WO2011130395A1 - Inhibitors of flip to treat cancer - Google Patents

Inhibitors of flip to treat cancer Download PDF

Info

Publication number
WO2011130395A1
WO2011130395A1 PCT/US2011/032299 US2011032299W WO2011130395A1 WO 2011130395 A1 WO2011130395 A1 WO 2011130395A1 US 2011032299 W US2011032299 W US 2011032299W WO 2011130395 A1 WO2011130395 A1 WO 2011130395A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
compound
bay
group
cancer
Prior art date
Application number
PCT/US2011/032299
Other languages
French (fr)
Inventor
Scott A. Kuwada
Original Assignee
University Of Utah Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Utah Research Foundation filed Critical University Of Utah Research Foundation
Publication of WO2011130395A1 publication Critical patent/WO2011130395A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline

Definitions

  • Cancer cells have long been known for their ability to grow in the absence of adhesion, a characteristic known as anchorage-independent proliferation (Martin, "Normal Cells and Cancer Cells.” In Molecular Oncology, J. M. Bishop, and R. A. Weinberg, eds., New York, Scientific American, pp. 13-40, 1996). This is of clinical relevance to metastasis, because cancer cells must transiently survive in the absence of adhesion as they travel to and migrate into distant tissues via the circulatory or lymphatic systems. Efforts to curb metastasis are critical since the presence of metastases is the single most important prognostic indicator for survival in cancer patients.
  • Iatrogenic seeding of cancer cells is particularly worrisome as the primary modality of therapy for most resectable solid tumors is surgery.
  • Intra-abdominal cancer seeding and peritoneal carcinomatosis complicate the medical and surgical therapy of abdominal cancers such as colorectal, gastric, pancreatic, and ovarian (De Vita et al, Cancer: Principles and Practice of Oncology. 5 ed.
  • Peritoneal seeding of tumor cells can occur in up to 35% of patients following curative resections of pancreatic head cancers (Johnstone et al., "Patterns of disease recurrence following definitive therapy of adenocarcinoma of the pancreas using surgery and adjuvant radiotherapy: correlations of a clinical trial,” Int J Radiat Oncol Biol Phys 27(4):831-4, 1993) or through metastasis of intraabdominal cancers (Yu et al., "Prospective randomized trial of early postoperative intraperitoneal chemotherapy as an adjuvant to resectable gastric cancer," Ann Surg
  • NF- ⁇ is a critical mediator of cancer cell survival and is commonly over-expressed in malignant cells making it a selective target for cancer therapy
  • Bosk et al "Nuclear factor-kappa B is upregulated in colorectal cancer," Surgery 30(2):363-9, 2001
  • Sovak et al "Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer” J Clin Invest 100(12):2952-60, 1997
  • Mori et al "Constitutive activation of NF-kappaB in primary adult T-cell leukemia cells," Blood 93(7):2360-8, 1999
  • Wang et al "The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells," Clin Cancer Res 5(1): 119-27, 1999
  • Nakshatri et al "Constitutive activation of NF-kappaB during progression of breast cancer to hormone -in
  • compositions and methods related to inhibiting the affect of NF- KB on cancer through inhibition of metastasis, readhesion, and cancer cell proliferation.
  • the disclosed subject matter in one aspect, relates to compositions and methods for preparing and using such compositions.
  • the disclosed subject matter relates to methods of using small molecules that inhibit NF- ⁇ and/or FLIP to treat cancer and, more specifically, prevent peritoneal seeding of cancer cells and cancer stem cells.
  • FIG. 1 is a group of graphs that shows the effect of NF- ⁇ inhibitors on colon cancer cell proliferation.
  • DLD-1, HCT-116, and HT-29 were cultured on 96-well plates in the presence or absence of BAY 11-7082 (FIG. 1A) or BAY 11-7085 (FIG. IB) for 8 days.
  • the cells were fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm.
  • control (DMSO) squares BAY 0.1 ⁇ diamonds, BAY 1.0 ⁇ triangles, BAY 10.0 ⁇ ovals.
  • the data points represent the mean values of experiments performed in triplicate and the error bars the standard error of the means.
  • FIG. 1 is a group of graphs that shows the effect of NF- ⁇ inhibitors on colon cancer cell proliferation.
  • FIG. 1C shows electrophoretic mobility shift assays showing inhibition of NF- KB binding to an NF- ⁇ DNA consensus oligonucleotide in nuclear lysates from HT-29 cells treated with BAY 11-7082 or BAY 11-7085 for 3 (left panel) or 6 (right panel) hours.
  • the FIG shows the typical results of four experiments.
  • the densitometry data of the gels are shown in graphic form at the bottom of the FIG. Excess cold competitor DNA oligonucleotide was used to demonstrate the specificity of the protein-DNA binding.
  • FIG. 2A is a graph that shows NF- ⁇ inhibitors, BAY 11 -7082 and BAY 11 - 7085, decrease anchorage-independent proliferation of colon cancer cells.
  • DLD-1, HCT- 116, and HT-29 cells were cultured for 6 days in soft agar in the presence of DMSO (control), 10 ⁇ BAY 11-7082, or 10 ⁇ BAY 11-7085.
  • the data points represent the means of colony counts per field (2 OX) performed in 5 different regions of the culture dishes. The data shown represent a typical result of two separate experiments.
  • FIG. 2B and FIG. 2C are graphs that show athymic mice received subcutaneous injections of HT- 29 (FIG. B) or HCT-116 (FIG.
  • FIG. 3 is a group of graphs that show NF- ⁇ inhibitors cause apoptosis of colon cancer cells.
  • FIG. 3A is a graph that shows adherent and HT-29 cells were treated with DMSO alone (control), 10 and 20 ⁇ BAY 11-7085, or 20 ⁇ MG-132 for 24 hours. The data points represent the average percent of annexin V-positive, propidium iodide-negative cells (of the total cells counted) from triplicate experiments. The error bars represent the standard error of the means.
  • FIG. 3B is a graph that shows adherent HT-29 cells were treated with BAY 11-7085 at various concentrations for 24 hours after which the nonadherent and adherent cells were collected separately.
  • FIG. 3C shows adherent HCT-116 cells were treated with 20 ⁇ MG-132 for 24 hours after which the cells were collected. An immunoblot of the lysates for cleaved PARP was performed. Each lane represents equal total protein concentrations.
  • FIG. 3D is a graph that shows confluent monolayers of HT-29 cells were treated with various concentrations of BAY 11-7085 for 3 hours after which they were dispersed with trypsin-PBS. The cells were allowed to adhere to plastic dishes for 1 hour after which they were washed gently in PBS and stained with crystal violet.
  • the crystal violet stained cells were solubilized in deoxycholate and the absorbance detected in a spectrophotometer at 590 nm.
  • the adherent cells were expressed as a fraction of the controls. Each data point represents the average of triplicate experiments and the error bars the standard errors of the means.
  • FIG. 4A is a graph that shows transient suspension of colon cancer cells greatly increases their susceptibility to apoptosis in the presence of BAY 11-7085.
  • HT-29 cells that were either transiently suspended with trypsin-EDTA or adherent for three days, were treated at time zero with DMSO or 10- 100 ⁇ BAY 11-7085. The percent of apoptotic cells was determined using the annexin V flow cytometry assay and graphed versus the dose of BAY 11-7085.
  • FIG. 4B are photographs showing DLD-1
  • FIG. 4C are photographs showing HCT-116
  • 4D are photographs showing HT-29 cells that were transiently suspended in trypsin-EDTA and allowed to readhere to glass coverslips for 1-24 hours. The cells were fixed and immunostained with an NF- ⁇ p65 subunit monoclonal antibody. Note the strong nuclear staining at 1 hour versus the markedly reduced nuclear staining of the cells.
  • FIG. 5A is a graph that shows NF- ⁇ binding activity assay (TransAM) of DLD-1 , HCT-116, and HT-29 cells that were transiently suspended by scraping or with trypsin-EDTA.
  • the transiently suspended cells were lysed and allowed to bind to DNA oligonucleotides, containing the NF- ⁇ consensus binding site, that were immobilized to 96-well plates.
  • NF- ⁇ protein bound to the oligonucleotides was detected by an antibody to the p65 subunit that only recognized p65 that is bound to DNA.
  • the positive control was HeLa cells that were stimulated with TNFa.
  • a control was performed using excess free NF- ⁇ oligonucleotides (Competitor).
  • FIG. 5B is a graph that shows HT-29 cells were transiently suspended and allowed to readhere in the presence or absence of BAY 11-7085 for three hours after which they were lysed. NF- ⁇ binding activity was determined by the TransAM assay as in FIG. 5A.
  • FIG. 5C is a graph that shows transiently suspended DLD-1, HCT-116, and HT-29 cells were allowed to readhere in the presence of DMSO or BAY 11-7085 for 8 hours after which the percent of apoptotic cells was determined by the flow cytometric annexin V assay. The data shown represent typical results of three separate experiments.
  • FIG. 5B is a graph that shows HT-29 cells were transiently suspended and allowed to readhere in the presence or absence of BAY 11-7085 for three hours after which they were lysed. NF- ⁇ binding activity was determined by the TransAM assay as in FIG. 5A.
  • FIG. 5C is a graph that shows transiently suspended DLD-1,
  • FIG. 5D shows 293-COX-2 cells were incubated in the presence or absence of 1 ⁇ g/mL of ponasterone for 48 hours after which they were lysed. An immunoblot for COX-2 was performed.
  • FIG. 5E is a graph that shows 293-COX-2 cells were incubated in the presence or absence of ponasterone for 48 hours followed by 8 hours of exposure to DMSO or BAY 11-7085. Note that induction of COX-2 resulted in an increased susceptibility to BAY 11-7085-induced apoptosis.
  • FIG. 6 is a group of photographs showing an intraabdominal seeding model.
  • FIGs. 6A, B, C show tumor implants of the liver of three athymic mice 21 days after receiving intraperitoneal injections of HT-29 cells. The mice were pretreated and treated for a total of 21 days with DMSO twice weekly.
  • FIG. 6D shows intestinal tumor implants (arrows) of an athymic mouse 21 days after intraperitoneal injections of HT-29 cells. The mouse was pretreated and treated for a total of 21 days with DMSO twice weekly.
  • FIG. 6E is a micrograph showing tumor invasion through the liver capsule. This section was obtained from the tumor implant from the mouse in FIG. 6A.
  • FIG. 6E is a micrograph showing tumor invasion through the liver capsule. This section was obtained from the tumor implant from the mouse in FIG. 6A.
  • FIG. 6F shows a peritoneal tumor implant taken from the bowel wall of the mouse in FIG. 6A.
  • FIG. 6G, (liver), FIG. 6H, (abdominal wall), and FIG. 61, (intestinal) show tumor implants in three athymic mice 21 days after receiving intraabdominal injections of HCT-116 cells. All of the mice were treated with 5 mg/kg of BAY 11-7085 twice weekly.
  • FIG. 7A is a graph that shows DLD- 1 HCT- 116, and HT-29 cells were cultured in 96-well plates for 10 days in the presence or absence of 10 or 50 ⁇ g/mL of cA2. At various time -points the cells were the cells were fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm. The data points represent the means of triplicate experiments and the error bars the standard errors of the means.
  • FIG. 7B is a graph that shows adherent HT-29 cells were pretreated with 50 ⁇ g/mL of cA2 for 48 hours followed by the addition of DMSO or 20 ⁇ BAY 11-7085 for another 24 hours.
  • FIG. 7C shows adherent or transiently suspended HT-29 cells were treated with 20 ⁇ BAY 11-7085 for 8 hours after which they were lysed.
  • FIG. 7D shows a FLIP immunoblot of DLD-1, HCT-116, and HT-29 lysates.
  • FIG. 8 is a graph that shows HT-29 cells that were transiently suspended and allowed to readhere in the presence or absence of various concentrations of the NF-KB inhibitors, PDTC, BAY 11-7085, and MG-132 for 4 hours. The percentage of apoptotic cells was determined using the annexin V assay. The data points represent the averages of three experiments; bars, +-.SE. E. DLD-1 and F. HT-29 cells were transduced with an adenovirus containing the ⁇ super-repressor construct. After 3 days, the cells were transiently suspended and allowed to readhere in the presence of subapoptotic
  • FIGs. 9A-C are graphs that shown BAY 11-7085 inhibits NF- ⁇ activation during pancreatic and colon cancer cell readhesion in vitro.
  • Transiently suspended pancreatic (Su.86, PL45, BxPC-3) and colon (HT-29) cancer cells were allowed to readhere in the presence of vehicle or various concentrations of BAY 11-7085 for 4 hours.
  • the levels of the p65 subunit of NF- ⁇ in nuclear lysates were performed with an ELISA-based kit.
  • the data points represent the average of triplicate results and the error bars the standard error of the means. The results for each
  • FIG. 10 is a graph showing that BAY 11-7085 induces apoptosis of pancreatic and colon cancer cells during readhesion.
  • Pancreatic (Su.86, PL-45) and colon cancer cells (HT-29) were transiently suspended and allowed to readhere in the presence or absence of vehicle or various concentrations of BAY 11-7085 for 8 hours.
  • the floating and adherent cells were collected and lysed.
  • Western blots for cleaved PARP were performed to determine if the cells were undergoing apoptosis (FIG. 10A).
  • Transiently suspended pancreatic cancer cells (105 per well) were allowed to readhere in the presence of vehicle or various concentrations of BAY 11-7085 for 24 hours.
  • Nonadherent apoptotic cells were washed away and the remaining adherent cells stained with crystal violet and then dried.
  • the crystal violet stained cells were lysed and the absorbance read in a plate spectrophotometer (FIG. 10B). Each data point represents the average of triplicate results and error bars, the standard error of the means. The results are expressed as a percentage of cells surviving compared with vehicle alone.
  • FIG. 11 are photographs showing bioluminescent detection of pancreatic cancer cells stably transfected with firefly luciferase.
  • FIGs. 11 A-B show an image of 5 athymic mice 9 days after IP injection of 106 Su.86 pancreatic cancer cells. The mice received IP firefly D-luciferin and then euthanized in order to visualize the intraabdominal tumor implants. The intraabdominal tumors were visualized before sacrificing the mice (A) or after euthanasia and exposure of the intraabdominal cavity (B).
  • FIG. 11C is a dissecting microscopic image of a barely visible tumor implant on the edge of the liver detected by bioluminescent imaging (arrow in FIG. 11 A).
  • FIG. 11 A is a dissecting microscopic image of a barely visible tumor implant on the edge of the liver detected by bioluminescent imaging
  • FIG. 12 shows thiazolidinediones inhibit NF-KB activation and induce apoptosis of pancreatic and colon cancer cells during cell readhesion.
  • FIG. 12A shows transiently suspended HT-29 cells were allowed to readhere in the presence of vehicle (DMSO) or various concentrations of rosiglitazone for 3 hours (dark bars). Untreated adherent monolayers (Control) and Jurkat cells treated with TNFa (Jurkat Control) served as controls. The cells were collected and analyzed for NF- ⁇ activation in an ELISA (see Methods).
  • FIG. 12B shows transiently suspended HT-29 cancer cells were allowed to readhere in the presence of DMSO, ciglitazone, rosiglitazone, or troglitazone for 8 hours. The floating and readherent cells were collected and lysed. Aliquots of the cell lysates were used to generate cleaved PARP immunoblots. All lanes contain equal total protein concentrations and the actin blot served as a loading control. The blots shown are typical of duplicate experiments. FIGs.
  • 12C-E show transiently suspended BxPC-3, Su.86, and HT-29 cancer cells were allowed to readhere in the presence of vehicle (DMSO) or various concentrations of ciglitazone, MCC-555, pioglitazone, rosiglitazone, and troglitazone for 8 hours.
  • vehicle DMSO
  • the floating cells were washed away and the adherent cells were then fixed and stained with crystal violet.
  • the cells were lysed in deoxycholate and the intensity of the staining quantified by spectrophotometry. The results are normalized to the controls. Each data point represents the mean of triplicate experiments and the error bars, the standard error of the mean.
  • FIG. 13 shows thiazolidinediones induce apoptosis of pancreatic and colon cancer cells during cell readhesion in a PPARy -independent fashion.
  • FIGs. 13A-B show transiently suspended Su.86 (PPARy -negative) and HT-29 (PPARy -positive) cancer cells were allowed to readhere in the presence of various concentrations of ciglitazone for 8 hours. The cells were also incubated with or without the irreversible PPARy inhibitor GW9662 during readhesion. The floating cells were washed away and the adherent cells were then fixed and stained with crystal violet. The cells were lysed in deoxycholate and the intensity of the staining quantified by spectrophotometry. The results are normalized to the controls. Each data point represents the mean of triplicate experiments and the error bars, the standard error of the mean.
  • FIG. 14 is a grap of tumor light output versus days of treatment.
  • FIG. 15 (A-D) shows expression of RANK and its ligand, RANKL, for most of the cancer cell lines.
  • FIG. 16 is a schematic illustration of a cancer cell.
  • FIG. 17 shows the results from a Cancer Stem Cell Longitudinal treatment Study in Nude Mice for BAY 11-7085 Su.86.86 versus Control.
  • Ranges can be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10" is also disclosed.
  • references in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed.
  • X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
  • a weight percent (wt. %) of a component is based on the total weight of the formulation or composition in which the component is included.
  • "Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • substituted is contemplated to include all permissible substituents of organic compounds.
  • permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described below.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms, such as nitrogen can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • substitution or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • a 1 ,” “A 2 ,” “A 3 ,” and “A 4 " are used herein as generic symbols to represent various specific substituents. These symbols can be any substituent, not limited to those disclosed herein, and when they are defined to be certain substituents in one instance, they can, in another instance, be defined as some other substituents.
  • alkyl as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like.
  • the alkyl group can also be substituted or unsubstituted.
  • the alkyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo- oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo- oxo, sulfonyl, sulfone, sulfoxide, or thiol, as
  • alkyl is generally used to refer to both unsubstituted alkyl groups and substituted alkyl groups; however, substituted alkyl groups are also specifically referred to herein by identifying the specific substituent(s) on the alkyl group.
  • halogenated alkyl specifically refers to an alkyl group that is substituted with one or more halide, e.g., fluorine, chlorine, bromine, or iodine.
  • alkoxyalkyl specifically refers to an alkyl group that is substituted with one or more alkoxy groups, as described below.
  • alkylamino specifically refers to an alkyl group that is substituted with one or more amino groups, as described below, and the like.
  • alkyl is used in one instance and a specific term such as “alkylalcohol” is used in another, it is not meant to imply that the term “alkyl” does not also refer to specific terms such as “alkylalcohol” and the like.
  • cycloalkyl refers to both unsubstituted and substituted cycloalkyl moieties
  • the substituted moieties can, in addition, be specifically identified herein; for example, a particular substituted cycloalkyl can be referred to as, e.g., an "alkylcycloalkyl.”
  • a substituted alkoxy can be specifically referred to as, e.g., a "halogenated alkoxy”
  • a particular substituted alkenyl can be, e.g., an "alkenylalcohol,” and the like.
  • a general term such as “cycloalkyl”
  • a specific term such as “alkylcycloalkyl”
  • alkoxy is an alkyl group bound through a single, terminal ether linkage; that is, an "alkoxy” group can be defined as— OA 1 where A 1 is alkyl as defined above.
  • alkoxylalkyl is an alkyl group that contains an alkoxy substituent and can be defined as— A ⁇ O-A 2 , where A 1 and A 2 are alkyl groups.
  • the alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described
  • alkynyl is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond.
  • the alkynyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • aryl as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like.
  • aryl also includes "heteroaryl,” which is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus.
  • non- heteroaryl which is also included in the term “aryl,” defines a group that contains an aromatic group that does not contain a heteroatom.
  • the aryl group can be substituted or unsubstituted.
  • the aryl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo- oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • bias is a specific type of aryl group and is included in the definition of aryl.
  • Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl.
  • cycloalkyl as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.
  • heterocycloalkyl is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus.
  • the cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted.
  • the cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like.
  • heterocycloalkenyl is a type of cycloalkenyl group as defined above, and is included within the meaning of the term “cycloalkenyl,” where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus.
  • the cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted.
  • the cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • cyclic group is used herein to refer to either aryl groups, non-aryl groups (i.e., cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl groups), or both. Cyclic groups have one or more ring systems that can be substituted or unsubstituted. A cyclic group can contain one or more aryl groups, one or more non-aryl groups, or one or more aryl groups and one or more non-aryl groups.
  • amine or "amino” as used herein are represented by the formula NA 1 A2 A 3 , where A 1 , A2 , and A 3 J can be, independently, hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • a “carboxylic acid” as used herein is represented by the formula— C(0)OH.
  • a “carboxylate” as used herein is represented by the formula— C(0)0 " .
  • the term “ester” as used herein is represented by the formula— OC(0)A 1 or where A 1 can be an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • ether as used herein is represented by the formula A 1 OA 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • ketone as used herein is represented by the formula ⁇ (0) ⁇ 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • halide refers to the halogens fluorine, chlorine, bromine, and iodine.
  • hydroxyl as used herein is represented by the formula— OH.
  • nitro as used herein is represented by the formula— N0 2 .
  • sil as used herein is represented by the formula— SiA ⁇ A 3 , where A 1 , A 2 , and A 3 can be, independently, hydrogen, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • halogenated alkyl alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl,
  • sulfonyl is used herein to refer to the sulfo-oxo group represented by the formula where A 1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • sulfonylamino or “sulfonamide” as used herein is represented by the formula— S(0) 2 NH-.
  • sulfone as used herein is represented by the formula A 1 S(0) 2 A 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • sulfoxide as used herein is represented by the formula A 1 S(0)A 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • R 1 is a straight chain alkyl group
  • one of the hydrogen atoms of the alkyl group can optionally be substituted with a hydroxyl group, an alkoxy group, an alkyl group, a halide, and the like.
  • a first group can be incorporated within second group or, alternatively, the first group can be pendant (i.e., attached) to the second group.
  • an alkyl group comprising an amino group the amino group can be incorporated within the backbone of the alkyl group.
  • the amino group can be attached to the backbone of the alkyl group. The nature of the group(s) that is (are) selected will determine if the first group is embedded or attached to the second group.
  • the starting materials and reagents used in preparing the disclosed compounds and compositions are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Acros Organics (Morris Plains, N.J.), Fisher Scientific (Pittsburgh, Pa.), or Sigma (St.
  • NF-KB is a heterodimeric transcription factor comprised of various protein subunits: p50/pl05, p65/RelA, p52/pl00, c-Rel, and RelB.
  • NF- ⁇ is localized to the cytoplasm by association with the ⁇ proteins, which inhibit translocation of NF- ⁇ into the nucleus thus preventing its transcriptional activity (Beg et al, " ⁇ interacts with the nuclear localization sequences of the subunits of NF- ⁇ : a mechanism for cytoplasmic retention," Genes Dev 6: 1899-913, 1992).
  • IKK ⁇ kinases
  • NF- ⁇ mediates the transcription of several survival genes, for example c-Myc, Bcl2, p53, p21, c-FLIP, c-IAP-1, c-IAP-2, XIAP, IEX-1L, COX- 2, TRAF-1, and TRAF-2 (Kreuz et al, " NF- ⁇ inducers upregulate cFLIP, a
  • inhibitors that can inhibit anchorage-dependent as well as anchorage-independent proliferation, and tumorigenicity, and can cause apoptosis of cancer cells, including pancreatic and colon cancer cells as well as cancer stem cells ⁇ e.g., pancreatic cancer stem cells).
  • the induction of apoptosis of cancer cells by NF-KB inhibition can occur in a cell adhesion-dependent fashion.
  • Readhesion following transient suspension of the cell lines causes a large activation of NF- ⁇ , which renders cells extraordinarly sensitive to NF- ⁇ inhibitor-induced apoptosis.
  • pretreatment of athymic mice with an NF- ⁇ inhibitor completely prevented liver metastasis following intraperitoneal delivery of a colon cancer cell line.
  • colon and pancreatic cancer cells, as well as cancer cells related to colon and pancreatic cancer cells utilize NF- ⁇ for mitogenesis and as a major survival factor during readhesion.
  • FLIP /FLICE /CASH /FLAME /CASPER is encoded by CFLAR, one of a number of pro-survival genes regulated by NF- ⁇ . FLIP is an important inhibitor of caspase 8 and thus death receptor-induced apoptosis. BAY 11- 7085 and anisomycin, a JNK activator, was shown to cause the rapid downregulation of FLIP. FLIP overexpression inhibited the pro-apoptotic activity of BAY 11-7085 during colon cancer cell readhesion. As shown herein, the disclosed inhibitors can also inhibit FLIP, thereby rendering cancer cells susceptible to death receptor mediated apoptosis.
  • the inhibitors disclosed herein can be any molecule that inhibits NF- ⁇ and FLIP, and that activates JNK. It is understood that NF- ⁇ function can be inhibited or altered in many ways by an inhibitor. For example, NF- ⁇ function can be inhibited by an inhibitor directly interacting with NF- ⁇ . Directly interacting with NF- ⁇ means that the inhibitor touches or binds with NF- ⁇ . An inhibitor can also indirectly inhibit NF-KB function. Indirectly inhibiting the function of NF- ⁇ means that an inhibitor does not touch or bind NF- ⁇ . A molecule would indirectly inhibit NF- ⁇ function by, for example, reducing the expression or activation or nuclear transport of NF-KB.
  • Examples of indirect inhibitors are those that inhibit NF- ⁇ transport into the nucleus. NF- ⁇ remains cytoplasmic if it interacts with ⁇ . When ⁇ is
  • compositions can, in certain examples, inhibit NF- KB transport to the nucleus through interactions with ⁇ , which prevent ⁇ from interacting with NF- ⁇ .
  • Other examples of indirect inhibitors that inhibit NF- ⁇ transport into the nucleus are inhibitors that inhibit ⁇ phosphorylation.
  • Still other examples of indirect inhibitors are inhibitors that inhibit expression of NF- ⁇ .
  • Still other examples of indirect inhibitors are inhibitors that inhibit translation of NF-KB.
  • the disclosed inhibitors can inhibit TNFa-induced NF- ⁇ activation.
  • TNFa typically causes an activation of NF- ⁇ and the disclosed inhibitors can decrease this TNFa-induced activation.
  • Many assays can be used to determine if inhibitors are decreasing TNFa-dependent activation.
  • inhibitors that can inhibit anchorage dependent and independent proliferation and tumorigenicity and can cause apoptosis. Still further, disclosed herein are inhibitors that inhibit induction of apoptosis by NF- ⁇ inhibition, which occurs in a cell-adhesion dependent fashion. Inhibitors that affect cells where readhesion of cells causes a large activation of NF- ⁇ that causes the cells to become sensitized to NF- ⁇ inhibitor induced apoptosis are also disclosed.
  • BAY 11-7082 and BAY I l7085 which inhibit ⁇ phosphorylation and TNFa induced NF- ⁇ activation.
  • BAY 11- 7082 also inhibits growth of DLD-1 and HCT-116.
  • Cells that contain APC mutations are susceptible to BAY 11-7085 and related molecules.
  • DLD-2 and HT-29 cells are susceptible to BAY 11-7085.
  • DLD-1 and HT-29 contain APC mutations.
  • HCT-116 cells have activating mutations on B-catenin, which is normally regulated by the APC product. Also HCT-116 cells do not express COX-2, which is commonly over-expressed in colorectal cancers.
  • Anchorage-independent inhibition of proliferation can be inhibited by both BAY 11-7082 and 7085 in DLD-1 and HT-29 both BAY 11-7082 and BAY 11-7085 reduce tumor volumes in vivo in athymic mice injected with HT-29 tumor cells.
  • MC-132 and PDTC cause an increase in apoptosis in adhered cells and BAY- 11-7085 and BAY- 11-7082 cause apoptosis in cells even when not adhered.
  • compositions can be used in a variety of ways as research tools.
  • the disclosed compositions such as BAY- 11-7085 and BAY- 11-7082, PDTC and MC-132 can be used as reagents and standards in cellular proliferation assays and as inhibitors for assays related to cancer.
  • compositions can be used for example as competitive inhibitors in combinatorial chemistry protocols or other screening protocols to isolate molecules that posses desired functional properties related to inhibition of cancer or metastasis of cell systems disclosed herein.
  • the disclosed inhibitors can be given to a subject. Any subject in need of the NF-KB /FLIP inhibitors as disclosed herein can be given the NF-KB/FLIP inhibitors. Any subject in need of the JNK activators can be given the INK activators.
  • the subject can, for example, be a mammal, such as a mouse, rat, rabbit, hamster, dog, cat, pig, cow, sheep, goat, horse, or primate, such as monkey, gorilla, orangutan, chimpanzee, or human.
  • the disclosed NF-KB/FLIP inhibitors and JNK activators can be used for inhibiting cancer cell proliferation.
  • Inhibiting cancer cell proliferation means reducing or preventing cancer cell growth.
  • Inhibitors can be determined by using a cancer cell assay. For example, either a DLD-1, HCT-116, HT-29, Su.86, or a BxPC-3 cell line cell line can be cultured on 96-well plates in the presence or absence of the inhibitor for 8 days. The cells can be fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm.
  • the inhibitors are those that will inhibit 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%o, 90%), or 95%) of the cell growth relative to a control as determined by
  • the disclosed inhibitors can also inhibit anchorage- dependent proliferation of cancer cells, such as colon cancer cells or pancreatic cancer cells, including cancer stem cells.
  • Inhibitors can be assayed using a soft agar colony formation assay.
  • DLD-1, HCT-116, HT-29, Su.86, and a BxPC-3 cells can be cultured for 6 days in soft agar in the presence of DMSO (control) or inhibitor. The number of colony counts can then be compared by, for example, taking a percentage of the cells formed relative to a control.
  • the inhibitors are those that will inhibit 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%), or 95% of the cell growth relative to a control as determined by counting colonies formed.
  • NF-KB/FLIP inhibitors can be used for promoting cancer cell apoptosis. Promoting cancer cell apoptosis means causing the cell to die.
  • An apoptosis assay can be used to determine if the inhibitors promote cancer cell apoptosis. For example, an apoptosis assay as described in Example 1 can be used. In certain examples, the percent of apoptosis was determined as the percent of annexin V-positive, propidium iodide-negative cells of the total cells counted in an Apoptosis assay, such as that described in Example 1.
  • the inhibitor can cause at least about 10%>, 15%, 20%>, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the cells to be apoptotic.
  • the apoptotic effect of the disclosed inhibitors is enhanced in cells that are transiently suspended, such as metastatic cells circulating through the blood stream, prior to readhesion.
  • Inhibitors that have this property can be determined by assaying their apoptotic effect as, for example described herein, in a transient suspension assay, as described in Examples 1 and 2. For example, cells can be put into suspension by scraping and then allowed to readhere. Inhibitors that cause an increase in apoptosis upon readhering to the substrate are disclosed. An increase upon readhering to the substrate can be determined by, for example, looking at multiples of cells that were apoptotic upon readhering to the substrate compared with cells that were apoptotic upon remaining in suspension.
  • NF-KB/FLIP inhibitors and JNK activators that can be used for inhibiting readhesion of cancer cells to a surface. Inhibiting readhesion of cancer cells to a surface means decreasing the number of cells capable of readhering to a surface after being transiently suspended as discussed herein.
  • the NF- ⁇ /FLIP inhibitors and JNK activators disclosed herein can also be used for inhibiting metastasis of cancer cells. Inhibiting metastasis of cancer cells means decreasing or lowering the amount of metastatic tumors that arise in an organism.
  • inhibitors that inhibit metastasis in an in vivo assay are to inject a cancer cell line, such as HT-29, into the abdominal cavity of an athymic mouse. Mice are pretreated with the inhibitor or a control intraperotneally, for example.
  • the mouse can then be treated regularly, for example, twice weekly with vehicle or BAY 11-7085 for a period of time, for example, 21 days.
  • the mouse can then be sacrificed and assayed for metastatic tumor formation.
  • NF- ⁇ inhibitors can inhibit intraabdominal metastasis arising from, for example, colon or rectal cancers.
  • NF- ⁇ /FLIP inhibitors can also inhibit hepatic, parietal or peritoneal metastasis arising from, for example, colon or rectal cancers.
  • the disclosed compositions can be used to treat, prevent, and/or inhibit metastasis.
  • metastasis For example, hemogenous or lymphatic metastis.
  • the disclosed NF- ⁇ /FLIP inhibitors and TNK activators can be used for inhibiting tumorigenesis.
  • Inhibiting tumorigenesis means decreasing or lowering the amount of tumors present in an organism.
  • One way of performing an in vivo assay to determine if an inhibitor inhibits tumorigenesis is to inject a cancer cell line
  • compositions which inhibit tumorigenesis in this type of assay disclosed herein as well as compositions that reduce tumorigenesis by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% relative to a control compound.
  • NF-KB/FLIP inhibitors and JNK activators can be administered to cells which utilize NF- ⁇ , FLIP and/or JNK for mitogenesis.
  • NF-KB inhibitors can be administered to cells which utilize NF- ⁇ for readhesion.
  • a cell can be determined to utilize NF- ⁇ for readhesion if in a transient suspension assay, upon readhesion NF- ⁇ is activated as determined by any assay that looks at NF- ⁇ activation.
  • NF- ⁇ binding assays to the consensus sequence can be used, such as TRANS-AMTM assay the discussed in Examples 1 and 2. If the NF- ⁇ activation is greater upon readhesion then in suspension, the cells can be considered to utilize NF-KB for readhesion.
  • the disclosed NF-KB/FLIP inhibitors and JNK activators can also be administered to cells to induce the apoptosis of the cells in a TNFa independent manner.
  • TNFa can activate apoptosis, but the NF- ⁇ inhibitors can be administered to cause apoptosis in a TNFa independent manner.
  • the disclosed inhibitors can promote apoptosis of cancer cells, such as colon cancer cells, without TNFa.
  • An inhibitor or cell can be shown to be apoptotic without needing the inhibitor by using TNFa inhibiting antibody, such as cA2, and seeing that apoptosis is still caused by the inhibitor even in the presence of the TNFa inhibiting antibody, such as cA2.
  • compositions When the compositions are administered, they typically cause a decrease in the expression of anti-apoptotic proteins.
  • Expression of the anti-apoptotic proteins can be determined by any means for determining expression. For example, standard
  • biotechnology methods such as PCR or Northern blots can be used to determine the expression levels of anti-apoptotic genes.
  • a decrease can be determined by assaying the expression levels of a desired anti-apoptotic gene in the presence of a potential inhibitor and comparing this level of expression to the level of expression in the absence of the inhibitor.
  • inhibitors which decrease the expression of the anti-apoptotic genes in such an assay.
  • NF-KB/FLIP inhibitors and JNK activators can, for example, be used to reduce the proliferation of cancer cells (including cancer stem cells), as well as to cause the apoptosis of cancer (stem) cells or inhibit the readhesion of cancer (stem) cells or inhibit the metastasis of cancer cells.
  • the inhibitors can, for example, can be administered to any cancer cell that uses NF- ⁇ to survive or metastize or adhere or which activates NF- ⁇ during its life cycle.
  • the inhibitors can be administered to cancer cells, e.g., pancreatic cancer stem cells, to restore the death receptor mediated apoptosis.
  • NF-KB/FLIP inhibitors and JNK activators can be administered to cancer cells that have a mutation in the adenomatous polyposis coli (APC) gene.
  • APC has been shown to be a tumor suppressor gene in, for example, colon cells (see for example, Groden et ah, Cancer Research, 55: 1531-1539, 1995, herein incorporated by reference at least for material related to APC mutations and assays of the same). A variety of different morphologies of the effect of the mutated APC can be seen.
  • Whether a cell contains an APC mutation can be assayed for using standard recombinant biotechnology protocols, for example, sequencing and PCR analysis or ligation mediated chain reaction (LCR) or other methods using (for example chip technology) capable of assaying and comparing DNA sequences.
  • the mutations can readily be assayed as being functional mutations, by for example, expressing the mutant protein in a cell and determining if the mutant protein induces an oncogenic phenotype.
  • Assays for determining the effect of an APC mutation can also be performed as discussed in Groden et al.
  • an activating mutation can be determined by assaying the mutation and comparing the effects to the effects of APC mutations in DLD-1 cells or HT29 cells.
  • a fully functional APC gene can be transfected into a DLD-1 or HT29 cell, and this decreases the oncogenic behavior of the DLD-1 or HT29 cells because the non-mutant APC gene rescues normal phenotype.
  • a given APC mutation can be assayed by, for example, transfecting the mutant APC into either a DLD-1 or HT29 cell line and comparing the level of rescue provided by the mutant APC to the level of rescue of the non-mutant APC.
  • the APC mutant will be considered an in-activating APC mutant, i.e., a mutant which causes oncogenic phenotype, if the rescue of the cells transfected by the mutant APC is less than 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the rescue of cells transfected with the non-mutant APC, as judged by any of the criteria used to judge oncogenic phenotype of DLD-1 or HT29 cells.
  • NF-KB/FLIP inhibitors and JNK actovaotrs are not administered to cancer cells that have an activating mutation on ⁇ -catenin. Whether a cell contains a ⁇ -catenin mutation can be assayed for using standard recombinant
  • NF-KB/FLIP inhibitors and JNK activators can be any suitable inhibitors that can be used for example, sequencing and PCR analysis. The mutations can be assayed for function as discussed herein.
  • NF-KB/FLIP inhibitors and JNK activators can be any suitable inhibitors that can be used for example, sequencing and PCR analysis. The mutations can be assayed for function as discussed herein.
  • a cell expresses the COX-2 gene if there are detectable transcripts of the COX-2 gene in the cell using an assay to detect transcripts, such as a hybridization assay, such as a northern blot or any of the chip type assays available, or an amplification based assay based on, for example, PCR or other amplification methods.
  • the inhibitors can be administered to cancer cells that over express the COX-2 gene. Over expression of COX-2 can be determined by using any of the methods and assays discussed for the expression of COX-2 and comparing the level of expression to that of a control population of cells.
  • a given cancer cell or cancer cell line can be assayed for COX-2 expression and this can be compared to the COX-2 expression of this cell type in the absence of the oncogenic phenotype.
  • a given cancer cell line can be considered to over express COX-2 if the expression is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% of the expression of COX-2 in DLD-1 cells or HT29 cells, when expression of COX-2 of the cell of interest and DLD-1 or HT29 are assayed in parallel.
  • the disclosed inhibitors can also be administered to cancer cells that express the COX-2 gene as well as having mutations in the APC gene.
  • cancer cell lines are cells that are maintained in cell culture, but that arose from a specific type of cancer.
  • Inhibitors can be used for a variety of cancers, but can, for example, be used for cancers that are related to the DLD-1 cancer cell line and the HT-29 cancer cell line.
  • the DLD-1 cancer cell line and the HT-29 cancer cell line arose from colon cancer cells.
  • cancer cell lines having the properties of the DLD-1 cancer cell line and the HT29 cell line.
  • compositions can be used to treat any disease where
  • lymphomas Hodgkins and non-Hodgkins
  • leukemias carcinomas, carcinomas of solid tissues
  • squamous cell carcinomas adenocarcinomas
  • sarcomas gliomas, high grade gliomas, blastomas, neuroblastomas, plasmacytomas, histiocytomas, melanomas, adenomas, hypoxic tumors, myelomas, AIDS-related lymphomas or sarcomas, metastatic cancers, or cancers in general.
  • compositions can be used to treat is the following: lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, cervical carcinoma, breast cancer, and epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, stomach cancer, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, prostatic cancer, pancreatic cancer, or cancer stem cells.
  • Compounds disclosed herein may also be used for the treatment of precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.
  • precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.
  • NF-KB/FLIP inhibitors NF-KB/FLIP inhibitors
  • An NF-KB/FLIP inhibitor can be any composition that causes a decrease in the expression of anti-apoptotic proteins or an increase in the expression of apoptotic proteins.
  • the inhibitors can also be any composition wherein the composition inhibits ⁇ phosphorylation.
  • the inhibitor can also be any composition, wherein the composition inhibits TNFa induced NF- ⁇ activation.
  • a suitable inhibitor also activates JNK and inhibits FLIP.
  • the inhibitors useful in any of the methods disclosed herein are olefins.
  • An "olefin” is defined herein as any compound or molecule possessing at least one carbon-carbon double bond. Each carbon atom of the carbon-carbon double bond may be unsubstituted or independently substituted with one or two different moieties.
  • the inhibitor is an olefin having at least one electron- withdrawing group.
  • the inhibitor is an olefin having at least two electron-withdrawing groups. In this case, when two electron-withdrawing groups are present, the electron-withdrawing groups can be present on the same olefmic carbon atom or one electron-withdrawing group can be on each olefmic carbon atom.
  • electron-withdrawing group is any group that has an affinity or attraction for electron density.
  • C a olefmic carbon atom
  • Cp olefmic carbon atom
  • Cp is more susceptible to nucleophilic attack
  • electron withdrawing groups possess one or more carbon-carbon multiple bonds, carbon- heteroatom multiple bonds, or heteroatom-heteroatom multiple bonds.
  • electron-withdrawing groups include, but are not limited to, a cyano group, a sulfo-oxy group, a phospho-oxy group, a carboxyl group, a nitro group, a halogen, a halogenated alkyl group, an unsubstituted aromatic ring, or a substituted aromatic ring having at least one cyano group, sulfo-oxy group, phospho-oxy group, carboxyl group, hydroxyl group, amino group, ether group, halogenated alkyl group, halogen, or nitro group.
  • phospho-oxy group is a group having one of the following structures
  • R 1 is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic.
  • sulfo-oxy group is a group having one of the following structures
  • the inhibitor is an olefin having a cyano group and a sulfo-oxy group having the structure
  • R 2 is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic.
  • the inhibitor has the structure I.
  • R 3 , R 4 and R 5 are, independently, hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic, wherein the compound is the E- or Z-isomer.
  • the stereochemistry about the carbon-carbon double bond will vary depending upon the relative positions of the cyano group (-CN) and the sulfonyl group (- S(0 2 )R 5 )).
  • the cyano group and sulfonyl group are cis to one another, then the compound is the Z-isomer, and when the cyano group and sulfonyl group are trans to one another, then the compound is the E-isomer.
  • the inhibitor having the Structure I is the E-isomer.
  • R 3 and R 4 are hydrogen.
  • R 5 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, substituted or unsubstituted phenyl, or benzyl.
  • R 5 is a phenyl group having at least one alkyl group.
  • the inhibitor having the Structure I is the ethyl ester of 2- cyano-3-(methylsulfonyl)-3-(l ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl-2-propenoic acid;
  • the inhibitor is (2E)-3-(tolylsulfonyl)-2-propenenitrile, which is also referred to herein as BAY-7082.
  • the inhibitor is (2E)-3-[4-(tertiary butyl phenyl)sulfonyl]- 2-propenenitrile, which is also referred to herein as BAY- 11-7085.
  • the inhibitors useful in any of the methods disclosed herein can comprise at least one amino acid residue.
  • An "amino acid residue” is produced when an amino acid is reacted with one or more compounds capable of reacting with the amino acid. For example, when an amino acid is reacted with two other amino acids to produce a tripeptide, the resultant tripeptide contains three amino acid residues.
  • the amino acid can react with other non-amino acids to produce a compound having an amino acid residue.
  • the inhibitor has at least one leucine residue. In another example, the inhibitor comprises three leucine residues. In a further example, the compound is N-[(phenylmethoxy)carbonyl]-L-leucyl-N-[(lS)-l-formyl-3-methylbutyl]-L- leucinamide, which has the structure
  • This compound is also referred to as Sigma MG-132.
  • the inhibitor is pyrrolidine dithiocarbamate, which has the structure II:
  • R ⁇ and R 7 are, independently, hydrogen, alkyl alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic, or 3 ⁇ 4 and R 7 together for an ring with the nitrogen atom, and X and Y are, independently oxyten or sulfur, or the pharmaceutically active salt thereo.
  • X and Y are both oxygen. In another embodiment, X and Y are both sulfur. In a further embodiment, and R 7 can form a ring system.
  • R 6 and R 7 can be collectively a methylene group such as (CH 2 )3.
  • the resultant ring structure would be a four-membered ring with one nitrogen atom.
  • the ring can be from a three- to 10-membered ring.
  • X and Y are sulfur
  • R 6 and R 7 is (CH 2 ) 4 . This compound is also referred to as PDTC.
  • Formula II also enompasses pharmaceutically acceptable esters, amides, and salts of such compounds.
  • Pharmaceutically acceptable salts are prepared by treating the free acid with an appropriate amount of a pharmaceutically acceptable base.
  • Representative pharmaceutically acceptable bases are ammonium hydroxide, sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, magnesium hydroxide, ferrous hydroxide, zinc hydroxide, copper hydroxide, aluminum hydroxide, ferric hydroxide, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, lysine, arginine, histidine, and the like.
  • the reaction is conducted in water, alone or in combination with an inert, water-miscible organic solvent, at a temperature of from about 0°C to about 100°C, such as at room temperature.
  • the molar ratio of compounds of structural formula II to base used are chosen to provide the ratio desired for any particular salts.
  • the starting material can be treated with approximately one equivalent of pharmaceutically acceptable base to yield a neutral salt.
  • Ester derivatives are typically prepared as precursors to the acid form of the compounds— as illustrated in the examples below— and accordingly can serve as prodrugs. Generally, these derivatives will be lower alkyl esters such as methyl, ethyl, and the like.
  • Amide derivatives -(CO)NH 2 , -(CO)NHR, and -(CO)NR 2 , where R is an alkyl group defined above, can be prepared by reaction of the carboxylic acid-containing compound with ammonia or a substituted amine.
  • inhibitors include include derivatives of glitazones or thiazolidinediones, such as, troglitazone; rosiglitazone and pioglitazone.
  • Other useful thiazolidinediones include ciglitazone, englitazone, darglitazone and BRL 49653 as disclosed in WO 98/05331 which is incorporated herein by reference.
  • inhibitors can be obtain from commercially available libraries, for example, kinase inhibitors libraries (BioMol), synthetic compound libraries (e.g., from Bayer or Sigma Aldrich), natural compound libraries (Specs, TimTec), or other libraries from companies such as Sigma-Aldrich (St. Louis, Mass.), Arqule (Woburn, Ma.), Enzymed (Iowa City, Iowa), Maybridge Chemical Co.(Trevillett, Cornwall, UK), MDS Panlabs (Bothell, Wash.), Pharmacopeia (Princeton, N.J.), and Trega (San Diego, Calif).
  • Preferred compounds are low molecular weight compounds.
  • Low molecular weight compounds i.e., with a molecular weight of 500 Dalton or less, are likely to have good absorption and permeation in biological systems and are consequently more likely to be successful drug candidates than compounds with a molecular weight above 500 Dalton.
  • Compounds including natural products, inorganic chemicals, and biologically active materials such as proteins and toxins can also be assayed using these methods for the ability to inhibit NF- ⁇ and/or FLIP.
  • Specific screening methods are known in the art and along with integrated robotic systems and collections of chemical compounds/natural products are extensively incorporated in high throughput screening so that large numbers of test compounds can be tested for antagonist or agonist activity within a short amount of time.
  • These methods include homogeneous assay formats such as fluorescence resonance energy transfer, fluorescence polarization, time-resolved fluorescence resonance energy transfer, scintillation proximity assays, reporter gene assays, fluorescence quenched enzyme substrate, chromogenic enzyme substrate and electrochemiluminescence, as well as more traditional heterogeneous assay formats such as enzyme-linked immunosorbant assays (ELISA) or radioimmunoassays.
  • ELISA enzyme-linked immunosorbant assays
  • Homogeneous assays are "mix and read” assays that are very amenable to robotic application, whereas heterogeneous assays require separation of bound analyte from free by more complex unit operations such as filtration, centrifugation or washing. These assays are utilized to detect a wide variety of specific biomolecular interactions and the inhibition thereof by small organic molecules, including protein- protein, receptor-ligand, enzyme-substrate, etc. These assay methods and techniques are well known in the art and are described more fully in the following: High Throughput Screening: The Discovery of Bioactive Substances, John P. Devlin (ed.), Marcel Dekker, New York, 1997.
  • the screening assays of the present invention are amenable to high throughput screening of chemical libraries and are suitable for the identification of small molecule drug candidates, antibodies, peptides, and other antagonists and/or agonists.
  • inhibitors which are direct inhibitors of NF- ⁇ and/or FLIP.
  • a direct inhibitor is an inhibitor that interacts directly with NF- ⁇ and/or FLIP.
  • a direct inhibitor touches in some way the NF- ⁇ molecule and/or the FLIP molecule such that the NF-KB and/or FLIP dependent activities, such as readhesion, metastasis, and apoptosis supression are inhibited.
  • Indirect inhibitors are direct inhibitors of NF- ⁇ and/or FLIP.
  • inhibitors which are indirect inhibitors of NF- ⁇ and.or FLIP.
  • An indirect inhibitor is an inhibitor that does not interact directly with NF- ⁇ and/or FLIP.
  • An indirect inhibitor touches in some way a molecule that is involved in a signal transduction pathway that NF- ⁇ and/or FLIP is involved in such that the NF- ⁇ and/or FLIP dependent activities, such as readhesion and metastasis are inhibited.
  • an indirect inhibitor of NF- ⁇ and/or FLIP would be an indirect inhibitor which inhibits the expression of NF- ⁇ and/or FLIP and thereby prevent NF-KB and/or FLIP from functioning.
  • Another example of an indirect inhibitor of NF-KB would be an indirect inhibitor that inhibits translation of a gene encoding NF- ⁇ and/or FLIP.
  • An example of a molecule that is involved in a signal transduction pathway that NF-KB is involved in is ⁇ .
  • Molecules that inhibit ⁇ from phosphorylating can be indirect inhibitors.
  • compositions can also be administered in vivo in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject, along with the nucleic acid or vector, without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • the carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject, as would be well known to one of skill in the art.
  • the compositions may be administered orally, parenterally ⁇ e.g.,
  • topical intranasal administration means delivery of the compositions into the nose and nasal passages through one or both of the nares and can comprise delivery by a spraying mechanism or droplet mechanism, or through aerosolization of the nucleic acid or vector. The latter may be effective when a large number of animals is to be treated simultaneously.
  • compositions by inhalant can be through the nose or mouth via delivery by a spraying or droplet mechanism. Delivery can also be directly to any area of the respiratory system (e.g. , lungs) via intubation.
  • the exact amount of the compositions required will vary from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the allergic disorder being treated, the particular nucleic acid or vector used, its mode of administration and the like. Thus, it is not possible to specify an exact amount for every composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein.
  • compositions if used, are generally characterized by injection.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution of suspension in liquid prior to injection, or as emulsions.
  • a more recently revised approach for parenteral administration involves use of a slow release or sustained release system such that a constant dosage is maintained. See, e.g., U.S. Patent 3,610,795, which is incorporated by reference herein.
  • the compositions disclosed herein can be administered
  • the disclosed compositions can be used as a wash. In this way, cancer cells that were not excised and/or cells that may be unattached but still present in the body, can be killed upon readhereing.
  • the disclosed compositions can be administered around the time of surgery (peri-operative), before surgery (pre-operative), or after surger (post-operatively).
  • the compounds can be administered a week, 2-5 days, 1-3 days, 1-18 hours, 1-12 hours, 1-6 hours, or less than an hour before or after tumor resection surgery.
  • compositions can be administered by I.V., by injection and/or an I.V. drip.
  • the materials may be in solution, suspension (for example, incorporated into microparticles, liposomes, or cells). These may be targeted to a particular cell type via antibodies, receptors, or receptor ligands.
  • the following references are examples of the use of this technology to target specific proteins to tumor tissue (Senter, et al,
  • Vehicles such as "stealth” and other antibody conjugated liposomes (including lipid mediated drug targeting to colonic carcinoma), receptor mediated targeting of DNA through cell specific ligands, lymphocyte directed tumor targeting, and highly specific therapeutic retroviral targeting of murine glioma cells in vivo.
  • the following references are examples of the use of this technology to target specific proteins to tumor tissue (Hughes et al, Cancer Research, 49:6214-6220, (1989); and Litzinger and Huang, Biochimica et Biophysica Acta, 1104: 179-187, (1992)).
  • receptors are involved in pathways of endocytosis, either constitutive or ligand induced.
  • receptors cluster in clathrin-coated pits, enter the cell via clathrin-coated vesicles, pass through an acidified endosome in which the receptors are sorted, and then either recycle to the cell surface, become stored intracellularly, or are degraded in lysosomes.
  • the internalization pathways serve a variety of functions, such as nutrient uptake, removal of activated proteins, clearance of macromolecules, opportunistic entry of viruses and toxins, dissociation and degradation of ligand, and receptor-level regulation. Many receptors follow more than one intracellular pathway, depending on the cell type, receptor concentration, type of ligand, ligand valency, and ligand concentration.
  • compositions including antibodies, can be used therapeutically in combination with a pharmaceutically acceptable carrier.
  • compositions are known to those skilled in the art. These most typically would be standard carriers for administration of drugs to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH. The compositions can be administered intramuscularly or subcutaneously. Other compounds will be administered according to standard procedures used by those skilled in the art. [00133] Pharmaceutical compositions may include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice. Pharmaceutical compositions may also include one or more active ingredients such as antimicrobial agents, antiinflammatory agents, anesthetics, and the like.
  • the pharmaceutical composition may be administered in a number of ways depending on whether local or systemic treatment is desired, and on the area to be treated. Administration may be topically (including ophthalmically, vaginally, rectally, intranasally), orally, by inhalation, or parenterally, for example by intravenous drip, subcutaneous, intraperitoneal or intramuscular injection.
  • the disclosed antibodies can be administered intravenously, intraperitoneally, intramuscularly, subcutaneous ly, intracavity, or transdermally.
  • Preparations for parenteral administration include sterile aqueous or nonaqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
  • Formulations for topical administration may include ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • compositions may be necessary or desirable.
  • compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, or tablets. Thickeners, flavorings, diluents, emulsifiers, dispersing aids or binders may be desirable..
  • compositions may potentially be administered as a
  • pharmaceutically acceptable acid- or base- addition salt formed by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases such as mono-, di-, trialkyl and aryl amines and substituted ethanolamines.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid
  • organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyru
  • the dosage ranges for the administration of the compositions are those large enough to produce the desired effect in which the symptoms disorder are effected.
  • the dosage should not be so large as to cause adverse side effects, such as unwanted cross- reactions, anaphylactic reactions, and the like.
  • the dosage will vary with the age, condition, sex and extent of the disease in the patient and can be determined by one of skill in the art.
  • the dosage can be adjusted by the individual physician in the event of any counterindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days.
  • compositions can be used as targets for any combinatorial technique to identify molecules or macromolecular molecules that interact with the disclosed compositions in a desired way. Also disclosed are the compositions that are identified through combinatorial techniques or screening techniques in which the compositions interact with NF- ⁇ , FLIP, JNK, and/or ⁇ such that the compositions decrease the cellular proliferative activity of NF- ⁇ , FLIP, and ⁇ or portions thereof, increase INK, where the compositions were identified using NF- ⁇ , FLIP, JNK, or ⁇ as targets in a screening or selection protocol.
  • Combinatorial chemistry includes but is not limited to all methods for isolating small molecules or macromolecules that are capable of binding either a small molecule or another macromolecule, typically in an iterative process.
  • Proteins, oligonucleotides, and sugars are examples of macromolecules.
  • oligonucleotide molecules with a given function, catalytic or ligand-binding can be isolated from a complex mixture of random oligonucleotides in what has been referred to as "in vitro genetics" (Szostak, TIBS 19:89, 1992).
  • Combinatorial techniques are particularly suited for defining binding interactions between molecules and for isolating molecules that have a specific binding activity, often called aptamers when the macromolecules are nucleic acids.
  • RNA molecule is generated in which a puromycin molecule is covalently attached to the 3 '-end of the RNA molecule.
  • An in vitro translation of this modified RNA molecule causes the correct protein, encoded by the RNA to be translated.
  • the puromycin a peptdyl acceptor which cannot be extended, the growing peptide chain is attached to the puromycin which is attached to the RNA.
  • the protein molecule is attached to the genetic material that encodes it. Normal in vitro selection procedures can now be done to isolate functional peptides. Once the selection procedure for peptide function is complete traditional nucleic acid manipulation procedures are performed to amplify the nucleic acid that codes for the selected functional peptides.
  • RNA is transcribed with puromycin at the 3'- end, new peptide is translated and another functional round of selection is performed.
  • protein selection can be performed in an iterative manner just like nucleic acid selection techniques.
  • the peptide which is translated is controlled by the sequence of the RNA attached to the puromycin. This sequence can be anything from a random sequence engineered for optimum translation (i.e. no stop codons etc.) or it can be a degenerate sequence of a known RNA molecule to look for improved or altered function of a known peptide.
  • the method utilizes a library of peptide molecules that attached to an acidic activation domain.
  • a peptide of choice for example a portion of NF- ⁇ , FLIP, JNK, or ⁇ is attached to a DNA binding domain of a transcriptional activation protein, such as Gal 4.
  • a transcriptional activation protein such as Gal 4.
  • Combinatorial libraries can be made from a wide array of molecules using a number of different synthetic techniques. For example, libraries containing fused 2,4- pyrimidinediones (U.S. Patent 6,025,371) dihydrobenzopyrans (U.S. Patent 6,017, 768and 5,821,130), amide alcohols (U.S. Patent 5,976,894), hydroxy-amino acid amides (U.S. Patent 5,972,719) carbohydrates (U.S. Patent 5,965,719), l,4-benzodiazepin-2,5-diones (U.S. Patent 5,962,337), cyclics (U.S. Patent 5,958,792), biaryl amino acid amides (U.S.
  • Patent 5,847,150 quinolines
  • PNA U.S. Patent 5,831,014
  • tags U.S. Patent 5 ,721 ,099
  • polyketides U. S . Patent 5,712,146
  • morpholino-subunits U. S . Patent 5,698,685 and 5,506,33
  • sulfamides U.S. Patent 5,618,825
  • benzodiazepines U.S. Patent 5,288,514).
  • combinatorial methods and libraries included traditional screening methods and libraries as well as methods and libraries used in iterative processes.
  • compositions can be used as targets for any molecular modeling technique to identify either the structure of the disclosed compositions or to identify potential or actual molecules, such as small molecules, which interact in a desired way with the disclosed compositions.
  • kits that are drawn to reagents that can be used in practicing the methods disclosed herein.
  • the kits can include any reagent or combination of reagent discussed herein or that would be understood to be required or beneficial in the practice of the disclosed methods.
  • the kits could include molecules, including for example, BAY 11-7082 or BAY 11-7085, for use in in vitro cell assays as standards for anti-proliferative activity.
  • compositions such as BAY 11-7082 and BAY 11- 7085, disclosed herein have certain functions, such as antimetastatic activities or antiproliferative activities.
  • certain structural requirements for performing the disclosed functions and it is understood that there are a variety of structures which can perform the same function which are related to the disclosed structures, and that these structures will ultimately achieve the same result, for example, inhibition of antiproliferative activities.
  • compositions disclosed herein and the compositions necessary to perform the disclosed methods can be made using any method known to those of skill in the art for that particular reagent or compound unless otherwise specifically noted.
  • compositions Disclosed are processes for making the compositions as well as making the intermediates leading to the compositions. There are a variety of methods that can be used for making these compositions, such as synthetic chemical methods and standard molecular biology methods. It is understood that the methods of making these and the other disclosed compositions are specifically disclosed.
  • animals produced by the process of transfecting a cell within the animal with any of the nucleic acid molecules disclosed herein Disclosed are animals produced by the process of transfecting a cell within the animal any of the nucleic acid molecules disclosed herein, wherein the animal is a mammal. Also disclosed are animals produced by the process of transfecting a cell within the animal any of the nucleic acid molecules disclosed herein, wherein the mammal is mouse, rat, rabbit, cow, sheep, pig, or primate.
  • NF- ⁇ inhibitors BAY 11-7082 (BioMol), BAY 11-7085, (BioMol) and N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132) (Sigma) were solubilized in DMSO.
  • the antagonistic and chimeric TNFa monoclonal antibody (cA2) was purchased from the clinical pharmacy at the University of Utah and reconstituted in sterile water.
  • Polyclonal antibodies to FLIP S / L , c-IAP-1, c-IAP-2, TRAF-1, and TRAF-2 were obtained from Santa Cruz Biotechnology.
  • PARP poly(ADP-ribose) polymerase
  • DLD-1, HCT-116, and HT-29 colon cancer cell lines were obtained from the ATCC collection and were cultured in Dulbecco's Modification of Eagle's Medium (DMEM) supplemented with 10% fetal bovine serum, glutamine, penicillin, and streptomycin.
  • DMEM Dulbecco's Modification of Eagle's Medium
  • 293 cells stably transfected with COX-2 (293 -COX-2) under the control of a ponasterone sensitive promoter were cultured in Dulbecco's Modification of Eagle's Medium supplemented with 10% fetal bovine serum, 400 ⁇ g/mL of zeocin, 400 ⁇ g/mL of G418, glutamine, penicillin, and streptomycin.
  • 293-COX-2 cells were cultured for 48 hours in media supplemented with ⁇ g/ml of ponasterone. All cells were cultured at 37°C in a humidified incubator with a 5%> C0 2 atmosphere.
  • cells were dispersed and plated at 40,000 cells/well in 96-well dishes. At various days in culture, the cells were gently washed twice with 100 ⁇ of ice-cold blocking buffer (1% radioimmunoassay grade BSA in PBS) and twice with 100 ⁇ /well of ice-cold PBS. The cells were fixed for 10 min in 100%) ice-cold methanol (100 ⁇ /well), then allowed to air-dry. The cells were stained with ⁇ of 0.1 % crystal violet in H 2 0 for 10 minutes, then washed gently four times with dd H 2 0 and four times with PBS. The plates were then air-dried completely. The stained cells were then solubilized in 1% sodium deoxycholate, and the plates read at 590 nm in a spectrophotometer. The absorption at 590 nm is proportional to the number of attached cells.
  • ice-cold blocking buffer 1% radioimmunoassay grade BSA in PBS
  • CYTOTOX96TM kit from Promega was used according to the manufacturer's instructions. Briefly, DLD-1, HCT-116, and HT-29 cells were incubated with 1, 2.5, 5, 7.5 and 10 ⁇ BAY 11-7082 or BAY 11-7085 in 96-well plates for 24 hours then lysed by adding 15 ⁇ , of lysis 10X Solution (9% (v/v)
  • the adherent cell monolayers were washed once with cold PBS after which 200 ⁇ iL of cold Buffer A (20 mM HEPES pH 7.8, 1 mM EDTA, 1 mM EGTA, 10% glycerol, 0.2% NP-40, 0.2 mM Na 3 V0 4 , 1 mM DTT, 0.5 mM PMSF,and, 1 ⁇ g/mL each of aprotinin and leupeptin) was added to the cells. The cells were scraped into microfuge tubes, kept on ice, and sonicated. The lysates were centrifuged at 500 g for 5 min at 4°C and the cytoplasmic fraction (supernatant) was removed.
  • cold Buffer A 20 mM HEPES pH 7.8, 1 mM EDTA, 1 mM EGTA, 10% glycerol, 0.2% NP-40, 0.2 mM Na 3 V0 4 , 1 mM DTT, 0.5 mM PM
  • Buffer B (20 mM HEPES pH 7.8, 1 mM EDTA, 1 mM EGTA, 0.42 M NaCl, 1.5 mM MgCl 2 , 25% glycerol, 0.2 mM Na 3 VO 4 , 1 mM DTT, 0.5 mM PMSF, and, 10 ⁇ g/mL each of aprotinin and leupeptin
  • the nuclear fraction pellet
  • the suspension was vortexed, kept on ice for 30 minutes, and centrifuged at 13,000 g for 10 minutes.
  • the supernatants (nuclear fraction) were normalized for total protein concentration using the BCA assay (Pierce et al. , "Novel inhibitors of cytokine-induced ⁇ phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo " J Biol Chem 272:21096-103, 1997).
  • Activation of NF- ⁇ was also determined using the TRANS-AMTM assay (Active Motif, Carlsbad, California) per the manufacturer's instructions. (Renard et al., "Development of a sensitive multi-well colorimetric assay for active NF- ⁇ ," Nucleic Acids Res 29, E21 , 2001). Briefly, cell monolayers on 60 mm dishes were washed with ice-cold PBS and removed by incubating in trypsin-PBS or scraping.
  • the cells were centrifuged for 10 minutes at 1 ,000 rpm at 4°C and resuspended for 10 minutes in ⁇ of 4°C lysis buffer (20 mM HEPES (pH 7.5), 350 mM NaCl, 20% glycerol, 1% Igepal- CA630, 1 mM MgCl 2 , 0.5 mM EDTA, 0.1 mM EGTA, 1 ⁇ , of 1 M dithiothreitol and 10 of protease inhibitor cocktail (proprietary) per mL of lysis buffer).
  • the lysates were centrifuged for 20 minutes at 14,000 g at 4°C.
  • Lysates containing 5 ⁇ g of total protein were added to 20 ⁇ , of lysis buffer per well in 96-well dishes containing immobilized oligonucleotides corresponding to the NF- ⁇ consensus DNA binding site (SEQ ID NO:2) (5 '-GGGACTTTCC-3 ').
  • the plate was covered with an adhesive film and incubated for 1 hour at room temperature on a rocker.
  • the wells were then washed three times with 200 ⁇ per well of washing buffer (100 mM phosphate buffer (pH 7.5), 500 mM NaCl, 1% Tween).
  • p65 subunit monoclonal antibody 100 ⁇ , of p65 subunit monoclonal antibody (the p65 antibody supplied with the kit only recognizes p65 -containing NF- ⁇ heterodimers that are bound to DNA containing the NF-KB consensus binding sequence) was diluted 1 : 1 ,000 in IX antibody binding buffer (4 mM HEPES (pH7.5), 120 mM KCl, 8% glycerol, 1% bovine serum albumin) and added to each well and incubated for 1 hour at room temperature. The wells were washed with 100 ⁇ , of washing buffer three times.
  • IX antibody binding buffer 4 mM HEPES (pH7.5), 120 mM KCl, 8% glycerol, 1% bovine serum albumin
  • Single cell suspensions were plated at 100,000 cells per well in 24-well plates in media containing DMSO, BAY 11-7082, BAY 11-7085, or MG-132 for 8-24 hours in an incubator.
  • DMSO, BAY 11-7082, BAY 11-7085, or MG-132 were added to cells that were confluent and adherent for several days.
  • the nonadherent cells were aspirated off with the media and spun for 3 minutes at 2,000 rpm in 15 mL polypropylene tubes.
  • the adherent cells were washed twice with PBS, dispersed in trypsin, and spun down for 3 minutes at 2,000 rpm in 15 mL polypropylene tubes.
  • the nonadherent and adherent cells were resuspended in IX binding buffer (10 mM
  • ⁇ of the cell solution were transferred to a 5 mL culture tube and incubated with 5 of annexin V-FITC-conjugated monoclonal antibody (Pharmingen) and 10 of propidium iodide (from a 50 ⁇ g/mL stock solution made in PBS) in the dark at 25°C for 15 minutes. 400 ⁇ , of binding buffer was added to each tube and the cells were analyzed by flow cytometry immediately. The percent of apoptosis was determined as the percent of annexin V-positive, propidium iodide -negative cells of the total cells counted.
  • DLD-1 and HT-29 cells that were approximately 60-70% confluent were transduced with Ad- ⁇ super-repressor by adding 1-50 ⁇ of purified virus to the cells. After 3 days, the expression of the ⁇ super-repressor was ascertained by immunoblot. In a parallel experiment, the transduced cells were resuspended and allowed to readhere in BAY 11-7085 for 8 h. The percentage of surviving (nonapoptotic) cells was determined using crystal violet staining.
  • the cells were washed twice in ice cold PBS and then lysed in 4°C lysis buffer (50 mM HEPES, 150 mM NaCl, 1.5 mM MgCl 2 , 1 mM EGTA, 100 mM NaF, 10 mM Na 2 P0 4 , 1 mM Na 3 V0 4 , 10% glycerol, 1% TRITONTM X-100, and ⁇ g/mL each of aprotinin, leupeptin, chymostatin, pepstatin) and clarified at 12,000 rpm at 4°C for 15 minutes.
  • the lysates were normalized for total protein concentration.
  • the blocked blots were probed with primary antibody for 2 hours at 4°C on a rocker.
  • the blots were washed twice for 10 minutes each in blocking buffer, then incubated with a 1 :2000 dilution of a rabbit-anti-mouse secondary antibody (for monoclonal primary antibodies; for polyclonal antibodies, this step was skipped).
  • HT-29 and HCT-116 cells were harvested in 0.25% trypsin-PBS-EDTA, washed once in media and PBS, and resuspended in PBS at 1 million cells per 200 ⁇ ,.
  • One million cells were injected either subcutaneous ly in the backs or intraperitoneally in 5 week old female nu/nu athymic mice (Charles River Labs). For the mice with
  • mice subcutaneous tumors, the tumors were allowed to establish themselves for 10 days after which they were randomized to BAY 11-7085 or DMSO.
  • mice were randomized to BAY 11-7085 or DMSO.
  • mice received DMSO or BAY 11-7085 (5 mg/kg) 24 hours before the cancer cells were injected intraperitoneally. All mice received BAY 11-7085 (5 mg/kg) or an equal volume of DMSO (about 200 ⁇ ,) twice weekly for 21-32 days.
  • Subcutaneous tumor sizes were determined by measuring the length and width with calipers. These studies were approved by the University of Utah Institutional Review Board and Institutional Animal Care and Use Committee and performed in the Animal Resource Center. Mice were euthanized when they experienced a greater than 10% loss in body weight or if they appeared ill. Post-mortem examinations included sectioning of kidney, lung, and liver tissues which were stained with hematoxylin and eosin followed by examination for tissue toxicity/damage by an experienced mouse pathologist (E.J.E.) who was blinded to therapy. Results
  • NF-KB inhibitors diminish colon cancer cell proliferation
  • BAY 11-7082 and BAY 11-7085 Two compounds, BAY 11-7082 and BAY 11-7085, known to inhibit NF-KB and arthritis in rodent models (Pierce et al, "Novel inhibitors of cytokine-induced ⁇ phosphorylation and endothelial cell adhesion molecule expression show antiinflammatory effects in vivo " J Biol Chem 272:21096-103, 1997) were tested on colon cancer cell lines.
  • BAY 11-7082 and BAY 11-7085 are soluble compounds that inhibit ⁇ phosphorylation and TNFa-induced NF- ⁇ activation (Id.). They were also found to activate JNK/SAPK and p38, members of the MAP kinase family of signal transduction proteins (Id.). Incubation of the colon cancer cell lines, DLD-1, HCT-116, HT-29, with 10 ⁇ of BAY 11-7085, but not BAY 11-7082 greatly inhibited cell proliferation (FIGs. 1A and
  • NF- ⁇ electrophoretic mobility shift assays were performed. Following 3 and 6 hour incubations of HT-29 cells with the two compounds at 10 ⁇ concentrations, a more profound inhibition of NF- ⁇ activity occurred with BAY 11-7085 than BAY 11-7082 (FIG. 1C). Consistent with previous reports in a variety of cancer cells, HT-29 colon cancer cells demonstrated constitutive activation of NF-KB. DLD-1 and HCT-116 cells demonstrated constitutive activation of NF-KB by EMS A as well (data not shown).
  • HT-29 colon cancer cells were treated with the NF- ⁇ inhibitors, BAY 11-7085 or MG-132 for 24 h, the percentage of apoptotic cells (annexin V-positive and propidium iodide-negative) was relatively greater for MG-132 than BAY 11-7085, and was proportional to the number of floating cells. It was confirmed in HT-29 cells that were treated with increasing concentrations of BAY 11-7085 for 24 h. After separating the HT-29 cells that remained adherent and those that became nonadherent after BAY 11- 7085 treatment, the percentage of apoptotic cells was determined for each group.
  • nonadherent apoptotic cells were apoptotic, only a minority of the adherent cells was apoptotic.
  • the percentage of nonadherent apoptotic cells decreased after treatment with 50 and 100 ⁇ BAY 11-7085 compared with lower concentrations of BAY 11-7085, because many of the cells were positive for both annexin V and propridium iodide indicating that they had completed apoptosis and were now necrotic.
  • HT-29 cells were treated with 20 ⁇ MG-132 for 8 h, after which the adherent and nonadherent cells were collected separately, and then lysed and immunoblotted for cleaved PARP, a product of caspase cleavage.
  • cleaved PARP a product of caspase cleavage.
  • the loss of colon cancer cell adhesion was associated with the induction of apoptosis by IFN and TNFa.
  • the inhibition of colon cancer cell adhesion appears to be related to the apoptotic effects of the NF-KB inhibitors.
  • NF-KB inhibitors inhibit colon cancer cell tumorigenicity
  • Anchorage-independent cell growth is a hallmark of cancer cells and is a good correlation to tumorigencity in vivo. Therefore, the effects of BAY 11-7082 and BAY 11- 7085 were examined on colon cancer cell growth using the soft agar colony formation assay. At 10 ⁇ concentrations of BAY 11-7082 or BAY 11-7085 for 6 days, anchorage- independent proliferation of the DLD-1 and HT-29, but not HCT-116, colon cancer cell lines was significantly inhibited (FIG. 2A).
  • HCT-116 cells While HT-29 cells readily formed tumors in the athymic mice, HCT-116 cells formed only very small tumors in most cases. There was a statistically significant overall reduction in the tumor volumes in the HT-29 xenografts in the BAY 11-7085 treatment group compared with the control group (FIG. 2B). There was no statistically significant overall difference in the tumor volumes in the HCT-116 xenografts in the treatment or control groups (FIG. 2C).
  • NF-KB inhibitor-induced apoptosis is associated with a loss and inhibition of cell adhesion
  • HT-29 cells were treated with increasing concentrations of BAY 11-7085 for 8 hours, after which the percentage of apoptotic cells was determined separately for the cells that remained adherent and those that became non-adherent. While the non-adherent cells were predominantly apoptotic, only a minority of the adherent cells was apoptotic (FIG. 3B). Note that the percentage of apoptosis in the non-adherent cell fraction decreased at 50 and 100 ⁇ concentrations of BAY 11-7085.
  • BAY 1 1-7085 Various concentrations of BAY 1 1-7085 were added to HT-29 cells that were transiently suspended in trypsin-PBS for approximately 15 minutes before replating. The transiently suspended cells were quickly allowed to readhere in order to prevent anoikis.
  • BAY 11-7085 was added to adherent HT-29 cell monolayers. BAY-11- 7085 at all concentrations tested caused a significant increase in apoptosis of the transiently suspended HT-29 cells (FIG. 4A). On the other hand, even the highest doses of BAY 11-7085 caused only a relatively small degree of apoptosis of adherent compared with transiently suspended HT-29 cells (FIG. 5).
  • the scraped cells were lysed and added to 96-well plates containing immobilized oligonucleotides corresponding to the NF-KB consensus binding sequence (TRANS-AMTM assay).
  • NF- ⁇ binding to the immobilized oligonucleotides was detected using a p65 monoclonal primary antibody and a horseradish peroxidase-conjugated anti-mouse secondary antibody.
  • the assay showed that there was relatively little activation of NF- ⁇ compared to the positive control (HeLa cells treated with TNF ) (FIG. 5 A).
  • the transiently suspended cells were allowed to readhere, a large induction of NF- ⁇ activation occurred (FIG. 5A).
  • NF- ⁇ binding assay was used to determine whether NF-KB inhibitors could diminish the activation of NF- ⁇ by readhesion.
  • HT-29 cells transiently suspended with trypsin were mixed with increasing concentrations of BAY 11-7085 then placed on culture dishes for 8 hours.
  • adherent HT-29 cells were treated with increasing concentrations of BAY 11-7085 for 8 hours.
  • the cells were scraped, and all the cells were collected and lysed.
  • the NF- ⁇ binding assay confirmed the strong activation of NF- ⁇ caused by the readhesion of transiently suspended HT-29 cells and demonstrated a dose-related inhibition of this NF- ⁇ activation by BAY 11-7085 (FIG. 5B, transiently suspended cells).
  • NF- ⁇ activation was determined in adherent and confluent DLD-1, HCT-116, Caco-2 and HT-29 cells that were removed from plastic dishes by trypsin or scraping. Scraping of the cells did not result in NF- ⁇ activation (FIG. 5C). In addition, cells that were transiently suspended in trypsin-PBS but not allowed to readhere, did not show significantly more activation of NF- ⁇ by the TRANS-AMTM assay (FIG. 5C).
  • DLD-1, HCT-116, Caco-2 and HT-29 colon cancer cell lines were transiently suspended, treated with BAY 11-7085 and allowed to readhere. While DLD-1 and HT-29 cells showed the same propensity for BAY 11-7085-induced apoptosis, Caco-2 cells showed a moderate increase, while HCT-116 cells did not (FIG. 5C).
  • HCT-116 cells do not, (Groden et al, "Response of colon cancer cell lines to the introduction of APC, a colon- specific tumor suppressor gene," Cancer Res 55: 1531-9, 1995); however, HCT-116 cells carry activating mutations in the ⁇ -catenin gene (CTNNB1) (Ilyas et al, " ⁇ -catenin mutations in cell lines established from human colorectal cancers," Proc Natl Acad Sci USA 94: 10330- 4, 1997) whose gene product is normally regulated by the APC gene product (Munemitsu et al, "Regulation of intracellular ⁇ -catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein," Proc Natl Acad Sci USA 92:3046-50, 1995; Rubinfeld et al.
  • APC adenomatous polyposis coli
  • HCT- 116 cells do not express COX-2 while DLD-1 and HT-29 cells do (His et al., "Lack of cyclooxygenase-2 activity in HT-29 human colorectal carcinoma cells," Exp Cell Res 256:563-70, 2000; Shao et al, "Regulation of constitutive cyclooxygenase-2 expression in colon carcinoma cells," J Biol Chem 275:33951-6, 2000; Tsuji et al, "Evidences for involvement of cyclooxygenase-2 in proliferation of two gastrointestinal cancer cell lines," Prostaglandins Leukot Ess ent Fatty Acids 55:179-83, 1996).
  • COX-2 is commonly overexpressed in colorectal tumors and plays roles in survival and metastasis in colorectal cancers.
  • COX-2 overexpression plays a role in the susceptibility of the colon cancer cells to BAY 11-7085-induced apoptosis.
  • 293 cells transfected with a ponasterone- inducible COX-2 construct (293-COX-2) were treated with BAY 11-7085.
  • Treatment of 293-COX-2 cells with 1 ⁇ g/mL of ponasterone for 48 hours led to a large induction of COX-2 protein, while the uninduced cells showed no measurable COX-2 protein expression by western blot (FIG. 5E)
  • Uninduced 293-COX-2 cells showed no increase in apoptosis following treatment with BAY 11-7085 (FIG. 5F).
  • MG-132 and PDTC Two other soluble NF- ⁇ inhibitors, MG-132 and PDTC, were used to test their activity in the induction of apoptosis of colon cancer cells during readhesion.
  • MG-132 more potently induced apoptosis of HT-29 colon cancer cells during readhesion.
  • PDTC caused apoptosis of HT-29 cells during readhesion as well.
  • IKB super-repressor in DLD-1 and HT-29 cells significantly lowered the apoptotic threshold of BAY 11-7085 compared with the controls.
  • the effect of the IKB super-repressor was much greater for DLD-1 than HT-29 cells because the former expressed higher levels of IKB super-repressor protein than the latter cells.
  • the IKB super-repressor protein was functional in both cell lines.
  • NF-KB inhibitor prevents intraabdominal metastasis in vivo
  • BAY 11-7085 induced apoptosis of readherent HT-29 cells
  • an in vivo model was used to test the ability of the drug to prevent metastasis by seeding of intraabdominal tissue with colon cancer cells.
  • seeding of the peritoneal cavity by tumor cells with tumor cell implantation of the peritoneal surfaces can occur. Seeding results from the transient suspension of cancer cells, through a loss of adhesion either naturally ro because of surgical displacement, followed by readhesion to other tissues.
  • the HT-29 and HCT-116 colon cancer cell lines were injected into the abdominal cavities of athymic mice that had been pretreated 24 hours earlier with either intraperitoneal vehicle alone (DMSO) or BAY 11-7085 (1 mg/kg). The mice were then treated twice weekly with vehicle or BAY 11-7085 for a total of 21 days. Mice sacrificed 7 days after the introduction of colon cancer cells intraabdominally showed no evidence of tumoral implantation of the parietal or visceral peritoneal surfaces.
  • DMSO intraperitoneal vehicle alone
  • BAY 11-7085 1 mg/kg
  • mice that had been injected intraabdominally with HT-29 cells and treated with vehicle, 5 of 6 developed metastases of the parietal peritoneum and all 6 developed hepatic metastases (FIG. 6C and Table 3). Interestingly, the majority of the metastases involved ventral and dependent areas of parietal and visceral peritoneum, suggesting that the colon cancer cells took some time to implant.
  • NF-KB inhibitors decrease expression of anti-apoptotic proteins
  • TNFa is expressed by a number of colon cancer cell lines including HT-29 cells (Jung et al., "A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion," J Clin Invest 95:55-65, 1995).
  • HT-29 cells were pretreated with a monoclonal antibody, cA2, which inhibits TNFa binding to TNFa receptors (D'Haens et al., "Endoscopic and histological healing with infliximab anti-tumor necrosis factor antibodies in Crohn's disease: A European multicenter trial," Gastroenterology 116: 1029- 34, 1999) followed by treatment with BAY 11-7085.
  • cA2 monoclonal antibody
  • NF-KB regulates the expression of a number of genes, including c-IAP-1, c- IAP-2, TRAF-1, and TRAF-2, that encode proteins that mediate cell survival (Stehlik et al., "Nuclear factor (NF)-KB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha- induced apoptosis," J Exp Med
  • NF- ⁇ inhibitors cause apoptosis of colon cancer cells in a two- step process.
  • the NF- ⁇ inhibitors inhibit cell adhesion of adherent colon cancer cells in vitro.
  • the readhesion of these floating cells causes a large and transient activation of NF-KB. This renders the readherent cells extremelyly susceptible to NF- ⁇ inhibitor- induced apoptosis.
  • NF- ⁇ is an important survival factor for certain cancer cells, such as colon cancer cells, during the process of readhesion, particularly cancer cells that express COX-2 and/or mutant APC genes.
  • compositions disclosed herein can be used for the prevention of metastasis.
  • the process of metastasis has been proposed to involve a number of sequential steps: invasion, dissociation, intavasation into the circulatory or lymphatic systems, dissemination, arrest in the microcirculation, extravasation, and invasion of distant tissues (Engers et ah,
  • metastasis is surprisingly inefficient with as few as 0.05% of circulating tumor cells producing stable metastases (Liotta et al., "Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation," Cancer Res 34:997-1004, 1974; Nicolson, “Gene expression, cellular diversification and tumor progression to the metastatic phenotype,” Bioessays 13:337-42, 1991; Weiss, “Metastatic inefficiency,” In Liver Metastasis, L. Weiss, and H. Gilbert, eds. (Boston, Hall), pp. 126-157, 1985).
  • micrometases was 50% versus 91% for those without micrometases. This means that the stage II patients with micrometastases behaved clinically more like TMN stage III patients with regards to survival. Thus, a greater proportion of newly diagnosed colorectal cancers already have metastases than previously thought. Indeed, breast cancer cells in the blood of patients with advanced breast cancer were found as frequently as 1 per 1000
  • BAY 11-7085 (Biomol), anisomycin, ciglitazone (Cayman), pioglitazone (Cayman), troglitazone (Cayman), rosiglitazone (Cayman), GW9662 (Cayman), MC-555 (Cayman) were solubilized in DMSO.
  • Cleaved PARP pAb New England Biolabs
  • FLIP Sura Cruz
  • actin mAb (Lab Vision) were used for the Western blots.
  • D-Luciferin was purchased from Xenogen. Ketamine was purchased from the University of Utah Pharmacy.
  • BxPc-3, Su.86.86, and PL-45 pancreatic and HT-29 colon cancer cell lines were purchased from ATCC and cultured in DMEM supplemented with 10% fetal bovine serum, glutamine, penicillin, and streptomycin. All cells were cultured at 37° C in a humidified incubator in a 5% C0 2 atmosphere.
  • the cancer cell lines were stably transfected with pLuc-puro, a constitutive luciferase reporter (a kind gift from Dr. Stephen Lessnick, University of Utah) and selected in puromycin-containing media.
  • Pancreatic cancer cell cultures (BxPc-3 and Su.86.86) and colon cancer (HT- 29) cells were transiently suspended and 105 cells were transferred to 96-well plates and treated in vitro with varying concentrations of BAY 11-7085, thiazolidinediones, or DMSO for eight hours. To gently remove nonadherent cells, the wells were carefully washed from the wells twice with 100 ⁇ , ⁇ of blocking buffer (1% BSA in PBS) and twice again with ice-cold PBS.
  • blocking buffer 1% BSA in PBS
  • Pancreatic cancer (BxPc-3, Su.86.86, and PL-45) and colon cancer (HT-29) cell cultures were transiently suspended and treated in vitro with varying concentrations of BAY 11-7085 or rosiglitazone for 4 hours. Nuclear lysates were generated, normalized for total protein concentration, and used to determine NF- ⁇ activity in an ELISA kit (Panomics) per the manufacturer's instructions.
  • Apoptosis assay [00209] BxPc-3, PL-45, Su.86.86 and HT-29 cells were transiently suspended and treated in vitro with varying concentrations of BAY 11-7085 or DMSO for four to eight hours. Adherent and nonadherent treated cells were collected, lysates prepared, and protein concentrations normalized prior to Western Blot analysis using the Pierce BCA protein assay (Pierce, Rockford, 111.). The degree of apoptosis was determined by western blotting for cleaved PARP.
  • mice were randomized to intraperitoneal (IP) treatment with either BAY 11- 7085 (5 mg/kg) or an equal volume of vehicle (DMSO) 24 hours prior to IP injection of 106 suspended Su.86.86, BxPc-3, or HT-29 cells.
  • IP intraperitoneal
  • DMSO vehicle
  • the mice continued to receive DMSO or BAY 11-7085 (5 mg/kg) IP every 3 days for 9 days, or, DMSO and rosiglitazone (5 mg/kg) IP every day for 9 days.
  • mice were euthanized by an overdose of inhaled enflurane followed by cervical dislocation. The abdominal cavity was exposed surgically and the intraabdominal tumor implants imaged with the bioluminescent imaging system. The tumor implants for each mouse was verified with a dissecting zoom microscope.
  • FIG. 10A Treatment of pancreatic and colon cancer cells with 10-20 ⁇ BAY 11-7085 during cell readhesion increased apoptosis (cleaved PARP levels) (FIG. 10A). Since the induction of apoptosis of cancer cells during cell readhesion results in cell detachment (not shown), the number of readherent cells that survived treatment (FIG. 10B) was inversely proportional to the degree of apoptosis.
  • Intra-abdominal cancer cells can seed the peritoneal cavity and do so by adhering to the peritoneum, a tough connective tissue lining the abdominal cavity and organs.
  • pancreatic cancer cells Twenty- four hours later, 10 6 transiently suspended Su.86 or BxPc- 3 pancreatic cancer cells were delivered into the intra-abdominal cavity by intraperitoneal (IP) injection.
  • IP intraperitoneal
  • the mice continued to receive BAY 11-7085 or vehicle every 3 days for 9 days (4 doses total).
  • the cell lines used were stably transfected with a firefly luciferase construct in order to allow visualization of small tumor implants in anesthetized mice using a bioluminescent imager. Small immoveable intra-abominal tumor implants were readily visualized as early as 3-4 days following intraperitoneal delivery of the cell lines (not shown).
  • the mice were injected with luciferin and then euthanized.
  • FIGs. 11 A-D The abdominal cavities of the mice were rapidly exposed and the tumor implants visualized and imaged with the bioluminescent imager (FIGs. 11 A-D). Bioluminescent detection during necropsies aided the detection of tiny peritoneal tumor implants (2-3 mm) that were only otherwise visible with a zoom dissecting microscope. Tumor implants were observed on the parietal and visceral peritoneum with a predilection for the surface of the liver compared with the intestines or spleen (FIGs. 11 A-B). There was no evidence of metastases within the liver, lungs, or spleen of the mice (not shown) indicating that hematogenous spread had not occurred.
  • BAY 11-7085 or anisomycin a known activator of JNK, readily induced JNK activity (phosphorylation of c-JUN) in colon and pancreatic cancer cell lines (FIG. 12A). Furthermore, the drug-induced activation of TNK was inhibited by increasing
  • pancreatic and colon cancer cell lines were treated with anisomycin for 1.5 and 4 hours.
  • Anisomycin caused a decrease in FLIP expression in pancreatic and colon cell lines tested (FIG. 14A).
  • anisomycin induced apoptosis in colon and pancreatic cancer cell lines during cell readhesion (FIG. 14B).
  • Anisomycin increased the pro-apoptotic activity of BAY 1 1-7085 on readhering colon and pancreatic cancer cells (FIG. 14C).
  • HT-29 cells were allowed to readhere in the presence of vehicle (DMSO) or increasing concentrations of rosiglitazone.
  • DMSO vehicle
  • Transiently suspended colon and pancreatic cancer cells that were allowed to readhere to plates for 3 hours showed a marked increase in NF- ⁇ activity (FIG. 15 A).
  • Rosiglitazone at 30 ⁇ significantly inhibited readhesion-induced NF- ⁇ activation within 3 hours (FIG. 15 A) albeit to a lesser degree than BAY 11-7085 (FIG. 9).
  • HT-29 cells that were allowed to readhere in the presence of 30 ⁇ rosiglitazone, ciglitazone, or troglitazone for 8 hours exhibited increased apoptosis, demonstrated by increased cleaved PARP levels (FIG. 15B).
  • rosiglitazone decreased the survival of readhering BxPc3 and HT-29 cells in a concentration dependent fashion
  • Su.86 cells were resistant to rosiglitazone (FIGs. 15D-G).
  • the ability of rosiglitazone to cause downregulation of FLIP correlated with its ability to diminish the survival of readhering colon and pancreatic cancer cell lines.
  • Ciglitazone decreased the survival of both cell lines in a dose-dependent fashion (FIGs. 16A-B). However, the addition of 30 ⁇ GW 9662 did not significantly affect the efficacy of ciglitazone in these assays (FIGs. 16A-B). These data suggest that PPARy agonist activity is not required for the pro-apoptotic activity of the thiazolidinediones during cancer cell readhesion.
  • Peritoneal carcinomatosis is a fatal disorder with limited treatment options (Fujiwara, "Intraperitoneal chemotherapy and intraperitoneal washing cytology in management of ovarian cancer," Gan To Kagaku Ryoho 27 Suppl 2:354-8, 2000; Kanellos et al., “Incidence and prognostic value of positive peritoneal cytology in colorectal cancer,” Dis Colon Rectum 46(4):535-9, 2003; Nakatsuka et al., "Positive washing cytology in patients with pancreatic cancer indicates a contraindication of
  • transiently suspended colon cancer cells exhibit a strong and transient (3-8 hours) activation of NF- ⁇ during cell readhesion. It is described herein that now treatment of readherent pancreatic as well as colon cancer cells with the BAY 11-7085 and thiazolidinediones inhibit the adhesion-induced NF- ⁇ activity and cause apoptosis. The ability to induce apoptosis in readhering cancer cells is not universal to all NF-KB inhibitors that were tested.
  • infliximab an TNFr antagonistic antibody, and, bortezomib, a 26S proteasome inhibitor that indirectly inhibits NF- ⁇ by preventing the degradation of ⁇ (Voorhees and Orlowski, "The proteasome and proteasome inhibitors in cancer therapy," Annu Rev Pharmacol Toxicol 46: 189-213, 2006), failed to induce apoptosis of readhering HT-29 colon cancer cells.
  • JNK antagonizes NF-KB-mediated cell survival signaling by causing the down- regulation of FLIP, an important inhibitor of caspase 8 activity.
  • BAY 11-7085 and thiazolidinediones activate JNK and cause the rapid downregulation of FLIP protein in colon and pancreatic cancer cells. This is not likely due to their inhibitory activity of the compounds on NF- ⁇ , which regulates the transcription of CFLAR, the gene encoding FLIP.
  • the ability of anisomycin, a JNK agonist, to cause down-regulation of FLIP supports the role of JNK in mediating the down-regulation of FLIP. It is shown herein that the pro-apoptotic effect of BAY 11-7085 on readhering pancreatic cancer cells was enhanced by JNK activation by anisomycin in a dose-dependent fashion.
  • BAY 11-7085 caused the down-regulation of FLIP in readhering colon cancer cells, and, over-expression of FLIP in colon cancer cells caused greatly increased resistance to BAY 11-7085-induced apoptosis during cell readhesion.
  • BAY 11-7085 and to a lesser extent thiazolidinediones inhibited NF- ⁇ during colon and pancreatic cancer readhesion.
  • BAY 11-7085 was a more potent NF-KB inhibitor than rosiglitazone, thus potentially explaining the greater reduction in peritoneal implants generated with pancreatic cancer cell lines.
  • thiazolidinedione rosiglitazone as an inhibitor of peritoneal seeding is important since this compound is in clinical use and has a relatively low toxicity profile.
  • Thiazolidinediones have been shown to inhibit NF- ⁇ and this may or may not be related to their PPARy agonist activity (Su et al., "A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response," J Clin Invest 104(4):383-9, 1999).
  • rosiglitazone inhibited the intraabdominal seeding of colon and pancreatic cancer cell lines as well.
  • the proapoptotic activity of the thiazolidinediones may be independent of PPARy agonist activity as they showed pro-apoptotic activity in Su.86 and PL45 pancreatic cancer cells, neither of which expresses PPARy (Galli et al., "Antidiabetic thiazolidinediones inhibit invasiveness of pancreatic cancer cells via PPARgamma independent mechanisms," Gut 53(11): 1688-97, 2004).
  • BAY 11-7085 inhibits intraabdominal implantation of readhering pancreatic cancer cells in vivo.
  • Adult female athymic mice were randomized to receive either vehicle (DMSO) or drug (BAY 11-7085 or rosiglitazone) IP.
  • DMSO vehicle
  • BAY 11-7085 or rosiglitazone drug
  • the mice continued to receive the BAY 11-7085 every 3 days or rosiglitazone daily for 9 days after the cells were injected into the mice.
  • the mice received luciferin IP and sacrificed.
  • mice with peritoneal and/or hepatic tumor implants were compared using a test of proportions (*p ⁇ 0.05).
  • SU.86 pancreatic cancer cells were sorted for CD 133 expression cancer stem cells (CSC) by FACS and cocultured with monolayers of mesothelial cells, which line the peritoneum, and, express FASL and TRAIL. 4xl0 4 CD133+ SU.86 cells stably expressing firefly luciferase were injected intraperitoneally (IP) into nude mice. Mice received BAY 11-7085 or vehicle (DMSO) IP. Peritoneal implants were quantified in situ by bioluminescent scans.
  • CSC CD 133 expression cancer stem cells
  • BAY 11-7085 rapidly ( ⁇ 1 hr) induced ⁇ -dependent FLIP proteasomal degradation in SU.86 cells.
  • Pre-treatment of the DR-resistant CSC with 10 ⁇ BAY 11-7085 (BAY) or of SU.86 unsorted cells with FLIP siRNA restored DR- mediated apoptosis during cell adhesion to mesothelial cells.
  • Inhibition of JNK, TRAIL, or FAS reversed BAY-mediated cell apoptosis during adhesion to mesothelial cells.
  • BAY targeted FLIP for proteasomal degradation via INK activation.
  • BAY 11-7085 In vivo, BAY 11-7085 (5mg/kg IP TIW) greatly inhibited CSC peritoneal implants over 3 months (FIG. 14) without overt toxicity. Even a single dose of BAY 11-7085 administered IP together with unsorted SU.86 cells dramatically inhibited peritoneal implants after 1 month with 6 control mice having a two fold greater light output than the 6 BAY treated mice (p ⁇ 0.05). Bortezomib or gemcitabine failed to restore DR-mediated apoptosis or inhibit peritoneal implants.
