WO2009076212A1 - Inhibiteurs catalytiques de la topoisomérase ii - Google Patents

Inhibiteurs catalytiques de la topoisomérase ii Download PDF

Info

Publication number
WO2009076212A1
WO2009076212A1 PCT/US2008/085697 US2008085697W WO2009076212A1 WO 2009076212 A1 WO2009076212 A1 WO 2009076212A1 US 2008085697 W US2008085697 W US 2008085697W WO 2009076212 A1 WO2009076212 A1 WO 2009076212A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
pyrrolidinyl
optionally substituted
morpholino
hydrogen
Prior art date
Application number
PCT/US2008/085697
Other languages
English (en)
Inventor
David M. Ferguson
John R. Goodell
Hiroshi Hiasa
Joel W. Slaton
Daniel D. Billadeau
Original Assignee
Regents Of The University Of Minnesota
Mayo Foundation For Medical Education And Research
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 Regents Of The University Of Minnesota, Mayo Foundation For Medical Education And Research filed Critical Regents Of The University Of Minnesota
Publication of WO2009076212A1 publication Critical patent/WO2009076212A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Topoisomerase (topo) inhibitors have found widespread applications in treating various forms of cancer. Most topoisomerase II anticancer drugs are referred to as poisons because they function by trapping the enzyme-DNA cleavable complex, terminating DNA replication. Well known poisons include doxorubicin, etoposide, mitoxantrone, and amsacrine. While these compounds have proven their utility in the war on cancer, they are highly toxic and mutagenic, greatly limiting their use.
  • a series of acridine-based anti-herpes agents with topoisomerase activity was disclosed by Goodell et al. (Goodell, J. R.; Madhok, A. A.; Hiasa, H.; Ferguson, D. M. Synthesis and evaluation of acridine- and acridone-based anti-herpes agents with topoisomerase activity. Bioorg. Med. Chem. 2006, 14, 5467-80).
  • these antiviral acridines may also be useful as anti-cancer agents.
  • anticancer agents that act by unique mechanisms, have higher selectivity for their target, and/or that produce less severe side-effects following administration.
  • anticancer agents particularly topoisomerase II inhibitors, that have enhanced metabolic stability, prolonged half-lives or improved oral bioavailability in animals.
  • topoisomerase II inhibitors that can cross the blood brain barrier or distribute within the central nervous system including the brain.
  • the invention provides compounds that intercalate into DNA and inhibit the catalytic activity of hTopo II.
  • Representative compounds of formula I have been shown to block the formation of etoposide-induced covalent hTopo II-DNA complexes, suggesting that the compounds inhibit an early step of the hTopo II catalytic cycle by preventing binding of the enzyme to DNA.
  • the compounds function as catalytic inhibitors. Unlike poisons, these catalytic inhibitors do not induce DNA cleavage and fragmentation and hence show lower toxicities.
  • Representative compounds of formula I were also found to be selective inhibitors of topoisomerase II and exhibit little or no effect on topoisomerase I activity.
  • the invention provides a method to induce apoptosis in a cell comprising contacting the cell with a compound of formula I:
  • R 4 is -NR c R d , or -OR 6 ;
  • R a and R b are each independently hydrogen or (C 1 -C 6 )alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (Ci-
  • Rc is hydrogen or (Ci-C 6 )alkyl
  • R 4 is -NR c R d , or -OR 5 ;
  • R 3 and R b are each independently hydrogen or (d-C 6 )alkyl, or R 3 and Rb together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R c is hydrogen or (Q-C ⁇ alkyl;
  • R 4 is -NR 0 Ra, or -OR 6 ;
  • R a and R b are each independently hydrogen or (Ci-C 6 )alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R 4 is -NR c R d , or -OR 6 ;
  • R a and R b are each independently hydrogen or (d-C 6 )alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R 4 is -NR 0 R d , or -OR 8 ;
  • R a and R b are each independently hydrogen or (Ci-C 6 )alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R c is hydrogen or (C 1 -C 6 )alkyl
  • R a and R b are each independently hydrogen or (C]-C 6 )alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R 4 is -NR 0 R d , or -OR 6 ;
  • R 3 and R b are each independently hydrogen or (CrC 6 )alkyl, or R 3 and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 -
  • R c is hydrogen or (CrC 6 )alkyl
  • C 6 cycloalkyl(C r C 6 )alkyl, (CrC 6 )alkoxy, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C r C 6 )alkanoyl, halo(C r C 6 )alkyl, hydroxy(C r C 6 )alkyl, (C r C 6 )alkoxycarbonyl, (C 1 -C 6 )alkylthio, aryl(C r
  • R 4 is -NRcR d , or -OR 6 ;
  • R a and R b are each independently hydrogen or (d-C 6 )alkyl, or R 3 and R b together with the nitrogen to which they are attached form a piperidine, thiomorphorine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (Ci- C 6 )alkyl;
  • R 4 is -NR 0 R d , or -OR 8 ;
  • R 3 and R b are each independently hydrogen or or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R c is hydrogen or (d-C 6 )alkyl; and
  • R d is hydrogen, piperidinyl, pyrrolidinyl, piperazinyl, heteroaryl, or (C !
  • R 4 is -NRcR d , or -OR 6 ;
  • R 3 and R b are each independently hydrogen or (C t -C ⁇ alkyl, or R a and R b together with the nitrogen to which they are attached form a piperidine, thiomorpholine, pyrrolidine, morpholino, or a piperazine ring that is optionally substituted at the 4-position nitrogen with (C 1 - C 6 )alkyl;
  • R c is hydrogen or (CrC 6 )alkyl
  • representative compounds of the invention are capable of crossing the blood brain barrier. No other current topo drugs possess this beneficial property. Accordingly, such compounds may be particularly useful for treating certain specific cancers (e.g. recurrent pediatric cancers, cancers in the central nervous system).
  • certain embodiments of the invention provide methods, uses, and compositions comprising compounds of formula I, and salts thereof, for inducing apoptosis of a cell, inhibiting topoisomerase II in a cell, and/or treating cancer, in the central nervous system (e.g., the brain) of a mammal (e.g., a human in need of such treatment), by administering the compound of formula I, or a salt thereof, to the mammal to a location outside the central nervous system of the mammal.
  • the central nervous system e.g., the brain
  • a mammal e.g., a human in need of such treatment
  • the invention also provides processes and intermediates disclosed herein that are useful for preparing compounds of formula (I) or salts thereof.
  • Representative compounds of formula I have been shown to inhibit pancreatic cancer cell growth, ultimately leading to apoptosis. The compounds have also been found to inhibit pancreatic cancer cell proliferation in mouse xenografts. Representative compounds of formula I were also found to inhibit human cancer growth in vitro including the cancer cell lines DU- 145 (Prostate), HCT-116 (colon), Hepa-lclc7 (liver), H460 (non-small lung), MCF-7 (breast), SU86.86 (Pancreatic), MEL (melanoma), OCL-3 (ovarian), and REH (kidney).
  • FIG. 1 illustrates the induction of apoptosis and PARP cleavage following treatment with compounds 1-4.
  • FIG. 2 illustrates in vivo inhibition of pancreatic tumor cell proliferation. Representative sections from SU86.86 xenografts treated with either diluent, compound 2 or 3 and stained for (A) BrdU or (B) Ki-67. One hundred tumor cells were counted per microscopic field. The percentage of positively stained cells from 5 randomly selected fields is graphically depicted.
  • FIG. 3 illustrates the results of the topoisomerase I assay from Example 7.
  • halo is fluoro, chloro, bromo, or iodo.
  • Alkyl, alkoxy, etc. denote both straight and branched groups; but reference to an individual radical such as propyl embraces only the straight chain radical, a branched chain isomer such as isopropyl being specifically referred to.
  • Aryl denotes a phenyl radical or an ortho-fused bicyclic carbocyclic radical having about nine to ten ring atoms in which at least one ring is aromatic.
  • Heteroaryl encompasses a radical of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X) wherein X is absent or is H, O, (C 1 - C 4 )alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms comprising one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X).
  • (Ci-C 6 )alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec- butyl, pentyl, 3-pentyl, or hexyl;
  • (C 3 -C 6 )cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
  • (C 3 -C 6 )cycloalkyl(CrC 6 )alkyl can be cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethyl, or 2-cyclohexylethyl;
  • (Ci-C 6 )alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, is
  • a specific value for R 3 and R b independently is (d-C 6 )alkyl.
  • a specific value for R a and for R b is ethyl.
  • R a and R b together with the nitrogen to which they are attached is a morpholino ring.
  • a specific value for R a and R b together with the nitrogen to which they are attached is a piperazine ring that is optionally substituted at the 4-position nitrogen with (d-C 6 )alkyl.
  • a specific value for R a and R b together with the nitrogen to which they are attached is a
  • R 4 A specific value for R 4 is -NR 0 R 11 .
  • R 0 is hydrogen or (d-C 6 )alkyl
  • R 4 is -NH 2
  • R 0 is hydrogen or (CrC 6 )alkyl
  • R c is hydrogen and for R d is piperidinyl, that is substituted with arylCCrCe ⁇ lkyl.
  • R 4 is l-benzylpiperidin-4-ylamino.
  • R ⁇ is hydrogen and for R d is (C r C 6 )alkyl that is substituted with one or more heteroaryl.
  • a specific value for R 0 is hydrogen and for Ra is (CrC 6 )alkyl that is substituted with an indolyl ring.
  • a specific value for R 4 is N-(2-indol-3-ylethyl)amino.
  • R 4 A specific value for R 4 is -OR e .
  • R e is phenyl
  • a specific value for each of R f and R g is hydrogen.
  • the cancer is pancreatic cancer, prostate cancer, breast cancer, ovarian cancer, lung cancer, colon cancer, a leukemia, liver cancer, melanoma, brain cancer or kidney cancer,
  • the cancer is pancreatic cancer, prostate cancer, breast cancer, ovarian cancer, lung cancer, colon cancer, or a leukemia.
  • the cancer is liver cancer, melanoma, kidney cancer, non-small lung cancer or non-refractory prostate cancer.
  • the cancer is liver cancer, melanoma, or kidney cancer.
  • the cancer is non-small lung cancer or non-refractory prostate cancer.
  • the cancer is refractory prostate cancer. In another embodiment the cancer is non-refractory prostate cancer.
  • the cancer is a cancer of the central nervous system (CNS).
  • CNS central nervous system
  • Another intermediate useful for preparing a compound of formula I is a compound of the following formula II
  • a salt of a compound of formula I can be useful as an intermediate for isolating or purifying a compound of formula I. Additionally, administration of a compound of formula I as a pharmaceutically acceptable acid or base salt may be appropriate.
  • Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, ⁇ - ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • the compounds of formula I can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form.
  • the amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • the active compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compounds of formula I to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compounds of formula I can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 1 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day. In one embodiment of the invention the dose is in the range of 1 to 10 mg/kg/day. In another embodiment of the invention the dose is in the range of 10 to 40 mg/kg/day
  • the compound is conveniently formulated in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
  • the invention provides a composition comprising a compound of the invention formulated in such a unit dosage form.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • Compounds of the invention can also be administered in combination with other therapeutic agents, for example, other agents that are useful for the treatment of cancer. Examples of such agents include taxanes, cyclophosphamide, docetaxel, epirubicin, and 5-FU.
  • the invention also provides a composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, and a pharmaceutically acceptable diluent or carrier.
  • the invention also provides a kit comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, packaging material, and instructions for administering the compound of formula I or the pharmaceutically acceptable salt thereof and the other therapeutic agent or agents to an animal to treat cancer.
  • the cancer treatment methods of the invention can also be used in conjunction with radioimmunotherapy, wherein a therapeutic radioisotope (e.g. iodine-131) is administered along with a targeting agent (e.g. a monoclonal antibody) to deliver radiation directly to cancer cells.
  • a therapeutic radioisotope e.g. iodine-131
  • a targeting agent e.g. a monoclonal antibody
  • pancreatic cancer cell lines MiaPaCa-2, SU86.86 and BXPC-3 were obtained from ATCC (Rockville, MD) and were grown in DMEM medium containing 10% fetal calf serum and L- glutamine.
  • MTT assays were carried out over the indicated timecourse in the presence or absence of varying concentrations of compounds as previously described (Ougolkov et al., Cancer Research, 2005, 65, 2076-81).
  • apoptosis assays the indicated cell lines were treated as described in the text, harvested, and nuclei scored for apoptosis as previously described (Ougolkov et al., Cancer Research, 2005, 65, 2076-81). A minimum of 300 nuclei was analyzed per sample.
  • the SU86.86 pancreatic cancer cell line was incubated with diluent or a range of concentrations of compounds 1-4.
  • pancreatic cancer cells were incubated for 36 hours with diluent or the indicated compounds 1 thru 4 (15 ⁇ M final concentration). Samples were then fixed in 50% ethanol, rehydrated, treated with RNase, stained with 50 ⁇ g/ml propidium iodide in 0.1% sodium citrate, and analyzed (at least 20,000 events per sample) using a Becton Dickinson FACScan flow cytometer. Very small debris (forward scatter less than 10% of intact cells) was excluded from the analysis by gating. Cell-cycle profiles were determined using Modfit Software (Verity, Topsham, ME) as previously described (Bible and Kaufmann, Cancer Research, 1997; 57, 3375- 80).
  • Figure 1 shows the results following treatment of BXPC-3 cells with diluent or the indicated concentrations of compounds 1-4. Forty-eight hours post exposure, cells were collected and stained with Hoechst. Three hundred nuclei were counted per field and scored for apoptotic changes (fragmentation of the nucleus into multiple discrete fragments) and are graphically represented (lower Panel). Cell lysates from the same treated cells were probed for
  • PARP cleavage (upper panel), a hallmark of apoptosis.
  • ⁇ -Actin was used as a loading control.
  • Subcutaneous flank xenografts of SU86.86 tumor cells were established in female athymic nude mice to determine the effect of compounds 2 and 3 on tumor cell proliferation.
  • Tumor bearing mice (2 in each group) were injected intraperitoneally (IP) with either 50 ⁇ L of diluent (DMSO) in 500 ⁇ l PBS, or compound 2 (5 mg/kg) or 3 (10 mg/kg) dissolved in 50 ⁇ L of diluent in 500 ⁇ L of PBS.
  • the injections were given twice daily (every 12 hrs) over two days for a total of 4 injections. Three hours prior to sacrifice, mice were injected with 1 mg of BrdU.
  • Tumors were subsequently removed from the mice, fixed in paraformaldehyde and paraffin- embedded. Tumor sections were analyzed for proliferating tumor cells by performing immunohistochemical staining for either BrdU or the nuclear-associated proliferation antigen, Ki-67. Consistent with our in vitro cell-based assays, compared to diluent-treated animals, administration of either compound 2 or compound 3 resulted in a significant loss of BrdU+ and Ki-67+ tumor cells ( Figure 2). These data indicate that compounds 2 and 3 can abrogate tumor cell proliferation in vivo.
  • Compounds 1-4 were evaluated utilizing a MTT assay against nine cancer cell lines in vitro to determine the EC 50 value for cell growth inhibition.
  • the cancer cell lines investigated were DU-145 (Prostate), HCT-116 (colon), Hepa-lclc7 (liver), H460 (non-small lung), MCF-7 (breast), SU86.86 (Pancreatic), MEL (melanoma), OCL-3 (ovarian), and REH (kidney).
  • the following table summarizes the activities of compounds 1-4. Cancer cell growth inhibition against human cancers. a compound DU- 145 HCT-116 Hepa H460 MCF-7 SU86.86 MEL REH OCL-3
  • reaction was terminated by the addition of 1 ⁇ l of 10% (w/v) SDS and 2 ⁇ l of 10 mg/ml proteinase K followed by an additional 15-minute incubation at 37 °C. After extraction with 1 : 1 phenol: chloroform, samples were applied to 1% (w/v) agarose gels in TPE buffer (36 mM Tris, 30 mM NaH 2 PO 4 , and 1 mM EDTA) containing 10 ⁇ g/ml chloroquine. Gels were run at 45 V for 4.5 h, stained with 0.5 ⁇ g/ml ethidium bromide, and photographed under UV light.
  • Example 8 Mouse Xenograft (PC3-LN4 non-refractory prostate tumors)
  • mice were treated with twice daily 10 mg/kg IP injections of compound 2 (dissolved in ethanol) for 7 days. Tumor sizes were approximately 0.5 cm3 at the start of the treatment. Tumors sizes were measured for the 7 day dosing period and 2 days after dosing was completed. Compound 2 treated mice displayed a significant reduction in tumor growth as compared to the control group over the 9 day measuring period.
  • Example 10 The following illustrate representative pharmaceutical dosage forms, containing a compound of formula I ('Compound X 1 ), for therapeutic or prophylactic use in humans.
  • compositions may be obtained by conventional procedures well known in the pharmaceutical art.
  • Representative compounds of formula (I) have been shown to be selective for topoisomerase II over topoisomerase I. Accordingly, these selective inhibitors may possess fewer side effects or less severe side effects than other topoisomerase inhibitors.
  • Compounds of formula (I) also displayed unexpected metabolic stability and unexpectedly long half-lives in vivo. These attributes contributed to an enhanced pharmacokinetic(PK) profile for representative compounds of formula (I). This enhanced PK profile may be a significant factor regarding the in vivo efficacy of these compounds as demonstrated in the mouse xenograft model of Example 5.
  • Representative compounds of formula (I) have also been shown to distribute within the central nervous system (CNS) including the brain.
  • CNS central nervous system
  • the invention includes a method of detecting the location of a tumor in an animal comprising administering a compound of formula (I) to the animal, irradiating the tissue in the animal with an appropriate irradiation source and detecting the resulting emission of the compound by a suitable method of emission detection to ascertain the location of the tumor.
  • the invention also provides a method in which a compound of formula (I) is used to both locate a tumor in an animal and to treat said tumor in the animal.

Abstract

L'invention porte sur des méthodes et des compositions de traitement du cancer, inhibant sélectivement la topoisomérase II, et induisant l'apoptose.
PCT/US2008/085697 2007-12-05 2008-12-05 Inhibiteurs catalytiques de la topoisomérase ii WO2009076212A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US99262107P 2007-12-05 2007-12-05
US60/992,621 2007-12-05

Publications (1)

Publication Number Publication Date
WO2009076212A1 true WO2009076212A1 (fr) 2009-06-18

Family

ID=40365336

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/085697 WO2009076212A1 (fr) 2007-12-05 2008-12-05 Inhibiteurs catalytiques de la topoisomérase ii

Country Status (1)

Country Link
WO (1) WO2009076212A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015085229A1 (fr) * 2013-12-08 2015-06-11 Van Andel Research Institute Inhibiteurs d'autophagie
US10633336B2 (en) 2014-12-19 2020-04-28 The Broad Institute, Inc. Dopamine D2 receptor ligands
US10752588B2 (en) 2014-12-19 2020-08-25 The Broad Institute, Inc. Dopamine D2 receptor ligands

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US20070270455A1 (en) * 2005-07-20 2007-11-22 Gudkov Andrei V INHIBITION OF NF-kB
US20080171765A1 (en) * 2007-01-11 2008-07-17 Academia Sinica Aniline or phenol mustards linked to DNA-affinic molecules or water-soluble aromatic rings and their use as cancer therapeutic agents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US20070270455A1 (en) * 2005-07-20 2007-11-22 Gudkov Andrei V INHIBITION OF NF-kB
US20080171765A1 (en) * 2007-01-11 2008-07-17 Academia Sinica Aniline or phenol mustards linked to DNA-affinic molecules or water-soluble aromatic rings and their use as cancer therapeutic agents

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ATWELL G J ET AL: "POTENTIAL ANTITUMOR AGENTS. 43. SYNTHESIS AND BIOLOGICAL ACTIVITY OF DIBASIC 9-AMINOACRIDINE-4-CARBOXAMIDES, A NEW CLASS OF ANTITUMOR AGENT", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON.; US, vol. 27, no. 11, 1 January 1984 (1984-01-01), pages 1481 - 1485, XP001094330, ISSN: 0022-2623 *
FINLAY G J ET AL: "From amsacrine to DACA (N-[2-(dimethylamino)ethyl]acridine-4-carboxam ide): Selectivity for topoisomerases I and II among acridine derivatives", EUROPEAN JOURNAL OF CANCER PART A 1996 GB, vol. 32, no. 4, 1996, pages 708 - 714, XP007907513, ISSN: 0959-8049 *
GOODELL J R ET AL: "Synthesis and evaluation of acridine- and acridone-based anti-herpes agents with topoisomerase activity", BIOORGANIC AND MEDICINAL CHEMISTRY 20060815 GB, vol. 14, no. 16, 15 August 2006 (2006-08-15), pages 5467 - 5480, XP025133447, ISSN: 0968-0896 *
GOODELL, JOHN R. ET AL: "Acridine-Based Agents with Topoisomerase II Activity Inhibit Pancreatic Cancer Cell Proliferation and Induce Apoptosis", JOURNAL OF MEDICINAL CHEMISTRY , 51(2), 179-182 CODEN: JMCMAR; ISSN: 0022-2623, 2008, XP002517676 *
HE Z ET AL: "DNA threading bis(9-aminoacridine-4-carboxamides): Effects of piperidine sidechains on DNA binding, cytotoxicity and cell cycle arrest", BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER SCIENCE LTD, GB, vol. 16, no. 8, 15 April 2008 (2008-04-15), pages 4390 - 4400, XP022617889, ISSN: 0968-0896, [retrieved on 20080223] *
OPPEGARD, LISA M. ET AL: "Novel acridine -based compounds that exhibit an anti-pancreatic cancer activity are catalytic inhibitors of human topoisomerase II", EUROPEAN JOURNAL OF PHARMACOLOGY , 602(2-3), 223-229 CODEN: EJPHAZ; ISSN: 0014-2999, 2009, XP002517677 *
PASTWA E ET AL: "Cytotoxic and DNA-damaging properties of N-(2-(dimethylamino)ethyl)ac ridine-4-carboxamide (DACA) and Its analogues", BIOCHEMICAL PHARMACOLOGY, PERGAMON, OXFORD, GB, vol. 56, no. 3, 1 August 1998 (1998-08-01), pages 351 - 359, XP007907439, ISSN: 0006-2952 *
WAKELIN L P G ET AL: "Bisintercalating Threading Diacridines: Relationships between DNA Binding, Cytotoxicity, and Cell Cycle Arrest", JOURNAL OF MEDICINAL CHEMISTRY 20031218 US, vol. 46, no. 26, 18 December 2003 (2003-12-18), pages 5790 - 5802, XP007907438, ISSN: 0022-2623 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015085229A1 (fr) * 2013-12-08 2015-06-11 Van Andel Research Institute Inhibiteurs d'autophagie
US10633336B2 (en) 2014-12-19 2020-04-28 The Broad Institute, Inc. Dopamine D2 receptor ligands
US10752588B2 (en) 2014-12-19 2020-08-25 The Broad Institute, Inc. Dopamine D2 receptor ligands
US11498896B2 (en) 2014-12-19 2022-11-15 The Broad Institute, Inc. Dopamine D2 receptor ligands

Similar Documents

Publication Publication Date Title
EP1123099B1 (fr) Nouvelles indeno-isoquinoleines utilisees en tant qu'agents antineoplasiques
US20200405723A1 (en) Wee1 kinase inhibitors and methods of treating cancer using the same
US20110275643A1 (en) Aroylquinoline compounds
EP1951235B1 (fr) Nouvelles formes cristallines de chlorhydrate d'irinotecan
US20080242692A1 (en) Synthesis of Indenoisoquinoliniums and Methods of Use
EP0714387B1 (fr) Nouveaux bis-naphtalimides utilises dans le traitement de cancers
WO2009076212A1 (fr) Inhibiteurs catalytiques de la topoisomérase ii
US5942518A (en) Water-soluble fluoroethylcamptothecin derivative and process for production thereof
Liu et al. Discovery of novel tacrine derivatives as potent antiproliferative agents with CDKs inhibitory property
EP2044080B1 (fr) Dérivés de camptothécine à activité antitumorale
CN114478561B (zh) 一种依帕司他石蒜碱偶联物及其制备方法和用途
RU2561118C2 (ru) Производные камптотецина, обладающие противоопухолевой активностью
US8124769B2 (en) Stereoselective process and crystalline forms of a camptothecin
CN101139292B (zh) 4'-氧代羧酸二苯乙烯烷氧基衍生物
EP0536208A1 (fr) Agents anticancereux a base de 1,2-dihydro-3h-dibenzisoquinoline-1,3-diones
EP1480984B1 (fr) Composes hexacycliques
CZ2000711A3 (cs) Opticky čisté analogy kamptothecinu, meziprodukty syntézy a způsob přípravy
EP1430059B1 (fr) Sels acceptables d'un point de vue pharmaceutique de 20(s)-camptothecines
CN116589405A (zh) 高效抑制自噬、逆转肿瘤多药耐药的异喹啉生物碱衍生物及制备方法和应用
JP2002535276A (ja) 4−デメチルペンクロメジンのアシル誘導体、その使用及び調製
EP0639183B1 (fr) Oxydes d'azote de derives d'aza- et diaza-anthracenedione, leur preparation et leur utilisation comme agents antitumoraux
CN112608327B (zh) 呋喃并喹啉衍生物、其制备方法及应用
KR100825530B1 (ko) (-)-시스-2-(2-클로로페닐)-5,7-디하이드록시-8-[4r-(3s-하이드록시-1-메틸)피페리디닐]-4h-1-벤조피란-4-온의 에탄올 용매화물

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08859941

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08859941

Country of ref document: EP

Kind code of ref document: A1