, filed under the Patent Cooperation Treaty on Mar. 6, 1998 and designating all PCT member states. Filed jointly by Baylor University, Arizona Disease Control Research Commission, and Arizona State University.
  • 20) Mullica, D. F.; Pinney, K. G.; Mocharla, V. P.; Dingeman, K. M.; Bounds, A. D.; Sappenfield, E. L., Characterization and Structural Analyses of Trimethoxy and Triethoxybenzo[b]thiophene, J. Chem. Cryst., 1998, 28, 289-295.
  • 21) Pmnney, K. G.; Dingeman, K. D.; Bounds, A. D.; Mocharla, V. P.; Pettit, G. R.; Bai, R.; Hamel, E., A New Anti-Tubulin Agent Containing the Benzo[b]thiophene Ring System, Bioorganic and Medicinal Chemistry Letters, 1999, 9, 1081-1086.
  • 22) (a) D'Amato, R. J.; Lin, C. M.; Flynn, E.; Folkman, J.; Hamel, E., 2Methoxyestradiol, an endogenous mamalian metabolite, inhibits tubulin polymerization by interacting at the colchicine site, Proc. Natl. Acad Sci. 1994, 91, 3964.
  • (b) Cushman, M.; He, H-M.; Katzenellenbogen, J. A.; Lin, C. M.; Hamel, E., Synthesis, Antitubulin and Antimitotic Activity, and Cytotoxicity of Analogs of 2-Methoxyestradiol, an Endogenous Mammalian Metabolite of Estradiol That Inhibits Tubulin Polymerization by Binding to the Colchicine Binding Site, J. Med Chem., 1995, 38, 2041.
  • (c) Hamel, E.; Lin, C. M.; Flynn, E.; D
    Figure US20050065217A1-20050324-P00900
    Amato, R. J. D., Interactions of 2-Methoxyestradiol, and Endogenous Mammalian Metabolite, with Unpolymerized Tubulin and with Tubulin Polymers, Biochemistry, 1996, 35, 1304.
  • (d) Cushman, M.; He, H.-M.; Katzenellenbogen, J. A.; Varma, R. K.; Hamel, E.; Lin, C. M.; Ram, S.; Sachdeva, Y. P., Synthesis of Analogs of 2-Methoxyestradiol with Enhanced Inhibitory Effects on Tubulin Polymerization and Cancer Cell Growth, J. Med. Chem., 1997, (in press).
  • 23) Boyd, M. R.; Paull, K. D., Some Practical Considerations and Applications of the National Cancer Institute In Vitro Anticancer Drug Discovery Screen, Drug Development Research, 1995, 34,91.
  • 24) Dark, G. G., Hill, S. A., Prise, V. G., Tozer, G. M., Pettit, G. R., Chaplin, D. J., Combretastatin A-4, an Agent That Displays Potent and Selective Toxicity Toward Tumor Vasculature, Cancer Res., 1997, 57, 1829-1834.
  • 25) Angerer, E.; Prekajac, J.; Strohmeier, J.; J. Med Chem. 1984, 27,1439-1447.
  • 26) Inion, H.; Vogelaer, H.; Bauthier, J.; Colot, M.; Richard, J.; Eur. J. Med. Chem. 1977, 5, 483-487.
  • 27) Gastpar, R.; Goldbrunner, M.; Marko, D.; von Angerer, E., MethoxySubstituted 3-Formyl-2-phenylindoles Inhibit Tubulin Polymerization, J. Med Chem. 1998, 41, 4965-4972.
  • 28) Taylor, S. D.; Chen, M,J,; Dinaut, A. N.; and Batey, R. A., Tetrahedron 1998, 54, 4223-4242.
  • 29) Fischer,B.; and Sheihet, L., J. Org. Chem. 1998, 63, 393-395.
  • 30) Cell line studies carried out through collaborative efforts with Prof. George R. Pettit; Arizona State University; Tempe; Arizona.
  • 31) Pettit, G. R.; Toki, B.; Herald, D. L.; Verdier-Pinard, P.; Boyd, M. R.;Hamel, E.; and Pettit, R. K., J. Med Chem. 1998, 41, 1688-1695.
  • 32) Tubulin polymerization studies carried out through collaborative efforts with Dr Ernest Hamel, National Institute of Health, National Cancer Institute.
  • 33) Pettit, G. R.; and Rhodes, M. R., Anti-Cancer Drug Des. 1998, 13, 183-191.
  • 34) Dark, G. G.; Hill, S. A.; Prise, V. E.; Tozer, G. M.; Pettit, G. R.; and Chaplin, D. J., Cancer Res. 1997, 57; 1829-1834.
  • 35) Iyer, S.; Chaplin, D. J.; Rosenthal, D. S.; Boulares, A. H.; Li, L.Y.; and Smulson, M. E., Cancer Res. 1998, 58, 4510-4514.
  • 36) Pinney et al., Tubulin Binding Ligands and Corresponding Prodrug Constructs, Provisional Patent Application, U.S. Ser. No. 60/188,295, filed on Mar. 10, 2000.
  • 37) Pinney et al., Synthesis of a Benzo[b]thiophene-based Vascular Targeting Prodrug and Related Anti-Tubulin Ligands, 220th American Chemical Society, National Meeting, Division of Organic Chemistry, Abstract No. 196, Washington, D.C., Aug. 20-24, 2000.

Claims (47)

1. A compound of the structure:
Figure US20050065217A1-20050324-C00001
wherein
R1 through R5 contain at least one phenolic moiety or at least one amine group (NH2, NHR1, or NR6R7 where R6 and R7 are the same or different alkyl having up to 8 carbon atoms), benzyl, or aryl while the remaining R1 through R5 are hydrogen.
2. A compound of the structure:
Figure US20050065217A1-20050324-C00002
wherein
R1 through R5 contain at least one phosphate ester moiety (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different, benzyl, or aryl while the remaining R1 through R5 are hydrogen.
3. A compound of the structure:
Figure US20050065217A1-20050324-C00003
wherein
R1 through R5 contain at least one phosphate ester moiety (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl while the remaining R1 through R5 are hydrogen, and R6 is hydrogen or alkyl.
4. A compound of the structure:
Figure US20050065217A1-20050324-C00004
wherein
R1 through R5 contain at least one phenolic moiety or at least one amine (NH2, NHR1, or NR6R7 where R6 and R7 the same or different alkyl having up to 8 carbon atoms, benzyl, or aryl groups) while the remaining R1 through R5 are a hydrogen.
5. A compound of the structure:
Figure US20050065217A1-20050324-C00005
wherein
R1 through R1 contain at least one phosphate ester moiety (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl while the remaining R1 through R5 are hydrogen.
6. A compound of the structure:
Figure US20050065217A1-20050324-C00006
wherein
R1 through R5 contain at least one phosphate ester moiety (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M=a cation or (-NP(O)(OR)2 ) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), or benzyl, or aryl groups, while the remaining R1 through R5 are a hydrogen and R6 is hydrogen or alkyl.
7. A compound of the structure:
Figure US20050065217A1-20050324-C00007
wherein
R1 through R5 contain at least one phenolic moiety or at least one amine group (NH2, NHR or NR6R7 where R6 and R7 are the same or different alkyl having up to 8 carbon atoms may be the same or different), or benzyl, or aryl groups) while the remaining R1 through R5 are a hydrogen.
8. A compound of the structure:
Figure US20050065217A1-20050324-C00008
wherein
R1 through R5 contain at least one phosphate ester (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+6l ) 2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl while the remaining R1 through R5 are hydrogen.
9. A compound of the structure:
Figure US20050065217A1-20050324-C00009
wherein
R1 through R5 contain at least one phosphate ester (-OP(O)(OM+)2) or phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl, while the remaining R1 through R5 are hydrogen, and R6 is hydrogen or alkyl.
10. A compound of the structure:
Figure US20050065217A1-20050324-C00010
wherein
R1 through R5 contain at least one phenolic moiety or at least one amine group (NH2, NHR1, or NR6R7 where R6 and R7 are the same or different alkyl having upt to 8 carbon atoms, benzyl, or aryl) while the remaining R1 through R5 are a hydrogen.
11. A compound of the structure:
Figure US20050065217A1-20050324-C00011
wherein
R1 through R5 contain at least one phosphate ester (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl, while the remaining R1 through R5 are hydrogen.
12. A compound of the structure:
Figure US20050065217A1-20050324-C00012
wherein
R1 through R1 contain at least one phosphate ester moiety (-OP(O)(OM+)2) or a phosphoramidate (-NP(O)(OM+)2) where M is a cation or (-NP(O)(OR)2) where R is an alkyl with up to 8 carbon atoms (the two R groups are the same or different), benzyl, or aryl while the remaining R1 through R5 are hydrogen, and R6 is hydrogen or alkyl.
13. A compound of the structure:
Figure US20050065217A1-20050324-C00013
14. A compound of the structure:
Figure US20050065217A1-20050324-C00014
15. A compound of the structure:
Figure US20050065217A1-20050324-C00015
16. A compound of the structure:
Figure US20050065217A1-20050324-C00016
17. A compound of the structure:
Figure US20050065217A1-20050324-C00017
18. A compound of the structure:
Figure US20050065217A1-20050324-C00018
19. A compound of the structure:
Figure US20050065217A1-20050324-C00019
20. A compound of the structure:
Figure US20050065217A1-20050324-C00020
21. A compound of the structure:
Figure US20050065217A1-20050324-C00021
22. A compound of the structure:
Figure US20050065217A1-20050324-C00022
23. A compound of the structure:
Figure US20050065217A1-20050324-C00023
24. A compound of the structure:
Figure US20050065217A1-20050324-C00024
25. A compound of the structure:
Figure US20050065217A1-20050324-C00025
26. A compound of the structure:
Figure US20050065217A1-20050324-C00026
27. A compound of the structure:
Figure US20050065217A1-20050324-C00027
28. A compound of the structure:
Figure US20050065217A1-20050324-C00028
29. A compound of the structure:
Figure US20050065217A1-20050324-C00029
30. A compound of the structure:
Figure US20050065217A1-20050324-C00030
31. A compound of the structure:
Figure US20050065217A1-20050324-C00031
32. A compound of the structure:
Figure US20050065217A1-20050324-C00032
33. A compound of the structure:
Figure US20050065217A1-20050324-C00033
34. A compound of the structure:
Figure US20050065217A1-20050324-C00034
35. A compound of the structure:
Figure US20050065217A1-20050324-C00035
36. A compound of the structure:
Figure US20050065217A1-20050324-C00036
37. A compound of the structure:
Figure US20050065217A1-20050324-C00037
38. A compound of the structure:
Figure US20050065217A1-20050324-C00038
wherein
R is chosen to be any appropriate alkyl or branched alkyl having up to 8 carbon atoms, the two R groups may be the same or different.
39. A compound of the structure:
Figure US20050065217A1-20050324-C00039
40. A compound of the structure:
Figure US20050065217A1-20050324-C00040
wherein
M+is a cation.
41. A method for inhibitiing tubulin polymerization by contacting a tubulin-containing system with an effective amount of a compound described in any of claims 1-40.
42. The method of claim 41 wherein said system is in a tumor cell.
43. A method of treating a host afflicted with a neoplastic disease by administering to said host a compound described in any of claims 1-40.
44. The method of claims 41, wherein the contacted system is located in a patient.
45. The method of claim 41 described further as for treating cancer, wherein said cancer may be chosen from the group containing leukemia, lung, colon, thyroid, CNS, melanoma, ovarian, renal, prostate, and breast cancers.
46. A preparation for pharmaceutical use containing a compound from any of claims 1-40 as an active component along with a pharmaceutically acceptable carrier.
47. A method for selectively targeting and destroying tumor vasculature comprising administering an effective amount of a compound described in any of claims 1-40.
US10/861,186 1999-09-17 2004-06-04 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents Abandoned US20050065217A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/861,186 US20050065217A1 (en) 1999-09-17 2004-06-04 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15463999P 1999-09-17 1999-09-17
US10/070,484 US6849656B1 (en) 1999-09-17 2000-09-15 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents
US10/861,186 US20050065217A1 (en) 1999-09-17 2004-06-04 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2000/025408 Continuation WO2001019794A2 (en) 1999-09-17 2000-09-15 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents
US10/070,484 Continuation US6849656B1 (en) 1999-09-17 2000-09-15 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents

Publications (1)

Publication Number Publication Date
US20050065217A1 true US20050065217A1 (en) 2005-03-24

Family

ID=34082548

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/070,484 Expired - Fee Related US6849656B1 (en) 1999-09-17 2000-09-15 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents
US10/861,186 Abandoned US20050065217A1 (en) 1999-09-17 2004-06-04 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/070,484 Expired - Fee Related US6849656B1 (en) 1999-09-17 2000-09-15 Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents

Country Status (1)

Country Link
US (2) US6849656B1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4187441B2 (en) * 1999-09-17 2008-11-26 ベイラー・ユニバーシテイ Indole-containing and combretastatin-related antimitotic and antitubulin polymerization drugs
EP1919882A2 (en) * 2005-02-17 2008-05-14 Synta Pharmaceuticals Corporation Isoxazole combretastatin derivatives for the treatment of proliferative disorders
ES2529434T3 (en) 2007-11-21 2015-02-20 Oxigene, Inc. Method to treat hematopoietic neoplasms
NZ598489A (en) 2009-08-27 2014-03-28 Bionomics Ltd Combination therapy for treating proliferative diseases
AU2013204313C1 (en) 2012-06-01 2016-04-07 Bionomics Limited Combination Therapy
US11419934B2 (en) 2015-08-18 2022-08-23 Oncotelic Therapeutics, Inc. Use of VDAS to enhance immunomodulating therapies against tumors

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4133814A (en) * 1975-10-28 1979-01-09 Eli Lilly And Company 2-Phenyl-3-aroylbenzothiophenes useful as antifertility agents
US4656187A (en) * 1981-08-03 1987-04-07 Eli Lilly And Company Treatment of mammary cancer
US5342547A (en) * 1992-01-14 1994-08-30 Marine Biotechnology Institute Co., Ltd. Agents for controlling underwater fouling organisms
US5514703A (en) * 1995-03-13 1996-05-07 Eli Lilly And Company Benzothiophene compounds useful for inhibiting lipoxygenase
US5514704A (en) * 1995-03-13 1996-05-07 Eli Lilly And Company Benzothiophenes to inhibit leukotrienes
US5525632A (en) * 1993-09-08 1996-06-11 Ajinomoto Co., Ltd. Stilbene derivatives and pharmaceutical compositions containing them
US5532382A (en) * 1995-03-13 1996-07-02 Eli Lilly And Company Benzothiophenes substituted at the 3-carbonyl
US5596106A (en) * 1994-07-15 1997-01-21 Eli Lilly And Company Cannabinoid receptor antagonists
US5674906A (en) * 1995-03-07 1997-10-07 Ajinomoto Co., Inc. Stilbene compounds and pharmaceutical compositions containing them
US5731353A (en) * 1993-09-08 1998-03-24 Ajinomoto Co., Inc. Stilbene derivatives and pharmaceutical compositions containing them
US5886025A (en) * 1997-03-06 1999-03-23 Baylor University Anti-mitotic agents which inhibit tubulin polymerization
US5958916A (en) * 1996-08-29 1999-09-28 Eli Lilly And Company Dihydronaphthalene and naphthalene compounds, intermediates, formulations, and methods
US6110963A (en) * 1998-05-12 2000-08-29 American Home Products Corporation Aryl-oxo-acetic acids useful in the treatment of insulin resistance and hyperglycemia
US6162930A (en) * 1998-03-06 2000-12-19 Baylor University Anti-mitotic agents which inhibit tubulin polymerization
US6166069A (en) * 1998-05-12 2000-12-26 American Home Products Corporation Phenyl oxo-acetic acids useful in the treatment of insulin resistance and hyperglycemia
US6232327B1 (en) * 1998-04-02 2001-05-15 Asta Medica Aktiengesellschaft Indolyl-3-glyoxylic acid derivatives having antitumor action
US6538038B1 (en) * 1999-02-18 2003-03-25 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6756388B1 (en) 1993-10-12 2004-06-29 Pfizer Inc. Benzothiophenes and related compounds as estrogen agonists
NZ501822A (en) 1997-10-02 2001-12-21 Yukijirushi Nyugyo Kabushiki K Therapeutic agents for male and female sex hormone dependent diseases
WO2000006556A1 (en) 1998-07-27 2000-02-10 Abbott Laboratories Substituted oxazolines as antiproliferative agents
CO5170498A1 (en) 1999-05-28 2002-06-27 Abbott Lab BIARIL SULFONAMIDS ARE USEFUL AS CELL PROLIFERATION INHIBITORS
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
US20020091148A1 (en) 2000-09-15 2002-07-11 Bamaung Nwe Y. 3-substituted indole antiproliferative angiogenesis inhibitors
US6323228B1 (en) 2000-09-15 2001-11-27 Abbott Laboratories 3-substituted indole angiogenesis inhibitors
IT1317926B1 (en) 2000-11-03 2003-07-15 Sigma Tau Ind Farmaceuti TRICYCLIC DERIVATIVES OF INDOLE FOR ANTIANGIOGENIC ACTIVITY.
AUPR283801A0 (en) 2001-02-01 2001-03-01 Australian National University, The Chemical compounds and methods

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4133814A (en) * 1975-10-28 1979-01-09 Eli Lilly And Company 2-Phenyl-3-aroylbenzothiophenes useful as antifertility agents
US4656187A (en) * 1981-08-03 1987-04-07 Eli Lilly And Company Treatment of mammary cancer
US5342547A (en) * 1992-01-14 1994-08-30 Marine Biotechnology Institute Co., Ltd. Agents for controlling underwater fouling organisms
US5731353A (en) * 1993-09-08 1998-03-24 Ajinomoto Co., Inc. Stilbene derivatives and pharmaceutical compositions containing them
US5525632A (en) * 1993-09-08 1996-06-11 Ajinomoto Co., Ltd. Stilbene derivatives and pharmaceutical compositions containing them
US5596106A (en) * 1994-07-15 1997-01-21 Eli Lilly And Company Cannabinoid receptor antagonists
US5674906A (en) * 1995-03-07 1997-10-07 Ajinomoto Co., Inc. Stilbene compounds and pharmaceutical compositions containing them
US5514704A (en) * 1995-03-13 1996-05-07 Eli Lilly And Company Benzothiophenes to inhibit leukotrienes
US5532382A (en) * 1995-03-13 1996-07-02 Eli Lilly And Company Benzothiophenes substituted at the 3-carbonyl
US5514703A (en) * 1995-03-13 1996-05-07 Eli Lilly And Company Benzothiophene compounds useful for inhibiting lipoxygenase
US5958916A (en) * 1996-08-29 1999-09-28 Eli Lilly And Company Dihydronaphthalene and naphthalene compounds, intermediates, formulations, and methods
US5886025A (en) * 1997-03-06 1999-03-23 Baylor University Anti-mitotic agents which inhibit tubulin polymerization
US6162930A (en) * 1998-03-06 2000-12-19 Baylor University Anti-mitotic agents which inhibit tubulin polymerization
US6232327B1 (en) * 1998-04-02 2001-05-15 Asta Medica Aktiengesellschaft Indolyl-3-glyoxylic acid derivatives having antitumor action
US6110963A (en) * 1998-05-12 2000-08-29 American Home Products Corporation Aryl-oxo-acetic acids useful in the treatment of insulin resistance and hyperglycemia
US6166069A (en) * 1998-05-12 2000-12-26 American Home Products Corporation Phenyl oxo-acetic acids useful in the treatment of insulin resistance and hyperglycemia
US6538038B1 (en) * 1999-02-18 2003-03-25 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction
US6350777B2 (en) * 1999-12-03 2002-02-26 Baylor University Description anti-mitotic agents which inhibit tubulin polymerization

Also Published As

Publication number Publication date
US6849656B1 (en) 2005-02-01

Similar Documents

Publication Publication Date Title
AU777725B2 (en) Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents
US6593374B2 (en) Tubulin binding ligands and corresponding prodrug constructs
AU2001243527A1 (en) Tubulin binding ligands and corresponding prodrug constructs
EP1751128B1 (en) Chromene-derivatives with anti-tubulin and vascular targeting activity
Wu et al. Synthesis and activity of combretastatin A-4 analogues: 1, 2, 3-thiadiazoles as potent antitumor agents
JP5315060B2 (en) Substituted benzofurans, benzothiophenes, benzoselenophenes and indoles and their use as tubulin polymerization inhibitors
ES2551086T3 (en) Combretastatin analogues with tubulin binding activity
US20060142252A1 (en) Tubulin binding agents and corresponding prodrug constructs
US6849656B1 (en) Indole-containing and combretastatin-related anti-mitotic and anti-tubulin polymerization agents
Liou et al. Structure–Activity Relationship Studies of 3‐Aroylindoles as Potent Antimitotic Agents
US7091240B2 (en) Tubulin binding ligands and corresponding prodrug constructs
Siddiqui et al. Discovery of 3-(benzofuran-2-ylmethyl)-1H-indole derivatives as potential autophagy inducers in cervical cancer cells
ES2349746T3 (en) COMPOUNDS DERIVED FROM THE CHROMENE WITH ANTI-TIBULIN AND VASCULAR DIANA ACTIVITY.
WO2004099139A1 (en) Indole-containing compounds with anti-tubulin and vascular targeting activity
Liang et al. Structure-activity relationships and antiproliferative effects of 1, 2, 3, 4-4H-quinoxaline derivatives as tubulin polymerization inhibitors
HU202510B (en) Process for producing biocide hetero-polycyclic compounds and pharmaceutical compositions containing them as active components
Paegle et al. Selenium analogues of rintodestrant (G1T48) as potent estrogen receptor modulators and downregulators
Kessler Synthesis and evaluation of new inhibitors of tubulin polymerization and their corresponding prodrugs as potential vascular targeting agents
AU2003270999A1 (en) Anti-mitotic agents which inhibit tubulin polymerization

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION