CA2155329C - Acylfulvene analogues as antitumor agents - Google Patents

Acylfulvene analogues as antitumor agents Download PDF

Info

Publication number
CA2155329C
CA2155329C CA002155329A CA2155329A CA2155329C CA 2155329 C CA2155329 C CA 2155329C CA 002155329 A CA002155329 A CA 002155329A CA 2155329 A CA2155329 A CA 2155329A CA 2155329 C CA2155329 C CA 2155329C
Authority
CA
Canada
Prior art keywords
illudin
tumor
compound
cells
alkyl
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.)
Expired - Fee Related
Application number
CA002155329A
Other languages
French (fr)
Other versions
CA2155329A1 (en
Inventor
Michael J. Kelner
Trevor C. Mcmorris
Raymond Taetle
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.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Publication of CA2155329A1 publication Critical patent/CA2155329A1/en
Application granted granted Critical
Publication of CA2155329C publication Critical patent/CA2155329C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/12Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/45Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by condensation
    • C07C45/455Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by condensation with carboxylic acids or their derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/45Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by condensation
    • C07C45/46Friedel-Crafts reactions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/65Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by splitting-off hydrogen atoms or functional groups; by hydrogenolysis of functional groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/673Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by change of size of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/657Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings
    • C07C49/665Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings a keto group being part of a condensed ring system
    • C07C49/67Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings a keto group being part of a condensed ring system having two rings, e.g. tetralones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/657Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings
    • C07C49/683Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings having unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/703Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups
    • C07C49/723Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic
    • C07C49/727Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic a keto group being part of a condensed ring system
    • C07C49/737Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic a keto group being part of a condensed ring system having three rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/703Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups
    • C07C49/747Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/753Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/753Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups
    • C07C49/755Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups a keto group being part of a condensed ring system with two or three rings, at least one ring being a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/007Esters of unsaturated alcohols having the esterified hydroxy group bound to an acyclic carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/93Spiro compounds
    • C07C2603/94Spiro compounds containing "free" spiro atoms

Abstract

A method of inhibiting tumor cell growth in a subject is provided comprising contacting the tumor with a therapeutic amount of an acylfulvene analog having structure (I), wherein die analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R is (a), where R1= alkyl, aryl, NH2, NHR, or NR2.

Description

ACYLFULUENE ANALOGUES AS ANTITUMOR AGENTS

BACKGROUND OF THE INVENTION
Multiple agent chemotherapy has curative potential in some hematologic malignancies and advanced rapidly proliferating solid tumors. Curative chemotherapy has benefitted from the discovery of new, relatively non-cross resistant agents, and more effective use of existing agents. Interventions which increase the efficacy of conventional agents include more effective regimens for multiple drug administration, minimization of drug toxicities and increased use of adjuvant, surgical or radiation therapy.
Despite recent advances, patients with many types of malignancies remain at significant risk for relapse and mortality. After relapse, some patients can be reinduced into remission with their initial treatment regimen.
However, higher doses of the initial chemotherapeutic agent or the use of additional agents are frequently required, indicating the development of at least partial drug resistance. Recent evidence indicates drug resistance can develop simultaneously to several agents, including ones to which the patient was not exposed. The development of multiple-drug resistant (mdr) tumors may be a function of tumor mass and constitutes a major cause of treatment failure. To overcome this drug resistance, high-dose chemotherapy with or without radiation and allogenic or autologous bone marrow transplantation is employed. The high-dose chemotherapy may employ the original drugs) or be altered to include additional agents. The feasibility of this approach has been demonstrated for hematopoietic and solid tumors. The development of new drugs non-cross resistant with mdr phenotypes is required to further the curative potential of current regimens and to facilitate curative interventions in previously treated patients.
Recently, the in vitro anti-tumor activity of a novel class of Natural products called illudins was examined in Kelner, M. et al., Cancer Res. 47:3186 (1987).
. Illudin S and M are two types of iliudins known to exist. Illudins have a chemical structure entirely different from other chemotherapeutic agents. Illudin compounds were previously purified and submitted for evaluation to the National Cancer Institute Division of Cancer Treatment (NCI DCT) in vivo drug screening program but had a low therapeutic index in other experimental tumor systems in accordance with NCI studies.
The extreme toxicity of illudins has prevented any applications in human tumor therapy.
Thus, there exists a need for chemotherapeutic agents which are toxic to tumors, and especially mdr tumors, and have an adequate therapeutic index to be effective for in vivo treatment. The subject invention satisfies this need and provides related advantages.
SUMMARY OF THE INVENTION
A method of inhibiting tumor cell growth in a subject is provided comprising contacting the tumor with a therapeutic amount of an illudin S or illudin M analog having the structure:

_ WO 94/18151 ~ PCT/US94101232 R~
F R:

O
or R, R, ' O
wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R1 is an alkyl or hydrogen;
R~ is an alkyl; and R, is an alcohol or ester.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows the sensitivity of breast carcinoma and myeloid leukemia cells versus other tumors to illudin S.
Figure 2 shows the active sites of illudin S.
' Figure 3 shows an NMR spectrum demonstrating the presence of a short acting intermediate in acid. Signal A
- is from the hydrogen on the double bond in the 5 membered ring ( illudin M) . Signal B is from the hydrogen atom on the short lived intermediate that results from the cyclopropane ring opening up (but before the double bond reacts). Signals marked at C are from the product that 2~~53~9 results when the double bond has reacted. With time, the signal peaks from illudin M will disappear and the peaks at position C will be the predominate signals. Signal H will disappear concurrently with Signal A confirming it is a short lived intermediate arising from illudin M.
Figure 4 shows the effect of illudin S on Molt-4 tumor growth in athymic mice (Balb/c).
Figure 5 shows the effect of dehydroilludin M on tumor growth.
Figure 6 shows the response of HL60/MRI xenograft to dehydroilludin M.
Figure 7 shows illudin S uptake using relatively sensitive HL60 cells and resistant H cells.
Figure 8 shows the rapid intracellular accumulation of illudin S by HL60 cells was saturated at high concentrations.
Figure 9 shows the analysis of the initial uptake of illudin S by HL60 cells at varying concentrations conformed to Michaelis-Menton saturation constants.
Figure 10 shows the effect of 6-hydroxymethylacylfulvene and other conventional anticancer agents on human lung adenocarcinoma MV522.
Figure 11 shows the effect of different doses of 6-hydroxymethylacylfulvene on human lung adenocarcinoma MV522.

DETAILED DESCRIPTION OF THE INVENTION
. A method of inhibiting tumor cell growth in a subject is provided comprising contacting the tumor with a 5 therapeutic amount of an illudin S or illudin M analog having the structure:
R~
1 R~
O
wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R1 is an alkyl or hydrogen;
RZ is an alkyl; and R, is an alcohol or ester.
The analog can be any compound having the stated structure. Examples of the effective analogs are:
HC
O
A method of inhibiting tumor cell growth in a subject is also provided comprising contacting the tumor with a therapeutic amount of an illudin S or illudin M
analog having the structure:
R_ O

~1553~~
wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R1 is an alkyl, alkoxyl or hydrogen;
Rz is an alkyl; and R, is an alcohol or ester.
The analog may be any compound having the stated structure. Two examples of effective analogs are:
O
HC H( O
HC
O
The analog may also be an acylfulvene analog having the structure:
H( O
wherein R is O
CH_,OH, CH,_OC CHI, CH= O H, O
n CH, O CH3, I , Br, CH=OCR (where R~ = alkyl, aryl, NH:, Nl-1R, or NR,).

2~.553~~

Hy "inhibiting" is meant either decreasing the tumor cell growth rate from the rate which would occur without treatment or causing the tumor cell mass to decrease in size. Inhibiting also includes a complete regression of the tumor. Thus, the analogs can either be cytostatic or cytotoxic to the tumor cells.
The subject can be any animal having a tumor.
The analogs are effective on human tumors in vivo as well as on human tumor cell lines in vitro.
The tumor can be contacted with the analog by any effective means, many of which are well known in the art.
The route of administration to the subject can include intravenous, oral, intraperitoneal, and oral and nasal inhalation. The preferred route of administration depends on the subject and type of tumor encountered.
Applicants have made the surprising discovery that analogs of illudin S and M can be made which are less toxic than illudin S and M but are a more effective chemotherapeutic agent in vivo. As noted above, illudin S
and M have a low therapeutic index due to the extreme toxicity and, therefore, cannot be used therapeutically in humans. Applicants have discovered that various modifications in illudin S and M inhibit nucleophiles from reacting with the compound. This results in less facile opening of the cyclopropane ring and reduces the toxicity of the compound in vivo while resulting in a high therapeutic index. The Applicants have made the further discovery that an acylfulvene analog, 6-hydroxymethylacylfulvene, is markedly more efficacious and nontoxic than previously known S and M illudin analogs.
The various R groups recited define areas which do not affect the vital functional groups of illudins or their analogs and, therefore, can be a wide variety of substituents. Thus applicants intend that the various R
groups recited be construed broadly, for example, alkyl includes any functional group attached to the alkyl group (i.e., an alkylfulvene or the functional group itself).
The R groups include nitro, halogen, alkyl halide, alcohol, peroxide, endoperoxide, primary, secondary or tertiary amine, ether, ester, sulfhydryl, thioalkyl, thioester, dithioester, disulfide, carboxylic acid, enamine, imine or oxime groups.
The therapeutically effective amount of analog varies with the subject. However, it has been found that relatively high doses of the analogs can be administered due to the decreased toxicity compared to illudin S and M.
A therapeutic amount between 30 to 112,000 Ng per kg of body weight has been found especially effective for intravenous administration while 300 to 112,000 Ng per kg of body weight is effective if administered intraperitoneally. As one skilled in the art would recognize, the amount can vary depending on the method of administration. Further, the amount can vary if the analog is linked to a toxin.
The analogs can be attached to a reagent to form a complex which binds to a tumor-associated antigen. Such methods are well known in the art and can include a linker which serves to connect the reagent to the analog. Such attachment can include any chemical bond, as for example a covalent bond. The reagent can be any reagent which specifically binds to a tumor-associated antigen on the tumor cell or in the tumor cell area. Typically such reagent is an antibody, either polyclonal or monoclonal.
These complexes can then be used in therapy. The methods of the invention can be practiced on any tumor cells but are especially effective against tumor cells of myeloid, epidermoid, T-cell leukemia, and lung, ovarian and breast carcinoma.

21~~3~9 Also disclosed is a compound having the structure:
R~
w wherein R = methyl; and R1, Rz and R, = methyl or alkyl.
Also disclosed is a compound having the structure:

RO
i R = H or methyl; and R1 RZ and R, = methyl or alkyl.
EXAMPLE I
Synthesis of Deh~droilludin M
A mixture of illudin M (200 mg) and pyridinium dichromate (1 g) in dry dichloromethane (60 ml) was stirred at room temperature in a flask equipped with a rubber septum so that an atmosphere of argon could be maintained.
After 20 hours, the reaction mixture was diluted with diethyl ether (20 ml) and filtered through a short column of silica gel. The column was further eluted with more diethyl ether and the combined filtrate was concentrated, giving a residue which was chromatographed on silica gel with hexane-ethyl acetate (10:1) as eluent. The desired compound was obtained in early fractions from the chromatography. The yield was 140 mg of white crystals melting at 64-65°C. NMR spectral data were recorded for this compound.

215~3~~
EXAMPLE II
Synthesis of Fulvene Illudin S (50 mg) was dissolved in water (2 mL) 5 and 3N hydrochloric acid (2 mL) added to the solution. The resulting solution soon became cloudy (within 30 min) and a yellow precipitate formed. The mixture was placed in the refrigerator overnight; then it was extracted with chloroform (10 m1,). The yellow chloroform solution was 10 dried (MgSO,) and the solvent was removed under reduced pressure leaving an orange-yellow gum. This material was chromatographed on silica gel with hexanes . ethyl acetate (6 :1) as eluent giving the fulvene (20 mg) and the bisfulvene (10 mg). NMR spectral data were recorded for these compounds.
Alternatively, a total synthesis of the fulvene can also be achieved in the following way:
O _ O \ -.. _ \ -.

O,T~ O'THP
(THP=tetrahydroxopyranyl) OH OH
\ -~ \ -. w \
OH C ~ N OH
OH
3 0 O~P O O
Reaction of the known 1, 1-diacetyl cyclopropane with the dianion of the cyclopentadiene derivative shown gives a diol which on mild acid treatment gives the diolketone. Selective elimination of a tertiary hydroxyl group gives the desired fulvene.

WO 94/18151 '~ ~ ~ ~ PCTIUS94/01232 EXAMPLE III
In Vitro Studies To assess cytotoxic effects, various concentrations of illudins were added to cultures of cells ' for 48 hours, then cell growth/viability was determined by trypan blue exclusion. As an alternative to 48 hour continuous exposure studies, cells were plated in liquid culture in 96 well plates, exposed to various concentrations of illudins for 2 hours, pulsed with ['H]-thymidine for one to two hours and harvested onto glass filters. The filter papers were added to vials containing scintillation fluid and residual radioactivity determined in a beta (scintillation) counter.
When screening the sensitivity of other solid tumor cell lines to illudin S, a breast cell line, MCF-7, was noted to be markedly sensitive (Figure 1). Another breast cell line maintained in our laboratory, MDA-231, was also found to be markedly sensitive to illudin S (Figure 1) .
Studies with dehydroilludin M indicated this analog also displayed selective toxicity towards myeloid leukemia cells and breast carcinoma lines MCF-7 and MDA-231 (Table 1).

1~V0 9>1iQ8151 ~ p~ PCT/US94/01232 TA8I~E 1 Histiospecific cytotoxicity of illudin S and dehydroilludin M as demonstrated by inhibition of thymidine after a two hour exposure to the toxins (N - 3).
ICSO ( nM/L ) Compound Illudin S Dehydroilludin M
HL60, myeloid 7 1 246 19 8392, B-cell 236 31 > 38,000 8402, T-cell 669 t 196 > 38,000 242, melanoma 607 70 > 38,000 547, ovarian 607 110 > 38,000 SL-2, murine (thymic) 142 t 15 5,235 277 MCF-7, breast 58 5 653 65 MDA-231, breast 2.0 0.2 112 17 Because previous studies showed that CEM mdr variants were not resistant to illudin S, several other mdr cell types were studied for susceptibility to illudin S and the dehydroilludin M. These mdr daughter cell lines demonstrate a 200 to 800 fold increase in resistance to multiple conventional chemotherapeutic agents, but showed minimal or no resistance to illudin S or dehydroilludin M
(Table 2). Thus, mdr cells associated with or without the gp170 protein were still susceptible to illudin toxicity.
These studies indicate that illudins' novel structure confers relative non-cross resistance in multidrug resistant hematopoietic cell lines. The derivative of illudins, dehydroilludin M, is slightly less toxic than the parent illudin compound, but results (Table 2) indicate that there is no cross-resistance to this compound in various mdr cell lines.
The effect of illudin S and dehydroilludin M on L1210, murine bone marrow CFU-gm, and C1498 (AML cell line) was studied. Illudin S was the most potent agent ever tested in this assay and displayed the largest differential effect ever noted between L1210 and AML leukemia lines and CFU-gm zone cites (Table 3). The derivative, WO 94/18151 ~' ~ PCTIUS94/01232 dehydroilludin M, while less toxic was markedly more selective towards the AML line. It inhibited AML colony formation at concentrations where it had no effect on the CFU-gm cells (Table 4).

Sensitivity of Different Mdr Lines to Illudin S
MDR cell line Illudin S Dehy droilludin available M

CEM Variants Parent 8.3 2.6 nt VM-1 16.2 6.4 nt AraC 14 nt VT,B100(gp170+)3.7 t 0.7 nt Dox (gp170+) 14 20MDA-232(Breast) Parent 0.85 0.23 54 7 3-1(gp170+) 0.89 t 0.38 58 t11 MCF7-wt(Hreast) Parent 0.88 0.11 92 15 ADR (GSH- 3.7 0.4 68 t15 transferase ) HL-60 Parent 3.1 1.1 163 11 ADR (gp150+) 1.9 0.8 191 44 30KB variant Parent 0.58 0.12 125 14 C-1 (gp170+) 0.69 0.15 80 t18 VBL(gp170+) 0.69 0.11 78 19 L1210 Parent 0.42 0.08 62 8 DDPt(cis-plat) 0.46 0.12 119 39 BCNU 0.58 0.08 100 31 PAM(melphalan) 0.62 0.15 73 t31 CPA(cyclophos) 0.46 0.12 38 15 Inhibition of Growth by Illudin S
Illudin S Concentration Zone of Inhibition (ug/disc) L1210 _Go Colon 38 2.50 500 240 30 1.25 400 70 0 0.63 320 30 0 21553'9 Effect of Illudins on Colony Formation Zone Size Compound Dilution L 1210 CFU-GM C1498 (AML1 Illudin S 1/1,000 850 400 > 1000 1/4,000 600 200 800 1/16,000 550 0 550 1/64,000 300 0 250 Dehydroilludin M 1/25 400 200 > 1000 1/125 (repeat) 300 50 700 EXAMPLE IV
Structure Function Studies The structure-function studies were performed by synthesizing derivatives of the illudins and examining their in vitro toxicity for HL60 leukemia cells (Table 5).
This study identified three critical sites for illudin toxicity. These include the cyclopropane ring (site A), the alpha/beta unsaturated bond site (site H), and the ketone group (site C) (Figure 2). Alteration of any of these sites resulted in up to a 4 log decrease in toxicity.
In contrast, the non-ring primary hydroxyl group (Figure 2, site D) does not contribute to toxicity. Various large chemical groups can be attached to this site without altering toxicity. Many of the derivatives with a marked decrease in toxicity (as compared to illudin S or M) are still more potent than conventional chemotherapeutic agents such as BCNU or cis-platinum (Table 5).

21~~32~
._ WO 94/18151 PCT/US94101232 5 ICso for Various Illudin Derivatives Versus Other Agents in HL-60 cells COMPOUNDS nM

10 _ Illudin S or M 10 Dihydroilludin S or M 100,000 Acylfulvene 500 Dehydroilludin M (diketone) 246 15 Isoilludin M 3,800 Ptaquiloside 7,700 Pterosin C 12,500 2,5,6,7-tetramethylindenone 475 Illudin tosylate 3g DNA polymerase inhibitor: Aphidocolin 2,100 Alkylating agent: BCNU 23,300 Crosslinking agent: cis-platinum 550 14 Alkylating agent: MNNG 15,000 Protein Synthesis Inhibitor: Ricin 0.2 EXAMPLE V
Structure-Function Studies: Chemical Illudin M is readily converted to stable aromatic compounds (on treatment with dilute HC1) which in cell culture studies are more than 1,000 fold less toxic. The chlorine-carbon bond formation, cyclopropane ring opening and extrusion of the tertiary hydroxyl (as water) are synchronous. The intermediate formed can be detected by NMR spectroscopy of the reaction mixture (Figure 3). The intermediate, however, is highly reactive and is quickly converted to a phenol by attack of a second nucleophile, i.e., water. Thus, under acidic conditions, illudin M is clearly bifunctional.
The above studies indicate that the toxicity of illudiris is related to the ease with which the tertiary hydroxyl can be removed and the cyclopropane ring opened.
It was found that illudin toxicity depends on the combined 21~~~~9 ,, , effects of the cyclopropane group (site A, Figure 2), the two double bonds (conjugated diene) (site B), and the ketone (site C). It was hypothesized that oxidation of the secondary hydroxyl group in the five membered ring to a ketone would alter the potency or selectivity of the molecule by contributing to further electron delocalization within the molecule. The new ketone group acts as an "electron sink" so that electrons of the cyclopropane C-C
bonds are delocalized towards the ketone rather than to the carbon atom bearing the tertiary hydroxyl. This means the incipient carbocation, forming as the carbon-oxygen (oxygen of the tertiary hydroxyl) bond breaks, is not as stable as in the case of illudin M. Therefore, carbon-oxygen bond breaking is less favorable and reactivity is reduced. This ketone derivative, termed dehydroilludin M, was synthesized and was less toxic to HL-60 cells in vitro than illudin S
or M (Table 4). As discussed above, the toxicity of dehydroilludin M appeared relatively selective for myeloid and breast carcinoma cells in vitro (Figure 1 and Table 1).
Consistent with the above hypothesis are the results of the kinetics of the reaction of illudin M and dehydroilludin M with dilute HC1. In dilute HC1, illudin M undergoes a pseudo first-order reaction ( k =4 . 7 x 10'' min '1, tl/2 = 148 minutes). Dehydroilludin M also demonstrated first-order kinetics but the reaction was considerably slower (k = 2 x 10'' mine, tl/2 = 2765 min) . In the reaction with dehydroilludin M, no intermediate could be detected by NMR spectroscopy. Presumably it formed too slowly and is too short-lived to be detected. The lower reactivity shown by dehydroilludin M suggests it is more selective in its reaction with nucleophiles and thus has a lower toxicity compared to illudin M.
The reaction of illudins with a naturally occurring nucleophile, glutathione has also been studied.
At a wide pH range, from pH 3 to pH 9, glutathione WO 94/18151 . PCTIUS94/01232 spontaneously reacts with illudin M, illudin S, or dehydoilludin M, producing products analogous to those,:from the reaction of illudin M and HC1. The reaction rate is optimized at a pH of 6.1 to 7.0, indicating the reaction could occur intracellularly.
The toxicity of illudins towards a breast cell carcinoma line MCF7-wt and its MDR resistant daughter line MCF/Adr was then studied. The gp170 negative daughter cell line is drug resistant on the basis of a 50 fold increase in glutathione transferase, which results in a 200 to 800 fold decrease in sensitivity to conventional chemotherapeutic agents. This line also shows a 4.1 fold decrease in qlutathione content. This daughter line showed a 4.2 fold decrease in sensitivity to illudin S (parent ICso 0.88 nmoles/1; daughter line 3.?0 nanomoles/1) versus the 200 to 800 fold seen with other agents. Kinetic studies on the ability of illudins to inhibit glutathione transferase indicated there was no direct inhibition of enzyme activity. These findings show that while illudin toxicity is inversely correlated with intracellular glutathione content it is not correlated with glutathione transferase activity.
EZAMPhE VI
Animal Studies Using procedures set forth in Leonard, J.E. et al., Cancer Res. 47:2899-02 (1987) and Dillman, R.O. et al, Cancer Res: 45:5632-36 (1985) , Molt-4 (human T-cell leukemia) xenografts were established in four week old athymic Halb/c nu/nu mice. After 3 weekly doses of total body radiation (600 cGy), mice were given subcutaneous flank injections of Molt-4 cells together with irradiated (6000 cGy) HT-1080 feeder cells. Two animals received only irradiated HT-1080 feeder cells to ensure these cells did not induce tumors.

21~532~
Animals were monitored for Molt-4 tumor development and when tumors were palpable (approximately 4 x 4 mm at 5 to 7 days), mice were randomized into groups of 5 as previously described. ~ Control mice received intraperitoneal saline and treated mice received either 300 ug/kg illudin S, 30 or 300 ~rg/kg dehydroilludin M, IP twice weekly. In mice given illudin S there was tumor growth delay (Figure 4).
In contrast, in nude mice which received the dehydroilludin M at the low dosage of 30 Ng/kg (the compound was subsequently found to be nontoxic to mice at 60,000 ~rg/kg IP twice a week), three of five tumors underwent complete regression, but two tumors failed to respond (figure 5). The two apparently resistant tumors were harvested and tested in vitro for resistance to illudin S and dehydroilludin M. There was no evidence of resistance to either compound. Two of the complete responders were followed for over twelve weeks without evidence of tumor regression.
Using a different source of athymic nude mice, these experiments were repeated. In these studies there was little effect of illudins on tumor growth. The reason for this variability in response to Molt-4 xenografts probably relates to the low doses of dehydroilludin M, interanimal variations in glutathione metabolism, or drug.
distribution.
The efficacy of dehydroilludin M was then screened in a syngeneic model using murine SL-2 cells. SL-2 leukemia/lymphoma cells are injected subcutaneously and metastasized to lymph nodes, spleen, and lungs, and drug efficacy in this model is determined by increased life span (ILS). The SL-2 cells were administered at 2.5 million cells per animal and treatment was delayed for 7 days until the tumors were palpable. This is a relatively stringent WO 94/18151 ~ PCT/L1S94/01232 test against established tumors and contrasts to general drug screens in the SL-2 model which normally use only 0.5 million cells and starting drug treatment at 3 days.
Dehydroilludin M had a little effect at 30 mg/kg IP twice a week, ILS 5~, and 60 mg/kg IP twice a week, ILS 18~.
When administered IV at 0.03 mg/kg, twice a week, the ILS
increased to 38~. This suggests the drug is metabolized by the liver and is likely more efficacious when administered IV.
During the course of these in vivo experiments, it became clear from in vitro experiments, that histiospecificity of illudins depends upon the presence of an active energy-dependent pump. The SL-2 and the Molt-4 cells were studied and it was determined that the uptake mechanism was not present. Therefore, the studies were redirected into xenograft models that used cells of myeloid lineage.
Human HL-60 cells capable of growing as xenografts in nude mice without animal radiation were obtained from Dr. Theodore Hrightman (NCI). These cells termed HL-60 MRI cells, were confirmed to have energy-dependent uptake pump, a not unexpected finding as their parental cells possess the pump. Dehydroilludin M induced dose related tumor inhibition when administered IP on a twice a week schedule (figure 6). The MTD IP dose for dehydroilludin M was reached in these studies on the 2 dosages per week IP dose schedule. Similar tumor regressions have been observed with IV dehydroilludin M.
In collaboration, the in vivo effects of dehydroilludin M was again studied. Initially the compound was studied against L1210 cells. A dose of 2.5 mg/kg IP
given daily for 5 days resulted in an ILS of only 9$. The dehydroilludin M was then administered as a 24 hour infusion (5.0 mg/kg); the ILS was 11~. After we became ~~~~3~9 aware of the presence of the energy-dependent uptake in human myelocytic cells, dehydroilludin M was screened for in vivo efficacy against a syngeneic mouse AMI, model using C1498 cells and a single bolus of:illudin S, 2.5 mg/kg IP, 5 produced an ILS of 35~. A second trial using the same dosage, administered IP once a day for 5 days resulted in a 44~ ILS. As the animals can tolerate 60 mg/kg IP or 1 mg/kg IV (tail vein) on a twice a week schedule for 4 weeks without demonstrating weight loss or a decrease in 10 food/water intake, it is possible to further optimize both dosage and treatment schedule.
EXAMPLE VII
HL60/MRI Mouse Experiment With 15 Acylfulvene and Dehydroilludin M
Thirty mice were injected subcutaneously, over the shoulder, with 500,000 HL60/MRI cells (human myeloid leukemia tumor cells). Treatment was begun on day 11, 20 rather than immediately. This delay in starting treatment is a stringent test to determine whether a compound is effective. Hy delaying treatment, the tumor cells become firmly established.
The mice were divided into 6 groups of 5 each.
One group was the control and these animals received on a placebo, the solution used to dilute the agent. The other groups received the following compounds and dosages: the dehydroilludin M compound at 1.0 mg/kg, the dehydroilludin M at 3.0 mg/kg, the Acylfulvene at 0.3 mg/kg, the Acylfulvene at 1.0 mg/kg, the Acylfulvene at 3.0 mg/kg.
All animals received the placebo or drugs by intravenous injection using a tail vein. The placebo or drugs were administered on a twice a week schedule.

Results are summarized in the accompanying table 6. Both the dehydroilludin M and the Acylfulvene compound were effective at inhibiting tumor growth and demonstrated dosage dependence inhibition (the more drug administered, the less the tumors grew). The animals receiving the highest amount of either drug did not display any evidence of adverse effect, such as a decrease in food or water intake, nor a statistically significant decrease in body weight. These results show that higher dosages of either drug can be administered. Also, that the drug could be administered on a more effective dosage schedule, such as on a daily basis.

Summary: HL60/MRI experiment, intravenous - # 1 BY TOTAL TUMOR WEIGHT [MgJ

CONTROL
No Drug 99~36 845~282 3299~1080 10162~4123 16747~5061 Dehydroilludin M
1 mg/kg 114~55 883~311 2274~992 6025~1772 11507~3707 IV
3 mg/kg 101~40 911~309 2127~1092 2854~1260 4784~2303 IV
Acylfulvene 0.3 mg/kg 73~38 540~167 13521520 3204~1147 9501~4605 IV
1 mg/kg 58~32 582~297 964~685 232111434 6275~2865 3 mg/kg 38~30 369~250 336~215 437~238 1201~501 ~1~5~~~

EXAMPLE VIII
General In Vitro Screening Procedures and Cell Uptake Studies In keeping with the suggestions of the previous examples and our concentration'on mechanisms of illudin action and tissue specificity, other myeloid leukemia cell lines can be screened for rapid illudin uptake (KG1, KGla, HEL, K562, OCI-M1, AML-193).
The procedures for in vitro screening of illudin compounds are detailed in the previous examples.
Cytotoxicity of new analogs for cell lines is initially evaluated over a 5 log range using growth or semi-solid colony forming assays, and inhibition of thymidine incorporation. Inhibition of thymidine incorporation is used because earlier studies indicate that thymidine incorporation is preferentially inhibited by illudins and correlates closely with cell death. Analogs are screened against normal bone marrow progenitors and a variety of cell lines involving various leukemias, B and T cell) and solid tumors (melanoma, ovarian).
In vitro testing of dehydroilludin M on various cell lines, including MDR lines, can also be performed on DNA-repair deficient cell lines and normal bone marrow progenitors. A variety of other analogs can be prepared.
Since these analogs will have alterations in the known active sites, they are expected to result in a similar tumor inhibition. Screening studies for these analogs can include various mdr cells (to ensure that no cross-resistance occurs) and DNA-repair deficient cell lines.
In vitro testing can also study sensitivity of other breast cell lines to determine if they are also preferentially sensitive to illudin S, dehydroilludin M, and the fulvene analog.

_ WO 94/18151 '~ ~ ~ PCT/US94/01232 EXAMPLE IX
Assessment of Illudin Uptake in Tumor Cells While human myeloid tumor cells are sensitive to illudins, their normal precursors, granulocyte/macrophage forming units, are relatively resistant to illudins by 1.5 to 2.0 logs, demonstrating that the transport system is absent from some normal marrow cells and providing a therapeutic margin of safety.
Specific illudin S uptake was assayed using relatively sensitive HL60 cells and resistant B cells. At 37°C, HL60 myeloid leukemia cells demonstrated rapid uptake of illudin S, while the relatively insensitive 8392 B-cells exhibited comparatively little drug incorporation (Figure 7). The intracellular accumulation of illudins in the B
cell line was slow and linear for 7 hours (r - 0.984), at which time the intracellular concentration approached that of the incubation mixture. HL60 cells, in contrast, rapidly accumulated the toxin and intracellular accumulation reach a plateau within one hour. HL60 cells exposed to 10 nM illudin S concentrated the toxin 19 fold, whereas B cells did not actively concentrate the toxin.
The rapid intracellular accumulation of illudin S by HL60 cells was saturated at high concentrations (Figure 8). In contrast, illudin S accumulation in 8392 B cells remained concentration dependent. Analysis of the initial uptake of illudin S by HL60 cells at varying concentrations revealed that the influx of illudin S conformed to Michaelis-Menton saturation kinetics (Figure 9). The Vmax for HL60 cells was 27 picomoles/minute/mg of protein and the Km was 4.2 NM. This indicates HL60 cells have a very high transport capacity for illudins as the Vmax for illudins is 5 times the Vmax for folate, a vitamin required by cells.

21~~~~~

Cold (4°C), 1~ azide, and the metabolic blockers 2-deoxyglucose and antimycin A, all block uptake of illudin S into HL60 cells but hade'little effect on the insensitive 8392 B-cells (Table 7). These studies indicate that illudin S is transported and concentrated into HL60 cells by an energy dependent transport system, whereas the transport into insensitive H-cells occurs only by diffusion (passive or nonenergy requiring transport). MCF7 breast tumor cells also demonstrated inhibition of uptake by cold.
The finding of an energy-dependent transport mechanism explains why myeloid and breast tumor cells are so sensitive to illudins with short exposure times, but H-cells are not.

Uptake of ['H] Illudin S by HL60 Myeloid versus 8392 B-cells Maximum uptake per hour (picomoles)' Conditions HL60 8392 MCF7 37C 75 16b 5.5 1.4 29 4 4C 4.3 t 0.9 3.4 1.0 4.0 2.1 1$ Azide 8.7 1.4 4.3 1.3 NTb 2-deoxyglucose & 16.7 3.5 3.6 1.4 NT

Antimycin A

Cells were exposed to 100 ng/ml of ['H]-labeled illudin S for one hour and harvested as described. Results are expressed as mean t SE and represent 3 experiments.
aper 10 million cells bNT = not tested EXAMPLE X
Synthesis and Structure of 2,5,6.7 Tetramethyl-1-Indenone and Dehydropterosin Compounds 5 First 2,4,5,6-tetramethyl-1,3-indanione was synthesized by preparing a solution of 1,2,3-trimethylbenzene and methylmalonylchloride in carbon disulfide and adding aluminum trichloride dropwise over two hours. The mixture was relfuxed for 2 more hours, crushed 10 ice added, and extracted three times with chloroform. The combined extract was washed with brine, dried, and solvent removed to leave a residue which was purified by chromatography with 1~ ethyl acetate in benzene. Removal of solvent and purification by sublimation gave the desired 15 product.
The 2,5,6,7-tetramethyl-1-indenone was prepared by reducing 2,4,5,6-tetramethyl-1,3-indanione with zinc dust at 50°C. Product was purified by chromatography with 20 1~ ethyl acetate in benzene to yield two isomers. The major isomer was treated with 10~ potassium hydroxide, then purified by sublimation. The compound has the structure:
O
Dehydropterosin 0 synthesis: 3-acetoxy-6(beta-methoxy) ethyl-2,5,7-trimethyl-1-indanone was dissolved in tetrahydrofuran and 10~ potassium hydroxide and refluxed for two hours. The solution was then extracted three times with ether and the combined extracts chromatographed with 2~ ethylacetate in benzene to yield the Dehydropterosin 0 compound. The compound has the structure:

2~~53~~g 26 O
RO
R = H Dehydropterosin B
R = CH3 Dehydropterosin 0 Hoth compounds were toxic to cells in vitro and have antifungal properties.
EXAMPLE XI
Synthesis of Acylfulvene Analogs Many acylfulvene analogs possessing the key structural features required for antitumor activity may be prepared starting from illudin S or by total synthesis from simple precursors. Illudin S is produced by fermentation of Omphalotus illudens. On dissolving this compound, in water and adding dilute HZSO, it is converted to the acylfulvene (R1=H, RZ=H, R3=CH3) in 55~ yield. A large number of analogs may be obtained from the acylfulvene by modifying the R2 substituent, e.g., Rz - hydroxymethyl, bromo, iodo, chloro, fluoro, vitro, p-hydroxybenzyl, p-methoxybenzyl. Rz can also be polynuclear or heterocyclic aromatic groups.
OH R, "1. \ '1'. \ \.
--. R~
HO~.... ~ ~~CH OH R~O~~...

The R1 group may be acyl or alkyl. Analogs with different R, groups (and also Rz groups) can be prepared by total synthesis as outlined.
~O
\ R' w w O
S S
OH RZ
R
HO ~ R' ~ HO ~ ' S S
O
Aldol condensation of 1,1-diacetylcyclopropane with the dianion derived from an appropriately substituted cyclopentadiene gives an intermediate which is readily converted to the acylfulvene. A wide variety of compounds are possible since R2 and R, may be alkyl, aryl or substituted alkyl or aryl groups.
Synthesis of acylfulvene. Illudin S 2 g (9.2 mmol) was dissolved in 700 mL water followed by addition of 4 M HZSO, (236 mL). The solution was stirred overnight, and extracted with ethyl acetate. The organic phase was washed with saturated NaHCO" water and brine, dried over MgSO, and concentrated. Chromatography on silica with hexane and ethyl acetate afforded 0.82 g acylfulvene (50$). 1H NMR S
0.73 to 1.50 (m, 4 H), 1.38 (s, 3 H), 2.00 (s, 3 H), 2.15 (s, 3 H), 3.93 (s, 1 H), 6.43 (s, 1 H), 7.16 (s, 1 H); MS
m/Z 216 (M+) , 202 (M+-CH2) , 188 (M+-CHZCHZ) , 173 (M+-2CHz-CH,), 170 (M+-2CH2-H~0). W 325 nm (8.3 x 10'), 235 nm (16.6 x 10'). The compound has the structure:

~15~3~~ 28 HC
O
Synthesis of 6-hydroxymethylfulvene. Acylfulvene (550 mg, 2.5 mmo1) was dissolved in 40 mL THF and 30%
formaldehyde-water solution (40 mL) was added. 4 N HzSOa solution (26.4 ml) was added to bring the final concentration of HZSOa to 1 N. The solution was stirred overnight and extracted with ethyl acetate. The organic phase was washed with saturated NaHCO, solution, water and brine and dried over MgS04. Chromatography on silica gel with hexane and ethyl acetate gave 400 mg hydroxymethylfulvene (64%). 1H NMR b 0.72 to 1.48 (m, 4 H), 1.38 (s, 3 H), 2.15 (s, 3 H), 2.19 (s, 3 H), 3.90 (s, 1 H), 4.66 (d, J=2.1 Hz, 2 H), 7.10 (s, 1 H). MS m/Z 246 (M+), 22 8 ( M+-HZO ) , 218 ( M+-CHZCH2 ) , 18 6 ( M+-CH3-CH2-CHZ-OH ) , 18 5 (M+-H20-CHZ-CHz-CH, ) . W 233 nm ( 1. 0 x 10° ) , 325 nm ( 7 . 7 x 10'). The compound has the structure:
CH,OH
,,, 3 0 HO "~
O
Iodofulvene. To a solution of acylfulvene (60 mg, 0 . 28 mmol ) in 15 ml CHzClz was added silver trifloroacetate (63 mg, 0.29 mmol). A solution of iodine (70.5 mg, 0.28 mmol) in 8 mL CHzCl2 was added dropwise at 0 ._ WO 94/18151 ~ ~ ~ PCT/US94I01232 °C. The mixture was stirred at that temperature for 3 hours then filtered through celite and eluted with ether.
Concentration of the filtrate gave iodofulvene as a red gum (73 mg, 77~). 1H NMR b 0.76 to 1.54 (m, 4 H), 1.38 (s, 3 H), 2.14 (s, 3 H), 2.36 (s, 3 H), 3.87 (s, 1 H), 7.16 (s, 1 H ) . MS m/ Z 3 4 2 ( M+ ) , 314 ( M+-CHZCH~ ) , 2 9 9 ( M+-CH2CHz-CH, ) , 2 9 6 ( M+-CHZCHz-H20 ) , 215 ( M+-I ) , 187 ( M+-I-CHZCHZ ) , 127 (I+). The compound has the structure:
H( O
Bromofulvene. Acylfulvene (60 mg, 0.28 mmol) was dissolved in 9 ml acetonitrile at 0 °C. N-bromosuccinimide (50 mg, 0.28 mmol) was added and the mixture was stirred at that temperature for 3.5 hours. Water was used to quench the reaction and ether to extract the product. The ether layer was washed with water and brine and dried over MgSO,.
Chromatography gave bromofulvene as orange crystals (77 mg, 94~; recrystalized from ether acetate-hexane, m.p.. 92-94 °C). 1H NMR b 0.75 to 1.55 (m, 4 H), 1.40 (s, 3 H), 2.12 (s, 3 H), 2.33 (s, 3 H), 3.89 (s, 1 H), 7.15 (s, 1 H). MS
m/Z 295 (M+) , 293 (M+-2 ) , 267, 265, (M+-CHZCHz) , 252, 250 ( M+-CHZCHz-CH, ) , 2 4 9 , 24 7 ( M+-CHzCH2-Hz0 ) , 215 ( fulvene-1 ) .
The compound has the structure:

O

21~~32~ 30 p-Hydroxybenzylfulvene. Phenol (40 mg, 0.4 mmol) was added to a solution of hydroxymethyl-fulvene (70 mg, 0.28 mmol) in dry CHzCl2 (25 mL). The mixture was cooled to -78 °C and boron triflouride etherate (0.3 mL, 2.7 mmol) was added dropwise. The reaction was stirred at that temperature for 1 hour and water was added to quench the reaction. The organic layer was washed with HBO, NaHC03 and brine, and dried over MgS04. Chromatography on silica gel with hexane-ethyl acetate yielded 90 mg (98$) of red crystals (m. p. 143-144 °C). 1H NMR b 0.59-1.43 (m, 4 H), 1.36 (s, 3 H), 1.76 (s, 3 H), 2.07 (s, 3 H), 3.95 (s, 1 H), 3.97 (d, J=7.2 Hz, 2 H), 4.83 (s, 1 H), 6.74 (d, J=8.4 Hz, 2 H), 6.91 (d, J=8.4 Hz, 2 H), 7.22 (s, 1 H). MS m/Z 322 (M+) , 294 (M+-2 CHZ) , 279 (M+-2 CH2-CH3) , 251 (M+-2 CHZ-CH3-CO), 215, 107. W 228 nm (1.2 x 10°, with inflections at 243 and 262 nm), 325 (7.1 x 10'), 410 nm (2.6 x 10'). The compound has the structure:
cx~ O OH
2 0 /~
HO ~~~~.
O
p-Methoxybenzylfulvene. Anisole (0.04 mL, 0.37 mmol) was added to a solution of hydroxymethylfulvene (10 mg, 0.04 mmol) in dry CHzClz (5 mL). The mixture was cooled to -78 °C and boron triflouride etherate (0.04 mL, 0.36 mmol) was added dropwise. The reaction was stirred at that temperature for 1 hour and water was added to quench the reaction. The organic layer was washed with H20, saturated NaHC03 and brine, and dried over MgSOa. Concentration of the solution gave a residue which was dried in vacuo, yielding the product in quantitative yield (14 mg). 1H NMR
d 0.59-1.40 (m, 4 H). 1.36 (s, 3 H), 1.76 (s, 3 H), 2.07 (s, 3 H), 3.78 (s, 3 H), 3.95 (s, 1 H), 3.99 (d, J=16.12 Hz, 2 H), 6.81 (d, J=8.8 Hz, 2 H), 6.96 (d, J=8.3 Hz, 2 H), 7.22 (s, 1 H). MS m/Z 336 (M+), 308 (M+-CHzCH2), 215, 121 ( ( CH2-Ph-OCH, ) + ) , W 410 nm ( 2 . 7 x 103 ) , 325 nm ( 7 . 0 x 10' ) , 267 nm (1.0 x 10°), 245 nm (inflection), 226 nm (1.9 x 10°), 203 nm (1.4 x 10°). The compound has the structure:
~ H,_ OCH3 n HO ~~~, EXAMPLE XII
In Vitro Cell Culture Studies of Acylfulvene Analogs In vitro testing using cell culture assays demonstrated the 6-hydroxymethylacylfulvene, bromoacylfulvene, and iodoacylfulvene analogs were markedly toxic to the target tumor cells HT,60 and MV522 at both 2 and 4 8 hour exposure periods ( Table 8 and Table 9 ) . The relative toxicity ratio (2 to 48 hour toxicity) suggested these analogs would be more efficacious in vivo than either the parent Illudin S compound or the analogs described in the original patent application.

a~

c -.a '~ N O ~' N O OJ
f~. ~ ' O

O I N tl CD O
? M' 1 v N vD :~ N G7~ .~ ~ N

N

1-1 ~ 01 ~Dt~ ~ ~OO 10 O tl1 O O -~ N N .~ t0W LI7 Ca er C7I~ ~-i O 'i C1 ~--~O

ri ~i .1 .~~ ,1 N

N

~ O
+~

~ r1 O

O O O ~p ~-.I p~ N
C ~ O 01 1l1 01 O1 .~ ro tG CO~ O 01O t~ w N

r-i N ~ f'~1. . p~ 01 ~-1 ri N C1rl N N N ep ~--If~

N U +I +I+I +I +I+I +I +I +I
.~

U

cn a N vo u n o, m n .w c '~ ri Q1 M M M lfl lf7~ M M
G

O ro M N M er M err~ M N r C .1 ,~ m w .~ '~ .~10 t~ ~ ~ O ~1 Ll1 ~Ot0 y -1 v-1 Q1 M

U

~! ~"~H

~i a a m ~s1 ro O ~ ~ N w N ~"., r-I -~~ .-~ 01N eh ro U1 Gl I O

~ N 1-i .-1m M M m N

H +I +~+~ +I +I+1 +i +I +I

o O n O O M M N

o ro a -~ ~ a n M o o vo vo :C M

'~
01 r-1 M CD N C1 ro r-I M N ~-1 a ro ro c .'.;
~

.v c ~' a~
,~, o a ' ~ a~ a~

a a -.~ ro w o ~

~ ~ U y i r -i O ~ ro O U
x U

O C "'~ ~ O '~ ~ N

. N
a.1 0 'Q i~ rar~ ' '~",' c ro c ~ a a ro ~ ~ ~ .

a~ ro~
N -.~~ x ~ ~ x x 0 U U ?, 0 0 a a rl H a H +~ro U to .C
0 N ' ' fl O W

?~ s 0 O 'D

a H A

a ~o ~ .a . a, a N ri ~l1 O tI1 O

N

215~~~~

c rl M M N .-In 10 v--1 1-I N ri N N 00 ey0 er 1l1N

N

C H

O O d' M 01 M 1Ct0 10 u1 OD

H +~ -i~r n cow o ro .,~ M M ~ M tn 10 tf7 a ,~ , N

N

r-~

G

o ro c ro N O N M O M O

0 v rl O Lf1 t0M 00 00 , y -1 N ~ ~ ~ .-1~ rl M N

ro a~ U +I +I+I +I +f+I +I +I +I
C

ro rl N C7 01M 1G ~'1D 10 CD M

b o~ ~ ~ M n M o~ o~ n c0 ~ M N of ~ rt ~O O rl OD

~I O c .-r,~ .-in .a U

H H

H

r~ O N

G~7 C ,...I

~3 " r-I ~Ger O ~-1N 01 O

Cn ~ ~ 10n ~-i M N ~ M r-1 1G M t0aT ~ 1CN O N '-I

a awo o ~ ov o, w I

U x N M W ~ N M O

ri CT

r~i ro C

ro w ~ ~

p r O O

x G

~r ~ I N O

r--I~I

U

0 O N ~ ~t c x ~ ~ a~ .-~r, ~.J rl .~ O '~ N N N

0 'fl +~ ~--r~

U a ~ ~ ~ ro ' ~ >, ~ x x -~

H 0 ~ O .-~I0 47U >, O 0 a CL .-1 H a la +~ro U f-1 .o ~ Ts 'vw v roo ro o ~ ~ ~ 0 U >, . 0 'D

U U I U t-tO I t c~ c~ t--tt:~ry o c9.L7ri t1 CL

m O vr1 0 N

EXAMPLE XIII
In Vivo Studies Based on these in vitro screening results, one of the analogs, the 6-hydroxymethylacylfulvene, was chosen for in vivo studies to determine if the analog was indeed more potent than the acylfulvene. The xenograft was again the human lung adenocarcinoma MV522 as it is a nonresponsive model to conventional anti-cancer agents and kills by metastasis (not local tumor invasion). The conventional anti-cancer agents cis-platinum, Taxol*, mitomycin C, adriamycin, as well as Illudin S ~ were chosen as ' pharmaceutical controls. A control group was included that received only the solvent used to dissolve the drugs, a 40%
dimethylsulfoxide/normal saline mixture (40% DMSO/NS). The 6-hydroxymethylacylfulvene analog actually induced tumor regression in animals. Actual regression of tumors by an anticancer agent has never been noted before in this model.
There was no inhibition of tumor growth by conventional anticancer agents or by Illudin S (Figure 10). Animals received only 9 doses of the 6-hydroxymethylacylfulvene analog. There was no evidence of toxic side effects in these animals as evidenced by a decrease in activity, weight gain, food intake, or water intake. There was no significant increase in the life span of cisplatinum, ~Taxol*, mitomycin C, and Adriamycin treated animals as compared control (DMSO/NS) treated animals. The Illudin S
actually caused premature death (drug toxicity) (Table 10).
The 6-hydroxymethylacylfulvene treated animals lived significantly longer than controls (DMSO/NS treated), cisplatinum,~Taxol*, mitomycin C, and Adriamycin treated animals (p < 0.001 for all groups) (Table 10).
* Trade-mark - ' CA 02155329 2005-08-03 TABLE 1~
Efficacy of 6-hydroxymethylacylfulvene analog, versus other agents, in the human lung adenocarcinoma MV522 metastatic lung tumor model - First experiment 10 drug life span controls DMSO/NS IP 3X/WK 100 +
7%

cis-platinum 3.2 mg/kg 1X/WK 102 +
IP 8%

Taxol* 4.0 mg/kg 5X/WK 100 10%
IP

15 mitomycin C 2.4 mg/kg 1X/WK 111 2%
IP

Adriamycin 2.6 mg/kg 1X/WK 98 +
IP 12%

Illudin S 2.5 mg/kg IP 3X/WK <26%
[5/5 dead at only 3 doses]
6-hydroxymethylacylfulvene 10 mg/kg IP.3X/WK 233 + 18%
The experiment was repeated with different dosages of the 6-hydroxymethylacylfulvene analog to determine if a dose-response pattern was present. Taxol~
and low dose Illudin S were again included as pharmaceutical controls (Table 11). A control group receiving only the pharmaceutical solvent (40% DMSO/NS) was again included. Tumor regression was again noted at 10 mg/kg, and inhibition of tumor growth was noted with 1 and 5 mg/kg treatment. The-Taxol*and low dose Illudin S again failed to inhibit tumor growth (Figure 11). In this second experiment there was again no significant increase in the life span of Taxol*or Illudin S treated animals as compared controls (40% DMSO/NS treated animals). The 10 mg/kg 6-hydroxymethylacylfulvene and 5 mg/kg 6-hydroxymethylacylfulvene treated animals lived significantly longer than controls (DMSO/NS treated), * Trade-mark WO 94!18151 PCT/US94/01232 'Taxol*,or Illudin S treated animals.' The probability (or significance) value for 10 mg/kg 6-hydroxymethylacylfulvene treated animals versus the controls, Taxol* treated, and Illudin S treated animals was less than 0.001 in each case (p < 0.001). The probability value for the 5 mg/kg 6-hydroxymethylacylfulvene treated animals versus the controls, Taxol* treated, and Illudin S treated animals was also less than 0.001 in each case (p < 0.001). The 1 mg/kg 6-hydroxymethylacylfulvene treated animals also lived significantly longer than the control group (p < 0.01).
TABhE 11 Efficacy of 6-hydroxymethylacylfulvene analog, versus other agents, in the human lung adenocarcinoma MV522 metastatic lung tumor model - Second experiment drug life s an controls DMSO/NS IP 3X/WK 100 _+ 16%
(100% by definition) Taxol* 4.0 mg/kg IP 5X/WK 120 _+ 10%
(not significant) Illudin S 0.25 mg/kg IP 3X/WK 104% + 20%
6-hydroxymethylacylfulvene 10 mg/kg IP 3X/WK 232 + 20%
6-hydroxymethylacylfulvene 5 mg/kg IP 3X/WK 154 + 13%
6-hydroxymethylacylfulvene 1 mg/kg IP 3X/WK 135 + 10%
The experiment was repeated for a third time (Table 12). The amount of Taxol* administered IP was increased to demonstrate maximum dosage, and a subcutaneous dosage~was added due to reports that this route may be more efficacious. Adriamycin and mitomycin C were again included. Two of the new acylfulvene derivatives * Trade-mark - WO 94/18151 215 5 3 2 g PCT/US94101232 (iodoacylfulvene and the p-hydroxybenzylfulvene) were included in this experiment. The diketone analog was included to demonstrate the marked improvement of the 6-hydroxymethylacylfulvene. The 6-hydroxymethylacylfulvene markedly increased the life span versus controls and all other drug treated animals. The probability (or significance) value for the 6-hydroxymethylacylfulvene versus all the control group, the mitomycin C treated group, the adriamycin treated group, and both of the taxol treated groups was less than 0.0005 (p < 0.0005). This is an extremely significant effect. It is important to note that based on the longevity of 2 particular animals, there is the possibility they may have been cured. Although the diketone also markedly increased the lifespan versus controls (p < 0.002) and other drug treated animals, it was not as effective as the 6-hydroxymethylacylfulvene. Note that the lifespan of the high dose taxol treated animals (6 mg/kg IP) not only decreased below that of the 4 mg/kg taxol treated animals in the previous experiment, but it was now less than the life span of untreated or control animals. This indicates that the maximum dose for taxol had been reached and drug toxicity was now killing the animals. The other new analogs, the bromoacylfulvene and the p-hydroxybenzylfulvene are also effective in this model.

Efficacy of 6-hydroxymethylacylfulvene analog, versus other agents, in the human lung adenocarcinoma MV522 metastatic lung tumor model - Third experiment drug life s an controls DMSO/NS IP 3X/WK 100 + 29%
taxol 6.0 mg/kg IP 5X/WK 93 + 22%
taxol 20.0 mg/kg IP 5X/WK 113 + 22%
mitomycin C 2.4 mg/kg IP 1X/WK 149 + 12%
adriamycin 2.6 mg/kg IP 1X/WK 105 + 25%
diketone 30 mg/kg IP 3X/WK 163 _+ 6%
(listed original application) iodoacylfulvene 20 mg/kg IP 3X/WK 120 _+ 34%
p-hydroxybenzylfulvene 15 mg/kg IP 3X/WK 125 _+ 16%
p-hydroxybenzylfulvene 20 mg/kg IP 3X/WK 126 + 22%
6-hydroxymethylacylfulvene 10 mg/kg IP 3X/WK > 204 ( 2 animals alive, ? cured) --Although the invention has been described with reference to the presently-preferred embodiment, it should be understood that various modifications can be made without departing from the spirit of the invention.
Accordingly, the invention is limited only by the following claims.

Claims (22)

WE CLAIM:
1. A use of a compound of the formula:

wherein R is selected from the group consisting of wherein R1 is alkyl aryl, NH2, NH(alkyl) or N(alkyl)2, for preparing a medicament effective to inhibit tumor growth in a subject.
2. The use according to claim 1 wherein the medicament is adapted for intravenous, oral or intraperitoneal administration or for administration by inhalation.
3. The use according to claim 1 or 2, wherein the medicament comprises about 30-100 mg of the compound and a pharmaceutically acceptable carrier.
4. The use according to claim 1, 2 or 3, wherein the tumor is a myeloid tumor, an epidermoid tumor, T-cell leukemia, or lung, ovarian or breast carcinoma.
5. The use according to any one of claims 1 to 4, wherein R is CH2OH, p-hydroxybenzyl, p-methoxybenzyl, acetoxymethyl, I or Br.
6. A pharmaceutical composition comprising an effective tumor-inhibiting amount of a compound of the formula:

wherein R is selected from the group consisting of wherein R1 is alkyl, aryl, NH2, NH(alkyl) or N(alkyl)2; in combination with a pharmaceutically acceptable carrier.
7. The pharmaceutical composition of claim 6 wherein R is CH2OH, p-hydroxybenzyl, p-methoxybenzyl, acetoxymethyl, I or Br.
8. The pharmaceutical composition of claim 6 which is adapted for intravenous, oral or intraperitoneal administration or for administration by inhalation.
9. The pharmaceutical composition of claim 6 wherein about 30-100 mg of the compound is combined with the pharmaceutically acceptable carrier.
10. The pharmaceutical composition of claim 6 wherein the tumor is a myeloid tumor, an epidermoid tumor, T-cell leukemia, or lung, ovarian or breast carcinoma.
11. A compound of the formula:

wherein R is selected from the group consisting of:
12. The compound of claim 11 wherein R is I or Br.
13. The compound of claim 11 wherein R is CH2OH.
14. The compound of claim 11 wherein R is CH2OC(O)CH3.
15. The compound of claim 11 wherein R is 4-hydroxybenzyl.
16. The compound of claim 11 wherein R is 4-methoxybenzyl.
17. The compound of claim 11 wherein R is CH2OC(O)NH2.
18. A use of a compound of the formula:

wherein R is selected from the group consisting of wherein R1 is alkyl aryl, NH2, NH(alkyl) or N(alkyl)2, for inhibiting tumor growth in a subject.
19. The use according to claim 18 wherein the compound is for intravenous, oral or intraperitoneal administration or for administration by inhalation.
20. The use according to claim 18 or 19, wherein the tumor is a myeloid tumor, an epidermoid tumor, T-cell leukemia, or lung, ovarian or breast carcinoma.
21. The use according to any one of claims 18, 19 or 20, wherein R is CH2OH, p-hydroxybenzyl, p-methoxybenzyl, acetoxymethyl, I or Br.
22. The use of the pharmaceutical composition of any one of claims 6 to 10, for inhibiting tumor growth in a subject.
CA002155329A 1993-02-09 1994-02-02 Acylfulvene analogues as antitumor agents Expired - Fee Related CA2155329C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/015,179 1993-02-09
US08/015,179 US5439936A (en) 1989-10-03 1993-02-09 Method of treating certain tumors using illudin analogs
PCT/US1994/001232 WO1994018151A1 (en) 1993-02-09 1994-02-02 Acylfulvene analogues as antitumor agents

