CA2067365C - Illudin analogs as anti-tumor agents - Google Patents

Illudin analogs as anti-tumor agents Download PDF

Info

Publication number
CA2067365C
CA2067365C CA002067365A CA2067365A CA2067365C CA 2067365 C CA2067365 C CA 2067365C CA 002067365 A CA002067365 A CA 002067365A CA 2067365 A CA2067365 A CA 2067365A CA 2067365 C CA2067365 C CA 2067365C
Authority
CA
Canada
Prior art keywords
illudin
analog
alkyl
group
tumor
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 - Lifetime
Application number
CA002067365A
Other languages
French (fr)
Other versions
CA2067365A1 (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
Priority to CA002530543A priority Critical patent/CA2530543C/en
Publication of CA2067365A1 publication Critical patent/CA2067365A1/en
Application granted granted Critical
Publication of CA2067365C publication Critical patent/CA2067365C/en
Anticipated expiration legal-status Critical
Expired - Lifetime 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
    • 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 illudin S or illudin M analog having structure (I) or (II), wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R1 is an alkyl or hydrogen; R2 is an alkyl; and R3 is an alcohol or ester.

Description

~1'O 91/(1-i7,-i PC.'T'/l'S40/Ui61-t . : .
ILLUDIN ANALOGS AS APlt'ITUMOR AGENT'S ~ ~ ' I ' ~ O O
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 s 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 illudins known to exist. Illudins have a chemical structure entirely different from other chemotherapeutic agents.
Illudin compounds were previously purified and submitted l0 for evaluation to the National Cancer Institute Division of Cancer Treatment (NCI DCT) i~r v v 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 j"p vivo treatment. The subject invention satisfies this need and provides related advantages.
1~ 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 H analog having the structure ~~ or R
wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein wo ~no.s~sa pcric:snoiosb~a y ~: ,y 3 2~~7~~J~ I, R~ is an alkyl or hydrogen5 ' RZ is an alkyl ; and R3 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 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 B 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 B cells.

WO 91/0475-t fCf/~'S9(1/OSG1=~
2os7~s~ ~'~' ' ' 4 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.
DETAILED DESCRIPTION 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 O
RI
"v .
o wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein .
R~ is an alkyl or hydrogen;
RZ is an alkyl: and R3 is an alcohol or ester.
The analog can be any compound having the stated structure.
Two examples of the effective analogs are:

O O
tst ii0~~
HOt» .
O O
A method of inhibiting tumor cell growth in a subject 4 PCT/C,'594/05614 ,.
,. 5 '~~~' ''2r0-~73fi~
is also provided comprising contacting the tumor with a therapeutic amount of an illudin S or illudin M analog having the structure wherein the analog is capable of inhibiting tumor cell growth without excessive toxicity to the subject and wherein R~ is an alkyl, alkoxyl or hydrogen;
RZ is an alkyl; and R3 is an alcohol or ester.
The analog may be any compound having the stated structure.
Two examples of effective anlogs are:
vN~
HO'~~
O
By "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 v'vo as well as on human tumor cell lines in vitro.

WO 91/0-l7s:i fCt/l.'S9U/U561~

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 still maintaining a high therapeutic index.
The various R groups recited define areas which do not affect the nucleophile reactivity of the 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 structure which is attached to the alkyl group, i.e. an alkylfulvene.
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 over illudin S and M. A
therapeutic amount between 30 to 1000 ~g per kg of body weight has been found especially effective for intravenous administration while 300 to 60,000 or 1,200,000 ~Cg per kg of body weight is effective if administered intraperitoneally. As one skilled in the art would «

rp c~f/pa7;~ fCf/L'S90/05614 ~, ;., ~ 2067365 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.
Also disclosed is a compound having the structure ' o R
~ RZ
R.3 wherein R = methyl or hydroxyls and R~, RZ and R3 = methyl or alkyl.
Also disclosed is a compound having the structure R~ o / Rz 5. fCT/l.'S9U/05614 . , R = H or methyl ; and R~ RZ and R3 = methyl or alkyl . .
EXAMPLE I
Synthesis of Dehydroilludin 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.
EXAMPLE II
Synthesis of Fulvene Illudin S (50 mg) was dissolved in water (2 mL) and 3N
hydrochloric acid (2 mL) added to the solution. The resulting solLtion 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 mL). The yellow chloroform solution was dried (MgS04) 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 W'O 91/Oa75a pcriusyo/osb t a ~;,:r:_ 9 . .. ~~ 2os73s~
these compounds. , Alternatively, a total synthesis of the fulvene can f also be achieved in the following way:
O , O
\O _ O
0.~~ ~~
to (THP. tetrahydropyrany~
OH
OH OH
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 followed by oxidation of the secondary hydroxyl, gives the diolketone. Selective elimination of a tertiary hydroxyl group gives the desired fulvene.
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 flludins for 2 hours, pulsed with [3H]-thymidine for one to two hours and harvested onto glass filters. The filter papers were ~

1'O 9!/O:175a fCT/~'S90/05614 673~i5 1~
20 .
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) .
Table 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).
I C5o ( nM/ L) Compound Illudin S , Dehydroilludin M
HL60, myeloid 7 1 246 19 8392, B-cell 236 t 31 > 38,000 8402, T-cell 669 196 > 38,000 242, melanoma 607 70 > 38,000 547, ovarian 607 110 > 38,000 SL-2, murine (thymic) 142 15 5,235 t 277' MCF-7, breast 58 t 5 653 65 MDA-231, breast 2.0 0.2 112 t 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 w( ~) s ~n-~W-~ PCt/t S9o/0>6 t.~
i;' 11 :;~~ ~ ~;-. , i confers relative non-cross resistance ' 'in ~~~1~~~~g ' resistant hematopoietic cell lines. The derivative of illudins, dehydroilludin M, is slightly less potent 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, marine 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, 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).
Table 2: Sensitivity of Different Mdr Lines to Illudin S

MDR cell line Illudin S Deh vdroilludin available M

CEM Variants Parent 8.3 2.6 nt VM-1 16.2 t 6.4 nt AraC 14 nt VLB100(gp170+)3.7 0.7 nt Dox (gp170+) 14 MDA-231(Breast)Parent 0.85 0.23 54 7 3-1(gp170+) 0.89 0.38 58 11 MCF7-wt(Breast)Parent 0.88 0.11 92 15 t ADR (GSH- 3.7 0.4 68 15 transferase) HL-60 Parent 3.1 1.1 163 11 ADR (gp150+) 1.9 0.8 191 44 KB variant Parent 0.58 0.12 125 14 C-1 (gp170+) 0.69 0.15 80 ?8 VBL(gp170+) 0.69 0.11 78 19 L1210 Parent 0.42 t 0.08 62 8 DDPt(cis-plat)0.46 0.12 119 t 39 BCNU 0.58 0.08 100 31 PAM(melphalan)0.62 ~ 0.15 73 31 CPA(cyclophos)0.46 0.12 38 15 WO 91/0475:1 PCf/t;S9()/OSbi-i 2os73s~ ~2 Table 3: 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 Table 4: Effect of Illudins on Colony Formation Zone Size Compound Dilution L1210 CFU-GM C1498 ~~AML) 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 B), and the 7-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, w0 91 /0-t75-~ , . ~, , PCT/L'S90/()5614 ., :,, ,:.- .
13 ~2os~~s~
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).
Table 5: ICSO for Various Illudin Derivatives Versus Other Agents in HL-60 cells COMPOUNDS nM

Illudin S or M 10 Dihydroilludin S or M 100,000 Acylfulvene 500 Dehydroilludin M (diketone) 46 Isoilludin M 3,800 Ptaquiloside 7,700 Pterosin C 12,500 2,5,6,7-tetramethylindenone 475 Illudin tosylate 38 DNA polymerase inhibitor: Aphidocolin 2,100 Alkylating agent: BCNU 23,300 Crosslinking agent: cis-platinum 17,000 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 tcs a phenol by attack of a second nucleophile, wo <> > ~o:~~>~ ~ 0 6 7 3 6 5 E~cri cls~oios~ ~~
- -. 14 i.e., water. Thus, under acidic conditions, illudin M is clearly bifunctional.
The above studies indicate that the toxicity of -illudins 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 effects of the cyclopropane group (site A, Figure 2), the two double bonds (conjugated diene) (site B), and the 7 ketone (site C) towards electron resonance (or, delocalization) in the illudin molecule. It was hypothesized that oxidation of the secondary 3'-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 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 i.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 10-3 min -~, tl/2 = 148 minutes). Dehydroilludin M also demonstrated first-order kinetics but the reaction was considerably slower (k = 2 x 104 mina tl/2 = 2765 min) . In the reaction wfth dehydroilludin M, no intermediate could be detected by tv0 91 /075.1 PC?/I:SgO/05614 15 , , 207365 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 spontaneously reacts with illudin M, illudin S, or dehydoilludin M, producing products analogous to that 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 glutathione content. This daughter line showed a 4.2 fold decrease in sensitivity to illudin S (parent ICSo 0.88 nmoles/1; daughter line 3.70 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 inhibiticn of enzyme activity. These findings show that illudin toxicity is inversely correlated with intracellular glutathione content but not with glutathione transferase activity.

EXAMPLE 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 Balb/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.
Animals were monitored for Molt-4 tumor development and when tumors were palpable (approximately 4 x 4 nm 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 ~g/kg illudin S, 30 ~g/kg, or 30 ~g/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 ~cg/kg (the compound was subsequently found to be nontoxic to mice at 1000 ug/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 lice, these experiments were repeated. In these studies there was ~VO 91 /(l.l?;-t PC'f/L590/0~614 ;
,.~s.;
.2~~~73~~
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 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 Brightman (NCI). These cells termed HL-60 MRI
cells, were confirmed to have energy-dependent uptake pump, a not unexpected finding as their parental cells posses the WO 91 /04754 fCT/L'590/05614 ~ 1$
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 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 AML model using C1498 cells and a single bolus of illudin S, 2.5 mg/kg IP, 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 30 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 food/water intake, it is possible to optimize both dosage and treatment schedule.
EXAMPLE VII
HL60/MRI Mouse Experiment With Acvlfulvene 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, rather than immediately. This delay in starting treatment is a stringent test to determine Whether a compound is effective. By delaying treatment, the tumor cells become firraly eEtablished.

wo <~no~~sa r~cric s o o f~ ~
. ~~~~~s5 .. 19 , ~ . , , .
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. .

vro 9no.~7sa r>crios~oiosma ...<.,, 2051365 2~

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

CONTROL
No Drug 99136 845282 329911080101624123 167475061 Dehydroilludin M

1 mg/kg 11455 883311 2274992 60251772 115073707 IV

3 mg/kg 10140 911309 21271092 28541260 47842303 IV

Acylfulvene 0.3 mg/kg7338 540167 1352520 32041147 95014605 IV

1 mg/kg 5832 582297 964685 23211434 62752865 3 mg/kg 3830 369250 336215 437238 12011501 EXAMPLE VIII
General In Vitro Screenina 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 WO 91 /U.i75-l PCT/l.'S90/O~G 14 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.
EXAMPLE IX
Assessment of Illudin Uptake in Tumor Cells ~n7hile 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 V'f> 91/(~.t7;.t PCt~/U590/OS614 ~r ,~os73s5 . , 22 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 E
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 ~M. 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.
Cold (4°C), 1% azide, and the metabolic blockers 2-deoxyglucose and antimycin A, all block uptake of illudin S into HL60 cells but have 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 B-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 B
cells are not.

wo 9noa7s4 nc-ri~s9oiosb~a i 23 ~ ~ - 20073fi5 . .

Uptake of (3H] Illudin S by HL60 Myeloid versus 8392 B-cells Maximum uptake per hour (picomoles)' . , Conditions HL60 8392 MCF7 37'C 75 16b 5.5 1.4 29 4 4C 4.3 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 (3H]-labeled illudin S for one hour and harvested as described. Results are expressed as mean ~ SE and represent 3 experiments.
°per 10 million cells eNT = not tested EXAMPLE X
Synthesis and Structure of 2 5 6 7 Tetramethvl-1-Indenone and Dehydropterosin Compounds First 2,4,6-trimethyl-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 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 product.
The 2,5,6,7-tetramethyl-1-indenone was prepared by w0 91/()47;. PCT/L:590/0561-i ~. 2067365 reducing 2,4,5,6-tetramethyl-1,3-indanione with zinc dust , at 50'C. Product was purified by chromatography with 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:
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 O
compound. The compound has the structure:

R = H Dehydropterosin O
R = CH3 Dehydropterosin B
Both compounds were toxic to cells in vitro and have antifungal properties.
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.
... . -a

Claims (28)

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. An illudin S or illudin M analog having the structure wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy.
2. An illudin S or illudin M analog having the structure wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy.
3. The complex of claim 1 wherein the reagent is an antibody.
4. The complex of claim 2 wherein the reagent is an antibody.
5. A use of a therapeutic amount of an illudin S or illudin M analog having the structure:

wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy; for inhibiting tumor cell growth in a subject.
6. A use of a therapeutic amount of an illudin S or illudin M analog having the structure wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy; for the preparation of a medicament for inhibiting tumor cell growth in a subject.
7. A use of a therapeutic amount of an illudin S or illudin M analog having the structure wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy; for inhibiting tumor cell growth in a subject.
8. A use of a therapeutic amount of an illudin S or illudin M analog having the structure:

wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen; and wherein R1 is alkyl, alkoxyl, or hydrogen; R2 is alkyl; and R3 is hydroxy;
for the preparation of a medicament for inhibiting tumor cell growth in a subject.
9. A therapeutic composition comprising 30 to 100 mg of the analog of claim 1 and a pharmaceutically acceptable carrier.
10. A therapeutic composition comprising 30 to 100 mg of the analog of claim 2 and a pharmaceutically acceptable carrier.
11. A method of synthesizing an illudin S or illudin M analog having the following basic structural formula:

wherein R1 is alkyl, alkoxyl, or hydrogen; and R2 is alkyl, comprising (1) reacting 1,1-diacetyl cyclopropane with a dianion of a derivative of cyclopentadiene, wherein the dianion is of the formula:

wherein R1 and R2 are as defined above, and Y is an acid-labile alcohol protecting group, to produce a diol of the formula:

wherein R1, R2 and Y are as defined above;
(2) treating the diol with acid to produce a triol of the formula:

wherein R1 and R2 are as defined above;
(3) oxidizing the secondary alcohol group of the triol to produce a diolketone of the formula:

wherein R1 and R2 are as defined above, and (4) selectively eliminating a tertiary alcohol group on the diolketone to produce the illudin S or illudin M analog.
12. The illudin S or illudin M analog of claim 1, wherein the analog is selected from the group consisting of:

13. An illudin S or illudin M analog selected from the group consisting of:

wherein the analog is attached to a reagent to form a complex which binds to a tumor antigen.
14. The use as defined by claim 5, wherein the therapeutic amount is between to 1,200,000 µg per kg of body weight.
15. The use as defined by claim 7, wherein the therapeutic amount is between to 60,000 pg per kg of body weight.
16. The use as defined by claim 6, wherein the analog is for intravenous, oral, intraperitoneal, or inhalation delivery.
17. The use as defined by claim 16, wherein the therapeutic amount is between to 1,000 pg per kg of body weight.
18. The use as defined by claim 8, wherein the analog is for intravenous, oral, intraperitoneal, or inhalation delivery.
19. The use as defined by claim 18, wherein the therapeutic amount is between to 1,000 pg per kg of body weight.
20. The use as defined by claim 5, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, lung, ovarian and breast carcinoma.
21. The use as defined by claim 5, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, and breast carcinoma.
22. The use as defined by claim 6, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, lung, ovarian and breast carcinoma.
23. The use as defined by claim 6, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, and breast carcinoma.
24. The use as defined by claim 7, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, lung, ovarian and breast carcinoma.
25. The use as defined by claim 7, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, and breast carcinoma.
26. The use as defined by claim 8, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, lung, ovarian and breast carcinoma.
27. The use as defined by claim 8, wherein the tumor cell is selected from the group consisting of myeloid, epidermoid, T-cell leukemia, and breast carcinoma.
28. The method of claim 11, wherein the analog has the formula:

CA002067365A 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents Expired - Lifetime CA2067365C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA002530543A CA2530543C (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US41639589A 1989-10-03 1989-10-03
US416,395 1989-10-03
PCT/US1990/005614 WO1991004754A2 (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA002530543A Division CA2530543C (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents

Publications (2)

Publication Number Publication Date
CA2067365A1 CA2067365A1 (en) 1991-04-04
CA2067365C true CA2067365C (en) 2006-01-31

Family

ID=23649800

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002067365A Expired - Lifetime CA2067365C (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents
CA002530543A Expired - Lifetime CA2530543C (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA002530543A Expired - Lifetime CA2530543C (en) 1989-10-03 1990-10-02 Illudin analogs as anti-tumor agents

Country Status (11)

Country Link
US (1) US5439942A (en)
EP (1) EP0565519B1 (en)
JP (1) JPH05503077A (en)
AT (1) ATE141802T1 (en)
AU (1) AU6729590A (en)
CA (2) CA2067365C (en)
DE (1) DE69028320T2 (en)
DK (1) DK0565519T3 (en)
ES (1) ES2091249T3 (en)
IL (1) IL95875A (en)
WO (1) WO1991004754A2 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439936A (en) * 1989-10-03 1995-08-08 The Regents Of The University Of California Method of treating certain tumors using illudin analogs
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
US7655695B2 (en) 2005-08-03 2010-02-02 The Regents Of The University Of California Illudin analogs useful as anticancer agents
US20080065083A1 (en) * 2006-09-07 2008-03-13 Csaba Truckai Bone treatment systems and methods
US11135182B2 (en) 2014-04-10 2021-10-05 Af Chemicals, Llc Affinity medicant conjugates
US10285955B2 (en) 2014-04-10 2019-05-14 Af Chemicals, Llc Affinity medicant conjugate
CA2944699C (en) 2014-04-10 2023-05-23 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
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
US11160807B1 (en) 2018-12-11 2021-11-02 Af Chemicals, Llc Methods, compositions and devices for treating cancer with illudofulvenes
US11591295B2 (en) 2019-11-25 2023-02-28 Af Chemicals Llc Affinity illudofulvene conjugates

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS62234040A (en) * 1986-04-02 1987-10-14 Kyowa Hakko Kogyo Co Ltd Substance dc1043 and production thereof

Also Published As

Publication number Publication date
CA2530543C (en) 2008-04-01
ATE141802T1 (en) 1996-09-15
US5439942A (en) 1995-08-08
IL95875A (en) 1994-10-21
JPH05503077A (en) 1993-05-27
CA2067365A1 (en) 1991-04-04
DK0565519T3 (en) 1996-09-23
ES2091249T3 (en) 1996-11-01
WO1991004754A3 (en) 1991-12-12
CA2530543A1 (en) 1991-04-18
EP0565519B1 (en) 1996-08-28
DE69028320T2 (en) 1997-03-06
AU6729590A (en) 1991-04-28
DE69028320D1 (en) 1996-10-02
IL95875A0 (en) 1991-07-18
EP0565519A1 (en) 1993-10-20
WO1991004754A2 (en) 1991-04-18

Similar Documents

Publication Publication Date Title
AU676889B2 (en) Acylfulvene analogues as antitumor agents
CA2067365C (en) Illudin analogs as anti-tumor agents
EP0735880A1 (en) Use of vanadium molybdenum or tungsten complexes for controlling cellular proliferation
JP4464679B2 (en) Novel dammaran sapogenin, method for its use as anticancer agent and method for its production
US20030078293A1 (en) Use of etodolac for the treatment of chronic lymphocytic leukemia
EP1454893B1 (en) Illudin analogs useful as antitumor agents
DE69907419T2 (en) ANTITUMOR SUBSTANCES
Kondo et al. Selective inhibition of T-cell-dependent immune responses by bisbenzylisoquinoline alkaloids in vivo
US6359000B1 (en) Anticancer agent
JPH0296523A (en) Platinum chemical remedy product
EP3988521A1 (en) Cellular senescence-activating compounds
US5414015A (en) Anti-skin tumor promoting composition
Jacquillat et al. Clinical Activity of Detorubicin: A New Anthracycline Derivative¹
JPS58144317A (en) Carcinostatic agent
Kudinova et al. Effect of the synthetic resorcinol derivative of caryophyllene on cholesterol biosynthesis and the functional activity of immune system cells
WO1992000740A2 (en) Bis-dioxopiperazines and their use as protection agents
JPH0840914A (en) Oncocyte proliferation inhibitor
JPH04505020A (en) Use of 1,2-benzopyrone derivatives to treat human malignant tumors
Gridley et al. Effects of radiolabelled monoclonal antibody infusion on blood leukocytes in cancer patients
JPH08291064A (en) Cytonecrotic agent for cancer
NZ753549A (en) Combination comprising benzoheterocyclic compound and androgen receptor pathway modulator, application thereof and treatment method
NZ753549B2 (en) Combination comprising benzoheterocyclic compound and androgen receptor pathway modulator, application thereof and treatment method
US20110306662A1 (en) Steroid-Derived Cyclopamine Analogs and Methods for Using the Same in the Prevention or Treatment of Cancer

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry