WO2012081039A1 - Molecules with anticancer activity and uses thereof - Google Patents

Molecules with anticancer activity and uses thereof Download PDF

Info

Publication number
WO2012081039A1
WO2012081039A1 PCT/IN2011/000866 IN2011000866W WO2012081039A1 WO 2012081039 A1 WO2012081039 A1 WO 2012081039A1 IN 2011000866 W IN2011000866 W IN 2011000866W WO 2012081039 A1 WO2012081039 A1 WO 2012081039A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cancer
pharmaceutically acceptable
formula
acceptable salt
Prior art date
Application number
PCT/IN2011/000866
Other languages
French (fr)
Inventor
Martin Annette
Shruti Patil
Nilesh Shridhar Malik
Kailash Dattatraya Panghavane
Original Assignee
Godavari Biorefineries Limited
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 Godavari Biorefineries Limited filed Critical Godavari Biorefineries Limited
Publication of WO2012081039A1 publication Critical patent/WO2012081039A1/en

Links

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/10Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings
    • C07D317/12Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/10Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings
    • C07D317/14Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D317/16Radicals substituted by halogen atoms or nitro radicals

Definitions

  • the present invention relates to molecules with anticancer activity and uses thereof. More particularly, the present invention relates to molecules with anticancer activity and their use for targeting cancer.
  • Chemotherapy is often used to treat patients with metastasized cancer). Chemotherapy destroys the main tumor and cells that travel through the blood or lymph systems to other parts of the body.
  • anticancer drugs used in chemotherapy are known to cause side effects. Most common side effects are myelosuppression (decreased production of blood cells), mucositis (inflammation of the lining of the digestive tract) and alopecia (hair loss).
  • cancer stem cells Besides toxicity, another failure of the known anticancer agents is their inability to kill cancer stem cells. In recent years, it is hypothesized that only a small fraction of the entire tumor mass are responsible for the tumorigenic activities within the tumor. These small fractions of tumorigenic cells, according to the new model, are with stem-cell-like qualities and are called "cancer stem cells". These cancer stem cells were shown to have similarcellular markers, CD34+/CD38 , as primitive hematopoietic stem cells. Since then, researchers have found cancer stem cells conclusively in various types of tumors including those of the brain, breast, skin, prostate, and so on.
  • cancer stem cells give rise both to more tumor stem cells and to the majority of cells in the tumor that have lost the capacity for extensive proliferation and the ability to give rise to new tumors that are slowly becoming resistant to drugs causing relapse. Since these cells having significant self- renewal potential,can cause relapse, to make it possible to treat patients with aggressive, non-resectable tumors, and refractory or recurrent cancers, as well as for preventing tumor metastasisthere is need in the art for newer anticancer agents, the use of which can target and inhibit, reduce, or eliminate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells.
  • the present invention provides agents which show capability for inhibition of proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs) thus offering a new opportunity for the treatment of cancers.
  • CSCs cancer stem cells
  • the present invention provides molecules represented by compounds having the general formula I or pharmaceutically acceptable salt thereof, and pharmaceutically acceptable compositions having ability to inhibit proliferation of and/or eradicate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
  • CSCs cancer stem cells
  • the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful in targeting cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for treating a variety of diseases, disorders or condition.
  • diseases, disorders, or conditions include those described herein.
  • composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, for inhibiting the proliferation of and/or eradicating cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
  • CSCs cancer stem cells
  • the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, or pharmaceutically acceptable compositions thereof in inhibiting the proliferation of and/or eradicating cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
  • CSCs cancer stem cells
  • the invention provides a compound of formula I, or a pharmaceutically acceptable salts thereof or pharmaceutically acceptable compositions thereof for the prophylactic or therapeutic treatment of cancer.
  • the invention provides a method of inhibiting proliferation of or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells in a patient comprising the step of administering to patient a compound of formula I, or a pharmaceutically acceptable salts thereof or pharmaceutically acceptable compositions thereof.
  • the present invention is directed towards molecules represented by compounds of Formula 1 having ability to inhibit proliferation of and/or eradicate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
  • CSCs cancer stem cells
  • the present invention relates to the use of such anticancer molecules that is compounds of Formula 1 in targeting cancer.
  • the present invention also relates to the use of anticancer molecules represented by Formula 1 for inhibition of proliferation and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells.
  • the present invention relates to the use of anticancer molecules represented by Formula 1 for prevention and/or treatment of cancer.
  • the present invention relates to the molecules having ability to inhibit proliferation and/or eradication of cancer cells and/or cancer stem cells.
  • Such molecules are represented by compounds of Formula 1:
  • Ring A is a 4-8 membered partially unsaturated, or an aromatic ring
  • n is 0-4;
  • each R, R', R" and R'" is independently hydrogen or an alkyl, optionally substituted group selected from Cl-6 aliphatic, alkenyl, alkynyl, a 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring, aryl, phenyl, an 8- 10 membered bicyclic aromatic carbocyclic ring; a 4-8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; and
  • X is H, F, CI, Br, or I;
  • Compounds of this invention include those described generally above, and further described herein below.
  • Ring A is a 5-6 membered partially unsaturated, or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, Ring A is a 5-6 membered aromatic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, Ring A is pyrido or thiopheno. In some embodiments, Ring A is benzo. As defined generally above, n is 0-4. In certain embodiments, n is 1-3. In some embodiments, n is 2.
  • each R is independently hydrogen or an optionally substituted group selected from Ci_ 6 aliphatic, phenyl, or a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each R is independently hydrogen or an optionally substituted Ci_6 aliphatic group.
  • each R is independently hydrogen or an optionally substituted Ci. 2 aliphatic group.
  • each R is hydrogen.
  • each R is an optionally substituted C a . 2 aliphatic group.
  • the compounds of the present invention are in the form pharmaceutically acceptable derivative including but not limited to any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • the compounds of the present invention are in the form of pharmaceutically acceptable salts.
  • Such salts are acid or base salts.
  • pharmaceutically acceptable acid 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, a-ketoglutarate, and a- glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts include adipate, alginate, aspartate, benzenesulfonate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci_4alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • salts may be obtained using standard procedures well known in the art, for example by reacting sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • 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.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 1 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
  • a warhead moiety, R 1 of a provided compound comprises one or more deuterium atoms.
  • the present invention provides a compound of formula
  • the present invention provides a compound of formula
  • R 3 is F, CI, Br, or I.
  • the present invention provides any compound selected from those depicted above, or a pharmaceutically acceptable salt.
  • the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for inhibiting proliferation and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for treating cancer in an animal.
  • the present invention further provides pharmaceutical compositions comprising the compounds represented by the Formula 1 for inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for prevention and/or treatment of cancer.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the compounds represented by the Formula 1 or their pharmaceutically acceptable salt or their composition can be formulated into dosage form to be administered to a mammalian host, such as a human patient adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the compound may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • compositions and preparations 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.
  • Such compositions and preparations should contain the active molecule in an amount such that an effective dosage level will be obtained.
  • the percentage of the compositions and preparations may, of course, be varied conveniently by a person skilled in the art.
  • the compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same are formulated into solid dosage forms for oral administration, such dosage form can be selected from but not limiting to tablets, troches, pills, capsules.
  • Pharmaceutically inert excipient that may be incorporated for formulation of such dosage form include 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.
  • the unit dosage form When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like.
  • Liquid dosage forms are another exemplary dosage form for formulating compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same.
  • Such liquid dosage form can be in the form of for example a syrup, emulsions, suspensions, or elixir containing the active molecule, and other pharmaceutically acceptable inert excipients commonly used in the art such as, for example sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor diluents.
  • Other adjuvants include solubilizing agents, wetting agents, emulsifying and suspending agents.
  • the compounds represented by the Formula. 1 or their pharmaceutically acceptable salt or composition comprising the same may also be formulated as pharmaceutical dosage forms to be administered intravenously or intraperitoneal ⁇ by injection or infusion.
  • 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.
  • Solutions of the active molecule 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 molecules represented by the Formula 1 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 molecules represented by the Formula 1 or their pharmaceutically acceptable salt or composition comprising the same may be applied in pure form, i.e., when they are liquids.
  • 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 molecules 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 molecules of Formula I to the skin are known to the art.
  • Ophthalmic formulations may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • transdermal patches which have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • Formulation for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • the compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same may also be formulated for nasal administration through aerosol or inhalation.
  • compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food.
  • the amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • Compounds and compositions described herein are generally useful for the inhibition of proliferation and/or eradication of cancer cells and/or cancer stem cells. Thus, provided compounds are useful for prevention and/or treatment of cancers, including, but not limited to hematological cancers and solid tumors.
  • the invention in one embodiment relates to the use of compounds represented by the Formula 1 for inhibition of proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells.
  • the use of compounds represented by the Formula 1 for inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells having significant self-renewal potential, such as cancer stem cells include stem cells within tumors or leukemic stem cells.
  • the invention relates to a method of inhibiting cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells in a patient, leading to remission of the cancer, comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the present invention provides a method for treating a disorder mediated by cancer cells cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof.
  • a disorder mediated by cancer cells cancer cells and/or cancer cells having significant self-renewal potential such as cancer stem cells
  • cancer stem cells for prevention and/or treatment of cancer
  • cancer of at least one type selected from the group consisting of cancer is selected from breast, prostate, ovary, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, or stomach.
  • Exemplary cancers include acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, soft-tissue sarcoma, squamous cell carcinomas, colon carcinoma, an adenocarcinoma, a sweat gland carcinoma, a sebaceous gland carcinoma, a papillary carcinoma, a papillary adenocarcinoma, a cystadenocarcinoma, a medullary carcinoma, a bronchogenic carcinoma, a renal cell carcinoma, a hepatocellular carcinoma, a bile duct carcinoma, a choriocarcinoma, a seminoma, an embryonal carcinoma, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervix cancer, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, Ewing's sarcoma, gallbladder cancer,
  • the invention relates to a method of inhibiting cancer cells cancer cells and/or cancer cells having significant self- renewal potential, such as cancer stem cells, " in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • cancer stem cells cancer cells and/or cancer cells having significant self-renewal potential such as cancer stem cells in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to biological assays, gene expression studies, and biological target identification.
  • Some of the breast, prostrate and ovarian cell lines selected include but not limited to DU145, PC3, LNCaP, MDA MB 231, MCF7, T47D, MCF10A, HeLa, or L929.
  • the activity of a compound utilized in this invention as an inhibitor of of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells may be assayed in vitro or in vivo.
  • An in vivo assessment of the inhibitory or cytotoxic activity of the compounds of the invention may be made using an animal model of cancer, e.g., a rodent or primate model.
  • Cell-based assays may be performed using, e.g., a cell line isolated from a tumor or blood- borne cancer.
  • Cell-based assays for activity against a specific protein or nucleic acid component of a cancer cell line e.g., an enzyme, structural protein, DNA or RNA, may also be performed.
  • biochemical or mechanism-based assays e.g., transcription assays using a purified protein, Northern blot, RT-PCR, etc., may be performed.
  • In vitro assays include assays that determine cell morphology, viability, cell count, or growth inhibition, and/or the cytotoxicity, enzyme inhibitory activity, and/or the subsequent functional consequences of treatment of cancer cells with compounds of the invention. Alternate in vitro assays quantitate the ability of the inhibitor to bind to protein or nucleic acid molecules within the cell. Inhibitor binding may be measured by radiolabelling the inhibitor prior to binding, isolating the inhibitor/target molecule complex and determining the amount of radiolabel bound. Alternatively, inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with purified proteins or nucleic acids bound to known radioligands.
  • chemotherapeutic agents include, but are not limited to agents such as kinase inhibitors, alkylating agents, antimetabolites, tubulin stabilizers, tubulin assembly inhibitors, DNA replication inhibitors, cell cycle inhibitors, topoisomerase inhibitors, cytotoxic antibiotics or nanoparticle or protein conjugates of any of the aforementioned agents.
  • the chemotherapeutic agents used in combination with compounds or compositions of the invention include, but are not limited to imatinib, nilotinib, gefitinib, sunitinib, carfilzomib, salinosporamide A, retinoic acid, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide, azathioprine, mercaptopurine, doxifluridine, fluorouracil, gemcitabine, methotrexate, tioguanine, vincristine, vinblastine, vinorelbine, vindesine, podophyllotoxin, etoposide, teniposide, tafluposide, paditaxel, docetaxel, irinotecan, topotecan, amsacrine, actinomycin, doxorubicin,
  • a combination of 2 or more chemotherapeutic agents may be administered together with compounds of the invention.
  • a combination of 3 or more chemotherapeutic agents may be administered with compounds of the invention.
  • the chemotherapeutic agents are selected from alkylating agents or antimetabolites.
  • the chemotherapeutic agent is cisplatin.
  • agents the inhibitors of this invention may also be combined with include, without limitation: vitamins and nutritional supplements, cancer vaccines, treatments for neutropenia (e.g. G-CSF, filgrastim, lenograstim), treatments for thrombocytopenia (e.g. blood transfusion, erythropoietin), antiemetics (e.g.
  • 5-HT 3 receptor antagonists dopamine antagonists, NK1 receptor antagonists, histamine receptor antagonists, cannabinoids, benzodiazepines, or anticholinergics
  • Alzheimer's Disease such as Aricept * and Excelon ®
  • Parkinson's Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine
  • agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex ® and Rebif ® ), Copaxone ® , and mitoxantrone
  • treatments for asthma such as albuterol and Singulair 8
  • agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol
  • antiinflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclo
  • compounds of the present invention are administered in combination with antisense agents, a monoclonal or polyclonal antibody or an siRNA therapeutic.
  • the compounds may also be used in combination with other drugs that are used in the treatment/prevention/suppression or amelioration of the diseases or conditions for which compounds as represent by Formula 1 are useful.
  • Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention.
  • Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another, normally within five hours from one another.
  • a pharmaceutical composition containing such other drugs in addition to the compound of the present invention is preferred.
  • compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of the present invention.
  • the compounds represented by the Formula 1 either alone or in combination with other active or supplementary agents may be used for inhibition of proliferation and/or eradication of cancer cells and/or cancer stem cells for targeting cancer as a standalone therapy or may be used either before or after or in conjunction with other therapies such as radiation, surgery or any other method for prevention and/or treatment of cancer.
  • compositions administered will be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • amount of both, an inventive compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • Dosage amount of the compounds represented by the Formula 1, other active agents and interval may be adjusted individually to provide plasma levels of the active species.
  • HPLC assays or bioassays can be used to determine plasma concentrations.
  • the therapeutically effective dose of molecules is an amount sufficient to cause cell death or inhibit proliferation and cause differentiation of stem cells in solid tumors or leukemias.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive can be administered to a patient receiving these compositions.
  • the compounds of the invention for oral or parenteral administration may be at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • the use of compounds represented by the Formula 1 for remission of cancer is a newer method for treatment of cancer which involves the inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells which would reduce the likelihood of clinical relapse after treatment.
  • Example 1 In certain embodiments, compounds of formula I where are prepared according to the procedure outlined in Scheme 1.
  • compounds and synthetic intermediates thereof of the present invention may be prepared according to methods known to one of ordinary skill in the art.
  • compounds of the present invention are assayed as inhibitors of CSCs using methods known in the art including an in vitro cell death assay, an in vitro soft agar growth inhibition assay, an in vitro 3D sphere forming stem cell assay, and in vivo toxicity assays in animals.
  • In- vitro colorimetric cell death assay An exemplary procedure for the in vitro colorimetric cell death assay follows. Cancer cells were plated in a 96 well plate as per predetermined plating efficiency, the plate was incubated for 24 hours in a 5% C0 2 atmosphere at 37 degrees Celsius, a range of concentrations of the compound of the present invention of formula 1 was added to the wells, the plates were incubated further for 48 hours in a 5% C0 2 atmosphere, the plate was centrifuged twice at 3000 rpm for 3 minutes, the supernatant fluid was discarded, 100 uL of 0.5mg/mL MTT solution was added, the plate was incubated for 4 hours in a 5% C0 2 atmosphere at 37 degrees Celsius, the plate was centrifuged twice at 3000 rpm for 3 minutes, supernatant was aspirated very carefully, 200 uL DMSO was added to each well to solubilize MTT crystals and mixed well by shaking the plate, the plate was incubated for 10 minutes in a
  • Cells were grown in two dimensions on a plastic substrate, harvested in suspension in serum-free media, then the cells in the sample were trypsinized and a single cell suspension was formed by passing through a cell strainer.
  • the cells were diluted according to the predetermined plating efficiency for the cell line being studied by suspending the cells in stem cell culture medium. 100 uL of this suspension was added into each well of a 96 well suspension plate, and the plate was incubated at 37 degrees Celsius in 5% C0 2 atmosphere for 24 hours, then 2 uL of appropriate concentrations of the drug were added into each respective well along with 100 uL of stem cell culture medium, and the plates incubated at 37 degrees Celsius under 5% C0 2 atmosphere for 72 hours.
  • Results of the in vitro 3D sphere forming stem cell assay for compound of formula 1-1 are set forth in Table 2.
  • the number in each box is the total number of spheres formed in the presence of compound of formula 1-1 at each drug concentration.
  • GC refers to a growth control performed in the absence of drug or solvent (DMSO).
  • GCEA refers to a growth control performed in the absence of drug or solvent ethyl acetate.
  • Compound of formula 1-1 showed significant sphere reduction. It was found to be the most effective compound on breast, prostrate and ovarian cancer cell lines.

Abstract

The present invention relates to molecules represented by compounds having the general formula I or pharmaceutically acceptable salt thereof, and pharmaceutically acceptable compositions for inhibiting proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs). Such compounds have the general formula (I): The invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, or pharmaceutically acceptable compositions thereof for inhibiton the proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs) for remission of cancer.

Description

Title
Molecules with anticancer activity and uses thereof, Field of invention
The present invention relates to molecules with anticancer activity and uses thereof. More particularly, the present invention relates to molecules with anticancer activity and their use for targeting cancer.
Background of the Invention
The main purpose of chemotherapy is to kill cancer cells. . Chemotherapy is often used to treat patients with metastasized cancer). Chemotherapy destroys the main tumor and cells that travel through the blood or lymph systems to other parts of the body.
Although chemotherapy is an important therapeutic strategy for cancer treatment, anticancer drugs used in chemotherapy are known to cause side effects. Most common side effects are myelosuppression (decreased production of blood cells), mucositis (inflammation of the lining of the digestive tract) and alopecia (hair loss).
Thus, for cancer patients on chemotherapy, sometimes the "cure" can be as deadly as the disease itself. Adverse drug reactions are one of the leading causes of death among patients receiving cancer treatment. Chemo drugs are supposed to be toxic to cancer cells, but due to very high concentrations of these drugs they are toxic to normal cells as well thereby damaging normal cells and their activities.
Besides toxicity, another failure of the known anticancer agents is their inability to kill cancer stem cells. In recent years, it is hypothesized that only a small fraction of the entire tumor mass are responsible for the tumorigenic activities within the tumor. These small fractions of tumorigenic cells, according to the new model, are with stem-cell-like qualities and are called "cancer stem cells". These cancer stem cells were shown to have similarcellular markers, CD34+/CD38 , as primitive hematopoietic stem cells. Since then, researchers have found cancer stem cells conclusively in various types of tumors including those of the brain, breast, skin, prostate, and so on.
These cancer stem cells give rise both to more tumor stem cells and to the majority of cells in the tumor that have lost the capacity for extensive proliferation and the ability to give rise to new tumors that are slowly becoming resistant to drugs causing relapse. Since these cells having significant self- renewal potential,can cause relapse, to make it possible to treat patients with aggressive, non-resectable tumors, and refractory or recurrent cancers, as well as for preventing tumor metastasisthere is need in the art for newer anticancer agents, the use of which can target and inhibit, reduce, or eliminate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells. Summary of the Invention
Accordingly, the present invention provides agents which show capability for inhibition of proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs) thus offering a new opportunity for the treatment of cancers.
In some of the embodiments the present invention provides molecules represented by compounds having the general formula I or pharmaceutically acceptable salt thereof, and pharmaceutically acceptable compositions having ability to inhibit proliferation of and/or eradicate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs). Such compounds have the general formula I:
(
Figure imgf000004_0001
In some embodiments the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful in targeting cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for treating a variety of diseases, disorders or condition. Such diseases, disorders, or conditions include those described herein.
In some embodiments the invention provides composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, for inhibiting the proliferation of and/or eradicating cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
In some embodiments the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, or pharmaceutically acceptable compositions thereof in inhibiting the proliferation of and/or eradicating cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs).
In some other embodiments the invention provides a compound of formula I, or a pharmaceutically acceptable salts thereof or pharmaceutically acceptable compositions thereof for the prophylactic or therapeutic treatment of cancer. In some other embodiments the invention provides a method of inhibiting proliferation of or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells in a patient comprising the step of administering to patient a compound of formula I, or a pharmaceutically acceptable salts thereof or pharmaceutically acceptable compositions thereof. Description of the Invention
Accordingly, the present invention is directed towards molecules represented by compounds of Formula 1 having ability to inhibit proliferation of and/or eradicate cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells (CSCs). The present invention relates to the use of such anticancer molecules that is compounds of Formula 1 in targeting cancer. The present invention also relates to the use of anticancer molecules represented by Formula 1 for inhibition of proliferation and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells. Further, the present invention relates to the use of anticancer molecules represented by Formula 1 for prevention and/or treatment of cancer.
In one embodiment the present invention relates to the molecules having ability to inhibit proliferation and/or eradication of cancer cells and/or cancer stem cells. Such molecules are represented by compounds of Formula 1:
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is a 4-8 membered partially unsaturated, or an aromatic ring;
n is 0-4; R1 is -CN, -N02, -R, -OR, -COR, -C02R', -OCOR', -C02R, -CONR'R", -COR'NR"2, -NR'R", -NR"COR', --NR"CONR'2, -NR"C02R', -C02NR, -SR, SOR, or -S02R, -S02R'NR"2, -NR'S02R", SO 2 NR'R" or (C=NR')NR'R"';
Wherein each R, R', R" and R'" is independently hydrogen or an alkyl, optionally substituted group selected from Cl-6 aliphatic, alkenyl, alkynyl, a 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring, aryl, phenyl, an 8- 10 membered bicyclic aromatic carbocyclic ring; a 4-8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; and
X is H, F, CI, Br, or I; Compounds of this invention include those described generally above, and further described herein below.
In certain embodiments, Ring A is a 5-6 membered partially unsaturated, or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, Ring A is a 5-6 membered aromatic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, Ring A is pyrido or thiopheno. In some embodiments, Ring A is benzo. As defined generally above, n is 0-4. In certain embodiments, n is 1-3. In some embodiments, n is 2.
In certain embodiments, each R is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, or a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, each R is independently hydrogen or an optionally substituted Ci_6 aliphatic group. In some embodiments, each R is independently hydrogen or an optionally substituted Ci.2 aliphatic group. In some embodiments, each R is hydrogen. In some embodiments, each R is an optionally substituted Ca.2 aliphatic group.
In certain embodiments, the compounds of the present invention are in the form pharmaceutically acceptable derivative including but not limited to any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
In certain embodiments, the compounds of the present invention are in the form of pharmaceutically acceptable salts. Such salts are acid or base salts. Examples of pharmaceutically acceptable acid 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, a-ketoglutarate, and a- glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts. Other pharmaceutically acceptable salts include adipate, alginate, aspartate, benzenesulfonate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p- toluenesulfonate, undecanoate, valerate salts, and the like.
Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci_4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting sufficiently basic compound such as an amine with 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.
It will be appreciated by those skilled in the art that compounds as represented by Formula 1 exhibit polymorphism. It is to be understood that the present invention encompasses, polymorphic forms, of a compounds represented by Formula 1, which possess the useful properties described herein.
Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 1 C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention. In certain embodiments, a warhead moiety, R1, of a provided compound comprises one or more deuterium atoms. In certain embodiments, the present invention provides a compound of formula
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is defined above and in classes and subclasses described above and herein for compounds of formula I.
In certain embodiments, the present invention provides a compound of formula
Figure imgf000011_0002
III or a pharmaceutically acceptable salt thereof, wherein each variable is defined above except R1 which is the same as R1 and in classes and subclasses described above and herein for compounds of formula I. In some of the preferred embodiments: R1 and R1' is independently -0(CH 2 )„ , -0(CH2 ) CH=CH 2, -OArOCOCH3, -CH3/ C2HS,-NHAC-N (CH2)n and
R 3 is F, CI, Br, or I.
Exemplary compounds of present invention are set forth herein, below: Exemplary Compounds of Formula I
Figure imgf000012_0001
1-7 1-8
Figure imgf000013_0001
1-9
In certain embodiments, the present invention provides any compound selected from those depicted above, or a pharmaceutically acceptable salt. In certain other embodiments the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for inhibiting proliferation and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for treating cancer in an animal. The present invention further provides pharmaceutical compositions comprising the compounds represented by the Formula 1 for inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells for prevention and/or treatment of cancer.
According to certain embodiments, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
The compounds represented by the Formula 1 or their pharmaceutically acceptable salt or their composition can be formulated into dosage form to be administered to a mammalian host, such as a human patient adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
The compound may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
The compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same 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. Such compositions and preparations should contain the active molecule in an amount such that an effective dosage level will be obtained. The percentage of the compositions and preparations may, of course, be varied conveniently by a person skilled in the art.
In certain embodiments the compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same are formulated into solid dosage forms for oral administration, such dosage form can be selected from but not limiting to tablets, troches, pills, capsules. Pharmaceutically inert excipient that may be incorporated for formulation of such dosage form include 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. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like.
Liquid dosage forms are another exemplary dosage form for formulating compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same. Such liquid dosage form can be in the form of for example a syrup, emulsions, suspensions, or elixir containing the active molecule, and other pharmaceutically acceptable inert excipients commonly used in the art such as, for example sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor diluents. Other adjuvants include solubilizing agents, wetting agents, emulsifying and suspending agents.
The compounds represented by the Formula. 1 or their pharmaceutically acceptable salt or composition comprising the same may also be formulated as pharmaceutical dosage forms to be administered intravenously or intraperitoneal^ by injection or infusion. 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. Solutions of the active molecule 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. In all cases, 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 molecules represented by the Formula 1 in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, 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.
For topical administration, the molecules represented by the Formula 1 or their pharmaceutically acceptable salt or composition comprising the same 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 molecules 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 molecules of Formula I to the skin are known to the art.
Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Ophthalmic formulations may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
Formulation for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. The compounds of the present invention or their pharmaceutically acceptable salt or composition comprising the same may also be formulated for nasal administration through aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
Compounds and compositions described herein are generally useful for the inhibition of proliferation and/or eradication of cancer cells and/or cancer stem cells. Thus, provided compounds are useful for prevention and/or treatment of cancers, including, but not limited to hematological cancers and solid tumors.
The invention in one embodiment relates to the use of compounds represented by the Formula 1 for inhibition of proliferation of and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells.
The use of compounds represented by the Formula 1 for inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells having significant self-renewal potential, such as cancer stem cells include stem cells within tumors or leukemic stem cells. According to certain embodiments, the invention relates to a method of inhibiting cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells in a patient, leading to remission of the cancer, comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In some embodiments the present invention provides a method for treating a disorder mediated by cancer cells cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.
The use of compounds represented by the Formula 1 for inhibition and/or eradication of cancer cells cancer cells and/or cancer cells having significant self- renewal potential, such as cancer stem cells for prevention and/or treatment of cancer, include cancer of at least one type selected from the group consisting of cancer is selected from breast, prostate, ovary, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, or stomach. Exemplary cancers include acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, soft-tissue sarcoma, squamous cell carcinomas, colon carcinoma, an adenocarcinoma, a sweat gland carcinoma, a sebaceous gland carcinoma, a papillary carcinoma, a papillary adenocarcinoma, a cystadenocarcinoma, a medullary carcinoma, a bronchogenic carcinoma, a renal cell carcinoma, a hepatocellular carcinoma, a bile duct carcinoma, a choriocarcinoma, a seminoma, an embryonal carcinoma, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervix cancer, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, Ewing's sarcoma, gallbladder cancer, hairy cell leukemia, head and neck cancer, Hodgkin's lymphoma, Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non-Hodgkin's lymphoma, osteosarcoma, ovary cancer, germ cell cancer, pancreatic cancer, penis cancer, prostate cancer, retinoblastoma, skin cancer, stomach cancer, testicular cancer, thyroid cancer, trophoblastic neoplasms, cancer of the uterus, vaginal cancer, cancer of the vulva, and Wilm's tumor.
According to another embodiment, the invention relates to a method of inhibiting cancer cells cancer cells and/or cancer cells having significant self- renewal potential, such as cancer stem cells," in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
Inhibition of cancer stem cells cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to biological assays, gene expression studies, and biological target identification.
Examples of cancer cell lines that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include, breast, prostrate and ovarian cell lines. Some of the breast, prostrate and ovarian cell lines selected include but not limited to DU145, PC3, LNCaP, MDA MB 231, MCF7, T47D, MCF10A, HeLa, or L929. The activity of a compound utilized in this invention as an inhibitor of of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells, may be assayed in vitro or in vivo. An in vivo assessment of the inhibitory or cytotoxic activity of the compounds of the invention may be made using an animal model of cancer, e.g., a rodent or primate model. Cell-based assays may be performed using, e.g., a cell line isolated from a tumor or blood- borne cancer. Cell-based assays for activity against a specific protein or nucleic acid component of a cancer cell line, e.g., an enzyme, structural protein, DNA or RNA, may also be performed. Additionally, biochemical or mechanism-based assays, e.g., transcription assays using a purified protein, Northern blot, RT-PCR, etc., may be performed. In vitro assays include assays that determine cell morphology, viability, cell count, or growth inhibition, and/or the cytotoxicity, enzyme inhibitory activity, and/or the subsequent functional consequences of treatment of cancer cells with compounds of the invention. Alternate in vitro assays quantitate the ability of the inhibitor to bind to protein or nucleic acid molecules within the cell. Inhibitor binding may be measured by radiolabelling the inhibitor prior to binding, isolating the inhibitor/target molecule complex and determining the amount of radiolabel bound. Alternatively, inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with purified proteins or nucleic acids bound to known radioligands. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of cancer cells or CSCs, are set forth in the Examples below. The aforementioned assays are exemplary and not intended to limit the scope of the invention. The skilled practitioner can appreciate that modifications can be made to conventional assays to develop equivalent assays that obtain the same result.
In certain embodiments, a provided compound, or composition thereof, is administered in combination with Such chemotherapeutic agents include, but are not limited to agents such as kinase inhibitors, alkylating agents, antimetabolites, tubulin stabilizers, tubulin assembly inhibitors, DNA replication inhibitors, cell cycle inhibitors, topoisomerase inhibitors, cytotoxic antibiotics or nanoparticle or protein conjugates of any of the aforementioned agents. In certain embodiments, the chemotherapeutic agents used in combination with compounds or compositions of the invention include, but are not limited to imatinib, nilotinib, gefitinib, sunitinib, carfilzomib, salinosporamide A, retinoic acid, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide, azathioprine, mercaptopurine, doxifluridine, fluorouracil, gemcitabine, methotrexate, tioguanine, vincristine, vinblastine, vinorelbine, vindesine, podophyllotoxin, etoposide, teniposide, tafluposide, paditaxel, docetaxel, irinotecan, topotecan, amsacrine, actinomycin, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, plicamycin, mitomycin, mitoxantrone, melphalan, busulfan, capecitabine, pemetrexed, epothilones, 13-cis-Retinoic Acid, 2-CdA, 2-Chlorodeoxyadenosine, 5-Azacitidine, 5-Fluorouracil, 5-FU, 6-Mercaptopurine, 6-MP, 6-TG, 6-Thioguanine, Abraxane, Accutane®, Actinomycin-D, Adriamycin®, Adrucil®, Afinitor®, Agrylin®, Ala-Cort®, Aldesleukin, Alemtuzumab, ALIMTA, Alitretinoin, Alkaban-AQ®, Alkeran®, All- transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine, Aminoglutethimide, Anagrelide, Anandron®, Anastrozole, Arabinosylcytosine, Ara-C, Aranesp®, Aredia®, Arimidex®, Aromasin®, Arranon®, Arsenic Trioxide, Arzerra™, Asparaginase, ATRA, Avastin®, Azacitidine, BCG, BCNU, Bendamustine, Bevacizumab, Bexarotene, BEXXAR®, Bicalutamide, BiCNU, Blenoxane®, Bleomycin, Bortezomib, Busulfan, Busulfex®, C225, Calcium Leucovorin, Campath®, Camptosar®, Camptothecin-11, Capecitabine, Carac™, Carboplatin, Carmustine, Carmustine Wafer, Casodex® CC-5013, CCI-779, CCNU, CDDP, CeeNU, Cerubidine® Cetuximab, Chlorambucil, Citrovorum Factor, Cladribine, Cortisone, Cosmegen® CPT-11, Cytadren® Cytosar-U® Cytoxan® Dacarbazine, Dacogen, Dactinomycin, Darbepoetin Alfa, Dasatinib, Daunomycin, Daunorubicin Hydrochloride, Daunorubicin Liposomal, DaunoXome® Decadron, Decitabine, Delta-Cortef® Deltasone® Denileukin, Diftitox, DepoCyt™, Dexamethasone, Dexamethasone Acetate, Dexamethasone Sodium Phosphate, Dexasone, Dexrazoxane, DHAD, DIC, Diodex, Docetaxel, Doxil® Doxorubicin, Doxorubicin Liposomal, Droxia™, DTIC, DTIC-Dome® Duralone® Efudex® Eligard™, Ellence™, Eloxatin™, Elspar® Emcyt® Epirubicin, Epoetin Alfa, Erbitux, Erlotinib, Erwinia L- asparaginase, Estramustine, Ethyol, Etopophos®, Etoposide, Etoposide Phosphate, Eulexin® Everolimus, Evista® Exemestane, Fareston® Faslodex® Femara® Filgrastim, Floxuridine, Fludara® Fludarabine, Fluoroplex® Fluorouracil, Fluorouracil (cream), Fluoxymesterone, Flutamide, Folinic Acid, FUDR® Fulvestrant, G-CSF, Gefitinib, Gemcitabine, Gemtuzumab, ozogamicin, ,Gemzar Gleevec™, Gliadel® Wafer, GM-CSF, Goserelin, Granulocyte - Colony Stimulating Factor, Granulocyte Macrophage Colony Stimulating Factor, Halotestin® Herceptin® Hexadrol, Hexalen® Hexamethylmelamine, HMM, Hycamtin® Hydrea® Hydrocort Acetate® Hydrocortisone, Hydrocortisone Sodium Phosphate, Hydrocortisone Sodium Succinate, Hydrocortone Phosphate, Hydroxyurea, Ibritumomab, Ibritumomab, Tiuxetan, Idamycin® Idarubicin Ifex® IFN-alpha, Ifosfamide, IL-11, IL-2, Imatinib mesylate, Imidazole Carboxamide, Interferon alfa, Interferon Alfa-2b (PEG Conjugate), lnterleukin-2, lnterleukin-11, Intron A® (interferon alfa-2b), Iressa® Irinotecan, Isotretinoin, Ixabepilone, Ixempra™, Kidrolase®, Lanacort® Lapatinib, L-asparaginase, LCR, Lenalidomide, Letrozole, Leucovorin, Leukeran, Leukine™, Leuprolide, Leurocristine, Leustatin™, Liposomal Ara-C, Liquid Pred® Lomustine, L-PAM, L-Sarcolysin, Lupron® Lupron Depot® Matulane® Maxidex, Mechlorethamine, Mechlorethamine Hydrochloride, Medralone® Medrol® Megace® Megestrol, Megestrol Acetate, Melphalan, Mercaptopurine, Mesna, Mesnex™, Methotrexate, Methotrexate Sodium, Methylprednisolone, Meticorten® Mitomycin, Mitomycin-C, Mitoxantrone, M- Prednisol® MTC, MTX, Mustargen® Mustine, Mutamycin® Myleran® Mylocel™, Mylotarg®, Navelbine®, Nelarabine, Neosar®, Neulasta™, Neumega® Neupogen® Nexavar® Nilandron® Nilotinib, Nilutamide, Nipent® Nitrogen Mustard, Novaldex® Novantrone® Nplate, Octreotide, Octreotide acetate, Ofatumumab, Oncospar® Oncovin® Ontak® Onxal™, Oprelvekin, Orapred® Orasone® Oxaliplatin, Paclitaxel, Paditaxel Protein-bound, Pamidronate, Panitumumab, Panretin® Paraplatin® Pazopanib, Pediapred® PEG Interferon, Pegaspargase, Pegfilgrastim, PEG-INTRON™, PEG-L-asparaginase, PEMETREXED, Pentostatin, Phenylalanine Mustard, Platinol® Platinol-AQ® Prednisolone, Prednisone, Prelone® Procarbazine, PROCRIT® Proleukin® Prolifeprospan 20 with Carmustine Implant, Purinethol® Raloxifene, Revlimid® Rheumatrex® Rituxan® Rituximab, Roferon-A® (Interferon Alfa-2a), Romiplostim, Rubex® Rubidomycin hydrochloride, Sandostatin® Sandostatin LAR® Sargramostim, Solu-Cortef® Solu-Medrol® Sorafenib, SPRYCEL™, STI-571, Streptozocin, SU11248, Sunitinib, Sutent® Tamoxifen, Tarceva® Targretin® Tasigna® Taxol® Taxotere® Temodar® Temozolomide, Temsirolimus, Teniposide, TESPA, Thalidomide, Thalomid® TheraCys® Thioguanine, Thioguanine Tabloid® Thiophosphoamide, Thioplex® Thiotepa, TICE® Toposar® Topotecan, Toremifene, Torisel® Tositumomab, Trastuzumab, Treanda® Tretinoin, Trexall™, Trisenox® TSPA, TYKERB® VCR, Vectibix™, Velban® Velcade® VePesid® Vesanoid® Viadur™, Vidaza® Vinblastine, Vinblastine Sulfate, Vincasar Pfs® Vincristine, Vinorelbine, Vinorelbine tartrate, VLB, VM-26, Vorinostat, Votrient, VP-16, Vumon® Xeloda® Zanosar® Zevalin™, Zinecard® Zoladex® Zoledronic acid, Zolinza, Zometa® or combinations of any of the above.
In certain embodiments, a combination of 2 or more chemotherapeutic agents may be administered together with compounds of the invention. In certain embodiments, a combination of 3 or more chemotherapeutic agents may be administered with compounds of the invention. In some embodiments, the chemotherapeutic agents are selected from alkylating agents or antimetabolites. In other embodiments, the chemotherapeutic agent is cisplatin. Other examples of agents the inhibitors of this invention may also be combined with include, without limitation: vitamins and nutritional supplements, cancer vaccines, treatments for neutropenia (e.g. G-CSF, filgrastim, lenograstim), treatments for thrombocytopenia (e.g. blood transfusion, erythropoietin), antiemetics (e.g. 5-HT3 receptor antagonists, dopamine antagonists, NK1 receptor antagonists, histamine receptor antagonists, cannabinoids, benzodiazepines, or anticholinergics), treatments for Alzheimer's Disease such as Aricept* and Excelon®; treatments for Parkinson's Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), Copaxone®, and mitoxantrone; treatments for asthma such as albuterol and Singulair8; agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; antiinflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, and anti-Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; and agents for treating immunodeficiency disorders such as gamma globulin.
In certain embodiments, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with antisense agents, a monoclonal or polyclonal antibody or an siRNA therapeutic.
The compounds may also be used in combination with other drugs that are used in the treatment/prevention/suppression or amelioration of the diseases or conditions for which compounds as represent by Formula 1 are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another, normally within five hours from one another. When a compound as per the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of the present invention is preferred. Accordingly, the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of the present invention. The compounds represented by the Formula 1 either alone or in combination with other active or supplementary agents may be used for inhibition of proliferation and/or eradication of cancer cells and/or cancer stem cells for targeting cancer as a standalone therapy or may be used either before or after or in conjunction with other therapies such as radiation, surgery or any other method for prevention and/or treatment of cancer.
The amount of a composition administered will be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc. The amount of both, an inventive compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
Dosage amount of the compounds represented by the Formula 1, other active agents and interval may be adjusted individually to provide plasma levels of the active species. HPLC assays or bioassays can be used to determine plasma concentrations.
The therapeutically effective dose of molecules is an amount sufficient to cause cell death or inhibit proliferation and cause differentiation of stem cells in solid tumors or leukemias.
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals. Preferably, compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive can be administered to a patient receiving these compositions. In certain embodiments, the compounds of the invention for oral or parenteral administration may be at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
The use of compounds represented by the Formula 1 for remission of cancer is a newer method for treatment of cancer which involves the inhibition and/or eradication of cancer cells and/or cancer cells having significant self-renewal potential, such as cancer stem cells which would reduce the likelihood of clinical relapse after treatment.
Examples
Synthesis: As depicted in the Example below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
Example 1: In certain embodiments, compounds of formula I where are prepared according to the procedure outlined in Scheme 1.
SCHEME 1
Figure imgf000032_0001
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the appended claims.
In certain embodiments, compounds and synthetic intermediates thereof of the present invention may be prepared according to methods known to one of ordinary skill in the art.
Assays:
In certain embodiments, compounds of the present invention are assayed as inhibitors of CSCs using methods known in the art including an in vitro cell death assay, an in vitro soft agar growth inhibition assay, an in vitro 3D sphere forming stem cell assay, and in vivo toxicity assays in animals.
Compounds of the present invention were evaluated in a colorimetric in vitro cell death assay as described by Carmichael, J, DeGraff, WG, Gazdar, AF, Minna, JD, and Mitchell, JB. Evaluation of a tetrazolium-based semiautomatic colorimetric assay: assessment of chemosensitivity testing. Cancer Res 1987; 47: 936-942.
Example 2
In- vitro colorimetric cell death assay An exemplary procedure for the in vitro colorimetric cell death assay follows. Cancer cells were plated in a 96 well plate as per predetermined plating efficiency, the plate was incubated for 24 hours in a 5% C02 atmosphere at 37 degrees Celsius, a range of concentrations of the compound of the present invention of formula 1 was added to the wells, the plates were incubated further for 48 hours in a 5% C02 atmosphere, the plate was centrifuged twice at 3000 rpm for 3 minutes, the supernatant fluid was discarded, 100 uL of 0.5mg/mL MTT solution was added, the plate was incubated for 4 hours in a 5% C02 atmosphere at 37 degrees Celsius, the plate was centrifuged twice at 3000 rpm for 3 minutes, supernatant was aspirated very carefully, 200 uL DMSO was added to each well to solubilize MTT crystals and mixed well by shaking the plate, the plate was incubated for 10 minutes in a 5% C02 atmosphere at 37 degrees Celsius, the plate placed on the shaker of an ELISA plate reader and the absorbance at 570 nm was measured, then the percentage of viable cells remaining was calculated by first subtracting the background absorbance then comparing to the absorbance of a non-drug-treated cell sample, and the results were plotted on a graph to determine the IC50 for the compound.
The results of the in vitro colorimetric cell death assay are set forth in Table 1. Table 1. Results of in vitro colorimetric cell death assay.
Figure imgf000034_0001
Example 3
In Vitro 3D Sphere Forming Stem Cell Assay
Compounds of the present invention were evaluated in an in vitro 3D sphere forming stem cell assay as described by Rajasekhar, VK, Analytical Methods for Cancer Stem Cells. Methods in Molecular Biology, 2007, Volume 407, 83-95.
An exemplary procedure for the in vitro 3D sphere forming stem cell assay follows. Preparation of Drugs stock and dilutions:
Considering the molecular weight of 330.28 g/mol of the compound of formula I- 1, 33 mg was dissolved in ΙΟΟμΙ. ethyl acetate (tissue culture grade) to attain a final concentration of 1M. To obtain range of concentrations for the experimental study, dilutions were carried out as follows: · 0.01M (10"2M): 50μί of 10"1 M solution of compound of formula 1-1 was added to 450μί of ethyl acetate
• 0.001M (10"3M): 50μΙ_ of 10"2M solution of compound of formula 1-1 was added to 450μί of ethyl acetate • 0.0001M (10~4M): 50μί of 10"3M solution of compound of formula 1-1 was added to 450μί of ethyl acetate
• 0.00001M (10"5M): 50μί of 10"4M solution of compound of formula 1-1 was added to 450μί of ethyl acetate · 0.000001M (10"6M): 50μί of 10"5M solution of compound of formula 1-1 was added to 450μί. of ethyl acetate
Cells were grown in two dimensions on a plastic substrate, harvested in suspension in serum-free media, then the cells in the sample were trypsinized and a single cell suspension was formed by passing through a cell strainer. The cells were diluted according to the predetermined plating efficiency for the cell line being studied by suspending the cells in stem cell culture medium. 100 uL of this suspension was added into each well of a 96 well suspension plate, and the plate was incubated at 37 degrees Celsius in 5% C02 atmosphere for 24 hours, then 2 uL of appropriate concentrations of the drug were added into each respective well along with 100 uL of stem cell culture medium, and the plates incubated at 37 degrees Celsius under 5% C02 atmosphere for 72 hours. After incubation, 2.5 uL of the appropriate drug concentration was then added to each respective well along with 50 uL of stem cell culture medium, then the plates incubated at 37 degrees Celsius under 5% C02 atmosphere for further 72 hours. 3 uL of the appropriate drug concentration was added to each respective well along with 50 uL of stem cell culture medium, then incubated at 37 degrees Celsius under 5% C02 atmosphere for further 72 hours, then the spheres formed were observed under a microscope then counted and scored by size.
Results of the in vitro 3D sphere forming stem cell assay for compound of formula 1-1 are set forth in Table 2. The number in each box is the total number of spheres formed in the presence of compound of formula 1-1 at each drug concentration. GC refers to a growth control performed in the absence of drug or solvent (DMSO). GCEA refers to a growth control performed in the absence of drug or solvent ethyl acetate.
Table 2
Figure imgf000036_0001
Compound of formula 1-1 showed significant sphere reduction. It was found to be the most effective compound on breast, prostrate and ovarian cancer cell lines.

Claims

Claims:
1. A compound of formula I:
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is a 4-8 membered partially unsaturated, or aromatic ring;
n is 0-4;
R1 is -CN, -N02, -R, -OR, -COR, -C02R', -OCOR', -C02R, -CONR'R", -COR'NR"2, -NR'R", -NR"COR', --NR"CONR'2, -NR"C02R', -C02NR, -SR, SOR, or -S02R, -S02R'NR"2, -NR'S02R", SO 2 R'R" or (C=NR')NR'R"';
Wherein each R, R', R" and R'" is independently hydrogen or an alkyl, optionally substituted group selected from Cl-6 aliphatic, alkenyl, alkynyl, a 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring, aryl, phenyl, an 8-10 membered bicyclic aromatic carbocyclic ring; a 4- 8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; and X is H, F, CI, Br, or I;
2. The compound of claim 1 wherein Ring A is a 4-8 membered partially unsaturated, or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
3. The compound of claim 1 wherein Ring A is Benzo.
4. The compound of claim 1 which is compound of formula II, III or IV or or a pharmaceutically acceptable salt thereof, wherein each variable is as per claim 1 and R1 is same as R1:
Figure imgf000038_0001
5. The compound of claim 1 in which compound is compound of formula 1-1, 1-2, 1-3, 1-4, 1-5 1-6, 1-7, 1-8 or 1-9 or a pharmaceutically acceptable salt thereof,
Figure imgf000038_0002
1-4
Figure imgf000039_0001
1-5
Figure imgf000039_0002
1-9
6. A use of a compound of formula I as described in claim 1, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for treating cancer in an animal.
7. A pharmaceutical composition comprising a compound of formula I as described in claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
8. A use of a compound of formula I as described in claim 1 or a pharmaceutically acceptable salt thereof or composition of claim 7 for inhibiting the proliferation or of or eradication of cancer cells and/or cancer stem cells in a patient.
9. A method of inhibiting proliferation of or eradication of cancer cells and/or cancer stem cells in a patient comprising the step of administering to said patient a compound of formula I of claim 1 or composition of claim 7.
10. The method according to claim 9, wherein said cancer stem cells are from the cell lines selected from but not limiting to breast, prostrate and ovarian cell lines.
11. The method according to claim 10, wherein said cell lines are selected from but not limiting to DU145, PC3, LNCaP, MDA MB 231, MCF7, T47D, MCF10A, HeLa, or L929.
12. The method according to claim 9, wherein said cancer is selected from breast, prostate, ovary, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, or stomach.
13. The method according to claim 9, wherein said compound is administered in combination with an additional at least one chemotherapeutic agent.
PCT/IN2011/000866 2010-12-17 2011-12-16 Molecules with anticancer activity and uses thereof WO2012081039A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN3242/MUM/2010 2010-12-17
IN3242MU2010 2010-12-17

Publications (1)

Publication Number Publication Date
WO2012081039A1 true WO2012081039A1 (en) 2012-06-21

Family

ID=46244186

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2011/000866 WO2012081039A1 (en) 2010-12-17 2011-12-16 Molecules with anticancer activity and uses thereof

Country Status (1)

Country Link
WO (1) WO2012081039A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014097318A3 (en) * 2012-12-18 2015-03-19 Godavari Biorefineries Limited Agents for eliminating tumour-initiating cells

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5534531A (en) * 1992-01-21 1996-07-09 Norsk Hydro A.S. Compounds
US20050227978A1 (en) * 2002-01-30 2005-10-13 Sumitomo Pharmaceuticals Co., Ltd. Fibrosis inhibitor
WO2010089778A2 (en) * 2009-02-05 2010-08-12 Godavari Biorefineries Ltd. Process for the synthesis of cleistanthin

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5534531A (en) * 1992-01-21 1996-07-09 Norsk Hydro A.S. Compounds
US20050227978A1 (en) * 2002-01-30 2005-10-13 Sumitomo Pharmaceuticals Co., Ltd. Fibrosis inhibitor
WO2010089778A2 (en) * 2009-02-05 2010-08-12 Godavari Biorefineries Ltd. Process for the synthesis of cleistanthin

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ALAOUI-JAMALI ET AL.: "A Novel Experimental Heme Oxygenase-1-Targeted Therapy for Hormone-Refractory Prostate Cancer.", CANCER RES, vol. 69, 2009, pages 8017 - 8024, XP009167475, DOI: doi:10.1158/0008-5472.CAN-09-0419 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014097318A3 (en) * 2012-12-18 2015-03-19 Godavari Biorefineries Limited Agents for eliminating tumour-initiating cells

Similar Documents

Publication Publication Date Title
US10004809B2 (en) Precise delivery of therapeutic agents to cell mitochondria for anti-cancer therapy
US11680078B2 (en) Anticancer compounds
US20150328245A1 (en) Agents for Eliminating Tumour-Initiating Cells
WO2014201111A1 (en) Treatment of mtor hyperactive related diseases and disorders
CA2854836C (en) Tricyclic amino containing compounds for treatment or prevention of symptoms associated with endocrine dysfunction
CA2901155A1 (en) Camkii inhibitors and uses thereof
US10556928B2 (en) Stabilized peptoid-peptide hybrids and uses thereof
US11390613B2 (en) Azole analogues and methods of use thereof
WO2012081038A2 (en) Anticancer compounds and targeting cancer with the same
US10316050B2 (en) PT (IV) complexes containing 4,4′-disubstituted-2,2′-bipyridyl and their use in cancer therapy
AU2023200287A1 (en) KRAS Antagonists
WO2012081039A1 (en) Molecules with anticancer activity and uses thereof
US20160214941A1 (en) Compounds for lnhibition of Unregulated Cell Growth
US8871983B2 (en) Lipid compounds for suppression of tumorigenesis
US10457700B2 (en) Platinum prodrugs and methods of making and using thereof
US20240018132A1 (en) 5-hydroxy-1,4-naphthalenedione for use in the treatment of cancer
WO2023203185A1 (en) Mitochondriotropic benzamide potassium channel k v1.3 inhibitors
WO2023086365A1 (en) Compositions comprising amino lipid compounds and methods of making and use thereof

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: 11847962

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: 11847962

Country of ref document: EP

Kind code of ref document: A1