Publications (2)

Publication Number Publication Date
CA2155329A1 CA2155329A1 (en) 1994-08-18
CA2155329C true CA2155329C (en) 2006-08-15

Family

ID=21769946

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002155329A Expired - Fee Related CA2155329C (en) 1993-02-09 1994-02-02 Acylfulvene analogues as antitumor agents

Country Status (22)

Country Link
US (3) US5439936A (en)
EP (1) EP0683762B1 (en)
JP (1) JP3908270B2 (en)
KR (1) KR100311327B1 (en)
CN (1) CN1046934C (en)
AT (1) ATE174894T1 (en)
AU (1) AU676889B2 (en)
BR (1) BR9405689A (en)
CA (1) CA2155329C (en)
CZ (1) CZ288596B6 (en)
DE (1) DE69415507T2 (en)
DK (1) DK0683762T3 (en)
ES (1) ES2125441T3 (en)
GR (1) GR3029736T3 (en)
HU (1) HU220059B (en)
MD (1) MD1418G2 (en)
NO (1) NO318648B1 (en)
NZ (1) NZ262282A (en)
PL (1) PL175024B1 (en)
RU (1) RU2145849C1 (en)
TJ (1) TJ266B (en)
WO (1) WO1994018151A1 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5932553A (en) 1996-07-18 1999-08-03 The Regents Of The University Of California Illudin analogs useful as antitumor agents
US5723632A (en) * 1996-08-08 1998-03-03 Mgi Pharma, Inc. Total synthesis of antitumor acylfulvenes
US7141603B2 (en) * 1999-02-19 2006-11-28 The Regents Of The University California Antitumor agents
US6025328A (en) * 1998-02-20 2000-02-15 The Regents Of The University Of California Antitumor agents
US6436916B1 (en) 2000-10-12 2002-08-20 Alvin Guttag Ibuprofen-aspirin and hydroxymethylacylfulvene analogs
US7015247B2 (en) * 2000-10-12 2006-03-21 Alvin Guttag Ibuprofen-aspirin, hydroxymethylacylfulvene analogs and L-sugar illudin analogs
CA2571421A1 (en) 2004-06-24 2006-01-05 Nicholas Valiante Compounds for immunopotentiation
JP4989648B2 (en) * 2005-08-03 2012-08-01 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Iludine analogues useful as anticancer agents
CN102333528B (en) * 2009-01-26 2013-12-18 台北医学大学 Use of pterosin compounds for treating diabetes and obesity
JP2017518359A (en) 2014-04-10 2017-07-06 エイエフ ケミカルス, エルエルシーAf Chemicals, Llc Affinity drug conjugate
US10285955B2 (en) 2014-04-10 2019-05-14 Af Chemicals, Llc Affinity medicant conjugate
US11135182B2 (en) 2014-04-10 2021-10-05 Af Chemicals, Llc Affinity medicant conjugates
CN106083953A (en) * 2016-06-15 2016-11-09 成都医学院 A kind of from fiddlehead, extract former pteroside and the method preparing high purity raw pteroside
US20210137850A1 (en) * 2017-03-15 2021-05-13 Memorial Sloan Kettering Cancer Center Diagnosis & treatment of ercc3-mutant cancer
JP2021536492A (en) * 2018-09-04 2021-12-27 ランタン ファーマ インコーポレイテッド Illudin analogs, their use, and how to synthesize them
EP3667323A1 (en) 2018-12-11 2020-06-17 Kelner, Michael Methods, compositions and devices for treating cancer with illudofulvenes
RU2738848C1 (en) * 2019-10-24 2020-12-17 Общество с ограниченной ответственностью "Научно-исследовательский институт ХимРар" (ООО "НИИ ХимРар") Hepatitis b virus (hbv) inhibitor, which is a derivative of n-{3-[6-(dialkylamino)pyridazine-3-yl]phenyl}aryl sulphonamide derivatives and derivatives of n-{4-[6-(dialkylamino)pyridazine-3-yl]phenyl}aryl sulphonamide
EP4035684A1 (en) 2019-11-25 2022-08-03 AF Chemical LLC Affinity illudofulvene conjugates
US11591295B2 (en) 2019-11-25 2023-02-28 Af Chemicals Llc Affinity illudofulvene conjugates
CN112972443A (en) * 2021-03-29 2021-06-18 杭州添帆生物科技有限公司 Anticancer medicine and its application
WO2023239821A2 (en) * 2022-06-07 2023-12-14 Lantern Pharma Inc. Treating cancers with combinations of acylfulvenes with ibrutinib or bortezomib
WO2024016014A2 (en) * 2022-07-15 2024-01-18 Lantern Pharma Inc. Method for treating breast cancers and parp resistant breast cancers

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2067365C (en) * 1989-10-03 2006-01-31 Michael J. Kelner Illudin analogs as anti-tumor agents
GB9017024D0 (en) * 1990-08-03 1990-09-19 Erba Carlo Spa New linker for bioactive agents
FI85318C (en) * 1990-08-14 1992-03-25 Tecnomen Oy COMPENSATION AV FELET I EN KLOCKAS GAONG.
JPH06223404A (en) * 1993-01-25 1994-08-12 Mitsubishi Kasei Corp Optical information recording medium

Also Published As

Publication number Publication date
NO318648B1 (en) 2005-04-25
CZ198695A3 (en) 1996-01-17
RU2145849C1 (en) 2000-02-27
KR100311327B1 (en) 2001-12-28
HU9502358D0 (en) 1995-10-30
NZ262282A (en) 1997-06-24
DE69415507T2 (en) 1999-08-12
CZ288596B6 (en) 2001-07-11
TJ266B (en) 2000-08-07
CA2155329A1 (en) 1994-08-18
AU6170094A (en) 1994-08-29
BR9405689A (en) 1995-11-21
AU676889B2 (en) 1997-03-27
PL310159A1 (en) 1995-11-27
DE69415507D1 (en) 1999-02-04
EP0683762A1 (en) 1995-11-29
WO1994018151A1 (en) 1994-08-18
NO953099L (en) 1995-10-09
NO953099D0 (en) 1995-08-07
JPH08506812A (en) 1996-07-23
US5563176A (en) 1996-10-08
GR3029736T3 (en) 1999-06-30
ATE174894T1 (en) 1999-01-15
HUT72450A (en) 1996-04-29
MD960212A (en) 1999-01-31
HU220059B (en) 2001-10-28
PL175024B1 (en) 1998-10-30
CN1119854A (en) 1996-04-03
JP3908270B2 (en) 2007-04-25
MD1418G2 (en) 2000-10-31
EP0683762B1 (en) 1998-12-23
US5523490A (en) 1996-06-04
US5439936A (en) 1995-08-08
ES2125441T3 (en) 1999-03-01
CN1046934C (en) 1999-12-01
DK0683762T3 (en) 1999-08-23
MD1418F2 (en) 2000-02-29

Similar Documents

Publication Publication Date Title
CA2155329C (en) Acylfulvene analogues as antitumor agents
CA2530543C (en) Illudin analogs as anti-tumor agents
KR910000854B1 (en) Process for the preparation of aromatic biocidal compounds
US8497299B2 (en) Compositions including quinonoid derivatives of cannabinoids for therapeutic use
EP1454893B1 (en) Illudin analogs useful as antitumor agents
DE60012750T2 (en) NEW XANTHON DERIVATIVES, THEIR MANUFACTURE AND THEIR USE AS DRUGS
CA2518748A1 (en) Antimicrobial agents and anticancer agents
AU2003297671A1 (en) Diterpenoid compounds, compositions thereof and their use as anti-cancer or anti-fungal agents
Prescott et al. Long-chain thiosemicarbazones as potential anticancer and antiviral agents
US20030229146A1 (en) Cyclopentenone derivatives for cancer therapy
KR810001489B1 (en) Process for preparing oxazolidinedione derivatives of vinca alkaloids
US3934024A (en) Method of producing analgesia and compositions useful therein
Karekezi Aspects of the biological activity of the schistosomicide oxamniquine

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed