WO2014160034A1 - Aldehyde dehydrogenase-1 modulators and methods of use thereof - Google Patents

Aldehyde dehydrogenase-1 modulators and methods of use thereof Download PDF

Info

Publication number
WO2014160034A1
WO2014160034A1 PCT/US2014/025679 US2014025679W WO2014160034A1 WO 2014160034 A1 WO2014160034 A1 WO 2014160034A1 US 2014025679 W US2014025679 W US 2014025679W WO 2014160034 A1 WO2014160034 A1 WO 2014160034A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
inhibitors
compound
compounds
agents
Prior art date
Application number
PCT/US2014/025679
Other languages
French (fr)
Inventor
Che-Hong Chen
Wenjin Yang
Daria Mochly-Rosen
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2014160034A1 publication Critical patent/WO2014160034A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Aldehyde dehydrogenase- 1 (ALDH1A1) is mitochondria nuclear encoded cytosolic enzyme.
  • ALDH1 Al has been widely recognized as a key biomarker for cancer stem cell (CSC). Strong support for the CSC hypothesis, which proposes that a relatively rare subpopulation of tumor cells have the unique ability to initiate and perpetuate tumor growth has been published in the past decade. These cells, called cancer stem cells or tumor-initiating cells, share various characteristics with embryonic and somatic stem cells including self-renewal and multi-potent differentiation. CSCs may be highly resistant to radiation or chemotherapy; therefore, the development of more effective therapies for cancer requires effective targeting of this cell population. These cells can be identified and isolated using biomarkers such as ALDH1A1.
  • ALDHlAl isozymes such as ALDH1A1 metabolize a wide variety of intracellular aldehydes and can thus provide resistance to reactive oxygen species (ROS)-derived products or alkylating agents such as cyclophosphamide.
  • ROS reactive oxygen species
  • High ALDHlAl activity has been reported in CSCs from many cancers including breast, lung, liver, colon, pancreas, ovary, head and neck, prostate cancers and some hematologic cancers.
  • silencing ALDHlAl by siRNA or shRNA leads to cell cycle arrest, apoptosis, decreased ceil viability in vitro, and reduced tumorigenesis in vivo.
  • Silencing ALDHlAl by siRNA also sensitizes cancer cells to drug- induced cell death.
  • silencing ALDHlAl using nanoliposomal siRNA sensitized both taxane- and platinum-resistant cell lines to chemotherapy significantly reducing tumor growth in mice compared with chemotherapy alone (74%-90% reduction; P ⁇ 0.015).
  • ALDHlAl is not only a biomarker of CSCs, but also an attractive therapeutic target for human cancer
  • Antagonists of ALDHlAl are useful for treating disorders such as cancer.
  • Compounds of the present disclosure are antagonist of ALDHlAl, or inhibitors of ALDHlAl and are described herein.
  • the present disclosure provides compounds that function as inhibitors or antagonists of aldehyde dehydrogenase- 1 (ALDHlAl) activity, pharmaceutical compositions containing them, and methods of preparing these compounds.
  • the present disclosure relates to a pharmaceutical composition comprising a compound of formula (I) and a cancer
  • R 1 and R 2 are each independently selected from hydrogen and C 1 -C6 alkyl
  • R 3 and R 4 are each independently selected from hydrogen and C 1 -C6 alkyl
  • each R 7 is independently selected from halogen, C 1 -C6 alkyl, and OCH 3 ;
  • n 0, 1, 2, 3, 4, or 5;
  • R 1 and R 2 is hydrogen and the other is Ci-C 6 alkyl.
  • one of R 3 and R 4 is hydrogen and the other is C 1 -C6 alkyl.
  • one of R 3 and R 4 is hydrogen and the other is methyl or ethyl.
  • n is 0, 1, 2, or 3.
  • n is 1, 2, or 3.
  • n is 1.
  • n is 2.
  • each R 7 is C 1 -C6 alkyl.
  • R 7 is methyl, ethyl, or isopropyl.
  • each R 7 is halogen.
  • R 7 is fluoro.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound selected from Table 1 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a cancer chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a pharmaceutical composition as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound is selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the method as described herein further comprises administering a cancer chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non- peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • HDAC inhibitors histone- deacetylase inhibitors
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound and the chemotherapeutic agent are administered
  • the method as described herein further comprises administering ionizing radiation.
  • the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
  • the present disclosure provides a method of treating or preventing cancer in a subject comprising administering to the subject a first amount of a compound as described herein in a first treatment procedure, and a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
  • the radiation is a radiopharmaceutical agent.
  • the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
  • the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
  • the method described herein provides a synergistic effect in the treatment of cancer.
  • the present disclosure relates to a method of increasing the sensitivity of a cancerous cells or cancer stem cells to a chemotherapeutic agent comprising administering to a subject a compound as described herein or a pharmaceutical composition as described herein.
  • the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone-deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral. In one embodiment, the compound or pharmaceutical composition is administered to the respiratory tract.
  • the present disclosure provides a use of a pharmaceutical composition as disclosed above for the manufacture of a medicament for treating or preventing cancer.
  • the present disclosure provides a use of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer.
  • the present disclosure provides a use of a compound selected from Table 1 herein, or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament for treating or preventing cancer.
  • the medicament further comprises a cancer chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound and the chemotherapeutic agent are administered
  • the use further comprises administering ionizing radiation.
  • the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
  • the present disclosure provides a use of a first amount of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer in a first treatment procedure, wherein the use further comprises a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
  • the radiation is a radiopharmaceutical agent.
  • the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
  • the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
  • the use provides a synergistic effect in the treatment of cancer.
  • the present disclosure provides a use of a compound of formula (I) or a pharmaceutical composition as disclosed above for the manufacture of a medicament for increasing the sensitivity of cancerous cells or cancer stem cells to a chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral.
  • the compound or pharmaceutical composition is administered to the subject.
  • Figure 1 is a graph showing the selectivity of Compound 5 against different ALDH isozymes.
  • Figure 2A is a graph showing that Compound 5 sensitizes cytotoxic effect of paclitaxel in human multi-drug resistant ovarian NCI/ADR-RES cancer cells.
  • Figure 2B is a graph showing that Compound 5 sensitizes cytotoxic effect of paclitaxel in human multi-drug resistant ovarian TOV-21G-RT cancer cells.
  • Figure 3A is a graph showing that Compound 5 sensitizes cytotoxic effect of doxorubicin in human multi-drug resistant ovarian NCI/ADR-RES cancer cells.
  • Figure 3B is a graph showing that Compound 5 sensitizes cytotoxic effect of doxorubicin in human multi-drug resistant ovarian TOV-21G-RT cancer cells.
  • R 1 and R 2 are each independently selected from hydrogen and Ci-C 6 alkyl
  • R 3 and R 4 are each independently selected from hydrogen and C 1 -C6 alkyl
  • each R 7 is independently selected from halogen, C 1 -C6 alkyl, and OCH 3 ;
  • n 0, 1, 2, 3, 4, or 5;
  • one of R 1 and R 2 is hydrogen and the other is Ci-C 6 alkyl.
  • one of R 1 and R 2 is hydrogen and the other is straight-chain Ci-C 6 alkyl or branched C 3 -C6 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is straight-chain d- C 4 alkyl or branched C3-C4 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is straight-chain C 1 -C4 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is branched C3-C4 alkyl. [0047] In one embodiment, one of R 3 and R 4 is hydrogen and the other is Ci-C 6 alkyl. In one embodiment, one of R 3 and R 4 is hydrogen and the other is methyl or ethyl.
  • one of R 1 and R 2 is hydrogen and the other is straight-chain C 1 -C4 alkyl; and one of R 3 and R 4 is hydrogen and the other is methyl or ethyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is branched C 3 -C 4 alkyl; and one of R 3 and R 4 is hydrogen and the other is methyl or ethyl.
  • n is 0, 1, 2, or 3. In one embodiment, n is 1, 2, or 3. In one embodiment, n is 1. In one embodiment, n is 2.
  • each R 7 is Ci-C 6 alkyl. In one embodiment, each R 7 is independently methyl, ethyl, or isopropyl. In one embodiment, each R 7 is halogen. In one embodiment, each R 7 is independently chloro or fluoro. In one embodiment, R 7 is chloro. In one embodiment, R 7 is fluoro. In one embodiment, one or more R 7 is Ci-C 6 alkyl and one or more R 7 is halogen. In one embodiment, one or more R 7 is independently methyl, ethyl, or isopropyl and one or more R 7 is independently chloro or fluoro. In one embodiment, one or more R 7 is Ci-C 6 alkyl and one or more R 7 is OCH 3 .
  • one or more R 7 is independently methyl, ethyl, or isopropyl and one or more R 7 is OCH 3 . In one embodiment, one or more R 7 is halogen and one or more R 7 is OCH 3 . In one example, one or more R 7 is independently chloro or fluoro and one or more R 7 is OCH 3 .
  • the present disclosure further provides assays for identifying inhibitors of ALDHlAl.
  • subjects to be treated are humans.
  • a human to be treated according to a subject method is one that has the wild type ALDHlAl allele or a mutated/variant ALDHlAl allele, e.g. a mutated/variant ALDHlAl gene with over-expression of the ALDHlAl transcripts/enzyme, or a mutated/variant ALDHlAl gene which encodes a product with higher enzymatic activity of ALDHlAl.
  • a compound can be readily ascertained whether it is an ALDHlAl inhibitor or not.
  • Assays for dehydrogenase activity of ALDHlAl are known in the art, and any known assay can be used. Examples of dehydrogenase assays are found in various publications, including, e.g., Russo and Hilton, 1998, Cancer Res. 48:2963-8; Ho et al., 2006, Biochemistry 45:9445-53; Sheikh et al., 1997, J. Biol. Chem. 272:18817-18822; Vallari and Pietruszko, 1984, J. Biol. Chem. 259:4922; and Farres et al., 1994, J. Biol. Chem. 269:13854-13860.
  • the assays generally comprise two parts; one is the preparation of human ALDHlAl recombinant enzyme; and the other is the determination of the enzyme inhibition by a compound in the present disclosure.
  • human ALDHlAl recombinant enzyme full-length human ALDHlAl cDNA may be synthesized based on published DNA sequence (e.g., GenBank ID: NM_000689).
  • the synthetic gene may be cloned into the Nhel/EcoRI sites of the His-tag vector, pTrcHis, for protein expression. All the vectors are transformed into BL21 E. coli host cells and subjected to 0.5 niM isopropyl ⁇ -D-l-thiogalactopyranoside (IPTG) induction for protein expression at 30 °C.
  • IPTG isopropyl ⁇ -D-l-thiogalactopyranoside
  • the present disclosure is intended to include all isotopes of atoms occurring in the present
  • isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium
  • isotopes of carbon include C-13 and C-14.
  • a group may optionally be substituted with up to two Ri moieties and Ri at each occurrence is selected independently from the definition of R ⁇
  • substituents and/or variables are permissible, but only if such combinations result in stable compounds.
  • N-oxides can be converted to N-oxides by treatment with an oxidizing agent (e.g. , 3-chloroperoxybenzoic acid (ra-CPBA) and/or hydrogen peroxides) to afford other compounds of the present disclosure.
  • an oxidizing agent e.g. , 3-chloroperoxybenzoic acid (ra-CPBA) and/or hydrogen peroxides
  • ra-CPBA 3-chloroperoxybenzoic acid
  • hydrogen peroxides hydrogen peroxides
  • N-hydroxy compounds can be prepared by oxidation of the parent amine by an oxidizing agent such as ra-CPBA. All shown and claimed nitrogen-containing compounds are also considered, when allowed by valency and structure, to cover both the compound as shown and its N-hydroxy (i.e. , N-OH) and N-alkoxy (i.e. , N-OR, wherein R is substituted or unsubstituted Ci_ 6 alkyl,
  • Ci_6 alkenyl, Ci_ 6 alkynyl, C 3 . i4 carbocycle, or 3-14-membered heterocycle) derivatives
  • Ci_ 6 alkyl is meant to include alkyl groups with 1, 2, 3, 4, 5, 6, 1-6, 1-5, 1-4, 1-3, 1-2, 2-6, 2-5, 2-4, 2-3, 3-6, 3-5, 3-4, 4-6, 4-5, and 5-6 carbons.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • Ci-6 alkyl is intended to include Ci , C2, C3, C4, C5, and Cg alkyl groups.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, s-pentyl, and n- hexyl.
  • alkyl also includes alkyl groups that have oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more hydrocarbon backbone carbon atoms, these are known as heteroalkyl,
  • a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g. , Ci- for straight chain, C3-C6 for branched chain), and in another embodiment, a straight chain or branched chain alkyl has four or fewer carbon atoms.
  • cycloalkyls have from three to eight carbon atoms in their ring structure, and in another embodiment, cycloalkyls have five or six carbons in the ring structure.
  • halo or halogen refers to fluoro, chloro, bromo, and iodo.
  • perhalogenated generally refers to a moiety wherein all hydrogens are replaced by halogen atoms.
  • the compounds of the present disclosure can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
  • hydrates include monohydrates, dihydrates, etc.
  • Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
  • Solvates means solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H 2 0, such combination being able to form one or more hydrate.
  • Tautomers refers to compounds whose structures differ markedly in arrangement of atoms, but which exist in easy and rapid equilibrium. It is to be understood that the compounds of the present disclosure may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be within the scope of the present disclosure, and the naming of the compounds does not exclude any tautomer form.
  • Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present disclosure includes all tautomers of the present compounds [0073]
  • a tautomer is one of two or more structural isomers that exist in equilibrium and are readily converted from one isomeric form to another. This reaction results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. The concept of tautomers that are interconvertible by tautomerizations is called tautomerism.
  • a "pharmaceutical composition” is a formulation containing the disclosed compounds in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler, or a vial.
  • the quantity of active ingredient (e.g. , a formulation of the disclosed compound or salt, hydrate, solvate, or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved.
  • active ingredient e.g. , a formulation of the disclosed compound or salt, hydrate, solvate, or isomer thereof
  • the dosage will also depend on the route of administration.
  • routes of administration A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of a compound of this present disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
  • a "subject” includes mammals, e.g. , humans, companion animals (e.g. , dogs, cats, birds, and the like), farm animals (e.g. , cows, sheep, pigs, horses, fowl, and the like) and laboratory animals (e.g. , rats, mice, guinea pigs, birds, and the like).
  • the subject is human.
  • the phrase "pharmaceutically acceptable” refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable excipient” means an excipient that is useful in preparing a
  • composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • a "pharmaceutically acceptable excipient" as used in the specification and claims includes both one and more than one such excipient.
  • “Pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2- acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxy maleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic,
  • Other examples include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo-[2.2.2]- oct-2-ene-l -carboxylic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like.
  • the present disclosure also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • the pharmaceutically acceptable salts of the present disclosure can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile can be used.
  • non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile can be used.
  • Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 18th ed. (Mack Publishing Company, 1990).
  • salts can include, but are not limited to, the hydrochloride and acetate salts of the aliphatic amine -containing, hydroxyl amine - containing, and imine
  • esters For example a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g. , a. methyl, ethyl, or other ester. Also, an alcohol group in a compound can be converted to its corresponding ester, e.g. , an acetate, propionate, or other ester.
  • the compounds of the present disclosure can also be prepared as prodrugs, for example
  • prodrug and prodrug are used interchangeably.
  • prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g. , solubility, bioavailability, manufacturing, etc.) the compounds of the present disclosure can be delivered in prodrug form.
  • the present disclosure is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same.
  • Prodrugs are intended to include any covalently bonded carriers that release an active parent drug of the present disclosure in vivo when such prodrug is administered to a subject.
  • Prodrugs the present disclosure are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Prodrugs include compounds of the present disclosure wherein a hydroxy, amino, sulfhydryl, carboxy, or carbonyl group is bonded to any group that, may be cleaved in vivo to form a free hydroxyl, free amino, free sulfhydryl, free carboxy or free carbonyl group, respectively.
  • prodrugs include, but are not limited to, esters (e.g. , acetate, dialkylaminoacetates, formates, phosphates, sulfates, and benzoate derivatives) and carbamates (e.g. , N,N- dimethylaminocarbonyl) of hydroxy functional groups, esters groups (e.g. ethyl esters, morpholinoethanol esters) of carboxyl functional groups, N-acyl derivatives (e.g.
  • N-acetyl) N- Mannich bases Schiff bases and enaminones of amino functional groups, oximes, acetals, ketals and enol esters of ketone and aldehyde functional groups in compounds of Formula I, and the like, See Bundegaard, H. "Design of Prodrugs" p. 1-92, Elesevier, New York-Oxford (1985).
  • the term "mitochondrial aldehyde dehydrogenase- 1" or "ALDHlAl” refers to an enzyme that oxidizes an aldehyde (e.g., a xenogenic aldehyde, a biogenic aldehyde, or an aldehyde produced from a compound that is ingested, inhaled, or absorbed) to its corresponding acid in an NAD + -dependent reaction.
  • ALDHlAl oxidizes aldehydes derived from the breakdown of compounds, e.g., toxic compounds that are ingested, that are absorbed, that are inhaled, or that are produced during normal metabolism.
  • ALDHlAl encompasses ALDHlAl from various species. Amino acid sequences of ALDHlAl are publicly available. For example, a human ALDHlAl amino acid sequence is found under GenBank Accession Nos. NM_000689; a mouse ALDHlAl amino acid sequence is found under GenBank Accession No. NM_013467; and a rat ALDHlAl amino acid sequence is found under GenBank Accession No. NM_022407.
  • ALDHlAl as used herein also encompasses fragments, fusion proteins, and variants (e.g., variants having one or more amino acid substitutions, addition, deletions, and/or insertions) that retain ALDHlAl enzymatic activity. Specific enzymatically active ALDHlAl variants, fragments, fusion proteins, and the like can be verified by adapting the methods described herein.
  • An example of an ALDHlAl variant is an ALDHlAl with a 17 base pair (bp) deletion (-416/-432) or a 3 bp insertion (-524) in the
  • ALDHlAl promoter region designated as ALDH1A1*2 and ALDH1A1*3, respectively
  • An ALDHlAl variant retains at least about 1% of the enzymatic activity of a corresponding wild-type ALDHlAl enzyme.
  • Chloride Product D then undergoes thiolation by thiourea (E) to yield Thiourea Compound F.
  • the desired final product G is formed by nucleophilic substitution of Thiourea Compound F with a halide (R 3 X).
  • the present disclosure provides various treatment and prevention methods, generally involving administering to a subject an effective amount of a compound of the present disclosure.
  • Diseases and conditions associated with ALDH1A1 include cancer.
  • Treating includes any effect, e.g., lessening, reducing, modulating, or eliminating, that results in the improvement of the condition, disease, disorder, etc.
  • Treating or “treatment” of a disease state includes: (1) inhibiting the disease state, i.e., arresting the development of the disease state or its clinical symptoms; (2) relieving the disease state, i.e., causing temporary or permanent regression of the disease state or its clinical symptoms; or (3) reducing or lessening the symptoms of the disease state.
  • Preventing refers to causing the clinical symptoms of the disease state not to develop in a subject that may be exposed to or predisposed to the disease state, but does not yet experience or display symptoms of the disease state.
  • the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a pharmaceutical composition as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound is selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the method as described herein further comprises administering a cancer chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non- peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • HDAC inhibitors histone- deacetylase inhibitors
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound and the chemotherapeutic agent are administered
  • the method as described herein further comprises administering ionizing radiation.
  • the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
  • the present disclosure provides a method of treating or preventing cancer in a subject comprising administering to the subject a first amount of a compound as described herein in a first treatment procedure, and a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
  • the radiation is a radiopharmaceutical agent.
  • the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
  • the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
  • the method described herein provides a synergistic effect in the treatment of cancer.
  • the present disclosure relates to a method of increasing the
  • the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma ( ⁇ ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the cancer is ovarian cancer.
  • the present disclosure provides methods of treating or preventing cancer in a subject.
  • the methods generally involve administering to a subject an effective amount of a compound of the present disclosure in conjunction with a standard cancer therapy.
  • Standard cancer therapies include surgery (e.g., surgical removal of cancerous tissue), radiation therapy, bone marrow transplantation, chemotherapeutic treatment, biological response modifier treatment, and certain combinations of the foregoing.
  • Radiation therapy includes, but is not limited to, x-rays or gamma rays that are delivered from either an externally applied source such as a beam, or by implantation of small radioactive sources.
  • Chemotherapeutic agents are compounds that reduce proliferation of cancer cells, and encompass cytotoxic agents and cytostatic agents.
  • Non-limiting examples of chemotherapeutic agents include alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids, and steroid hormones.
  • Agents that act to reduce cellular proliferation are known in the art and widely used.
  • Such agents include alkylating agents, such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (Cytoxan.TM.), melphalan (L-sarcolysin), carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine, trie thylenethiophosphor amine, busulfan, dacarbazine, and temozolomide.
  • alkylating agents such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfon
  • Antimetabolite agents include folic acid analogs, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors, including, but not limited to, cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FudR), 6-thioguanine, 6-mercaptopurine (6-MP), pentostatin, 5 -fluorouracil (5-FU), methotrexate, 10-propargyl-5,8-dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid (DDATHF), leucovorin, fludarabine phosphate, pentostatine, and gemcitabine.
  • CYTOSAR-U cytarabine
  • cytosine arabinoside including, but not limited to, fluorouracil (5-FU), floxuridine (FudR), 6-thioguanine, 6-
  • Suitable natural products and their derivatives e.g., vinca alkaloids, antitumor
  • antibiotics include, but are not limited to, Ara- C, paclitaxel (Taxol®), docetaxel (Taxotere®), deoxycoformycin, mitomycin-C, L-asparaginase, azathioprine; brequinar; alkaloids, e.g. vincristine, vinblastine, vinorelbine, vindesine, etc.;
  • podophyllotoxins e.g. etoposide, teniposide, etc.
  • antibiotics e.g. anthracycline, daunorubicin hydrochloride (daunomycin, rubidomycin, cerubidine), idarubicin, doxorubicin, epirubicin and morpholino derivatives, etc.
  • phenoxizone biscyclopeptides e.g. dactinomycin
  • glycopeptides e.g. bleomycin
  • anthraquinone glycosides e.g. plicamycin (mithramycin)
  • anthracenediones e.g. mitoxantrone
  • azirinopyrrolo indolediones e.g. mitomycin
  • macrocyclic immunosuppressants e.g. cyclosporine, FK-506 (tacrolimus, prograi), rapamycin, etc.; and the like.
  • anti-proliferative cytotoxic agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
  • Microtubule affecting agents that have antiproliferative activity are also suitable for use and include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolstatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®), Taxol® derivatives, docetaxel (Taxotere®), thiocolchicine (NSC 361792), trityl cysterin, vinblastine sulfate, vincristine sulfate, natural and synthetic epothilones including but not limited to, eopthilone A, epothilone B, discodermolide; estramustine, nocodazole, and the like.
  • Hormone modulators and steroids that are suitable for use include, but are not limited to, adrenocorticosteroids, e.g. prednisone, dexamethasone, etc.;
  • estrogens and pregestins e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.; and adrenocortical suppressants, e.g.
  • estradiosteroids may inhibit T cell proliferation.
  • chemotherapeutic agents include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine;
  • epidophyllotoxin a topoisomerase inhibitor
  • procarbazine mitoxantrone
  • leucovorin tegafur
  • Other anti-proliferative agents of interest include immunosuppressants, e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-(3-(4- morpholinyl)propoxy)qu- inazoline); etc.
  • immunosuppressants e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)
  • Taxanes include paclitaxel, as well as any active taxane derivative or pro-drug.
  • “Paclitaxel” (which should be understood herein to include analogues, formulations, and derivatives such as, for example, docetaxel, TAXOLTM, TAXOTERETM (a formulation of docetaxel), 10-desacetyl analogs of paclitaxel and 3 'N-desbenzoyl-3 'N-t-butoxycarbonyl analogs of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see also WO 94/07882, WO 94/07881, WO 94/07880, WO 94/07876, WO 93/23555, WO 93/10076; U.S. Pat. Nos.
  • Biological response modifiers suitable for use in connection with the methods of the present disclosure include, but are not limited to, (1) inhibitors of tyrosine kinase (RTK) activity; (2) inhibitors of serine/threonine kinase activity; (3) tumor-associated antigen antagonists, such as antibodies that bind specifically to a tumor antigen; ( 4) apoptosis receptor agonists; (5) interleukin-2; (6) interferon- a.; (7) interferon - ⁇ ; (8) colony-stimulating factors; (9) inhibitors of angiogenesis; and (10) antagonists of tumor necrosis factor.
  • RTK tyrosine kinase
  • the present disclosure provides a use of a pharmaceutical
  • composition as disclosed herein for the manufacture of a medicament for treating or preventing cancer.
  • the present disclosure provides a use of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer.
  • the present disclosure provides a use of a compound selected from
  • Table 1 herein or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament for treating or preventing cancer.
  • the medicament further comprises a cancer chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound and the chemotherapeutic agent are administered simultaneously, staggered, alternating, or sequentially.
  • the use further comprises administering ionizing radiation.
  • the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
  • the present disclosure provides a use of a first amount of a
  • compound of formula (I) for the manufacture of a medicament for treating or preventing cancer in a first treatment procedure, wherein the use further comprises a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
  • the radiation is a radiopharmaceutical agent.
  • the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
  • the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
  • the use provides a synergistic effect in the treatment of cancer.
  • the present disclosure provides a use of a compound of formula (I) or a pharmaceutical composition as disclosed above for the manufacture of a medicament for increasing the sensitivity of cancerous cells or cancer stem cells to a chemotherapeutic agent.
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral.
  • the compound or pharmaceutical composition is administered to the respiratory tract.
  • the present disclosure relates to a pharmaceutical composition
  • R 1 and R 2 are each independently selected from hydrogen and C 1 -C6 alkyl
  • R 3 and R 4 are each independently selected from hydrogen and C 1 -C6 alkyl
  • each R 7 is independently selected from halogen, C 1 -C6 alkyl, and OCH 3 , and
  • n 0, 1, 2, 3, 4, or 5;
  • one of R 1 and R 2 is hydrogen and the other is Ci-C 6 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is straight-chain Ci-C 6 alkyl or branched C3-C6 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is straight-chain d- C 4 alkyl or branched C3-C4 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is straight-chain C 1 -C 4 alkyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is branched C3-C4 alkyl.
  • one of R 3 and R 4 is hydrogen and the other is Ci-C 6 alkyl. In one embodiment, one of R 3 and R 4 is hydrogen and the other is methyl or ethyl.
  • one of R 1 and R 2 is hydrogen and the other is straight-chain C 1 -C 4 alkyl; and one of R 3 and R 4 is hydrogen and the other is methyl or ethyl. In one embodiment, one of R 1 and R 2 is hydrogen and the other is branched C3-C4 alkyl; and one of R 3 and R 4 is hydrogen and the other is methyl or ethyl.
  • n is 0, 1, 2, or 3. In one embodiment, n is 1, 2, or 3. In one
  • n is 1. In one embodiment, n is 2.
  • each R 7 is Ci-C 6 alkyl. In one embodiment, each R 7 is independently methyl, ethyl, or isopropyl. In one embodiment, each R 7 is halogen. In one embodiment, each R 7 is independently chloro or fluoro. In one embodiment, R 7 is chloro. In one embodiment, R 7 is fluoro. In one embodiment, one or more R 7 is Ci-C 6 alkyl and one or more R 7 is halogen. In one embodiment, one or more R 7 is independently methyl, ethyl, or isopropyl and one or more R 7 is independently chloro or fluoro. In one embodiment, one or more R 7 is Ci-C 6 alkyl and one or more R 7 is OCH 3 .
  • one or more R 7 is independently methyl, ethyl, or isopropyl and one or more R 7 is OCH 3 . In one embodiment, one or more R 7 is halogen and one or more R 7 is OCH 3 . In one example, one or more R 7 is independently chloro or fluoro and one or more R 7 is OCH 3 .
  • a compound of the present disclosure is of one of the following formulae:
  • the present disclosure relates to a pharmaceutical composition
  • the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
  • non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells
  • cytotoxic agents cytostatic agents
  • alkylating agents nitrosoureas
  • antimetabolites antitumor antibiotics
  • taxanes taxanes
  • microtubule affecting agents metal complexes
  • steroid hormones e.g., tyrosine kinase and/
  • the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma ( ⁇ ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
  • the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral. In one embodiment, the compound or pharmaceutical composition is administered to the respiratory tract.
  • the present disclosure provides pharmaceutical compositions comprising a compound of the present disclosure (where a compound of the present disclosure is also referred to herein as "active agent” of the present disclosure, or “subject active agent”).
  • the compound of the present disclosure can be formulated with one or more pharmaceutically acceptable excipients.
  • a wide variety of pharmaceutically acceptable excipients are known in the art and need not be discussed in detail herein.
  • Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy," 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H. C.
  • compositions such as vehicles, adjuvants, carriers or diluents
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • a subject ALDH1A1 activity inhibitor may be administered to the host using any convenient means capable of treating cancer.
  • a subject ALDH1A1 activity inhibitor can be incorporated into a variety of formulations for therapeutic administration. More particularly, a subject ALDH1A1 activity inhibitor can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • a subject active agent may be administered in the form of their pharmaceutically acceptable salts, or a subject active agent may be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • a subject active agent can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • a subject active agent can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol
  • solubilizers such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • a subject active agent can be utilized in aerosol formulation to be administered via
  • a subject active agent can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
  • a subject active agent can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • An active agent can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycol monomethyl ethers, which melt at body temperature, yet are solidified at room temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and
  • each dosage unit for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the subject active agent.
  • unit dosage forms for injection or intravenous administration may comprise a subject active agent in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a subject active agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for a subject active agent depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • a subject active agent can be formulated for administration by injection.
  • injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation may also be emulsified or the active ingredient encapsulated in liposome vehicles.
  • a subject active agent is delivered by a continuous delivery system.
  • continuous delivery system is used interchangeably herein with “controlled delivery system” and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985.
  • the composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
  • the subject compounds may be administered in dosages of, for example, 0.1 ⁇ g to 10 mg/kg body weight per day.
  • the range is broad, since in general the efficacy of a therapeutic effect for different mammals varies widely with doses typically being 20, 30 or even 40 times smaller (per unit body weight) in man than in the rat.
  • the mode of administration can have a large effect on dosage.
  • oral dosages may be about ten times the injection dose. Higher doses may be used for localized routes of delivery.
  • a subject ALDH1A1 activity modulator can be administered in an amount of from about 1 mg to about 1000 mg per dose, e.g., from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 50 mg, from about 50 mg to about 75 mg, from about 75 mg to about 100 mg, from about 100 mg to about 125 mg, from about 125 mg to about 150 mg, from about 150 mg to about 175 mg, from about 175 mg to about 200 mg, from about 200 mg to about 225 mg, from about 225 mg to about 250 mg, from about 250 mg to about 300 mg, from about 300 mg to about 350 mg, from about 350 mg to about 400 mg, from about 400 mg to about 450 mg, from about 450 mg to about 500 mg, from about 500 mg to about 750 mg, or from about 750 mg to about 1000 mg per dose.
  • An exemplary dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time -release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient, etc.
  • the time -release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
  • dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and
  • each dosage unit for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more compounds of the present disclosure.
  • unit dosage forms for injection or intravenous administration may comprise the compound (s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • multiple doses of a subject compound are administered.
  • a subject compound is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (qid), or three times a day (tid).
  • a subject compound is administered continuously.
  • the duration of administration of a subject compound can vary, depending on any of a variety of factors, e.g., patient response, etc.
  • a subject compound can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • a subject compound is administered for the lifetime of the individual.
  • a subject ALDH1A1 activity inhibitor is administered to an individual using any combination
  • Administration can be acute (e.g., of short duration, e.g., a single administration, administration for one day to one week), or chronic (e.g., of long duration, e.g., administration for longer than one week, e.g., administration over a period of time from about 2 weeks to about one month, from about one month to about 3 months, from about 3 months to about 6 months, from about 6 months to about 1 year, or longer than one year).
  • routes of administration include intranasal, intramuscular, intratracheal, subcutaneous, intradermal, transdermal, sublingual, topical application, intravenous, rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the agent and/or the desired effect. The compound can be administered in a single dose or in multiple doses.
  • An active agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes.
  • routes of administration contemplated by the present disclosure include, but are not necessarily limited to, enteral, parenteral, or inhalational routes.
  • Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular, intraorbital, intracapsular, intraspinal, intrasternal, and intravenous routes, i.e., any route of administration other than through the alimentary canal.
  • Parenteral administration can be carried to effect systemic or local delivery of the agent. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
  • the agent can also be delivered to the subject by enteral administration.
  • Enteral routes of administration include, but are not necessarily limited to, oral and rectal (e.g., using a suppository) delivery.
  • Methods of administration of the agent through the skin or mucosa include, but are not necessarily limited to, topical application of a suitable pharmaceutical preparation, transdermal transmission, injection and epidermal administration.
  • a suitable pharmaceutical preparation for transdermal transmission, absorption promoters or iontophoresis are suitable methods.
  • Iontophoretic transmission may be
  • Example 1 Inhibitory activity of compounds against ALDH1A1
  • NM_000689 The synthetic gene was cloned into the Nhel/EcoRI sites of the His-tag vector, pTrcHis, for protein expression. All the vectors were transformed into BL21 E. coli host cells and subjected to 0.5 mM isopropyl ⁇ -D-l-thiogalactopyranoside (IPTG) induction for protein expression at 30°C. Purifications of the recombinant proteins by affinity nickel columns (HisTrap, GE Healthy Science, USA) were carried out using standard protocols according to manufacturer's instructions (Novagen, USA).
  • ALDH2 ALDH3A1, ALDH4A1, ALDH7A1, and ADH1B1 was determined.
  • Example 3 In vitro anti-cancer activity of the compounds in combination with paclitaxel and doxorubicin
  • Method Cell cultures (5000 cells/well) in medium containing 5% fetal bovine serum
  • FBS fetal bovine serum
  • Basal cells were fixed with 25 ⁇ of 50% (w/v) trichloroacetic acid (TCA) (final 10%) for 30-min as Tz.
  • Compound 5 at various concentrations was added to the cell cultures and then the suction of the basal cells with TCA was performed.
  • the basal wells were washed with phosphate buffer saline (PBS, 100 ⁇ ), and incubated for 48-h or 72-h treatment before reaction was terminated, followed by 30-min incubation at RT. After suction of the cells with TCA, all wells (including basal wells) were washed with PBS (100 ⁇ ) and air-dried for 20 min in the hood.
  • PBS phosphate buffer saline
  • SRB Sulforhodamine B

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides compounds that function as inhibitors of cytosolic aldehyde dehydrogenase- 1 (ALDHlAl) activity.

Description

ALDEHYDE DEHYDROGENASE-1 MODULATORS AND METHODS OF USE THEREOF
CROSS-REFERENCE
[0001] This application claims the benefit of U.S. Provisional Patent Application No. 61/781,793, filed March 14, 2013, which application is incorporated herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[0002] This invention was made with Government support under contract AA011147 awarded by the National Institutes of Health. The Government has certain rights in the invention.
INTRODUCTION
[0003] Aldehyde dehydrogenase- 1 (ALDH1A1) is mitochondria nuclear encoded cytosolic enzyme.
ALDH1 Al has been widely recognized as a key biomarker for cancer stem cell (CSC). Strong support for the CSC hypothesis, which proposes that a relatively rare subpopulation of tumor cells have the unique ability to initiate and perpetuate tumor growth has been published in the past decade. These cells, called cancer stem cells or tumor-initiating cells, share various characteristics with embryonic and somatic stem cells including self-renewal and multi-potent differentiation. CSCs may be highly resistant to radiation or chemotherapy; therefore, the development of more effective therapies for cancer requires effective targeting of this cell population. These cells can be identified and isolated using biomarkers such as ALDH1A1.
[0004] ALDHlAl isozymes, such as ALDH1A1, metabolize a wide variety of intracellular aldehydes and can thus provide resistance to reactive oxygen species (ROS)-derived products or alkylating agents such as cyclophosphamide. High ALDHlAl activity has been reported in CSCs from many cancers including breast, lung, liver, colon, pancreas, ovary, head and neck, prostate cancers and some hematologic cancers. In the literature, silencing ALDHlAl by siRNA or shRNA leads to cell cycle arrest, apoptosis, decreased ceil viability in vitro, and reduced tumorigenesis in vivo. Silencing ALDHlAl by siRNA also sensitizes cancer cells to drug- induced cell death. For example, in an in vivo orthotopic mouse model of ovarian cancer, silencing ALDHlAl using nanoliposomal siRNA sensitized both taxane- and platinum-resistant cell lines to chemotherapy, significantly reducing tumor growth in mice compared with chemotherapy alone (74%-90% reduction; P < 0.015). [Landen, C. N. et al. Mo! Cancer Ther. 2010 Dec;9(12):3186-99; Moreb, J. S. et al., Molecular Cancer 2008, 7:87.] [0005] ALDHlAl is not only a biomarker of CSCs, but also an attractive therapeutic target for human cancer,
[0006] Antagonists of ALDHlAl are useful for treating disorders such as cancer. Compounds of the present disclosure are antagonist of ALDHlAl, or inhibitors of ALDHlAl and are described herein.
SUMMARY
[0007] The present disclosure provides compounds that function as inhibitors or antagonists of aldehyde dehydrogenase- 1 (ALDHlAl) activity, pharmaceutical compositions containing them, and methods of preparing these compounds. In one embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound of formula (I) and a cancer
Figure imgf000003_0001
R1 and R2 are each independently selected from hydrogen and C1-C6 alkyl;
R3 and R4 are each independently selected from hydrogen and C1-C6 alkyl;
each R7 is independently selected from halogen, C1-C6 alkyl, and OCH3; and
n is 0, 1, 2, 3, 4, or 5;
provided that when one of R1 and R2 is hydrogen and the other is isopropyl,
Figure imgf000003_0002
[0008] In one embodiment, one of R1 and R2 is hydrogen and the other is Ci-C6 alkyl. In one embodiment, one of R3 and R4 is hydrogen and the other is C1-C6 alkyl. In one embodiment, one of R3 and R4 is hydrogen and the other is methyl or ethyl. In one embodiment, n is 0, 1, 2, or 3. In one embodiment, n is 1, 2, or 3. In one embodiment, n is 1. In one embodiment, n is 2. In one embodiment, each R7 is C1-C6 alkyl. In one embodiment, R7 is methyl, ethyl, or isopropyl. In one embodiment, each R7 is halogen. In one embodiment, R7 is fluoro.
[0009] In one embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound selected from Table 1 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a cancer chemotherapeutic agent.
[0010] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[0011] In another embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[0012] In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a pharmaceutical composition as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound is selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0013] In another embodiment, the method as described herein further comprises administering a cancer chemotherapeutic agent. In one embodiment, the chemotherapeutic agent is selected from non- peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor. In one embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[0014] In one embodiment, the compound and the chemotherapeutic agent are administered
simultaneously, staggered, alternating, or sequentially.
[0015] In one embodiment, the method as described herein further comprises administering ionizing radiation. In one embodiment, the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
[0016] In one embodiment, the present disclosure provides a method of treating or preventing cancer in a subject comprising administering to the subject a first amount of a compound as described herein in a first treatment procedure, and a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount. In one embodiment, the radiation is a radiopharmaceutical agent. In one embodiment, the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
[0017] In one embodiment, the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
[0018] In another embodiment, the method described herein provides a synergistic effect in the treatment of cancer.
[0019] In one embodiment, the present disclosure relates to a method of increasing the sensitivity of a cancerous cells or cancer stem cells to a chemotherapeutic agent comprising administering to a subject a compound as described herein or a pharmaceutical composition as described herein. In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone-deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor. In one embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor. [0020] In one embodiment, the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral. In one embodiment, the compound or pharmaceutical composition is administered to the respiratory tract.
[0021] In one embodiment, the present disclosure provides a use of a pharmaceutical composition as disclosed above for the manufacture of a medicament for treating or preventing cancer.
[0022] In one embodiment, the present disclosure provides a use of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer.
[0023] In one embodiment, the present disclosure provides a use of a compound selected from Table 1 herein, or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament for treating or preventing cancer.
[0024] In one embodiment, the medicament further comprises a cancer chemotherapeutic agent.
[0025] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[0026] In one embodiment, the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[0027] In one embodiment, the compound and the chemotherapeutic agent are administered
simultaneously, staggered, alternating, or sequentially.
[0028] In one embodiment, the use further comprises administering ionizing radiation.
[0029] In one embodiment, the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
[0030] In one embodiment, the present disclosure provides a use of a first amount of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer in a first treatment procedure, wherein the use further comprises a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
[0031] In one embodiment, the radiation is a radiopharmaceutical agent. [0032] In one embodiment, the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
[0033] In one embodiment, the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
[0034] In one embodiment, the use provides a synergistic effect in the treatment of cancer.
[0035] In one embodiment, the present disclosure provides a use of a compound of formula (I) or a pharmaceutical composition as disclosed above for the manufacture of a medicament for increasing the sensitivity of cancerous cells or cancer stem cells to a chemotherapeutic agent.
[0036] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[0037] In one embodiment, the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[0038] In one embodiment, the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral.
[0039] In one embodiment, the compound or pharmaceutical composition is administered to the
respiratory tract.
BRIEF DESCRIPTION OF THE DRAWINGS
[0040] Figure 1 is a graph showing the selectivity of Compound 5 against different ALDH isozymes.
[0041] Figure 2A is a graph showing that Compound 5 sensitizes cytotoxic effect of paclitaxel in human multi-drug resistant ovarian NCI/ADR-RES cancer cells.
[0042] Figure 2B is a graph showing that Compound 5 sensitizes cytotoxic effect of paclitaxel in human multi-drug resistant ovarian TOV-21G-RT cancer cells.
[0043] Figure 3A is a graph showing that Compound 5 sensitizes cytotoxic effect of doxorubicin in human multi-drug resistant ovarian NCI/ADR-RES cancer cells. [0044] Figure 3B is a graph showing that Compound 5 sensitizes cytotoxic effect of doxorubicin in human multi-drug resistant ovarian TOV-21G-RT cancer cells.
DETAILED DESCRIPTION
1. Compounds of the Present Disclosure
[0045]
Figure imgf000008_0001
R1 and R2 are each independently selected from hydrogen and Ci-C6 alkyl;
R3 and R4 are each independently selected from hydrogen and C1-C6 alkyl;
each R7 is independently selected from halogen, C1-C6 alkyl, and OCH3; and
n is 0, 1, 2, 3, 4, or 5;
provided that when one of R1 and R2 is hydrogen and the other is isopropyl,
Figure imgf000008_0002
[0046] In one embodiment, one of R1 and R2 is hydrogen and the other is Ci-C6 alkyl. In one
embodiment, one of R1 and R2 is hydrogen and the other is straight-chain Ci-C6 alkyl or branched C3-C6 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain d- C4 alkyl or branched C3-C4 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain C1-C4 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is branched C3-C4 alkyl. [0047] In one embodiment, one of R3 and R4 is hydrogen and the other is Ci-C6 alkyl. In one embodiment, one of R3 and R4 is hydrogen and the other is methyl or ethyl.
[0048] In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain C1-C4 alkyl; and one of R3 and R4 is hydrogen and the other is methyl or ethyl. In one embodiment, one of R1 and R2 is hydrogen and the other is branched C3-C4 alkyl; and one of R3 and R4 is hydrogen and the other is methyl or ethyl.
[0049] In one embodiment, n is 0, 1, 2, or 3. In one embodiment, n is 1, 2, or 3. In one embodiment, n is 1. In one embodiment, n is 2.
[0050] In one embodiment, each R7 is Ci-C6 alkyl. In one embodiment, each R7 is independently methyl, ethyl, or isopropyl. In one embodiment, each R7 is halogen. In one embodiment, each R7 is independently chloro or fluoro. In one embodiment, R7 is chloro. In one embodiment, R7 is fluoro. In one embodiment, one or more R7 is Ci-C6 alkyl and one or more R7 is halogen. In one embodiment, one or more R7 is independently methyl, ethyl, or isopropyl and one or more R7 is independently chloro or fluoro. In one embodiment, one or more R7 is Ci-C6 alkyl and one or more R7 is OCH3. In one example, one or more R7 is independently methyl, ethyl, or isopropyl and one or more R7 is OCH3. In one embodiment, one or more R7 is halogen and one or more R7 is OCH3. In one example, one or more R7 is independently chloro or fluoro and one or more R7 is OCH3.
[0051] It will be apparent to one skilled in the art based one the present disclosure that any of the
embodiments described above for each of R1, R2, R3, R4, R7, and n can be combined in any manner.
[0052] ae:
Figure imgf000009_0001
Figure imgf000010_0001
Representative compounds of the present disclosure include compounds listed in Table 1. Table 1
Figure imgf000010_0002
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
[0054] The present disclosure further provides assays for identifying inhibitors of ALDHlAl.
[0055] In some embodiments, subjects to be treated are humans. In some embodiments, a human to be treated according to a subject method is one that has the wild type ALDHlAl allele or a mutated/variant ALDHlAl allele, e.g. a mutated/variant ALDHlAl gene with over-expression of the ALDHlAl transcripts/enzyme, or a mutated/variant ALDHlAl gene which encodes a product with higher enzymatic activity of ALDHlAl. [Spence, J. P. et al., Alcohol Clin Exp Res. 2003 Sep;27(9): 1389-94.]
[0056] A compound can be readily ascertained whether it is an ALDHlAl inhibitor or not. Assays for dehydrogenase activity of ALDHlAl are known in the art, and any known assay can be used. Examples of dehydrogenase assays are found in various publications, including, e.g., Russo and Hilton, 1998, Cancer Res. 48:2963-8; Ho et al., 2006, Biochemistry 45:9445-53; Sheikh et al., 1997, J. Biol. Chem. 272:18817-18822; Vallari and Pietruszko, 1984, J. Biol. Chem. 259:4922; and Farres et al., 1994, J. Biol. Chem. 269:13854-13860. The assays generally comprise two parts; one is the preparation of human ALDHlAl recombinant enzyme; and the other is the determination of the enzyme inhibition by a compound in the present disclosure.
[0057] To prepare human ALDHlAl recombinant enzyme, full-length human ALDHlAl cDNA may be synthesized based on published DNA sequence (e.g., GenBank ID: NM_000689). The synthetic gene may be cloned into the Nhel/EcoRI sites of the His-tag vector, pTrcHis, for protein expression. All the vectors are transformed into BL21 E. coli host cells and subjected to 0.5 niM isopropyl β-D-l-thiogalactopyranoside (IPTG) induction for protein expression at 30 °C.
Purifications of the recombinant proteins by affinity nickel columns (e.g., HisTrap, GE Healthy Science, USA) are carried out using standard protocols according to manufacturer's instructions (e.g., Novagen, USA).
[0058] An example of the enzymatic assay is described as follows. The reaction cocktails are added into the wells in a 96-well plate and then put on a shaker for mixing. After 5 minutes of mixing, 2 of recombinant ALDHlAl enzyme is added to each well with DMSO as blank control in the plate. The plate is again put on shaker for another 5 minutes of shaking to enhance bonding between testing compounds and the enzyme. 2μί of 1M acetyladehyde in water is then added to each of wells containing the above reaction mixture. After a short vigorous mixing, the UV plate is fed into a plate reader to measure kinetic optical density (O.D.) at wavelength 340 nm at 25 °C over 4 minutes. Each point of measurement is taken at a time interval of 40 seconds.
[0059] The term "compounds of the present disclosure" refers to a compound according to formula I.
[0060] With respect to the chemical compounds useful in the present disclsoure, the following terms can be applicable:
[0061] The present disclosure is intended to include all isotopes of atoms occurring in the present
compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include C-13 and C-14. [0062] When any variable (e.g. , Ri) occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 Ri moieties, then the group may optionally be substituted with up to two Ri moieties and Ri at each occurrence is selected independently from the definition of R^ Also, combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds.
[0063] When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom in the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent. Combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds.
[0064] Compounds of the present disclosure that contain nitrogens can be converted to N-oxides by treatment with an oxidizing agent (e.g. , 3-chloroperoxybenzoic acid (ra-CPBA) and/or hydrogen peroxides) to afford other compounds of the present disclosure. Thus, all shown and claimed nitrogen-containing compounds are considered, when allowed by valency and structure, to include both the compound as shown and its N-oxide derivative (which can be designated as N— >0 or N+-0"). Furthermore, in other instances, the nitrogens in the compounds of the present disclosure can be converted to N-hydroxy or N-alkoxy compounds. For example, N-hydroxy compounds can be prepared by oxidation of the parent amine by an oxidizing agent such as ra-CPBA. All shown and claimed nitrogen-containing compounds are also considered, when allowed by valency and structure, to cover both the compound as shown and its N-hydroxy (i.e. , N-OH) and N-alkoxy (i.e. , N-OR, wherein R is substituted or unsubstituted Ci_6 alkyl,
Ci_6 alkenyl, Ci_6 alkynyl, C3.i4 carbocycle, or 3-14-membered heterocycle) derivatives.
[0065] When an atom or chemical moiety is followed by a subscripted numeric range (e.g. , Ci- ), the present disclosure is meant to encompass each number within the range as well as all intermediate ranges. For example, "Ci_6 alkyl" is meant to include alkyl groups with 1, 2, 3, 4, 5, 6, 1-6, 1-5, 1-4, 1-3, 1-2, 2-6, 2-5, 2-4, 2-3, 3-6, 3-5, 3-4, 4-6, 4-5, and 5-6 carbons.
[0066] As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, Ci-6 alkyl is intended to include Ci , C2, C3, C4, C5, and Cg alkyl groups. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, s-pentyl, and n- hexyl. When the term "alkyl" also includes alkyl groups that have oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more hydrocarbon backbone carbon atoms, these are known as heteroalkyl, In certain embodiments, a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g. , Ci- for straight chain, C3-C6 for branched chain), and in another embodiment, a straight chain or branched chain alkyl has four or fewer carbon atoms. Likewise, cycloalkyls have from three to eight carbon atoms in their ring structure, and in another embodiment, cycloalkyls have five or six carbons in the ring structure.
[0067] As used herein, "halo" or "halogen" refers to fluoro, chloro, bromo, and iodo. The term
"perhalogenated" generally refers to a moiety wherein all hydrogens are replaced by halogen atoms.
[0068] Additionally, the compounds of the present disclosure, for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Nonlimiting examples of hydrates include monohydrates, dihydrates, etc.
Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
[0069] "Solvates" means solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H20, such combination being able to form one or more hydrate.
[0070] "Tautomers" refers to compounds whose structures differ markedly in arrangement of atoms, but which exist in easy and rapid equilibrium. It is to be understood that the compounds of the present disclosure may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be within the scope of the present disclosure, and the naming of the compounds does not exclude any tautomer form.
[0071] Some compounds of the present disclosure can exist in a tautomeric form which are also intended to be encompassed within the scope of the present disclosure.
[0072] The compounds, salts and prodrugs of the present disclosure can exist in several tautomeric
forms, including the enol and imine form, and the keto and enamine form and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the present disclosure. Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present disclosure includes all tautomers of the present compounds [0073] A tautomer is one of two or more structural isomers that exist in equilibrium and are readily converted from one isomeric form to another. This reaction results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. The concept of tautomers that are interconvertible by tautomerizations is called tautomerism.
[0074] A "pharmaceutical composition" is a formulation containing the disclosed compounds in a form suitable for administration to a subject. In one embodiment, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler, or a vial. The quantity of active ingredient (e.g. , a formulation of the disclosed compound or salt, hydrate, solvate, or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of administration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for the topical or transdermal administration of a compound of this present disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In one embodiment, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
[0075] A "subject" includes mammals, e.g. , humans, companion animals (e.g. , dogs, cats, birds, and the like), farm animals (e.g. , cows, sheep, pigs, horses, fowl, and the like) and laboratory animals (e.g. , rats, mice, guinea pigs, birds, and the like). In one embodiment, the subject is human.
[0076] As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0077] "Pharmaceutically acceptable excipient" means an excipient that is useful in preparing a
pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the specification and claims includes both one and more than one such excipient.
[0078] The compounds of the present disclosure are capable of further forming salts. All of these forms are also contemplated within the scope of the present claims.
[0079] "Pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically
acceptable and that possesses the desired pharmacological activity of the parent compound.
[0080] As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed
compounds wherein the parent compound is modified by making acid or base salts thereof.
Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2- acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxy maleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic, phosphoric, polygalacturonic, propionic, salicyclic, stearic, subacetic, succinic, sulfamic, sulfanilic, sulfuric, tannic, tartaric, toluene sulfonic, and the commonly occurring amine acids, e.g. , glycine, alanine, phenylalanine, arginine, etc.
[0081] Other examples include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo-[2.2.2]- oct-2-ene-l -carboxylic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like. The present disclosure also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
[0082] It should be understood that all references to pharmaceutically acceptable salts include solvent addition forms (solvates) or crystal forms (polymorphs) as defined herein, of the same salt.
[0083] The pharmaceutically acceptable salts of the present disclosure can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile can be used. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 18th ed. (Mack Publishing Company, 1990). For example, salts can include, but are not limited to, the hydrochloride and acetate salts of the aliphatic amine -containing, hydroxyl amine - containing, and imine -containing compounds of the present disclosure.
[0084] The compounds of the present disclosure can also be prepared as esters, for example
pharmaceutically acceptable esters. For example a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g. , a. methyl, ethyl, or other ester. Also, an alcohol group in a compound can be converted to its corresponding ester, e.g. , an acetate, propionate, or other ester.
[0085] The compounds of the present disclosure can also be prepared as prodrugs, for example
pharmaceutically acceptable prodrugs. The terms "pro-drug" and "prodrug" are used
interchangeably herein and refer to any compound which releases an active parent drug in vivo. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g. , solubility, bioavailability, manufacturing, etc.) the compounds of the present disclosure can be delivered in prodrug form. Thus, the present disclosure is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same. "Prodrugs" are intended to include any covalently bonded carriers that release an active parent drug of the present disclosure in vivo when such prodrug is administered to a subject. Prodrugs the present disclosure are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present disclosure wherein a hydroxy, amino, sulfhydryl, carboxy, or carbonyl group is bonded to any group that, may be cleaved in vivo to form a free hydroxyl, free amino, free sulfhydryl, free carboxy or free carbonyl group, respectively.
[0086] Examples of prodrugs include, but are not limited to, esters (e.g. , acetate, dialkylaminoacetates, formates, phosphates, sulfates, and benzoate derivatives) and carbamates (e.g. , N,N- dimethylaminocarbonyl) of hydroxy functional groups, esters groups (e.g. ethyl esters, morpholinoethanol esters) of carboxyl functional groups, N-acyl derivatives (e.g. N-acetyl) N- Mannich bases, Schiff bases and enaminones of amino functional groups, oximes, acetals, ketals and enol esters of ketone and aldehyde functional groups in compounds of Formula I, and the like, See Bundegaard, H. "Design of Prodrugs" p. 1-92, Elesevier, New York-Oxford (1985). [0087] As used herein, the term "mitochondrial aldehyde dehydrogenase- 1" or "ALDHlAl" refers to an enzyme that oxidizes an aldehyde (e.g., a xenogenic aldehyde, a biogenic aldehyde, or an aldehyde produced from a compound that is ingested, inhaled, or absorbed) to its corresponding acid in an NAD+-dependent reaction. For example, ALDHlAl oxidizes aldehydes derived from the breakdown of compounds, e.g., toxic compounds that are ingested, that are absorbed, that are inhaled, or that are produced during normal metabolism.
[0088] The term "ALDHlAl" encompasses ALDHlAl from various species. Amino acid sequences of ALDHlAl are publicly available. For example, a human ALDHlAl amino acid sequence is found under GenBank Accession Nos. NM_000689; a mouse ALDHlAl amino acid sequence is found under GenBank Accession No. NM_013467; and a rat ALDHlAl amino acid sequence is found under GenBank Accession No. NM_022407. The term "ALDHlAl" as used herein also encompasses fragments, fusion proteins, and variants (e.g., variants having one or more amino acid substitutions, addition, deletions, and/or insertions) that retain ALDHlAl enzymatic activity. Specific enzymatically active ALDHlAl variants, fragments, fusion proteins, and the like can be verified by adapting the methods described herein. An example of an ALDHlAl variant is an ALDHlAl with a 17 base pair (bp) deletion (-416/-432) or a 3 bp insertion (-524) in the
ALDHlAl promoter region, designated as ALDH1A1*2 and ALDH1A1*3, respectively
(Alcohol Clin Exp Res. 2003, 27: 1389-94.) An ALDHlAl variant retains at least about 1% of the enzymatic activity of a corresponding wild-type ALDHlAl enzyme.
[0089] In the specification, the singular forms also include the plural, unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this present disclosure belongs. In the case of conflict, the present specification will control.
[0090] All percentages and ratios used herein, unless otherwise indicated, are by weight.
2. Synthesis of Compounds
[0091] The general procedure for preparing compounds of the present disclosure is described as follows.
Briefly, animation of 2,4-dichloroquinazoline (A) by amino ester (B) to form Compound C. Compound C undergoes intermolecular cyclization by heat to afford Chloride Product D.
Chloride Product D then undergoes thiolation by thiourea (E) to yield Thiourea Compound F. The desired final product G is formed by nucleophilic substitution of Thiourea Compound F with a halide (R3X).
Figure imgf000024_0001
Figure imgf000024_0002
2,2-dimethyl-5 hioxo-5,6-dihydroimidazo[1 ,2-c]quinazolin-3(2/-/)-one
3. Methods of Treatment and Prevention
[0092] The present disclosure provides various treatment and prevention methods, generally involving administering to a subject an effective amount of a compound of the present disclosure. Diseases and conditions associated with ALDH1A1 include cancer.
[0093] "Treating", includes any effect, e.g., lessening, reducing, modulating, or eliminating, that results in the improvement of the condition, disease, disorder, etc. "Treating" or "treatment" of a disease state includes: (1) inhibiting the disease state, i.e., arresting the development of the disease state or its clinical symptoms; (2) relieving the disease state, i.e., causing temporary or permanent regression of the disease state or its clinical symptoms; or (3) reducing or lessening the symptoms of the disease state.
[0094] "Preventing", refers to causing the clinical symptoms of the disease state not to develop in a subject that may be exposed to or predisposed to the disease state, but does not yet experience or display symptoms of the disease state.
Methods of Treating and Preventing Cancer
[0095] In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a pharmaceutical composition as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound as described herein. In one embodiment, the present disclosure provides a method of treating or preventing cancer comprising administering to a subject a compound is selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0096] In another embodiment, the method as described herein further comprises administering a cancer chemotherapeutic agent. In one embodiment, the chemotherapeutic agent is selected from non- peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor. In one embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[0097] In one embodiment, the compound and the chemotherapeutic agent are administered
simultaneously, staggered, alternating, or sequentially.
[0098] In one embodiment, the method as described herein further comprises administering ionizing radiation. In one embodiment, the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
[0099] In one embodiment, the present disclosure provides a method of treating or preventing cancer in a subject comprising administering to the subject a first amount of a compound as described herein in a first treatment procedure, and a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount. In one embodiment, the radiation is a radiopharmaceutical agent. In one embodiment, the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
[00100] In one embodiment, the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
[00101] In another embodiment, the method described herein provides a synergistic effect in the treatment of cancer.
[00102] In one embodiment, the present disclosure relates to a method of increasing the
sensitivity of a cancerous cells or cancer stem cells to a chemotherapeutic agent comprising administering to a subject a compound as described herein or a pharmaceutical composition as described herein. In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor. In one embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma (γ), colony- stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[00103] In one embodiment, the cancer is ovarian cancer.
[00104] The present disclosure provides methods of treating or preventing cancer in a subject.
The methods generally involve administering to a subject an effective amount of a compound of the present disclosure in conjunction with a standard cancer therapy. Standard cancer therapies include surgery (e.g., surgical removal of cancerous tissue), radiation therapy, bone marrow transplantation, chemotherapeutic treatment, biological response modifier treatment, and certain combinations of the foregoing.
[00105] Radiation therapy includes, but is not limited to, x-rays or gamma rays that are delivered from either an externally applied source such as a beam, or by implantation of small radioactive sources.
[00106] Chemotherapeutic agents are compounds that reduce proliferation of cancer cells, and encompass cytotoxic agents and cytostatic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids, and steroid hormones.
[00107] Agents that act to reduce cellular proliferation are known in the art and widely used. Such agents include alkylating agents, such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (Cytoxan.TM.), melphalan (L-sarcolysin), carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine, trie thylenethiophosphor amine, busulfan, dacarbazine, and temozolomide.
[00108] Antimetabolite agents include folic acid analogs, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors, including, but not limited to, cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FudR), 6-thioguanine, 6-mercaptopurine (6-MP), pentostatin, 5 -fluorouracil (5-FU), methotrexate, 10-propargyl-5,8-dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid (DDATHF), leucovorin, fludarabine phosphate, pentostatine, and gemcitabine.
[00109] Suitable natural products and their derivatives, (e.g., vinca alkaloids, antitumor
antibiotics, enzymes, lymphokines, and epipodophyllotoxins), include, but are not limited to, Ara- C, paclitaxel (Taxol®), docetaxel (Taxotere®), deoxycoformycin, mitomycin-C, L-asparaginase, azathioprine; brequinar; alkaloids, e.g. vincristine, vinblastine, vinorelbine, vindesine, etc.;
podophyllotoxins, e.g. etoposide, teniposide, etc.; antibiotics, e.g. anthracycline, daunorubicin hydrochloride (daunomycin, rubidomycin, cerubidine), idarubicin, doxorubicin, epirubicin and morpholino derivatives, etc.; phenoxizone biscyclopeptides, e.g. dactinomycin; basic
glycopeptides, e.g. bleomycin; anthraquinone glycosides, e.g. plicamycin (mithramycin);
anthracenediones, e.g. mitoxantrone; azirinopyrrolo indolediones, e.g. mitomycin; macrocyclic immunosuppressants, e.g. cyclosporine, FK-506 (tacrolimus, prograi), rapamycin, etc.; and the like.
[00110] Other anti-proliferative cytotoxic agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
[00111] Microtubule affecting agents that have antiproliferative activity are also suitable for use and include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolstatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®), Taxol® derivatives, docetaxel (Taxotere®), thiocolchicine (NSC 361792), trityl cysterin, vinblastine sulfate, vincristine sulfate, natural and synthetic epothilones including but not limited to, eopthilone A, epothilone B, discodermolide; estramustine, nocodazole, and the like.
[00112] Hormone modulators and steroids (including synthetic analogs) that are suitable for use include, but are not limited to, adrenocorticosteroids, e.g. prednisone, dexamethasone, etc.;
estrogens and pregestins, e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.; and adrenocortical suppressants, e.g. aminoglutethimide; 17a-ethinylestradiol; diethylstilbestrol, testosterone, fluoxymesterone, dromostanolone propionate, testolactone, methylprednisolone, methyl-testosterone, prednisolone, triamcinolone, chlorotrianisene, hydroxyprogesterone, aminoglutethimide, estramustine, medroxyprogesterone acetate, leuprolide, Flutamide (Drogenil), Toremifene (Fareston), and Zoladex®. Estrogens stimulate proliferation and differentiation; therefore, compounds that bind to the estrogen receptor are used to block this activity. Corticosteroids may inhibit T cell proliferation. [00113] Other chemotherapeutic agents include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine;
epidophyllotoxin; a topoisomerase inhibitor; procarbazine; mitoxantrone; leucovorin; tegafur; etc. Other anti-proliferative agents of interest include immunosuppressants, e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-(3-(4- morpholinyl)propoxy)qu- inazoline); etc.
[00114] "Taxanes" include paclitaxel, as well as any active taxane derivative or pro-drug.
"Paclitaxel" (which should be understood herein to include analogues, formulations, and derivatives such as, for example, docetaxel, TAXOL™, TAXOTERE™ (a formulation of docetaxel), 10-desacetyl analogs of paclitaxel and 3 'N-desbenzoyl-3 'N-t-butoxycarbonyl analogs of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see also WO 94/07882, WO 94/07881, WO 94/07880, WO 94/07876, WO 93/23555, WO 93/10076; U.S. Pat. Nos. 5,294,637; 5,283,253; 5,279,949; 5,274,137; 5,202,448; 5,200,534; 5,229,529; and EP 590,267), or obtained from a variety of commercial sources, including for example, Sigma Chemical Co., St. Louis, Mo. (T7402 from Taxus brevifolia; or T-1912 from Taxus yannanensis).
[00115] Biological response modifiers suitable for use in connection with the methods of the present disclosure include, but are not limited to, (1) inhibitors of tyrosine kinase (RTK) activity; (2) inhibitors of serine/threonine kinase activity; (3) tumor-associated antigen antagonists, such as antibodies that bind specifically to a tumor antigen; ( 4) apoptosis receptor agonists; (5) interleukin-2; (6) interferon- a.; (7) interferon -γ; (8) colony-stimulating factors; (9) inhibitors of angiogenesis; and (10) antagonists of tumor necrosis factor.
[00116] In one embodiment, the present disclosure provides a use of a pharmaceutical
composition as disclosed herein for the manufacture of a medicament for treating or preventing cancer.
[00117] In one embodiment, the present disclosure provides a use of a compound of formula (I) for the manufacture of a medicament for treating or preventing cancer.
[00118] In one embodiment, the present disclosure provides a use of a compound selected from
Table 1 herein, or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament for treating or preventing cancer.
[00119] In one embodiment, the medicament further comprises a cancer chemotherapeutic agent.
[00120] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[00121] In one embodiment, the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[00122] In one embodiment, the compound and the chemotherapeutic agent are administered simultaneously, staggered, alternating, or sequentially.
[00123] In one embodiment, the use further comprises administering ionizing radiation.
[00124] In one embodiment, the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
[00125] In one embodiment, the present disclosure provides a use of a first amount of a
compound of formula (I) for the manufacture of a medicament for treating or preventing cancer in a first treatment procedure, wherein the use further comprises a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
[00126] In one embodiment, the radiation is a radiopharmaceutical agent.
[00127] In one embodiment, the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
[00128] In one embodiment, the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
[00129] In one embodiment, the use provides a synergistic effect in the treatment of cancer.
[00130] In one embodiment, the present disclosure provides a use of a compound of formula (I) or a pharmaceutical composition as disclosed above for the manufacture of a medicament for increasing the sensitivity of cancerous cells or cancer stem cells to a chemotherapeutic agent.
[00131] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[00132] In one embodiment, the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
[00133] In one embodiment, the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral.
[00134] In one embodiment, the compound or pharmaceutical composition is administered to the respiratory tract.
4. Pharmaceutical Compositions
[00135] In one embodiment, the present disclosure relates to a pharmaceutical composition
and a cancer chemotherapeutic agent:
Figure imgf000030_0001
(I),
or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein
Figure imgf000030_0002
R1 and R2 are each independently selected from hydrogen and C1-C6 alkyl,
R3 and R4 are each independently selected from hydrogen and C1-C6 alkyl;
each R7 is independently selected from halogen, C1-C6 alkyl, and OCH3, and
n is 0, 1, 2, 3, 4, or 5;
provided that when one of R1 and R2 is hydrogen and the other is isopropyl,
Figure imgf000031_0001
[00136] In one embodiment, one of R1 and R2 is hydrogen and the other is Ci-C6 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain Ci-C6 alkyl or branched C3-C6 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain d- C4 alkyl or branched C3-C4 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain C1-C4 alkyl. In one embodiment, one of R1 and R2 is hydrogen and the other is branched C3-C4 alkyl.
[00137] In one embodiment, one of R3 and R4 is hydrogen and the other is Ci-C6 alkyl. In one embodiment, one of R3 and R4 is hydrogen and the other is methyl or ethyl.
[00138] In one embodiment, one of R1 and R2 is hydrogen and the other is straight-chain C1-C4 alkyl; and one of R3 and R4 is hydrogen and the other is methyl or ethyl. In one embodiment, one of R1 and R2 is hydrogen and the other is branched C3-C4 alkyl; and one of R3 and R4 is hydrogen and the other is methyl or ethyl.
[00139] In one embodiment, n is 0, 1, 2, or 3. In one embodiment, n is 1, 2, or 3. In one
embodiment, n is 1. In one embodiment, n is 2.
[00140] In one embodiment, each R7 is Ci-C6 alkyl. In one embodiment, each R7 is independently methyl, ethyl, or isopropyl. In one embodiment, each R7 is halogen. In one embodiment, each R7 is independently chloro or fluoro. In one embodiment, R7 is chloro. In one embodiment, R7 is fluoro. In one embodiment, one or more R7 is Ci-C6 alkyl and one or more R7 is halogen. In one embodiment, one or more R7 is independently methyl, ethyl, or isopropyl and one or more R7 is independently chloro or fluoro. In one embodiment, one or more R7 is Ci-C6 alkyl and one or more R7 is OCH3. In one example, one or more R7 is independently methyl, ethyl, or isopropyl and one or more R7 is OCH3. In one embodiment, one or more R7 is halogen and one or more R7 is OCH3. In one example, one or more R7 is independently chloro or fluoro and one or more R7 is OCH3.
[00141] It will be apparent to one skilled in the art based one the present disclosure that any of the embodiments described above for each of R1, R2, R3, R4, R7, and n can be combined in any manner.
[00142] In one embodiment, a compound of the present disclosure is of one of the following formulae:
Figure imgf000032_0001
[00143] In one embodiment, the present disclosure relates to a pharmaceutical composition
comprising a compound selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a cancer chemotherapeutic agent.
[00144] In one embodiment, the chemotherapeutic agent is selected from non-peptidic (non- proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone- deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
[00145] In another embodiment, the proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor. [00146] In one embodiment, the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral. In one embodiment, the compound or pharmaceutical composition is administered to the respiratory tract.
[00147] The present disclosure provides pharmaceutical compositions comprising a compound of the present disclosure (where a compound of the present disclosure is also referred to herein as "active agent" of the present disclosure, or "subject active agent"). The compound of the present disclosure can be formulated with one or more pharmaceutically acceptable excipients. A wide variety of pharmaceutically acceptable excipients are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy," 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H. C. Ansel et al., eds., 7th ed., Lippincott, Williams, & Wilkins; and Handbook of Pharmaceutical Excipients (2000) A. H. Kibbe et al., eds., 3rd ed. Amer.
Pharmaceutical Assoc.
[00148] The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
[00149] In the subject methods, a subject ALDH1A1 activity inhibitor may be administered to the host using any convenient means capable of treating cancer. Thus, a subject ALDH1A1 activity inhibitor can be incorporated into a variety of formulations for therapeutic administration. More particularly, a subject ALDH1A1 activity inhibitor can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
[00150] In pharmaceutical dosage forms, a subject active agent may be administered in the form of their pharmaceutically acceptable salts, or a subject active agent may be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.
[00151] For oral preparations, a subject active agent can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
[00152] A subject active agent can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
[00153] A subject active agent can be utilized in aerosol formulation to be administered via
inhalation. A subject active agent can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
[00154] Furthermore, a subject active agent can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. An active agent can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycol monomethyl ethers, which melt at body temperature, yet are solidified at room temperature.
[00155] Unit dosage forms for oral or rectal administration such as syrups, elixirs, and
suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the subject active agent. Similarly, unit dosage forms for injection or intravenous administration may comprise a subject active agent in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
[00156] The term "unit dosage form," as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a subject active agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for a subject active agent depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
[00157] A subject active agent can be formulated for administration by injection. Typically, injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation may also be emulsified or the active ingredient encapsulated in liposome vehicles. [00158] In some embodiments, a subject active agent is delivered by a continuous delivery system. The term "continuous delivery system" is used interchangeably herein with "controlled delivery system" and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
[00159] Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985. The composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
[00160] Depending on the subject and condition being treated and on the administration route, the subject compounds may be administered in dosages of, for example, 0.1 μg to 10 mg/kg body weight per day. The range is broad, since in general the efficacy of a therapeutic effect for different mammals varies widely with doses typically being 20, 30 or even 40 times smaller (per unit body weight) in man than in the rat. Similarly the mode of administration can have a large effect on dosage. Thus, for example, oral dosages may be about ten times the injection dose. Higher doses may be used for localized routes of delivery.
[00161] For example, a subject ALDH1A1 activity modulator can be administered in an amount of from about 1 mg to about 1000 mg per dose, e.g., from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 50 mg, from about 50 mg to about 75 mg, from about 75 mg to about 100 mg, from about 100 mg to about 125 mg, from about 125 mg to about 150 mg, from about 150 mg to about 175 mg, from about 175 mg to about 200 mg, from about 200 mg to about 225 mg, from about 225 mg to about 250 mg, from about 250 mg to about 300 mg, from about 300 mg to about 350 mg, from about 350 mg to about 400 mg, from about 400 mg to about 450 mg, from about 450 mg to about 500 mg, from about 500 mg to about 750 mg, or from about 750 mg to about 1000 mg per dose.
[00162] An exemplary dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time -release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient, etc. The time -release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release. [00163] Those of skill in the art will readily appreciate that dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
[00164] Unit dosage forms for oral or rectal administration such as syrups, elixirs, and
suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more compounds of the present disclosure. Similarly, unit dosage forms for injection or intravenous administration may comprise the compound (s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
[00165] In some embodiments, multiple doses of a subject compound are administered. The
frequency of administration of a subject compound can vary depending on any of a variety of factors, e.g., severity of the symptoms, etc. For example, in some embodiments, a subject compound is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (qid), or three times a day (tid). As discussed above, in some embodiments, a subject compound is administered continuously.
[00166] The duration of administration of a subject compound, e.g., the period of time over which a subject compound is administered, can vary, depending on any of a variety of factors, e.g., patient response, etc. For example, a subject compound can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more. In some embodiments, a subject compound is administered for the lifetime of the individual.
[00167] A subject ALDH1A1 activity inhibitor is administered to an individual using any
available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration. Administration can be acute (e.g., of short duration, e.g., a single administration, administration for one day to one week), or chronic (e.g., of long duration, e.g., administration for longer than one week, e.g., administration over a period of time from about 2 weeks to about one month, from about one month to about 3 months, from about 3 months to about 6 months, from about 6 months to about 1 year, or longer than one year). [00168] Conventional and pharmaceutically acceptable routes of administration include intranasal, intramuscular, intratracheal, subcutaneous, intradermal, transdermal, sublingual, topical application, intravenous, rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the agent and/or the desired effect. The compound can be administered in a single dose or in multiple doses.
[00169] An active agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes. In general, routes of administration contemplated by the present disclosure include, but are not necessarily limited to, enteral, parenteral, or inhalational routes.
[00170] Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular, intraorbital, intracapsular, intraspinal, intrasternal, and intravenous routes, i.e., any route of administration other than through the alimentary canal. Parenteral administration can be carried to effect systemic or local delivery of the agent. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
[00171] The agent can also be delivered to the subject by enteral administration. Enteral routes of administration include, but are not necessarily limited to, oral and rectal (e.g., using a suppository) delivery.
[00172] Methods of administration of the agent through the skin or mucosa include, but are not necessarily limited to, topical application of a suitable pharmaceutical preparation, transdermal transmission, injection and epidermal administration. For transdermal transmission, absorption promoters or iontophoresis are suitable methods. Iontophoretic transmission may be
accomplished using commercially available "patches" which deliver their product continuously via electric pulses through unbroken skin for periods of several days or more.
5. Examples
Example 1: Inhibitory activity of compounds against ALDH1A1
[00173] Method: To prepare human ALDH1A1 recombinant enzyme, full-length human
ALDH1 Al cDNA was synthesized based on published DNA sequence (GenBank ID:
NM_000689). The synthetic gene was cloned into the Nhel/EcoRI sites of the His-tag vector, pTrcHis, for protein expression. All the vectors were transformed into BL21 E. coli host cells and subjected to 0.5 mM isopropyl β-D-l-thiogalactopyranoside (IPTG) induction for protein expression at 30°C. Purifications of the recombinant proteins by affinity nickel columns (HisTrap, GE Healthy Science, USA) were carried out using standard protocols according to manufacturer's instructions (Novagen, USA).
[00174] An example of the enzymatic assay is described as follows. The reaction cocktails were added into the wells in a 96-well plate and then put on a shaker for mixing. After 5 minutes of mixing, 2 μΕ of recombinant ALDH1 Al enzyme was added to each well with DMSO as blank control in the plate. The plate was again put on shaker for another 5 minutes of shaking to enhance bonding between testing compounds and the enzyme. 2μΕ of 1M acetyladehyde in water was then added to each of wells containing the above reaction mixture. After a short vigorous mixing, the UV plate was fed into a plate reader to measure kinetic optical density (O.D.) at wavelength 340 nm at 25°C over 4 minutes. Each point of measurement was taken at a time interval of 40 seconds.
[00175] Results: The half maximal inhibitory concentration (IC50) values for compounds of the present disclosure against ALDH1A1 are listed in Table 2.
Table 2: Half Maximal Inhibitory Concentration (IC50) of Compounds Against ALDH1A1
Figure imgf000038_0001
Compound no. ic50
ALDH1A1 Inhibition (μΜ)
82 0.69
85 0.48
86 0.44
88 0.41
91 0.37
93 0.39
95 0.49
106 0.57
107 1.18
114 0.59
Example 2: Selectivity of Compound 5 with Other ALDH Isozymes
[00176] The specificity of Compound 5 against ALDH1A1, and other ALDH isozymes including
ALDH2, ALDH3A1, ALDH4A1, ALDH7A1, and ADH1B1 was determined.
[00177] Method: Isozyme-specific inhibition of Compound 5 was evaluated using standard
spectrometric ALDH enzymatic assays as described previously (e.g. Chen et. al, (2008) Science 321: 1493-1495, Sheikh et al. (1997) J. Biol. Chem. 272:18817-18822). Compound 5 (20 μΜ) was tested against purified human recombinant ALDH isozymes. In each assay, 10-50 μg of each isozyme were used with 10 mM of acetaldehyde as the substrate and 2.5 mM of NAD+ as the cofactor. Enzymatic activities were standardized using DMSO alone as the control (expressed as 100% activity).
[00178] Results: As shown in Figure 1, Compound 5 is a highly selective ALDH1A1 inhibitor
(99% inhibition at 20 μΜ) while no inhibitory activity against other five ALDH isozymes was observed.
Example 3: In vitro anti-cancer activity of the compounds in combination with paclitaxel and doxorubicin
[00179] Method: Cell cultures (5000 cells/well) in medium containing 5% fetal bovine serum
(FBS) were incubated in a 96-well plate for 24 h at 37 °C supplemented with 5% C02. Basal cells were fixed with 25 μί of 50% (w/v) trichloroacetic acid (TCA) (final 10%) for 30-min as Tz. Compound 5 at various concentrations was added to the cell cultures and then the suction of the basal cells with TCA was performed. The basal wells were washed with phosphate buffer saline (PBS, 100 μί), and incubated for 48-h or 72-h treatment before reaction was terminated, followed by 30-min incubation at RT. After suction of the cells with TCA, all wells (including basal wells) were washed with PBS (100 μί) and air-dried for 20 min in the hood.
Sulforhodamine B (SRB) solution (50 μί) at 0.4% (w/v) in 1% acetic acid was added to each well, followed by 10-min incubation at RT. Unbound dye was removed by washing twice with 1% acetic acid (100 μΐ^), and the plates were air dried for 30 min or overnight. Cells were solubilized with 10 mM trizma base (100 μΐ^). The plate was read at 515 nm detection in ELISA reader. Percentage growth inhibition was appropriately calculated.
[00180] Results: Compound 5 demonstrated significant synergistic effects (or sensitization) with paclitaxel (Figure 2) or doxorubicin (Figure 3) in two MDR ovarian cell lines, NCI/ ADR RES and TOV-21G-RT cells. In addition, analysis of the Combination Index (CI) was also performed, which showed strong synergistic effects of Compound 5 with pacitaxel and doxorubicin in those cancer cell lines (see Table 3).
[00181] Figure 2: (A) NCI/ADR-RES cells were treated with the indicated concentration of
paclitaxel in the absence or presence of Compound 5(3 μΜ) for 48 h; (B) TOV-21G-RT cells were treated with the indicated concentration of paclitaxel in the absence or presence of
Compound 5 (10 μΜ) for 48 h. Then, the cell growth was examined using SRB assay, and mean IC50 values were subsequently determined. Mean ± SE of five independent experiments (each in triplicate).
[00182] Figure 3: (A) NCI/ADR-RES cells were treated with the indicated concentration of
doxorubicin in the absence or presence of Compound 5 (3 μΜ) for 48 h; (B) TOV-21G-RT cells were treated with the indicated concentration of doxorubicin in the absence or presence of Compound 5 (10 μΜ) for 48 h. The cell growth was detected by SRB assay, and mean IC50 values were subsequently determined. Mean ± SE of five independent experiments (each in triplicate).
Table 3: Calculation of Combination Index for Compound 5 with Chemotherapies
Figure imgf000040_0001
Note: The mean combination index value, resulting from separate experiments at multiple effect levels, CI <1 indicates synergism, CI = 1 indicates additive effect, and CI >1 indicates antagonism.
INCORPORATION BY REFERENCE
[00183] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. OTHER EMBODIMENTS
While the present disclosure has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the present disclosure, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the present disclosure encompassed by the appended claims.

Claims

CLAIMS What is claimed is:
1. A pharmaceutical composition comprising a compound of formula (I) and a
chemo
Figure imgf000042_0001
or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein
Figure imgf000042_0002
is selected from , , and
Figure imgf000042_0003
R1 and R2 are each independently selected from hydrogen and C1-C6 alkyl;
R3 and R4 are each independently selected from hydrogen and C1-C6 alkyl;
each R7 is independently selected from halogen, C1-C6 alkyl, and OCH3; and
n is 0, 1, 2, 3, 4, or 5;
provided that when one of R1 and R2 is hydrogen and the other is isopropyl,
Figure imgf000042_0004
2. The composition of claim 1, wherein one of R1 and R2 is hydrogen and the other is C1-C6 alkyl.
3. The composition of any one of claims 1-2, wherein one of R3 and R4 is hydrogen and the other is C1-C6 alkyl.
4. The composition of claim 3, wherein one of R3 and R4 is hydrogen and the other is methyl or ethyl.
5. The composition of any one of claims 1-4, wherein n is 0, 1, 2, or 3.
6. The composition of claim 5, wherein n is 1, 2, or 3.
7. The composition of claim 6, wherein n is 1.
8. The composition of claim 6, wherein n is 2.
9. The composition of any one of claims 6-8, wherein each R7 is Ci-C6 alkyl.
10. The composition of claim 9, wherein each R7 is methyl, ethyl, or isopropyl.
11. The composition of any one of claims 6-8, wherein each R7 is halogen.
12. The composition of claim 11, wherein R7 is fluoro.
13. A pharmaceutical composition comprising a compound selected from Table 1, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a cancer chemotherapeutic agent.
14. The pharmaceutical composition of any one of claims 1-13, wherein the
chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony- stimulating factors, histone-deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
15. The pharmaceutical composition of claim 14, wherein the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
16. Use of a pharmaceutical composition of any one of claims 1 -15 for the manufacture of a medicament for treating or preventing cancer.
17. Use of a compound of formula (I) of claim 1 for the manufacture of a medicament for treating or preventing cancer.
18. Use of a compound selected from Table 1 , or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament for treating or preventing cancer.
19. The use of any one of claims 17-18, wherein the medicament further comprises a cancer chemotherapeutic agent.
20. The use of claim 19, wherein the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone-deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
21. The use of claim 20, wherein the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen, apoptosis receptor agonists, interleukin-2, interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
22. The use of any one of claims 17-21 , wherein the compound and the chemotherapeutic agent are administered simultaneously, staggered, alternating, or sequentially.
23. The use of any one of claims 17-18 further comprising administering ionizing radiation.
24. The use of claim 23, wherein the ionizing radiation is administered via external beam radiation therapy or brachytherapy.
25. Use of a first amount of a compound of formula (I) of claim 1 for the manufacture of a medicament for treating or preventing cancer in a first treatment procedure, wherein the use further comprises a second amount of radiation in a second treatment procedure wherein, the first and second amounts together comprise a therapeutically effective amount.
26. The use of claim 25, wherein the radiation is a radiopharmaceutical agent.
27. The use of claim 25, wherein the radiation is an ionizing radiation administered via external beam radiation therapy or brachytherapy.
28. The use of any one of claims 16-27, wherein the cancer is selected from solid tumors, skin cancers, myeloma, lymphoma, leukemia, ovarian cancer, lung cancer, brain cancer, pancreatic cancer, prostate cancer, colon cancer, colorectal cancer, breast cancer, liver cancer, kidney cancer, oral cancer, head cancer, neck cancer, throat cancer, and thyroid cancer.
29. The use of any one of claims 16 or 19-28, wherein the use provides a synergistic effect in the treatment of cancer.
30. Use of a compound of formula (I) of claim 1 or a pharmaceutical composition of any one of claims 1-15 for the manufacture of a medicament for increasing the sensitivity of cancerous cells or cancer stem cells to a chemotherapeutic agent.
31. The use of claim 30, wherein the chemotherapeutic agent is selected from non-peptidic (non-proteinaceous) compounds that reduce proliferation of cancer cells, cytotoxic agents, cytostatic agents, alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, taxanes, microtubule affecting agents, metal complexes, steroid hormones, tyrosine kinase and/or serine/threonine inhibitors, inhibitors of angiogenesis, colony-stimulating factors, histone-deacetylase inhibitors (HDAC inhibitors), and antagonists of tumor necrosis factor.
32. The use of claim 31 , wherein the non-proteinaceous compounds that reduce proliferation of cancer cells is selected from tumor-associated antigen antagonists, antibodies that bind specifically to a tumor antigen apoptosis receptor agonists, interleukin-2 interferon-alpha (a), interferon-gamma (γ), colony-stimulating factors, inhibitors of angiogenesis, and antagonists of tumor necrosis factor.
33. The use of any one of claims 16-32, wherein the compound or pharmaceutical composition is administered by a route selected from topical, intramuscular, intravenous, subcutaneous, and oral.
34. The use of claim 33, wherein the compound or pharmaceutical composition is administered to the respiratory tract.
PCT/US2014/025679 2013-03-14 2014-03-13 Aldehyde dehydrogenase-1 modulators and methods of use thereof WO2014160034A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361781793P 2013-03-14 2013-03-14
US61/781,793 2013-03-14

Publications (1)

Publication Number Publication Date
WO2014160034A1 true WO2014160034A1 (en) 2014-10-02

Family

ID=50736144

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/025679 WO2014160034A1 (en) 2013-03-14 2014-03-13 Aldehyde dehydrogenase-1 modulators and methods of use thereof

Country Status (1)

Country Link
WO (1) WO2014160034A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111053778A (en) * 2019-11-05 2020-04-24 徐州医科大学 Application of imidazoquinazoline derivative and salt thereof in preparation of PIM1 enzyme inhibitor, PIM1 enzyme inhibitor and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010062308A1 (en) * 2008-10-28 2010-06-03 The Board Of Trustees Of The Leland Stanford Junior University Modulators of aldehyde dehydrogenase and methods of use thereof
WO2011128407A2 (en) * 2010-04-16 2011-10-20 Bayer Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline-containing combinations
WO2012116010A2 (en) * 2011-02-22 2012-08-30 The General Hospital Corporation Antibiotic tolerance inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010062308A1 (en) * 2008-10-28 2010-06-03 The Board Of Trustees Of The Leland Stanford Junior University Modulators of aldehyde dehydrogenase and methods of use thereof
WO2011128407A2 (en) * 2010-04-16 2011-10-20 Bayer Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline-containing combinations
WO2012116010A2 (en) * 2011-02-22 2012-08-30 The General Hospital Corporation Antibiotic tolerance inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 25 September 2008 (2008-09-25), XP002727114, Database accession no. 1052659-30-9 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111053778A (en) * 2019-11-05 2020-04-24 徐州医科大学 Application of imidazoquinazoline derivative and salt thereof in preparation of PIM1 enzyme inhibitor, PIM1 enzyme inhibitor and application thereof
CN111053778B (en) * 2019-11-05 2021-07-23 徐州医科大学 Application of imidazoquinazoline derivative and salt thereof in preparation of PIM1 enzyme inhibitor, PIM1 enzyme inhibitor and application thereof

Similar Documents

Publication Publication Date Title
US10227304B2 (en) Mitochondrial aldehyde dehydrogenase-2 modulators and methods of use thereof
US11945803B2 (en) Modulators of RAS GTPase
JP6615769B2 (en) Polycyclic amide binding to mitochondrial aldehyde dehydrogenase 2 (ALDH2)
AU2023201669A1 (en) Compositions and methods for modulating hair growth
CA2417064A1 (en) Cyclic polyamine compounds for cancer therapy
US20220227740A1 (en) Modulators of RAS GTPase
AU2021202619B2 (en) Multifunctional inhibitors of MEK/PI3K and mTOR/MEK/PI3K biological pathways and therapeutic methods using the same
US11267817B2 (en) Substituted pyrrolo[1,2-a]quinoxalin-4(5H)-ones as CX3CR1 antagonists
US20160129030A1 (en) Treatment of mtor hyperactive related diseases and disorders
US9622993B2 (en) Thiosemicarbazones inhibitors of lysophosphatidic acid acyltransferase and uses thereof
US20040110727A1 (en) Aldehyde-releasing compounds
WO2014160034A1 (en) Aldehyde dehydrogenase-1 modulators and methods of use thereof
US20150080632A1 (en) Marinopyrrole derivatives as anticancer agents
US10682367B2 (en) Antitumor arylnaphthalene ligand glycosides
US11453696B2 (en) Adenosine analogs as methyltransferase inhibitors for treating cancer
WO2015084731A1 (en) Aldehyde dehydrogenase inhibitors and methods of use thereof
JP2006282653A (en) New compound to be selectively activated at target site and its use
Jiangzhou et al. Discovery of proqodine A derivatives with antitumor activity targeting NAD (P) H: quinone oxidoreductase 1 and nicotinamide phosphoribosyltransferase
US20120226031A1 (en) Palmarumycin based inhibitors of thioredoxin and methods of using same
CN115536637A (en) S-triazine derivative and its synthesis method and use
WO2020033377A1 (en) Histone demethylase 5 inhibitors and uses thereof
Barber Jones, RJ et al. Assessment of aldehyde dehydrogenase in viable cells. Blood 85, 2742-2746

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: 14725265

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: 14725265

Country of ref document: EP

Kind code of ref document: A1