  • Interleukin 15 protects against toxicity and potentiates antitumor activity of 5-fluorouracil alone and in combination with leucovorin in rats bearing colorectal cancer, Cancer Res 58, 1695-9.
  • COX-2 cyclooxygenase-2
  • PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs, Cell 99, 335-45.
  • Prostaglandin H synthase 2 is expressed abnormally in human colon cancer: evidence for a transcriptional effect, Proc Natl Acad Sci U S A 93, 4816-20.
  • Insulin-like growth factor-I protects colon cancer cells from death factor-induced apoptosis by potentiating tumor necrosis factor alpha- induced mitogen- activated protein kinase and nuclear factor kappaB signaling pathways, Cancer Res 60, 2007-17.
  • Rosette, C, and Karin, M. (1996). Ultraviolet light and osmotic stress: activation of the JNK cascade through multiple growth factor and cytokine receptors, Science 274, 1194-7.
  • Antisense oligonucleotides to the p65 subunit of NF- B block CD1 lb expression and alter adhesion properties of differentiated HL-60 granulocytes, Blood 82, 625-32.

Abstract

Dissclosed compositions and methods for inhibiting NF-κΒ and FLIP for the treatment of cellular proliferation and metastasis. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.

Description

INHIBITORS OF FLIP TO TREAT CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of United States Application No. 61/274,132, with a filing date of April 13, 2010, which is hereby incorporated herein by reference in its entirety.
BACKGROUND
[0002] Many adherent cells undergo apoptosis when converted to suspension culture or when detached from the underlying extracellular matrix (Strater et al., "Rapid onset of apoptosis in vitro follows disruption of βΐ-integrin/matrix interactions in human colonic crypt cells," Gastroenterology 110: 1776-84, 1996). This process has been termed anoikis and can be an important mechanism of tissue homeostasis (Frisch et al., "Disruption of epithelial cell-matrix interactions induces apoptosis," J Cell Biol 124:619-26, 1994). Cancer cells, on the other hand, have long been known for their ability to grow in the absence of adhesion, a characteristic known as anchorage-independent proliferation (Martin, "Normal Cells and Cancer Cells." In Molecular Oncology, J. M. Bishop, and R. A. Weinberg, eds., New York, Scientific American, pp. 13-40, 1996). This is of clinical relevance to metastasis, because cancer cells must transiently survive in the absence of adhesion as they travel to and migrate into distant tissues via the circulatory or lymphatic systems. Efforts to curb metastasis are critical since the presence of metastases is the single most important prognostic indicator for survival in cancer patients. Preventing metastasis of primary tumors is hampered by the apparent ease by which cancer cells gain access to the circulatory system either naturally or at the time of surgical resection (Hansen et al., "Blood irradiation for intraoperative autotransfusion in cancer surgery: demonstration of efficient elimination of contaminating tumor cells," Transfusion 39:608- 15, 1999; Hansen et al., "Tumor cells in blood shed from the surgical field," Arch Surg 130:387-93, 1995; Mehes et al, "Circulating breast cancer cells are frequently apoptotic," Am J Pathol 159: 17-20, 2001). Iatrogenic seeding of cancer cells is particularly worrisome as the primary modality of therapy for most resectable solid tumors is surgery. [0003] Intra-abdominal cancer seeding and peritoneal carcinomatosis complicate the medical and surgical therapy of abdominal cancers such as colorectal, gastric, pancreatic, and ovarian (De Vita et al, Cancer: Principles and Practice of Oncology. 5 ed.
Philadelphia: Lippincott Williams and Wilkins; 2001). Peritoneal seeding of tumor cells can occur in up to 35% of patients following curative resections of pancreatic head cancers (Johnstone et al., "Patterns of disease recurrence following definitive therapy of adenocarcinoma of the pancreas using surgery and adjuvant radiotherapy: correlations of a clinical trial," Int J Radiat Oncol Biol Phys 27(4):831-4, 1993) or through metastasis of intraabdominal cancers (Yu et al., "Prospective randomized trial of early postoperative intraperitoneal chemotherapy as an adjuvant to resectable gastric cancer," Ann Surg
228(3):347-54, 1998). The detection of intraperitoneal cancer cells during the resection of intraabdominal cancers portends a poor survival (Fujiwara, "Intraperitoneal chemotherapy and intraperitoneal washing cytology in management of ovarian cancer," Gan To Kagaku Ryoho 27 Suppl 2:354-8, 2000; Kanellos et al, "Incidence and prognostic value of positive peritoneal cytology in colorectal cancer," Dis Colon Rectum 46(4):535-9, 2003; Nakatsuka et al, "Positive washing cytology in patients with pancreatic cancer indicates a contraindication of pancreatectomy," Int J Surg Investig 1 (4) : 311-7, 1999; Santala et al, "Peritoneal cytology and preoperative serum CA 125 level are important prognostic indicators of overall survival in advanced endometrial cancer," Anticancer Res
23(3C):3097-103, 2003; Terauchi et al, "Combination chemotherapy with paclitaxel and intraperitoneal cisplatin for ovarian cancer with disseminated lesions in the peritoneum and the diaphragm," Int J Clin Oncol 7(6):356-60, 2002; Yachida et al, "Implications of peritoneal washing cytology in patients with potentially resectable pancreatic cancer," Br JSwrg 89(5):573-8, 2002). [0004] The factors involved in tumor implantation during intraabdominal spread are not well understood. However, it has been shown that during readhesion of colon cancer cells, NF-KB activity is transiently and strongly increased (Scaife et al, "Nuclear factor kappaB inhibitors induce adhesion-dependent colon cancer apoptosis: implications for metastasis," Cancer Res 62(23):6870-8, 2002). NF-κΒ is a critical mediator of cancer cell survival and is commonly over-expressed in malignant cells making it a selective target for cancer therapy (Lind et al, "Nuclear factor-kappa B is upregulated in colorectal cancer," Surgery 30(2):363-9, 2001; Sovak et al, "Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer," J Clin Invest 100(12):2952-60, 1997; Mori et al, "Constitutive activation of NF-kappaB in primary adult T-cell leukemia cells," Blood 93(7):2360-8, 1999; Wang et al, "The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells," Clin Cancer Res 5(1): 119-27, 1999; Nakshatri et al, "Constitutive activation of NF-kappaB during progression of breast cancer to hormone -independent growth," Mol Cell Biol 17(7):3629- 39, 1997).
[0005] Disclosed are compositions and methods related to inhibiting the affect of NF- KB on cancer, through inhibition of metastasis, readhesion, and cancer cell proliferation. SUMMARY
[0006] In accordance with the purposes of the disclosed materials, compounds, compositions, articles, and methods, as embodied and broadly described herein, the disclosed subject matter, in one aspect, relates to compositions and methods for preparing and using such compositions. In a further aspect, the disclosed subject matter relates to methods of using small molecules that inhibit NF-κΒ and/or FLIP to treat cancer and, more specifically, prevent peritoneal seeding of cancer cells and cancer stem cells.
[0007] Additional advantages will be set forth in part in the description that follows, and in part will be obvious from the description, or may be learned by practice of the aspects described below. The advantages described below will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive.
BRIEF DESCRIPTION OF THE DRAWINGS
[0008] The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate several embodiments of the invention and together with the description, serve to explain the principles of the invention.
[0009] FIG. 1 is a group of graphs that shows the effect of NF-κΒ inhibitors on colon cancer cell proliferation. DLD-1, HCT-116, and HT-29 were cultured on 96-well plates in the presence or absence of BAY 11-7082 (FIG. 1A) or BAY 11-7085 (FIG. IB) for 8 days. The cells were fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm. Legend: control (DMSO) squares, BAY 0.1 μΜ diamonds, BAY 1.0 μΜ triangles, BAY 10.0 μΜ ovals. The data points represent the mean values of experiments performed in triplicate and the error bars the standard error of the means. FIG. 1C shows electrophoretic mobility shift assays showing inhibition of NF- KB binding to an NF-κΒ DNA consensus oligonucleotide in nuclear lysates from HT-29 cells treated with BAY 11-7082 or BAY 11-7085 for 3 (left panel) or 6 (right panel) hours. The FIG shows the typical results of four experiments. The densitometry data of the gels are shown in graphic form at the bottom of the FIG. Excess cold competitor DNA oligonucleotide was used to demonstrate the specificity of the protein-DNA binding.
[0010] FIG. 2A is a graph that shows NF-κΒ inhibitors, BAY 11 -7082 and BAY 11 - 7085, decrease anchorage-independent proliferation of colon cancer cells. DLD-1, HCT- 116, and HT-29 cells were cultured for 6 days in soft agar in the presence of DMSO (control), 10 μΜ BAY 11-7082, or 10 μΜ BAY 11-7085. The data points represent the means of colony counts per field (2 OX) performed in 5 different regions of the culture dishes. The data shown represent a typical result of two separate experiments. FIG. 2B and FIG. 2C are graphs that show athymic mice received subcutaneous injections of HT- 29 (FIG. B) or HCT-116 (FIG. C) cells, followed by 5 mg/mL of BAY 11-7085 (dark bars) or DMSO (white bars) intraperitoneally twice weekly. The bars represent the 25- 75% ranges of the tumor volumes and the horizontal lines in the bars are the medians.
Using the Kruskal-Wallace statistical test to compare the median tumor volumes between BAY 11-7085 and DMSO yielded a p-value of 0.005 for HT-29 cells but >0.05 for HCT- 116 cells.
[0011] FIG. 3 is a group of graphs that show NF-κΒ inhibitors cause apoptosis of colon cancer cells. FIG. 3A is a graph that shows adherent and HT-29 cells were treated with DMSO alone (control), 10 and 20 μΜ BAY 11-7085, or 20 μΜ MG-132 for 24 hours. The data points represent the average percent of annexin V-positive, propidium iodide-negative cells (of the total cells counted) from triplicate experiments. The error bars represent the standard error of the means. FIG. 3B is a graph that shows adherent HT-29 cells were treated with BAY 11-7085 at various concentrations for 24 hours after which the nonadherent and adherent cells were collected separately. The results shown are a graphic representation of individual flow cytometry results expressed as the percentage of cells that were annexin V-positive of those that excluded propidium iodide. FIG. 3C shows adherent HCT-116 cells were treated with 20 μΜ MG-132 for 24 hours after which the cells were collected. An immunoblot of the lysates for cleaved PARP was performed. Each lane represents equal total protein concentrations. FIG. 3D is a graph that shows confluent monolayers of HT-29 cells were treated with various concentrations of BAY 11-7085 for 3 hours after which they were dispersed with trypsin-PBS. The cells were allowed to adhere to plastic dishes for 1 hour after which they were washed gently in PBS and stained with crystal violet. The crystal violet stained cells were solubilized in deoxycholate and the absorbance detected in a spectrophotometer at 590 nm. The adherent cells were expressed as a fraction of the controls. Each data point represents the average of triplicate experiments and the error bars the standard errors of the means.
[0012] FIG. 4A is a graph that shows transient suspension of colon cancer cells greatly increases their susceptibility to apoptosis in the presence of BAY 11-7085. HT-29 cells that were either transiently suspended with trypsin-EDTA or adherent for three days, were treated at time zero with DMSO or 10- 100 μΜ BAY 11-7085. The percent of apoptotic cells was determined using the annexin V flow cytometry assay and graphed versus the dose of BAY 11-7085. FIG. 4B are photographs showing DLD-1, FIG. 4C are photographs showing HCT-116, and FIG. 4D are photographs showing HT-29 cells that were transiently suspended in trypsin-EDTA and allowed to readhere to glass coverslips for 1-24 hours. The cells were fixed and immunostained with an NF-κΒ p65 subunit monoclonal antibody. Note the strong nuclear staining at 1 hour versus the markedly reduced nuclear staining of the cells.
[0013] FIG. 5A is a graph that shows NF-κΒ binding activity assay (TransAM) of DLD-1 , HCT-116, and HT-29 cells that were transiently suspended by scraping or with trypsin-EDTA. The transiently suspended cells were lysed and allowed to bind to DNA oligonucleotides, containing the NF-κΒ consensus binding site, that were immobilized to 96-well plates. NF-κΒ protein bound to the oligonucleotides was detected by an antibody to the p65 subunit that only recognized p65 that is bound to DNA. The positive control was HeLa cells that were stimulated with TNFa. A control was performed using excess free NF-κΒ oligonucleotides (Competitor). FIG. 5B is a graph that shows HT-29 cells were transiently suspended and allowed to readhere in the presence or absence of BAY 11-7085 for three hours after which they were lysed. NF-κΒ binding activity was determined by the TransAM assay as in FIG. 5A. FIG. 5C is a graph that shows transiently suspended DLD-1, HCT-116, and HT-29 cells were allowed to readhere in the presence of DMSO or BAY 11-7085 for 8 hours after which the percent of apoptotic cells was determined by the flow cytometric annexin V assay. The data shown represent typical results of three separate experiments. FIG. 5D shows 293-COX-2 cells were incubated in the presence or absence of 1 μg/mL of ponasterone for 48 hours after which they were lysed. An immunoblot for COX-2 was performed. FIG. 5E is a graph that shows 293-COX-2 cells were incubated in the presence or absence of ponasterone for 48 hours followed by 8 hours of exposure to DMSO or BAY 11-7085. Note that induction of COX-2 resulted in an increased susceptibility to BAY 11-7085-induced apoptosis.
[0014] FIG. 6 is a group of photographs showing an intraabdominal seeding model. FIGs. 6A, B, C show tumor implants of the liver of three athymic mice 21 days after receiving intraperitoneal injections of HT-29 cells. The mice were pretreated and treated for a total of 21 days with DMSO twice weekly. FIG. 6D shows intestinal tumor implants (arrows) of an athymic mouse 21 days after intraperitoneal injections of HT-29 cells. The mouse was pretreated and treated for a total of 21 days with DMSO twice weekly. FIG. 6E is a micrograph showing tumor invasion through the liver capsule. This section was obtained from the tumor implant from the mouse in FIG. 6A. FIG. 6F shows a peritoneal tumor implant taken from the bowel wall of the mouse in FIG. 6A. FIG. 6G, (liver), FIG. 6H, (abdominal wall), and FIG. 61, (intestinal) show tumor implants in three athymic mice 21 days after receiving intraabdominal injections of HCT-116 cells. All of the mice were treated with 5 mg/kg of BAY 11-7085 twice weekly.
[0015] FIG. 7A is a graph that shows DLD- 1 HCT- 116, and HT-29 cells were cultured in 96-well plates for 10 days in the presence or absence of 10 or 50 μg/mL of cA2. At various time -points the cells were the cells were fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm. The data points represent the means of triplicate experiments and the error bars the standard errors of the means. FIG. 7B is a graph that shows adherent HT-29 cells were pretreated with 50 μg/mL of cA2 for 48 hours followed by the addition of DMSO or 20 μΜ BAY 11-7085 for another 24 hours. The percent of apoptotic cells was then determined by the annexin V assay. Similar results were obtained for DLD-1 cells. The results shown are typical of two separate experiments. FIG. 7C shows adherent or transiently suspended HT-29 cells were treated with 20 μΜ BAY 11-7085 for 8 hours after which they were lysed.
Immunoblots were performed for c-IAP-2, TRAF-1, TRAF-2, and FLIP. All lanes contain equal total protein. FIG. 7D shows a FLIP immunoblot of DLD-1, HCT-116, and HT-29 lysates. [0016] FIG. 8 is a graph that shows HT-29 cells that were transiently suspended and allowed to readhere in the presence or absence of various concentrations of the NF-KB inhibitors, PDTC, BAY 11-7085, and MG-132 for 4 hours. The percentage of apoptotic cells was determined using the annexin V assay. The data points represent the averages of three experiments; bars, +-.SE. E. DLD-1 and F. HT-29 cells were transduced with an adenovirus containing the ΙκΒ super-repressor construct. After 3 days, the cells were transiently suspended and allowed to readhere in the presence of subapoptotic
concentrations of BAY 11-7085 or DMSO (vehicle) for 8 hours. The data are expressed as fractions of cells surviving compared with the controls (no adenovirus) and were quantitatively determined by staining the remaining adherent cells with crystal violet. [0017] FIGs. 9A-C are graphs that shown BAY 11-7085 inhibits NF-κΒ activation during pancreatic and colon cancer cell readhesion in vitro. Transiently suspended pancreatic (Su.86, PL45, BxPC-3) and colon (HT-29) cancer cells (105 per well (96-well plate)) were allowed to readhere in the presence of vehicle or various concentrations of BAY 11-7085 for 4 hours. The levels of the p65 subunit of NF-κΒ in nuclear lysates were performed with an ELISA-based kit. The data points represent the average of triplicate results and the error bars the standard error of the means. The results for each
concentration of BAY 11-7085 were normalized to the levels obtained with vehicle alone. The experiments were performed 2-3 times.
[0018] FIG. 10 is a graph showing that BAY 11-7085 induces apoptosis of pancreatic and colon cancer cells during readhesion. Pancreatic (Su.86, PL-45) and colon cancer cells (HT-29) were transiently suspended and allowed to readhere in the presence or absence of vehicle or various concentrations of BAY 11-7085 for 8 hours. The floating and adherent cells were collected and lysed. Western blots for cleaved PARP were performed to determine if the cells were undergoing apoptosis (FIG. 10A). Transiently suspended pancreatic cancer cells (105 per well) were allowed to readhere in the presence of vehicle or various concentrations of BAY 11-7085 for 24 hours. Nonadherent apoptotic cells were washed away and the remaining adherent cells stained with crystal violet and then dried. The crystal violet stained cells were lysed and the absorbance read in a plate spectrophotometer (FIG. 10B). Each data point represents the average of triplicate results and error bars, the standard error of the means. The results are expressed as a percentage of cells surviving compared with vehicle alone.
[0019] FIG. 11 are photographs showing bioluminescent detection of pancreatic cancer cells stably transfected with firefly luciferase. FIGs. 11 A-B show an image of 5 athymic mice 9 days after IP injection of 106 Su.86 pancreatic cancer cells. The mice received IP firefly D-luciferin and then euthanized in order to visualize the intraabdominal tumor implants. The intraabdominal tumors were visualized before sacrificing the mice (A) or after euthanasia and exposure of the intraabdominal cavity (B). FIG. 11C is a dissecting microscopic image of a barely visible tumor implant on the edge of the liver detected by bioluminescent imaging (arrow in FIG. 11 A). FIG. 1 ID is a magnified view of the liver implant. [0020] FIG. 12 shows thiazolidinediones inhibit NF-KB activation and induce apoptosis of pancreatic and colon cancer cells during cell readhesion. FIG. 12A shows transiently suspended HT-29 cells were allowed to readhere in the presence of vehicle (DMSO) or various concentrations of rosiglitazone for 3 hours (dark bars). Untreated adherent monolayers (Control) and Jurkat cells treated with TNFa (Jurkat Control) served as controls. The cells were collected and analyzed for NF-κΒ activation in an ELISA (see Methods). FIG. 12B shows transiently suspended HT-29 cancer cells were allowed to readhere in the presence of DMSO, ciglitazone, rosiglitazone, or troglitazone for 8 hours. The floating and readherent cells were collected and lysed. Aliquots of the cell lysates were used to generate cleaved PARP immunoblots. All lanes contain equal total protein concentrations and the actin blot served as a loading control. The blots shown are typical of duplicate experiments. FIGs. 12C-E show transiently suspended BxPC-3, Su.86, and HT-29 cancer cells were allowed to readhere in the presence of vehicle (DMSO) or various concentrations of ciglitazone, MCC-555, pioglitazone, rosiglitazone, and troglitazone for 8 hours. The floating cells were washed away and the adherent cells were then fixed and stained with crystal violet. The cells were lysed in deoxycholate and the intensity of the staining quantified by spectrophotometry. The results are normalized to the controls. Each data point represents the mean of triplicate experiments and the error bars, the standard error of the mean.
[0021] FIG. 13 shows thiazolidinediones induce apoptosis of pancreatic and colon cancer cells during cell readhesion in a PPARy -independent fashion. FIGs. 13A-B show transiently suspended Su.86 (PPARy -negative) and HT-29 (PPARy -positive) cancer cells were allowed to readhere in the presence of various concentrations of ciglitazone for 8 hours. The cells were also incubated with or without the irreversible PPARy inhibitor GW9662 during readhesion. The floating cells were washed away and the adherent cells were then fixed and stained with crystal violet. The cells were lysed in deoxycholate and the intensity of the staining quantified by spectrophotometry. The results are normalized to the controls. Each data point represents the mean of triplicate experiments and the error bars, the standard error of the mean.
[0022] FIG. 14 is a grap of tumor light output versus days of treatment. BAY treated mice (n=20) showed a significantly lower tumor burden than control mice (n=23), indicating BAY 11-7085 is capable of preventing implantation of the highly tumorigenic cancer stem cells.
[0023] FIG. 15 (A-D) shows expression of RANK and its ligand, RANKL, for most of the cancer cell lines.
[0024] FIG. 16 is a schematic illustration of a cancer cell. [0025] FIG. 17 shows the results from a Cancer Stem Cell Longitudinal treatment Study in Nude Mice for BAY 11-7085 Su.86.86 versus Control.
DETAILED DESCRIPTION
[0026] The materials, compounds, compositions, articles, devices, and methods described herein may be understood more readily by reference to the following detailed description of specific aspects of the disclosed subject matter and the Examples included therein and to the FIGs.
[0027] Before the present materials, compounds, compositions, articles, devices, and methods are disclosed and described, it is to be understood that the aspects described below are not limited to specific synthetic methods or specific reagents, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting.
[0028] Also, throughout this specification, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which the disclosed matter pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon. General Definitions
[0029] In this specification and in the claims that follow, reference will be made to a number of terms, which shall be defined to have the following meanings:
[0030] Throughout the description and claims of this specification the word
"comprise" and other forms of the word, such as "comprising" and "comprises," means including but not limited to, and is not intended to exclude, for example, other additives, components, integers, or steps.
[0031] As used in the description and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a composition" includes mixtures of two or more such compositions, reference to "an agent" includes mixtures of two or more such agents, reference to "the component" includes mixtures of two or more such component, and the like.
[0032] Ranges can be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as "about" that particular value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also understood that when a value is disclosed that "less than or equal to" the value, "greater than or equal to the value" and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value "10" is disclosed, then "less than or equal to 10" as well as "greater than or equal to 10" is also disclosed. It is also understood that throughout the application data are provided in a number of different formats and that this data represent endpoints and starting points and ranges for any combination of the data points. For example, if a particular data point "10" and a particular data point "15" are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed. [0033] References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
[0034] A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included. [0035] "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
Chemical Definitions
[0036] As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds.
Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms "substitution" or "substituted with" include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
[0037] "A1," "A2," "A3," and "A4" are used herein as generic symbols to represent various specific substituents. These symbols can be any substituent, not limited to those disclosed herein, and when they are defined to be certain substituents in one instance, they can, in another instance, be defined as some other substituents.
[0038] The term "alkyl" as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like. The alkyl group can also be substituted or unsubstituted. The alkyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo- oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
[0039] Throughout the specification "alkyl" is generally used to refer to both unsubstituted alkyl groups and substituted alkyl groups; however, substituted alkyl groups are also specifically referred to herein by identifying the specific substituent(s) on the alkyl group. For example, the term "halogenated alkyl" specifically refers to an alkyl group that is substituted with one or more halide, e.g., fluorine, chlorine, bromine, or iodine. The term "alkoxyalkyl" specifically refers to an alkyl group that is substituted with one or more alkoxy groups, as described below. The term "alkylamino" specifically refers to an alkyl group that is substituted with one or more amino groups, as described below, and the like. When "alkyl" is used in one instance and a specific term such as "alkylalcohol" is used in another, it is not meant to imply that the term "alkyl" does not also refer to specific terms such as "alkylalcohol" and the like.
[0040] This practice is also used for other groups described herein. That is, while a term such as "cycloalkyl" refers to both unsubstituted and substituted cycloalkyl moieties, the substituted moieties can, in addition, be specifically identified herein; for example, a particular substituted cycloalkyl can be referred to as, e.g., an "alkylcycloalkyl."
Similarly, a substituted alkoxy can be specifically referred to as, e.g., a "halogenated alkoxy," a particular substituted alkenyl can be, e.g., an "alkenylalcohol," and the like. Again, the practice of using a general term, such as "cycloalkyl," and a specific term, such as "alkylcycloalkyl," is not meant to imply that the general term does not also include the specific term. [0041] The term "alkoxy" as used herein is an alkyl group bound through a single, terminal ether linkage; that is, an "alkoxy" group can be defined as— OA1 where A1 is alkyl as defined above.
[0042] The term alkoxylalkyl as used herein is an alkyl group that contains an alkoxy substituent and can be defined as— A^O-A2, where A1 and A2 are alkyl groups. [0043] The term "alkenyl" as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond. Asymmetric structures such as (A1A2)C=C(A3A4) are intended to include both the E and Z isomers. This may be presumed in structural formulae herein wherein an asymmetric alkene is present, or it may be explicitly indicated by the bond symbol C=C. The alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
[0044] The term "alkynyl" as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond. The alkynyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below. [0045] The term "aryl" as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like. The term "aryl" also includes "heteroaryl," which is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus. Likewise, the term "non- heteroaryl," which is also included in the term "aryl," defines a group that contains an aromatic group that does not contain a heteroatom. The aryl group can be substituted or unsubstituted. The aryl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo- oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein. The term "biaryl" is a specific type of aryl group and is included in the definition of aryl. Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl. [0046] The term "cycloalkyl" as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc. The term
"heterocycloalkyl" is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein. [0047] The term "cycloalkenyl" as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one double bound, i.e., C=C. Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like. The term "heterocycloalkenyl" is a type of cycloalkenyl group as defined above, and is included within the meaning of the term "cycloalkenyl," where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted. The cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
[0048] The term "cyclic group" is used herein to refer to either aryl groups, non-aryl groups (i.e., cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl groups), or both. Cyclic groups have one or more ring systems that can be substituted or unsubstituted. A cyclic group can contain one or more aryl groups, one or more non-aryl groups, or one or more aryl groups and one or more non-aryl groups.
[0049] The term "aldehyde" as used herein is represented by the formula— C(0)H. Throughout this specification "C(O)" is a short hand notation for C=0.
[0050] The terms "amine" or "amino" as used herein are represented by the formula NA 1 A2 A 3 , where A 1 , A2 , and A 3J can be, independently, hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
[0051] The term "carboxylic acid" as used herein is represented by the formula— C(0)OH. A "carboxylate" as used herein is represented by the formula— C(0)0". [0052] The term "ester" as used herein is represented by the formula— OC(0)A1 or
Figure imgf000016_0001
where A1 can be an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above. [0053] The term "ether" as used herein is represented by the formula A1OA2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above. [0054] The term "ketone" as used herein is represented by the formula Α^(0)Α2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
[0055] The term "halide" as used herein refers to the halogens fluorine, chlorine, bromine, and iodine.
[0056] The term "hydroxyl" as used herein is represented by the formula— OH.
[0057] The term "nitro" as used herein is represented by the formula— N02.
[0058] The term "silyl" as used herein is represented by the formula— SiA^A3, where A1, A2, and A3 can be, independently, hydrogen, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
[0059]
— S(0)2A1,
Figure imgf000017_0001
halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl,
heterocycloalkyl, or heterocycloalkenyl group described above. Throughout this specification "S(O)" is a short hand notation for S=0.
[0060] The term "sulfonyl" is used herein to refer to the sulfo-oxo group represented by the formula
Figure imgf000017_0002
where A1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
[0061] The term "sulfonylamino" or "sulfonamide" as used herein is represented by the formula— S(0)2NH-. [0062] The term "sulfone" as used herein is represented by the formula A1S(0)2A2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above. [0063] The term "sulfoxide" as used herein is represented by the formula A1S(0)A2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
[0064] The term "thiol" as used herein is represented by the formula— SH. [0065] "R1," "R2," "R3," "Rn," where n is an integer, as used herein can,
independently, possess one or more of the groups listed above. For example, if R1 is a straight chain alkyl group, one of the hydrogen atoms of the alkyl group can optionally be substituted with a hydroxyl group, an alkoxy group, an alkyl group, a halide, and the like. Depending upon the groups that are selected, a first group can be incorporated within second group or, alternatively, the first group can be pendant (i.e., attached) to the second group. For example, with the phrase "an alkyl group comprising an amino group," the amino group can be incorporated within the backbone of the alkyl group. Alternatively, the amino group can be attached to the backbone of the alkyl group. The nature of the group(s) that is (are) selected will determine if the first group is embedded or attached to the second group.
[0066] Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer and diastereomer, and a mixture of isomers, such as a racemic or scalemic mixture. [0067] Certain materials, compounds, compositions, and components disclosed herein can be obtained commercially or readily synthesized using techniques generally known to those of skill in the art. For example, the starting materials and reagents used in preparing the disclosed compounds and compositions are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Acros Organics (Morris Plains, N.J.), Fisher Scientific (Pittsburgh, Pa.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991); March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition); and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
[0068] Reference will now be made in detail to specific aspects of the disclosed materials, compounds, compositions, articles, and methods, examples of which are illustrated in the accompanying Examples and FIGs. Inhibitors
[0069] NF-KB is a heterodimeric transcription factor comprised of various protein subunits: p50/pl05, p65/RelA, p52/pl00, c-Rel, and RelB. NF-κΒ is localized to the cytoplasm by association with the ΙκΒ proteins, which inhibit translocation of NF-κΒ into the nucleus thus preventing its transcriptional activity (Beg et al, "ΙκΒ interacts with the nuclear localization sequences of the subunits of NF-κΒ: a mechanism for cytoplasmic retention," Genes Dev 6: 1899-913, 1992). Under the influence of cytokines, reactive oxygen species, growth factors, and other stimuli, ΙκΒ kinases (IKK) phosphorylate IKB proteins on two critical serine residues (DiDonato et al, "Mapping of the inducible IkappaB phosphorylation sites that signal its ubiquitination and degradation," Mol Cell Biol 16: 1295-304, 1996; Traenckner et al, "Phosphorylation of human ΙκΒ-α on serines 32 and 36 controls ΙκΒ-α proteolysis and NF-κΒ activation in response to diverse stimuli," Embo J 14:2876-83, 1995) This then targets the IKB proteins for ubiquitination and subsequent degradation by the proteasome, which results in free NF-κΒ that translocates into the nucleus and activates the transcription of various genes possessing κΒ consensus DNA binding sites in their promoters (Alkalay et al, "Stimulation-dependent ΙκΒ a- phosphorylation marks the NF-κΒ inhibitor for degradation via the ubiquitin-proteasome pathway," Proc Natl Acad Sci USA 92: 10599-603, 1995; Chen et al, "Signal-induced site- specific phosphorylation targets ΙκΒ-α to the ubiquitin-proteasome pathway," Genes Dev 9: 1586-97, 1995; Henkel et al, "Rapid proteolysis of I kappa B-a is necessary for activation of transcription factor NF-κΒ," Nature 365: 182-5, 1993). Many of the genes regulated by NF-κΒ encode proteins that promote inflammation, including COX-2, (Crofford et al, "Involvement of nuclear factor kappa B in the regulation of
cyclooxygenase-2 expression by interleukin-1 in rheumatoid synoviocytes," Arthritis Rheum 40:226-36, 1997; Newton et al, "Evidence for involvement of NF-κΒ in the transcriptional control of COX-2 gene expression by IL-Ι β," Biochem Biophys Res Commun 237:28-32, 1997; Newton et al., "Superinduction of COX-2 mRNA by cycloheximide and interleukin-1 β involves increased transcription and correlates with increased NF-κΒ and JNK activation," FEB S Lett 418:135-8, 1997; Schmedtje et al., "Hypoxia induces cyclooxygenase-2 via the NF-κΒ p65 transcription factor in human vascular endothelial cells," J Biol Chem 272:601, 1997) and angiogenesis factors such as VEGF (Huang et al., "Blockade of NF-κΒ activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis," Oncogene 20:4188-97, 2001). In addition, NF-κΒ mediates the transcription of several survival genes, for example c-Myc, Bcl2, p53, p21, c-FLIP, c-IAP-1, c-IAP-2, XIAP, IEX-1L, COX- 2, TRAF-1, and TRAF-2 (Kreuz et al, " NF-κΒ inducers upregulate cFLIP, a
cycloheximide-sensitive inhibitor of death receptor signaling," Mol Cell Biol 21 :3964-73, 2001; Schwartz et al., "The role of NF-κΒ / ΙκΒ proteins in cancer: implications for novel treatment strategies," Surgical Oncology 8:143-153, 1999; Stehlik et al, "Nuclear factor (NF)-kappaB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha- induced apoptosis," J Exp Med 188:211-6, 1998; Wang et al., "NF-κΒ antiapoptosis: induction of TRAF1 and TRAF2 and c-IAPl and c- IAP2 to suppress caspase-8 activation," Science 281 : 1680-3, 1998; Wu et al, "IEX-1L, an apoptosis inhibitor involved in NF-KB-mediated cell survival," Science 281 :998- 1001, 1998).
[0070] Disclosed herein are inhibitors that can inhibit anchorage-dependent as well as anchorage-independent proliferation, and tumorigenicity, and can cause apoptosis of cancer cells, including pancreatic and colon cancer cells as well as cancer stem cells {e.g., pancreatic cancer stem cells). The induction of apoptosis of cancer cells by NF-KB inhibition can occur in a cell adhesion-dependent fashion. Readhesion following transient suspension of the cell lines causes a large activation of NF-κΒ, which renders cells exquisitely sensitive to NF-κΒ inhibitor-induced apoptosis. Furthermore, pretreatment of athymic mice with an NF-κΒ inhibitor completely prevented liver metastasis following intraperitoneal delivery of a colon cancer cell line. Thus, colon and pancreatic cancer cells, as well as cancer cells related to colon and pancreatic cancer cells, utilize NF-κΒ for mitogenesis and as a major survival factor during readhesion.
[0071] As shown herein, treatment of readhering cancer cells, like colon and pancreatic cancer cells, including cancer stem cells, with an inhibitor as disclosed herein greatly induced apoptosis. In vivo studies in mice showed that BAY 11-7085, an NF-KB inhibitor, inhibited colon and pancreatic cancer cell implantation of the intraabdominal cavity in mice following intraperitoneal injection of colon cancer cells. Further, not all NF-KB inhibitors induce apoptosis of readhering colon and pancreatic cancer cells, suggesting additional mechanisms of action for inhibitors like BAY 11-7085. For example, BAY 11-7085 also activated JNK/SAPK activity, which was recently shown to mediate the rapid down-regulation of FLIP. FLIP /FLICE /CASH /FLAME /CASPER is encoded by CFLAR, one of a number of pro-survival genes regulated by NF-κΒ. FLIP is an important inhibitor of caspase 8 and thus death receptor-induced apoptosis. BAY 11- 7085 and anisomycin, a JNK activator, was shown to cause the rapid downregulation of FLIP. FLIP overexpression inhibited the pro-apoptotic activity of BAY 11-7085 during colon cancer cell readhesion. As shown herein, the disclosed inhibitors can also inhibit FLIP, thereby rendering cancer cells susceptible to death receptor mediated apoptosis.
[0072] The inhibitors disclosed herein can be any molecule that inhibits NF-κΒ and FLIP, and that activates JNK. It is understood that NF-κΒ function can be inhibited or altered in many ways by an inhibitor. For example, NF-κΒ function can be inhibited by an inhibitor directly interacting with NF-κΒ. Directly interacting with NF-κΒ means that the inhibitor touches or binds with NF-κΒ. An inhibitor can also indirectly inhibit NF-KB function. Indirectly inhibiting the function of NF-κΒ means that an inhibitor does not touch or bind NF-κΒ. A molecule would indirectly inhibit NF-κΒ function by, for example, reducing the expression or activation or nuclear transport of NF-KB.
[0073] Examples of indirect inhibitors are those that inhibit NF-κΒ transport into the nucleus. NF-κΒ remains cytoplasmic if it interacts with ΙκΒ. When ΙκΒ is
phosphorylated, it causes ΙκΒ to be ubiquinated, and degraded, which allows NF-κΒ to transport to the nucleus. The disclosed compositions can, in certain examples, inhibit NF- KB transport to the nucleus through interactions with ΙκΒ, which prevent ΙκΒ from interacting with NF-κΒ. Other examples of indirect inhibitors that inhibit NF-κΒ transport into the nucleus are inhibitors that inhibit ΙκΒ phosphorylation. Still other examples of indirect inhibitors are inhibitors that inhibit expression of NF-κΒ. Still other examples of indirect inhibitors are inhibitors that inhibit translation of NF-KB.
[0074] Typically the disclosed inhibitors can inhibit TNFa-induced NF-κΒ activation. TNFa typically causes an activation of NF-κΒ and the disclosed inhibitors can decrease this TNFa-induced activation. Many assays can be used to determine if inhibitors are decreasing TNFa-dependent activation.
[0075] Also, disclosed are inhibitors that can inhibit anchorage dependent and independent proliferation and tumorigenicity and can cause apoptosis. Still further, disclosed herein are inhibitors that inhibit induction of apoptosis by NF-κΒ inhibition, which occurs in a cell-adhesion dependent fashion. Inhibitors that affect cells where readhesion of cells causes a large activation of NF-κΒ that causes the cells to become sensitized to NF-κΒ inhibitor induced apoptosis are also disclosed.
[0076] Some specific examples of suitable inhibitors are BAY 11-7082 and BAY I l7085, which inhibit ΙκΒ phosphorylation and TNFa induced NF-κΒ activation. BAY 11- 7082 also inhibits growth of DLD-1 and HCT-116. Cells that contain APC mutations are susceptible to BAY 11-7085 and related molecules. For example, cells obtained from cell lines DLD-2 and HT-29 cells are susceptible to BAY 11-7085. DLD-1 and HT-29 contain APC mutations. HCT-116 cells have activating mutations on B-catenin, which is normally regulated by the APC product. Also HCT-116 cells do not express COX-2, which is commonly over-expressed in colorectal cancers. Anchorage-independent inhibition of proliferation can be inhibited by both BAY 11-7082 and 7085 in DLD-1 and HT-29 both BAY 11-7082 and BAY 11-7085 reduce tumor volumes in vivo in athymic mice injected with HT-29 tumor cells. MC-132 and PDTC cause an increase in apoptosis in adhered cells and BAY- 11-7085 and BAY- 11-7082 cause apoptosis in cells even when not adhered.
Methods of using the compositions
[0077] The disclosed compositions can be used in a variety of ways as research tools. For example, the disclosed compositions, such as BAY- 11-7085 and BAY- 11-7082, PDTC and MC-132 can be used as reagents and standards in cellular proliferation assays and as inhibitors for assays related to cancer.
[0078] The compositions can be used for example as competitive inhibitors in combinatorial chemistry protocols or other screening protocols to isolate molecules that posses desired functional properties related to inhibition of cancer or metastasis of cell systems disclosed herein.
Methods of treating cancer
[0079] The disclosed inhibitors can be given to a subject. Any subject in need of the NF-KB /FLIP inhibitors as disclosed herein can be given the NF-KB/FLIP inhibitors. Any subject in need of the JNK activators can be given the INK activators. The subject can, for example, be a mammal, such as a mouse, rat, rabbit, hamster, dog, cat, pig, cow, sheep, goat, horse, or primate, such as monkey, gorilla, orangutan, chimpanzee, or human.
[0080] The disclosed NF-KB/FLIP inhibitors and JNK activators can be used for inhibiting cancer cell proliferation. Inhibiting cancer cell proliferation means reducing or preventing cancer cell growth. Inhibitors can be determined by using a cancer cell assay. For example, either a DLD-1, HCT-116, HT-29, Su.86, or a BxPC-3 cell line cell line can be cultured on 96-well plates in the presence or absence of the inhibitor for 8 days. The cells can be fixed, stained with crystal violet, solubilized in deoxycholate, and read in a spectrophotometer at 590 nm. In certain examples, the inhibitors are those that will inhibit 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%o, 90%), or 95%) of the cell growth relative to a control as determined by
spectrophotometry.
[0081] In certain examples the disclosed inhibitors can also inhibit anchorage- dependent proliferation of cancer cells, such as colon cancer cells or pancreatic cancer cells, including cancer stem cells. Inhibitors can be assayed using a soft agar colony formation assay. For example, DLD-1, HCT-116, HT-29, Su.86, and a BxPC-3 cells can be cultured for 6 days in soft agar in the presence of DMSO (control) or inhibitor. The number of colony counts can then be compared by, for example, taking a percentage of the cells formed relative to a control. In certain examples, the inhibitors are those that will inhibit 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%), or 95% of the cell growth relative to a control as determined by counting colonies formed.
[0082] Disclosed herein NF-KB/FLIP inhibitors can be used for promoting cancer cell apoptosis. Promoting cancer cell apoptosis means causing the cell to die. An apoptosis assay can be used to determine if the inhibitors promote cancer cell apoptosis. For example, an apoptosis assay as described in Example 1 can be used. In certain examples, the percent of apoptosis was determined as the percent of annexin V-positive, propidium iodide-negative cells of the total cells counted in an Apoptosis assay, such as that described in Example 1. The inhibitor can cause at least about 10%>, 15%, 20%>, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the cells to be apoptotic.
[0083] In certain examples, the apoptotic effect of the disclosed inhibitors is enhanced in cells that are transiently suspended, such as metastatic cells circulating through the blood stream, prior to readhesion. Inhibitors that have this property can be determined by assaying their apoptotic effect as, for example described herein, in a transient suspension assay, as described in Examples 1 and 2. For example, cells can be put into suspension by scraping and then allowed to readhere. Inhibitors that cause an increase in apoptosis upon readhering to the substrate are disclosed. An increase upon readhering to the substrate can be determined by, for example, looking at multiples of cells that were apoptotic upon readhering to the substrate compared with cells that were apoptotic upon remaining in suspension. Disclosed are inhibitors wherein the inhibitor causes at least about 1.5, 2, 3, 4, 5, 6, 7, 10, 15, 20, 30, 50, or 100 fold more apoptosis of cells that readhere then apoptosis of cells that remain in suspension (see for example FIG. 3). In certain examples, the disclosed inhibitor can cause at least about 17% of the cells to be apoptotic. [0084] Disclosed herein are NF-KB/FLIP inhibitors and JNK activators that can be used for inhibiting readhesion of cancer cells to a surface. Inhibiting readhesion of cancer cells to a surface means decreasing the number of cells capable of readhering to a surface after being transiently suspended as discussed herein.
[0085] The NF- κΒ/FLIP inhibitors and JNK activators disclosed herein can also be used for inhibiting metastasis of cancer cells. Inhibiting metastasis of cancer cells means decreasing or lowering the amount of metastatic tumors that arise in an organism. For example, disclosed are inhibitors that inhibit metastasis in an in vivo assay. One way of performing an in vivo assay to determine if an inhibitor inhibits metastasis is to inject a cancer cell line, such as HT-29, into the abdominal cavity of an athymic mouse. Mice are pretreated with the inhibitor or a control intraperotneally, for example. The mouse can then be treated regularly, for example, twice weekly with vehicle or BAY 11-7085 for a period of time, for example, 21 days. The mouse can then be sacrificed and assayed for metastatic tumor formation. Disclosed are compositions which inhibit metastatic tumor formation in this type of assay disclosed herein, as well as compositions that reduce metastatic tumor formation by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% relative to a control compound. For example, NF-κΒ inhibitors can inhibit intraabdominal metastasis arising from, for example, colon or rectal cancers. NF- κΒ/FLIP inhibitors can also inhibit hepatic, parietal or peritoneal metastasis arising from, for example, colon or rectal cancers. In certain example, the disclosed compositions can be used to treat, prevent, and/or inhibit metastasis. For example, hemogenous or lymphatic metastis.
[0086] The disclosed NF- κΒ/FLIP inhibitors and TNK activators can be used for inhibiting tumorigenesis. Inhibiting tumorigenesis means decreasing or lowering the amount of tumors present in an organism. For example, disclosed are inhibitors that inhibit tumorigenesis in an in vivo assay. One way of performing an in vivo assay to determine if an inhibitor inhibits tumorigenesis is to inject a cancer cell line
subcutaneously, such as HT-29, into an athymic mouse, such as a female mouse. The mouse can then be treated regularly, for example, twice weekly with vehicle or BAY 11- 7085 for a period of time, for example, 21 days or 28 days. The mouse can then be sacrificed and assayed for tumor formation and size. Disclosed are compositions which inhibit tumorigenesis in this type of assay disclosed herein, as well as compositions that reduce tumorigenesis by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% relative to a control compound.
[0087] The disclosed NF-KB/FLIP inhibitors and JNK activators can be administered to cells which utilize NF-κΒ, FLIP and/or JNK for mitogenesis. For example, NF-KB inhibitors can be administered to cells which utilize NF-κΒ for readhesion. A cell can be determined to utilize NF-κΒ for readhesion if in a transient suspension assay, upon readhesion NF-κΒ is activated as determined by any assay that looks at NF-κΒ activation. For example, NF-κΒ binding assays to the consensus sequence can be used, such as TRANS-AM™ assay the discussed in Examples 1 and 2. If the NF-κΒ activation is greater upon readhesion then in suspension, the cells can be considered to utilize NF-KB for readhesion. [0088] The disclosed NF-KB/FLIP inhibitors and JNK activators can also be administered to cells to induce the apoptosis of the cells in a TNFa independent manner. TNFa can activate apoptosis, but the NF-κΒ inhibitors can be administered to cause apoptosis in a TNFa independent manner. The disclosed inhibitors can promote apoptosis of cancer cells, such as colon cancer cells, without TNFa. An inhibitor or cell can be shown to be apoptotic without needing the inhibitor by using TNFa inhibiting antibody, such as cA2, and seeing that apoptosis is still caused by the inhibitor even in the presence of the TNFa inhibiting antibody, such as cA2.
[0089] When the compositions are administered, they typically cause a decrease in the expression of anti-apoptotic proteins. Expression of the anti-apoptotic proteins can be determined by any means for determining expression. For example, standard
biotechnology methods such as PCR or Northern blots can be used to determine the expression levels of anti-apoptotic genes. A decrease can be determined by assaying the expression levels of a desired anti-apoptotic gene in the presence of a potential inhibitor and comparing this level of expression to the level of expression in the absence of the inhibitor. Disclosed are inhibitors, which decrease the expression of the anti-apoptotic genes in such an assay.
[0090] As discussed herein NF-KB/FLIP inhibitors and JNK activators can, for example, be used to reduce the proliferation of cancer cells (including cancer stem cells), as well as to cause the apoptosis of cancer (stem) cells or inhibit the readhesion of cancer (stem) cells or inhibit the metastasis of cancer cells. The inhibitors can, for example, can be administered to any cancer cell that uses NF-κΒ to survive or metastize or adhere or which activates NF-κΒ during its life cycle.
[0091] The inhibitors can be administered to cancer cells, e.g., pancreatic cancer stem cells, to restore the death receptor mediated apoptosis. [0092] NF-KB/FLIP inhibitors and JNK activators can be administered to cancer cells that have a mutation in the adenomatous polyposis coli (APC) gene. APC has been shown to be a tumor suppressor gene in, for example, colon cells (see for example, Groden et ah, Cancer Research, 55: 1531-1539, 1995, herein incorporated by reference at least for material related to APC mutations and assays of the same). A variety of different morphologies of the effect of the mutated APC can be seen. Whether a cell contains an APC mutation can be assayed for using standard recombinant biotechnology protocols, for example, sequencing and PCR analysis or ligation mediated chain reaction (LCR) or other methods using (for example chip technology) capable of assaying and comparing DNA sequences. The mutations can readily be assayed as being functional mutations, by for example, expressing the mutant protein in a cell and determining if the mutant protein induces an oncogenic phenotype. Assays for determining the effect of an APC mutation can also be performed as discussed in Groden et al. In certain examples an activating mutation can be determined by assaying the mutation and comparing the effects to the effects of APC mutations in DLD-1 cells or HT29 cells. For example, as in Groden a fully functional APC gene can be transfected into a DLD-1 or HT29 cell, and this decreases the oncogenic behavior of the DLD-1 or HT29 cells because the non-mutant APC gene rescues normal phenotype. In certain examples, a given APC mutation can be assayed by, for example, transfecting the mutant APC into either a DLD-1 or HT29 cell line and comparing the level of rescue provided by the mutant APC to the level of rescue of the non-mutant APC. In certain examples the APC mutant will be considered an in-activating APC mutant, i.e., a mutant which causes oncogenic phenotype, if the rescue of the cells transfected by the mutant APC is less than 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the rescue of cells transfected with the non-mutant APC, as judged by any of the criteria used to judge oncogenic phenotype of DLD-1 or HT29 cells.
[0093] In certain examples, NF-KB/FLIP inhibitors and JNK actovaotrs are not administered to cancer cells that have an activating mutation on β-catenin. Whether a cell contains a β-catenin mutation can be assayed for using standard recombinant
biotechnology protocols, for example, sequencing and PCR analysis. The mutations can be assayed for function as discussed herein. [0094] Disclosed herein NF-KB/FLIP inhibitors and JNK activators can be
administered to cancer cells that express the COX-2 gene. A cell expresses the COX-2 gene if there are detectable transcripts of the COX-2 gene in the cell using an assay to detect transcripts, such as a hybridization assay, such as a northern blot or any of the chip type assays available, or an amplification based assay based on, for example, PCR or other amplification methods. In certain examples, the inhibitors can be administered to cancer cells that over express the COX-2 gene. Over expression of COX-2 can be determined by using any of the methods and assays discussed for the expression of COX-2 and comparing the level of expression to that of a control population of cells. In general cells do not show activated COX-2 expression, even if there is a basal amount of COX-2 expression, but upon additions of mitogens, for example, COX-2 expression, relative to expression in the absence of mitogens, increases. Thus, a given cancer cell or cancer cell line can be assayed for COX-2 expression and this can be compared to the COX-2 expression of this cell type in the absence of the oncogenic phenotype. In certain examples a given cancer cell line can be considered to over express COX-2 if the expression is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% of the expression of COX-2 in DLD-1 cells or HT29 cells, when expression of COX-2 of the cell of interest and DLD-1 or HT29 are assayed in parallel. [0095] The disclosed inhibitors can also be administered to cancer cells that express the COX-2 gene as well as having mutations in the APC gene.
[0096] It is understood that certain cancers can give rise to cancer cell lines.
Typically, cancer cell lines are cells that are maintained in cell culture, but that arose from a specific type of cancer. Inhibitors can be used for a variety of cancers, but can, for example, be used for cancers that are related to the DLD-1 cancer cell line and the HT-29 cancer cell line. The DLD-1 cancer cell line and the HT-29 cancer cell line arose from colon cancer cells. Also disclosed are cancer cell lines having the properties of the DLD-1 cancer cell line and the HT29 cell line.
[0097] The disclosed compositions can be used to treat any disease where
uncontrolled cellular proliferation occurs such as cancers. A non-limiting list of different types of cancers is as follows: lymphomas (Hodgkins and non-Hodgkins), leukemias, carcinomas, carcinomas of solid tissues, squamous cell carcinomas, adenocarcinomas, sarcomas, gliomas, high grade gliomas, blastomas, neuroblastomas, plasmacytomas, histiocytomas, melanomas, adenomas, hypoxic tumors, myelomas, AIDS-related lymphomas or sarcomas, metastatic cancers, or cancers in general. [0098] A representative but non-limiting list of cancers that the disclosed
compositions can be used to treat is the following: lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, cervical carcinoma, breast cancer, and epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, stomach cancer, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, prostatic cancer, pancreatic cancer, or cancer stem cells.
[0099] Compounds disclosed herein may also be used for the treatment of precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias. Compositions
[00100] Disclosed are the components to be used to prepare the disclosed compositions as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when
combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular composition such as BAY 11-7082 is disclosed and discussed and a number of modifications that can be made to a number of molecules including BAY 11-7082 are discussed, specifically contemplated is each and every combination and permutation of BAY 11-7082 and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods.
NF-KB/FLIP inhibitors and JNK activators
[00101] The disclosed methods involve NF-κΒ and FLIP inhibitors ("NF-KB/FLIP inhibitors"). An NF-KB/FLIP inhibitor can be any composition that causes a decrease in the expression of anti-apoptotic proteins or an increase in the expression of apoptotic proteins. The inhibitors can also be any composition wherein the composition inhibits ΙκΒ phosphorylation. The inhibitor can also be any composition, wherein the composition inhibits TNFa induced NF-κΒ activation. Still further, a suitable inhibitor also activates JNK and inhibits FLIP.
[00102] In certain examples, the inhibitors useful in any of the methods disclosed herein are olefins. An "olefin" is defined herein as any compound or molecule possessing at least one carbon-carbon double bond. Each carbon atom of the carbon-carbon double bond may be unsubstituted or independently substituted with one or two different moieties.
[00103] In certain examples, the inhibitor is an olefin having at least one electron- withdrawing group. In another example, the inhibitor is an olefin having at least two electron-withdrawing groups. In this case, when two electron-withdrawing groups are present, the electron-withdrawing groups can be present on the same olefmic carbon atom or one electron-withdrawing group can be on each olefmic carbon atom.
[00104] The term "electron-withdrawing group" is any group that has an affinity or attraction for electron density. For example, when an electron- withdrawing group is attached to an olefmic carbon atom (Ca), then the other olefmic carbon atom (Cp) is more susceptible to nucleophilic attack (i.e., Cp is more electropositive) when compared to an olefin that does not possess an electron- withdrawing group. Generally, electron withdrawing groups possess one or more carbon-carbon multiple bonds, carbon- heteroatom multiple bonds, or heteroatom-heteroatom multiple bonds. Examples of electron-withdrawing groups include, but are not limited to, a cyano group, a sulfo-oxy group, a phospho-oxy group, a carboxyl group, a nitro group, a halogen, a halogenated alkyl group, an unsubstituted aromatic ring, or a substituted aromatic ring having at least one cyano group, sulfo-oxy group, phospho-oxy group, carboxyl group, hydroxyl group, amino group, ether group, halogenated alkyl group, halogen, or nitro group.
[00105] The term "phospho-oxy group" is a group having one of the following structures
Figure imgf000031_0001
wherein R1 is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic.
[00106] The term "sulfo-oxy group" is a group having one of the following structures
Figure imgf000031_0002
wherein R is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic. In one example, the inhibitor is an olefin having a cyano group and a sulfo-oxy group having the structure
Figure imgf000031_0003
wherein R2 is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic.
[00107] In another example, the inhibitor has the structure I.
Figure imgf000032_0001
wherein R3, R4 and R5 are, independently, hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic, wherein the compound is the E- or Z-isomer. The stereochemistry about the carbon-carbon double bond will vary depending upon the relative positions of the cyano group (-CN) and the sulfonyl group (- S(02)R5)). When the cyano group and sulfonyl group are cis to one another, then the compound is the Z-isomer, and when the cyano group and sulfonyl group are trans to one another, then the compound is the E-isomer. In one example, the inhibitor having the Structure I is the E-isomer.
[00108] In one example, when the inhibitor has the Structure I, R3 and R4 are hydrogen. In another example, R5 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, substituted or unsubstituted phenyl, or benzyl. In a further example, R5 is a phenyl group having at least one alkyl group.
[00109] In another example, the inhibitor having the Structure I is the ethyl ester of 2- cyano-3-(methylsulfonyl)-3-(l ,2,4,5-tetrahydro-3H-3-benzazepin-3-yl-2-propenoic acid;
[2,2-bis[p-(dimethylamino)phenyl]vinyl] (methylsulfonyl)-fumaronitrile; [2,2-bis[p- dimethylamino)phenyl] vinyl] [(hydroxymethyl)sulfonyll)- fumaronitrile,
p-toluenesulfonate; [2,2-bis[p-dimethylamino)phenyl]vinyl] (ethylsulfonyl)-fumaronitrile; (benzylsulfonyl) [2,2-bis[p-(dimethylamino)phenyl]vinyl]-fumaronitrile; (allylsulfonyl) [2,2-bis[p-dimethylamino)phenyl]vinyl]-fumaronitrile; benzyl (benzylsulfonyl)- fumaronitrile; l ,7-bis(allylsulfonyl)-4-hydroxy-l,3,5-Heptatriene-l,2,6,7-tetracarbonitrile; 2-cyano-3-(methylsulfonyl)-3-(l,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-2-propenoic acid ethyl ester; a-[[[(lE)-l-cyano-2-(3,4-dihydroxyphenyl)ethenyl]sulfonyl]methylene]- 3,4-dihydroxy-P-oxo-(a E)-benzenepropanenitrile; 3-(methylsulfonyl)-(2E)-2- propenenitrile; 4,4,4-trifluoro-3-(hexylsulfonyl)-(2Z)-2-butenenitrile; a-[[[l-cyano-3- (3 ,4-dihydroxyphenyl)-3 -oxo- 1 -propenyl] sulfonyljmethylene] -3 ,4-dihydroxy-(E,E)- benzeneacetonitrile; 2,2'-[ 1 ,3-propanediylbis[(cyclohexylimino)-4, 1 -phenylene]]bis[3- (ethylsulfonyl)-2-butenedinitrile; a-(methylsulfonyl)methylene)-(Z)-benzeneacetonitrile; a- [ [( 1 , 1 -dimethylethyl)sulfonyl]methylene]-(Z)-benzeneacetonitrile; a- [methylsulfonyl)methylene] -(E)-benzeneacetonitrile; a-
[[(phenylmethyl)sulfonyl]methylene]-benzeneacetonitrile; a-[(butylsulfonyl)methylene]- (E)-benzeneacetonitrile; a-[(butylsulfonyl)methylene]-(Z)-benzeneacetonitrile; a-[[(l - methylethyl)sulfonyl]methylene]-(E)-benzeneacetonitrile; -[[(l- methylethyl)sulfonyl]methylene]-(Z)-benzeneacetonitrile; a-[[(l , 1- dimethylethyl)sulfonyl]methylene]-(E)-benzeneacetonitrile; -[[[4- chlorophenyl)methyl]sulfonyl]methylene]-(E)-benzeneacetonitrile; -[[[4- chlorophenyl)methyl]sulfonyl]methylene]-(Z)-benzeneacetonitrile; a-[[3- chloropropyl)sulfonyl]methylene]-(E)-benzeneacetonitrile; a-[[3- chloropropyl)sulfonyl]methylene] -(Z)-benzeneacetonitrile; 3 -(methy lsulfonyl)-(2E)-2- propenenitrile; 3-(methylsulfonyl)-(Z)-2-propenenitrile; a-[(methylsulfonyl)methylene]-
2- nitro-benzeneacetonitrile; 4-(dimethylamino)-a-[[(trifluoromethyl)sulfonyl]methylene]- benzeneacetonitrile; 2-chloro-3-(methylsulfonyl)-2 -propenenitrile; 2,3-dichloro-3- (methylsulfonyl)-2-propenenitrile; 2,3-dichloro-3-[(l-methylethyl)sulfonyl]-2- propenenitrile; 2,3-dichloro-3-[(l-methylpropyl)sulfonyl]-2-propenenitrile; 2,3-dichloro-
3 - [(3 -methy lbutyl)sulfonyl] -2-propenenitrile; 2,3 -dichloro-3 -(octylsulfonyl)-2- propenenitrile; 2,3 -dichloro-3 -(nonylsulfonyl)-2-propenenitrile; 3 - [(2- phenylethenyl)sulfony 1 )-2 -propenenitrile; 3 - [(2-phenylethenyl)sulfony 1 ] -2- propenenitrile; a-[ methylsulfonyl)phenylmethylene]-(Z)-benzeneacetonitrile; 3- (benzylsulfonyl)-acrylonitrile; 3 -(methylsulfony l)-2-propenenitrile; 3 -(ethylsulfonyl)- acrylonitrile; 3 ,3 '-(tetramethylenedisulfony l)di-acrylonitrile; 3 - [(2-cyanovinyl)sulfonyl)- propionitrile; 3-((4-(2,2-dichloro- 1 , 1 -difluoroethoxy)phenyl)sulfonyl)-2-propenenitrile; 3-((4-(2,2-dichloro- 1 , 1 -difluoroethoxy)-2-methyl-5-nitropheny)sulfonyl)-2-propenenitrile; or 3 -((3 -trifluoromethyl)phenyl)sulfonyl)-2-propenenitrile .
[00110] In another exampple, the inhibitor is (2E)-3-(tolylsulfonyl)-2-propenenitrile, which is also referred to herein as BAY-7082.
Figure imgf000034_0001
[00111] In another example, the inhibitor is (2E)-3-[4-(tertiary butyl phenyl)sulfonyl]- 2-propenenitrile, which is also referred to herein as BAY- 11-7085.
Figure imgf000034_0002
[00112] In an alternative example, the inhibitors useful in any of the methods disclosed herein can comprise at least one amino acid residue. An "amino acid residue" is produced when an amino acid is reacted with one or more compounds capable of reacting with the amino acid. For example, when an amino acid is reacted with two other amino acids to produce a tripeptide, the resultant tripeptide contains three amino acid residues.
Alternatively, the amino acid can react with other non-amino acids to produce a compound having an amino acid residue.
[00113] In one example, the inhibitor has at least one leucine residue. In another example, the inhibitor comprises three leucine residues. In a further example, the compound is N-[(phenylmethoxy)carbonyl]-L-leucyl-N-[(lS)-l-formyl-3-methylbutyl]-L- leucinamide, which has the structure
Figure imgf000035_0001
[00114] This compound is also referred to as Sigma MG-132.
[00115] In another example, the inhibitor is pyrrolidine dithiocarbamate, which has the structure II:
Figure imgf000035_0002
wherein R^ and R7 are, independently, hydrogen, alkyl alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic, or ¾ and R7 together for an ring with the nitrogen atom, and X and Y are, independently oxyten or sulfur, or the pharmaceutically active salt thereo.
[00116] In one embodiment, X and Y are both oxygen. In another embodiment, X and Y are both sulfur. In a further embodiment, and R7 can form a ring system. For example, R6 and R7 can be collectively a methylene group such as (CH2)3. The resultant ring structure would be a four-membered ring with one nitrogen atom. When R6 and R7 form a ring, the ring can be from a three- to 10-membered ring. In one embodiment, X and Y are sulfur, and R6 and R7 is (CH2)4. This compound is also referred to as PDTC.
[00117] Formula II also enompasses pharmaceutically acceptable esters, amides, and salts of such compounds. Pharmaceutically acceptable salts are prepared by treating the free acid with an appropriate amount of a pharmaceutically acceptable base.
Representative pharmaceutically acceptable bases are ammonium hydroxide, sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, magnesium hydroxide, ferrous hydroxide, zinc hydroxide, copper hydroxide, aluminum hydroxide, ferric hydroxide, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, lysine, arginine, histidine, and the like. In one aspect, the reaction is conducted in water, alone or in combination with an inert, water-miscible organic solvent, at a temperature of from about 0°C to about 100°C, such as at room temperature. The molar ratio of compounds of structural formula II to base used are chosen to provide the ratio desired for any particular salts. For preparing, for example, the ammonium salts of the free acid starting material, the starting material can be treated with approximately one equivalent of pharmaceutically acceptable base to yield a neutral salt. [00118] Ester derivatives are typically prepared as precursors to the acid form of the compounds— as illustrated in the examples below— and accordingly can serve as prodrugs. Generally, these derivatives will be lower alkyl esters such as methyl, ethyl, and the like. Amide derivatives -(CO)NH2, -(CO)NHR, and -(CO)NR2, where R is an alkyl group defined above, can be prepared by reaction of the carboxylic acid-containing compound with ammonia or a substituted amine.
[00119] Still further examples of inhibitors include include derivatives of glitazones or thiazolidinediones, such as, troglitazone; rosiglitazone and pioglitazone. Other useful thiazolidinediones include ciglitazone, englitazone, darglitazone and BRL 49653 as disclosed in WO 98/05331 which is incorporated herein by reference. [00120] Still further examples of inhibitors can be obtain from commercially available libraries, for example, kinase inhibitors libraries (BioMol), synthetic compound libraries (e.g., from Bayer or Sigma Aldrich), natural compound libraries (Specs, TimTec), or other libraries from companies such as Sigma-Aldrich (St. Louis, Mass.), Arqule (Woburn, Ma.), Enzymed (Iowa City, Iowa), Maybridge Chemical Co.(Trevillett, Cornwall, UK), MDS Panlabs (Bothell, Wash.), Pharmacopeia (Princeton, N.J.), and Trega (San Diego, Calif). Preferred compounds are low molecular weight compounds. Low molecular weight compounds, i.e., with a molecular weight of 500 Dalton or less, are likely to have good absorption and permeation in biological systems and are consequently more likely to be successful drug candidates than compounds with a molecular weight above 500 Dalton. Compounds including natural products, inorganic chemicals, and biologically active materials such as proteins and toxins can also be assayed using these methods for the ability to inhibit NF-κΒ and/or FLIP.
[00121] Specific screening methods are known in the art and along with integrated robotic systems and collections of chemical compounds/natural products are extensively incorporated in high throughput screening so that large numbers of test compounds can be tested for antagonist or agonist activity within a short amount of time. These methods include homogeneous assay formats such as fluorescence resonance energy transfer, fluorescence polarization, time-resolved fluorescence resonance energy transfer, scintillation proximity assays, reporter gene assays, fluorescence quenched enzyme substrate, chromogenic enzyme substrate and electrochemiluminescence, as well as more traditional heterogeneous assay formats such as enzyme-linked immunosorbant assays (ELISA) or radioimmunoassays. Homogeneous assays are "mix and read" assays that are very amenable to robotic application, whereas heterogeneous assays require separation of bound analyte from free by more complex unit operations such as filtration, centrifugation or washing. These assays are utilized to detect a wide variety of specific biomolecular interactions and the inhibition thereof by small organic molecules, including protein- protein, receptor-ligand, enzyme-substrate, etc. These assay methods and techniques are well known in the art and are described more fully in the following: High Throughput Screening: The Discovery of Bioactive Substances, John P. Devlin (ed.), Marcel Dekker, New York, 1997. The screening assays of the present invention are amenable to high throughput screening of chemical libraries and are suitable for the identification of small molecule drug candidates, antibodies, peptides, and other antagonists and/or agonists.
Direct inhibitors
[00122] Disclosed are inhibitors which are direct inhibitors of NF-κΒ and/or FLIP. A direct inhibitor is an inhibitor that interacts directly with NF-κΒ and/or FLIP. A direct inhibitor touches in some way the NF-κΒ molecule and/or the FLIP molecule such that the NF-KB and/or FLIP dependent activities, such as readhesion, metastasis, and apoptosis supression are inhibited. Indirect inhibitors
[00123] Disclosed are inhibitors which are indirect inhibitors of NF-κΒ and.or FLIP. An indirect inhibitor is an inhibitor that does not interact directly with NF-κΒ and/or FLIP. An indirect inhibitor touches in some way a molecule that is involved in a signal transduction pathway that NF-κΒ and/or FLIP is involved in such that the NF-κΒ and/or FLIP dependent activities, such as readhesion and metastasis are inhibited.
[00124] One example of an indirect inhibitor of NF-κΒ and/or FLIP would be an indirect inhibitor which inhibits the expression of NF-κΒ and/or FLIP and thereby prevent NF-KB and/or FLIP from functioning. Another example of an indirect inhibitor of NF-KB would be an indirect inhibitor that inhibits translation of a gene encoding NF-κΒ and/or FLIP.
[00125] An example of a molecule that is involved in a signal transduction pathway that NF-KB is involved in is ΙκΒ. Molecules that inhibit ΙκΒ from phosphorylating can be indirect inhibitors. Pharmaceutical carriers/Delivery of pharmaceutical products
[00126] As described above, the compositions can also be administered in vivo in a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject, along with the nucleic acid or vector, without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained. The carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject, as would be well known to one of skill in the art. [00127] The compositions may be administered orally, parenterally {e.g.,
intravenously), by intramuscular injection, by intraperitoneal injection, transdermally, extracorporeally, topically or the like, although topical intranasal administration or administration by inhalant is typically preferred. As used herein, "topical intranasal administration" means delivery of the compositions into the nose and nasal passages through one or both of the nares and can comprise delivery by a spraying mechanism or droplet mechanism, or through aerosolization of the nucleic acid or vector. The latter may be effective when a large number of animals is to be treated simultaneously.
Administration of the compositions by inhalant can be through the nose or mouth via delivery by a spraying or droplet mechanism. Delivery can also be directly to any area of the respiratory system (e.g. , lungs) via intubation. The exact amount of the compositions required will vary from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the allergic disorder being treated, the particular nucleic acid or vector used, its mode of administration and the like. Thus, it is not possible to specify an exact amount for every composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein.
[00128] Parenteral administration of the composition, if used, is generally characterized by injection. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution of suspension in liquid prior to injection, or as emulsions. A more recently revised approach for parenteral administration involves use of a slow release or sustained release system such that a constant dosage is maintained. See, e.g., U.S. Patent 3,610,795, which is incorporated by reference herein. In a preferred aspect, the compositions disclosed herein can be administered
intraperitoneally. For example, during intraabdominal surgery (e.g., to resection of a tumor), the disclosed compositions can be used as a wash. In this way, cancer cells that were not excised and/or cells that may be unattached but still present in the body, can be killed upon readhereing. Alternatively, the disclosed compositions can be administered around the time of surgery (peri-operative), before surgery (pre-operative), or after surger (post-operatively). The compounds can be administered a week, 2-5 days, 1-3 days, 1-18 hours, 1-12 hours, 1-6 hours, or less than an hour before or after tumor resection surgery.
[00129] In another preferred example, the disclosed compositions can be administered by I.V., by injection and/or an I.V. drip.
[00130] The materials may be in solution, suspension (for example, incorporated into microparticles, liposomes, or cells). These may be targeted to a particular cell type via antibodies, receptors, or receptor ligands. The following references are examples of the use of this technology to target specific proteins to tumor tissue (Senter, et al,
Bioconjugate Chem., 2:447-451, (1991); Bagshawe, K.D., Br. J. Cancer, 60:275-281, (1989); Bagshawe, et al, Br. J. Cancer, 58:700-703, (1988); Senter, et al, Bioconjugate Chem., 4:3-9, (1993); Battelli, et al, Cancer Immunol. Immunother., 35:421-425, (1992); Pietersz and McKenzie, Immunolog. Reviews, 129:57-80, (1992); and Roffler, et al, Biochem. Pharmacol, 42:2062-2065, (1991)). Vehicles such as "stealth" and other antibody conjugated liposomes (including lipid mediated drug targeting to colonic carcinoma), receptor mediated targeting of DNA through cell specific ligands, lymphocyte directed tumor targeting, and highly specific therapeutic retroviral targeting of murine glioma cells in vivo. The following references are examples of the use of this technology to target specific proteins to tumor tissue (Hughes et al, Cancer Research, 49:6214-6220, (1989); and Litzinger and Huang, Biochimica et Biophysica Acta, 1104: 179-187, (1992)). In general, receptors are involved in pathways of endocytosis, either constitutive or ligand induced. These receptors cluster in clathrin-coated pits, enter the cell via clathrin-coated vesicles, pass through an acidified endosome in which the receptors are sorted, and then either recycle to the cell surface, become stored intracellularly, or are degraded in lysosomes. The internalization pathways serve a variety of functions, such as nutrient uptake, removal of activated proteins, clearance of macromolecules, opportunistic entry of viruses and toxins, dissociation and degradation of ligand, and receptor-level regulation. Many receptors follow more than one intracellular pathway, depending on the cell type, receptor concentration, type of ligand, ligand valency, and ligand concentration.
Molecular and cellular mechanisms of receptor-mediated endocytosis has been reviewed (Brown and Greene, DNA and Cell Biology 10:6, 399-409 (1991)).
Pharmaceutically Acceptable Carriers [00131] The compositions, including antibodies, can be used therapeutically in combination with a pharmaceutically acceptable carrier.
[00132] Pharmaceutical carriers are known to those skilled in the art. These most typically would be standard carriers for administration of drugs to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH. The compositions can be administered intramuscularly or subcutaneously. Other compounds will be administered according to standard procedures used by those skilled in the art. [00133] Pharmaceutical compositions may include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice. Pharmaceutical compositions may also include one or more active ingredients such as antimicrobial agents, antiinflammatory agents, anesthetics, and the like. [00134] The pharmaceutical composition may be administered in a number of ways depending on whether local or systemic treatment is desired, and on the area to be treated. Administration may be topically (including ophthalmically, vaginally, rectally, intranasally), orally, by inhalation, or parenterally, for example by intravenous drip, subcutaneous, intraperitoneal or intramuscular injection. The disclosed antibodies can be administered intravenously, intraperitoneally, intramuscularly, subcutaneous ly, intracavity, or transdermally.
[00135] Preparations for parenteral administration include sterile aqueous or nonaqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
[00136] Formulations for topical administration may include ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional
pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
[00137] Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, or tablets. Thickeners, flavorings, diluents, emulsifiers, dispersing aids or binders may be desirable..
[00138] Some of the compositions may potentially be administered as a
pharmaceutically acceptable acid- or base- addition salt, formed by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases such as mono-, di-, trialkyl and aryl amines and substituted ethanolamines.
Therapeutic Uses
[00139] The dosage ranges for the administration of the compositions are those large enough to produce the desired effect in which the symptoms disorder are effected. The dosage should not be so large as to cause adverse side effects, such as unwanted cross- reactions, anaphylactic reactions, and the like. Generally, the dosage will vary with the age, condition, sex and extent of the disease in the patient and can be determined by one of skill in the art. The dosage can be adjusted by the individual physician in the event of any counterindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days.
Combinatorial chemistry
[00140] The disclosed compositions can be used as targets for any combinatorial technique to identify molecules or macromolecular molecules that interact with the disclosed compositions in a desired way. Also disclosed are the compositions that are identified through combinatorial techniques or screening techniques in which the compositions interact with NF-κΒ, FLIP, JNK, and/or ΙκΒ such that the compositions decrease the cellular proliferative activity of NF-κΒ, FLIP, and ΙκΒ or portions thereof, increase INK, where the compositions were identified using NF-κΒ , FLIP, JNK, or ΙκΒ as targets in a screening or selection protocol.
[00141] It is understood that when using the disclosed compositions in combinatorial techniques or screening methods, molecules, such as macromolecular molecules, will be identified that have particular desired properties such as inhibition or stimulation or the target molecule's function. The molecules identified and isolated when using the disclosed compositions, such as, NF-κΒ, FLIP, JNK, or ΙκΒ are used as targets, or when the disclosed compositions like BAY- 11-7082 or BAY- 11-7085 are used in competitive inhibition assays are also disclosed. Thus, the products produced using the combinatorial or screening approaches that involve the disclosed compositions are also considered herein disclosed.
[00142] Combinatorial chemistry includes but is not limited to all methods for isolating small molecules or macromolecules that are capable of binding either a small molecule or another macromolecule, typically in an iterative process. Proteins, oligonucleotides, and sugars are examples of macromolecules. For example, oligonucleotide molecules with a given function, catalytic or ligand-binding, can be isolated from a complex mixture of random oligonucleotides in what has been referred to as "in vitro genetics" (Szostak, TIBS 19:89, 1992). One synthesizes a large pool of molecules bearing random and defined sequences and subjects that complex mixture, for example, approximately 1015 individual sequences in 100 μg of a 100 nucleotide RNA, to some selection and enrichment process. Through repeated cycles of affinity chromatography and PCR amplification of the molecules bound to the ligand on the column, Ellington and Szostak (1990) estimated that 1 in 1010 RNA molecules folded in such a way as to bind a small molecule dyes. DNA molecules with such ligand-binding behavior have been isolated as well (Ellington and Szostak, 1992; Bock et al, 1992). Techniques aimed at similar goals exist for small organic molecules, proteins, antibodies and other macromolecules known to those of skill in the art. Screening sets of molecules for a desired activity whether based on small organic libraries, oligonucleotides, or antibodies is broadly referred to as combinatorial chemistry. Combinatorial techniques are particularly suited for defining binding interactions between molecules and for isolating molecules that have a specific binding activity, often called aptamers when the macromolecules are nucleic acids.
[00143] There are a number of methods for isolating proteins which either have de novo activity or a modified activity. For example, phage display libraries have been used to isolate numerous peptides that interact with a specific target. (See for example, U.S. Patent 6,031,071; 5,824,520; 5,596,079; and 5,565,332 which are herein incorporated by reference at least for their material related to phage display and methods relate to combinatorial chemistry) [00144] A preferred method for isolating proteins that have a given function is described by Roberts and Szostak (Roberts R.W. and Szostak J.W. Proc. Natl. Acad. Sci. USA, 94(23): 12997-302, 1997). This combinatorial chemistry method couples the functional power of proteins and the genetic power of nucleic acids. An RNA molecule is generated in which a puromycin molecule is covalently attached to the 3 '-end of the RNA molecule. An in vitro translation of this modified RNA molecule causes the correct protein, encoded by the RNA to be translated. In addition, because of the attachment of the puromycin, a peptdyl acceptor which cannot be extended, the growing peptide chain is attached to the puromycin which is attached to the RNA. Thus, the protein molecule is attached to the genetic material that encodes it. Normal in vitro selection procedures can now be done to isolate functional peptides. Once the selection procedure for peptide function is complete traditional nucleic acid manipulation procedures are performed to amplify the nucleic acid that codes for the selected functional peptides. After
amplification of the genetic material, new RNA is transcribed with puromycin at the 3'- end, new peptide is translated and another functional round of selection is performed. Thus, protein selection can be performed in an iterative manner just like nucleic acid selection techniques. The peptide which is translated is controlled by the sequence of the RNA attached to the puromycin. This sequence can be anything from a random sequence engineered for optimum translation (i.e. no stop codons etc.) or it can be a degenerate sequence of a known RNA molecule to look for improved or altered function of a known peptide. The conditions for nucleic acid amplification and in vitro translation are well known to those of ordinary skill in the art and are preferably performed as in Roberts and Szostak (Roberts R.W. and Szostak J.W., Proc. Natl. Acad. Sci. USA, 94(23): 12997-302, 1997).
[00145] Another preferred method for combinatorial methods designed to isolate peptides is described in Cohen et al. (Cohen et al., Proc Natl Acad Sci USA 95(24): 14272- 7, 1998). This method utilizes and modifies two-hybrid technology. Yeast two-hybrid systems are useful for the detection and analysis of protein:protein interactions. The two- hybrid system, initially described in the yeast Saccharomyces cerevisiae, is a powerful molecular genetic technique for identifying new regulatory molecules, specific to the protein of interest (Fields and Song, Nature 340:245-6, 1989). Cohen et al. modified this technology so that novel interactions between synthetic or engineered peptide sequences could be identified which bind a molecule of choice. The benefit of this type of technology is that the selection is done in an intracellular environment. The method utilizes a library of peptide molecules that attached to an acidic activation domain. A peptide of choice, for example a portion of NF-κΒ, FLIP, JNK, or ΙκΒ is attached to a DNA binding domain of a transcriptional activation protein, such as Gal 4. By performing the two-hybrid technique on this type of system, molecules that bind desired fragments of NF-κΒ, FLIP,JNK, or ΙκΒ can be identified.
[00146] Using methodology well known to those of skill in the art, in combination with various combinatorial libraries, one can isolate and characterize those small molecules or macromolecules, which bind to or interact with the desired target. The relative binding affinity of these compounds can be compared and optimum compounds identified using competitive binding studies, which are well known to those of skill in the art.
[00147] Techniques for making combinatorial libraries and screening combinatorial libraries to isolate molecules which bind a desired target are well known to those of skill in the art. Representative techniques and methods can be found in but are not limited to U.S. Patents 5,084,824, 5,288,514, 5,449,754, 5,506,337, 5,539,083, 5,545,568, 5,556,762, 5,565,324, 5,565,332, 5,573,905, 5,618,825, 5,619,680, 5,627,210, 5,646,285, 5,663,046, 5,670,326, 5,677,195, 5,683,899, 5,688,696, 5,688,997, 5,698,685, 5,712,146, 5,721,099, 5,723,598, 5,741,713, 5,792,431, 5,807,683, 5,807,754, 5,821,130, 5,831,014, 5,834,195, 5,834,318, 5,834,588, 5,840,500, 5,847,150, 5,856,107, 5,856,496, 5,859,190, 5,864,010, 5,874,443, 5,877,214, 5,880,972, 5,886,126, 5,886,127, 5,891,737, 5,916,899, 5,919,955, 5,925,527, 5,939,268, 5,942,387, 5,945,070, 5,948,696, 5,958,702, 5,958,792, 5,962,337, 5,965,719, 5,972,719, 5,976,894, 5,980,704, 5,985,356, 5,999,086, 6,001,579, 6,004,617, 6,008,321, 6,017,768, 6,025,371, 6,030,917, 6,040,193, 6,045,671, 6,045,755, 6,060,596, and 6,061,636.
[00148] Combinatorial libraries can be made from a wide array of molecules using a number of different synthetic techniques. For example, libraries containing fused 2,4- pyrimidinediones (U.S. Patent 6,025,371) dihydrobenzopyrans (U.S. Patent 6,017, 768and 5,821,130), amide alcohols (U.S. Patent 5,976,894), hydroxy-amino acid amides (U.S. Patent 5,972,719) carbohydrates (U.S. Patent 5,965,719), l,4-benzodiazepin-2,5-diones (U.S. Patent 5,962,337), cyclics (U.S. Patent 5,958,792), biaryl amino acid amides (U.S. Patent 5,948,696), thiophenes (U.S. Patent 5,942,387), tricyclic Tetrahydroquinolines (U.S. Patent 5,925,527), benzofurans (U.S. Patent 5,919,955), isoquinolines (U.S. Patent 5,916,899), hydantoin and thiohydantoin (U.S. Patent 5,859,190), indoles (U.S. Patent 5,856,496), imidazol-pyrido-indole and imidazol-pyrido-benzothiophenes (U.S. Patent 5,856,107) substituted 2-methylene-2, 3-dihydrothiazoles (U.S. Patent 5,847,150), quinolines (U.S. Patent 5,840,500), PNA (U.S. Patent 5,831,014), containing tags (U.S. Patent 5 ,721 ,099), polyketides (U. S . Patent 5,712,146), morpholino-subunits (U. S . Patent 5,698,685 and 5,506,337), sulfamides (U.S. Patent 5,618,825), and benzodiazepines (U.S. Patent 5,288,514).
[00149] As used herein combinatorial methods and libraries included traditional screening methods and libraries as well as methods and libraries used in iterative processes.
Computer assisted drug design
[00150] The disclosed compositions can be used as targets for any molecular modeling technique to identify either the structure of the disclosed compositions or to identify potential or actual molecules, such as small molecules, which interact in a desired way with the disclosed compositions.
[00151] It is understood that when using the disclosed compositions in modeling techniques, molecules, such as macromolecular molecules, will be identified that have particular desired properties such as inhibition or stimulation or the target molecule's function. The molecules identified and isolated when using the disclosed compositions are also disclosed. Thus, the products produced using the molecular modeling approaches that involve the disclosed compositions are also considered herein disclosed.
Kits
[00152] Disclosed herein are kits that are drawn to reagents that can be used in practicing the methods disclosed herein. The kits can include any reagent or combination of reagent discussed herein or that would be understood to be required or beneficial in the practice of the disclosed methods. For example, the kits could include molecules, including for example, BAY 11-7082 or BAY 11-7085, for use in in vitro cell assays as standards for anti-proliferative activity. Compositions with similar functions
[00153] It is understood that the compositions, such as BAY 11-7082 and BAY 11- 7085, disclosed herein have certain functions, such as antimetastatic activities or antiproliferative activities. Disclosed herein are certain structural requirements for performing the disclosed functions, and it is understood that there are a variety of structures which can perform the same function which are related to the disclosed structures, and that these structures will ultimately achieve the same result, for example, inhibition of antiproliferative activities.
Methods of making the compositions [00154] The compositions disclosed herein and the compositions necessary to perform the disclosed methods can be made using any method known to those of skill in the art for that particular reagent or compound unless otherwise specifically noted.
[00155] Disclosed are processes for making the compositions as well as making the intermediates leading to the compositions. There are a variety of methods that can be used for making these compositions, such as synthetic chemical methods and standard molecular biology methods. It is understood that the methods of making these and the other disclosed compositions are specifically disclosed.
[00156] Disclosed are cells produced by the process of transforming the cell with any of the disclosed nucleic acids. Disclosed are cells produced by the process of
transforming the cell with any of the non-naturally occurring disclosed nucleic acids.
[00157] Disclosed are any of the disclosed peptides produced by the process of expressing any of the disclosed nucleic acids. Disclosed are any of the non-naturally occurring disclosed peptides produced by the process of expressing any of the disclosed nucleic acids. Disclosed are any of the disclosed peptides produced by the process of expressing any of the non-naturally disclosed nucleic acids.
[00158] Disclosed are animals produced by the process of transfecting a cell within the animal with any of the nucleic acid molecules disclosed herein. Disclosed are animals produced by the process of transfecting a cell within the animal any of the nucleic acid molecules disclosed herein, wherein the animal is a mammal. Also disclosed are animals produced by the process of transfecting a cell within the animal any of the nucleic acid molecules disclosed herein, wherein the mammal is mouse, rat, rabbit, cow, sheep, pig, or primate.
[00159] Also disclose are animals produced by the process of adding to the animal any of the cells disclosed herein.
EXAMPLES
[00160] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in °C or is at ambient temperature, and pressure is at or near atmospheric.
Example 1:
Materials:
[00161] The NF-κΒ inhibitors, BAY 11-7082 (BioMol), BAY 11-7085, (BioMol) and N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132) (Sigma) were solubilized in DMSO. The antagonistic and chimeric TNFa monoclonal antibody (cA2) was purchased from the clinical pharmacy at the University of Utah and reconstituted in sterile water. Polyclonal antibodies to FLIPS/L, c-IAP-1, c-IAP-2, TRAF-1, and TRAF-2 were obtained from Santa Cruz Biotechnology. Antibodies to cleaved poly(ADP-ribose) polymerase (PARP) were obtained from New England Biolabs. The monoclonal antibody recognizing the p65 NF-KB subunit was obtained from Santa Cruz Biotechnology. Alkaline phosphatase-conjugated goat-anti-rabbit antibody was obtained from Jackson
Laboratories. Cell Culture, Proliferation, and Toxicity:
[00162] DLD-1, HCT-116, and HT-29 colon cancer cell lines were obtained from the ATCC collection and were cultured in Dulbecco's Modification of Eagle's Medium (DMEM) supplemented with 10% fetal bovine serum, glutamine, penicillin, and streptomycin. 293 cells stably transfected with COX-2 (293 -COX-2) under the control of a ponasterone sensitive promoter were cultured in Dulbecco's Modification of Eagle's Medium supplemented with 10% fetal bovine serum, 400 μg/mL of zeocin, 400μg/mL of G418, glutamine, penicillin, and streptomycin. For induction of COX-2 protein expression, 293-COX-2 cells were cultured for 48 hours in media supplemented with ^g/ml of ponasterone. All cells were cultured at 37°C in a humidified incubator with a 5%> C02 atmosphere.
[00163] For the cell proliferation assay, cells were dispersed and plated at 40,000 cells/well in 96-well dishes. At various days in culture, the cells were gently washed twice with 100 μΕΛνεΙΙ of ice-cold blocking buffer (1% radioimmunoassay grade BSA in PBS) and twice with 100 μΕ/well of ice-cold PBS. The cells were fixed for 10 min in 100%) ice-cold methanol (100 μΕ/well), then allowed to air-dry. The cells were stained with ΙΟΟμΕΛνεΙΙ of 0.1 % crystal violet in H20 for 10 minutes, then washed gently four times with dd H20 and four times with PBS. The plates were then air-dried completely. The stained cells were then solubilized in 1% sodium deoxycholate, and the plates read at 590 nm in a spectrophotometer. The absorption at 590 nm is proportional to the number of attached cells.
[00164] For the cell toxicity assay, the CYTOTOX96™ kit from Promega was used according to the manufacturer's instructions. Briefly, DLD-1, HCT-116, and HT-29 cells were incubated with 1, 2.5, 5, 7.5 and 10 μΜ BAY 11-7082 or BAY 11-7085 in 96-well plates for 24 hours then lysed by adding 15 μΐ, of lysis 10X Solution (9% (v/v)
TRITON™ X-100 in water) per 100 μΐ^ of culture medium, followed by incubation at 37°C for 45-60 minutes. Sample supernatants (50 μί) were transferred to a fresh 96 well enzymatic assay plate and incubated with reconstituted Substrate Mix (50 μΐ, per sample) for 30 minutes at room temperature in the dark. The enzymatic assay was then stopped by adding 50 μΙ,ΛνεΙΙ of Stop Solution. The plate was read at 490 nm and the absorbance values plotted as a ratio to the controls (DMSO alone). Electrophoretic Mobility Shift Assay:
[00165] The adherent cell monolayers were washed once with cold PBS after which 200 \iL of cold Buffer A (20 mM HEPES pH 7.8, 1 mM EDTA, 1 mM EGTA, 10% glycerol, 0.2% NP-40, 0.2 mM Na3V04, 1 mM DTT, 0.5 mM PMSF,and, 1 μg/mL each of aprotinin and leupeptin) was added to the cells. The cells were scraped into microfuge tubes, kept on ice, and sonicated. The lysates were centrifuged at 500 g for 5 min at 4°C and the cytoplasmic fraction (supernatant) was removed. 25-50 of Buffer B (20 mM HEPES pH 7.8, 1 mM EDTA, 1 mM EGTA, 0.42 M NaCl, 1.5 mM MgCl2, 25% glycerol, 0.2 mM Na3VO 4, 1 mM DTT, 0.5 mM PMSF, and, 10 μg/mL each of aprotinin and leupeptin) was added to the nuclear fraction (pellet). The suspension was vortexed, kept on ice for 30 minutes, and centrifuged at 13,000 g for 10 minutes. The supernatants (nuclear fraction) were normalized for total protein concentration using the BCA assay (Pierce et al. , "Novel inhibitors of cytokine-induced ΙκΒα phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo " J Biol Chem 272:21096-103, 1997).
[00166] 5 pmol of a DNA oligonucleotide containing the NF-κΒ consensus binding site (SEQ ID NO: l) (5 ' -AGTTGAGGGGACTTTCCC AGGC-3 '), 5 μΕ 5X forward reaction buffer, 10 units T4 polynucleotide kinase, 2.5 μΕ [32P]ATP (10 μα/μΕ, 300 Ci/mmol), and water (to 25μί total volume) were incubated for 10 minutes at 37°C. The reaction was stopped by heating the mixture for 10 min at 65°C and the labeled oligonucleotide was separated from unincorporated [32P]ATP by centrifuging the mixtures at 12,000- 16,000 g for 30 seconds in G-25 Sepharose columns. The labeled nucleic acid was recovered in the collection tube in approximately 25 μί of TE buffer.
[00167] For the binding assays, equal amounts of nuclear extracts (approx 2-5 μg of protein) were incubated on ice for 15 minutes with 4 μΕ 5X gel shift binding buffer (50 mM Tris HC1 pH 7.5, 250 mM KC1, 5 mM DTT, 1 mg/mL bovine serum albumin, 25% glycerol, H20 (volume adjusted to 5 mL)), 1.5 μg poly (dl-dC), 1 \xL 32P-labeled probe, and enough H20 to bring the total reaction volume to 20 μί. 2 μΕ 10X loading buffer (20% Glycerol, 0.1 M Na2EDTA pH 8, 0.25% bromphenol blue, 0.25% xylene cyanol) was added to each sample and then loaded onto a native 8% polyacrylamide gel. The gel was run in 0.5X TBE buffer for about 2 hours, dried, and subjected to autoradiography. The bands on the blots were quantified using the NIH Image program.
[00168] Activation of NF-κΒ was also determined using the TRANS-AM™ assay (Active Motif, Carlsbad, California) per the manufacturer's instructions. (Renard et al., "Development of a sensitive multi-well colorimetric assay for active NF-κΒ," Nucleic Acids Res 29, E21 , 2001). Briefly, cell monolayers on 60 mm dishes were washed with ice-cold PBS and removed by incubating in trypsin-PBS or scraping. The cells were centrifuged for 10 minutes at 1 ,000 rpm at 4°C and resuspended for 10 minutes in ΙΟΟμί of 4°C lysis buffer (20 mM HEPES (pH 7.5), 350 mM NaCl, 20% glycerol, 1% Igepal- CA630, 1 mM MgCl2, 0.5 mM EDTA, 0.1 mM EGTA, 1 μΐ, of 1 M dithiothreitol and 10 of protease inhibitor cocktail (proprietary) per mL of lysis buffer). The lysates were centrifuged for 20 minutes at 14,000 g at 4°C. Lysates containing 5 μg of total protein were added to 20 μΐ, of lysis buffer per well in 96-well dishes containing immobilized oligonucleotides corresponding to the NF-κΒ consensus DNA binding site (SEQ ID NO:2) (5 '-GGGACTTTCC-3 '). The plate was covered with an adhesive film and incubated for 1 hour at room temperature on a rocker. The wells were then washed three times with 200 μΕ per well of washing buffer (100 mM phosphate buffer (pH 7.5), 500 mM NaCl, 1% Tween). 100 μΐ, of p65 subunit monoclonal antibody (the p65 antibody supplied with the kit only recognizes p65 -containing NF-κΒ heterodimers that are bound to DNA containing the NF-KB consensus binding sequence) was diluted 1 : 1 ,000 in IX antibody binding buffer (4 mM HEPES (pH7.5), 120 mM KCl, 8% glycerol, 1% bovine serum albumin) and added to each well and incubated for 1 hour at room temperature. The wells were washed with 100 μΐ, of washing buffer three times. 100 μΐ, of horseradish peroxidase-conjugated secondary antibody diluted 1 : 1 ,000 in IX antibody binding buffer was added to each well and incubated for 1 hour at room temperature without agitation. The wells were washed four times with 200 μΐ, per well of IX washing buffer. 100 μΐ, of developing solution (tetramethylbenzidine in 1% DMSO) was added to each well and incubated for 10 minutes at room temperature. 100 μΐ, of stop solution (0.5M H2S04) was added to each well after which the absorbance was determined on a plate spectrophotometer at 450 nm. Specificity of binding was determined using 200-fold excess wildtype NF-κΒ oligonucleotides added at the time the cell lysates were added to the wells. Apoptosis Assay:
[00169] Single cell suspensions were plated at 100,000 cells per well in 24-well plates in media containing DMSO, BAY 11-7082, BAY 11-7085, or MG-132 for 8-24 hours in an incubator. Alternatively, DMSO, BAY 11-7082, BAY 11-7085, or MG-132 were added to cells that were confluent and adherent for several days. The nonadherent cells were aspirated off with the media and spun for 3 minutes at 2,000 rpm in 15 mL polypropylene tubes. The adherent cells were washed twice with PBS, dispersed in trypsin, and spun down for 3 minutes at 2,000 rpm in 15 mL polypropylene tubes. The nonadherent and adherent cells were resuspended in IX binding buffer (10 mM
HEPES/NaOH, pH 7.4, 140 mM NaCl, and 2.5 mM CaCl2) to a final concentration of 106 cells/mL. ΙΟΟμί of the cell solution were transferred to a 5 mL culture tube and incubated with 5 of annexin V-FITC-conjugated monoclonal antibody (Pharmingen) and 10 of propidium iodide (from a 50 μg/mL stock solution made in PBS) in the dark at 25°C for 15 minutes. 400 μΐ, of binding buffer was added to each tube and the cells were analyzed by flow cytometry immediately. The percent of apoptosis was determined as the percent of annexin V-positive, propidium iodide -negative cells of the total cells counted.
[00170] DLD-1 and HT-29 cells that were approximately 60-70% confluent were transduced with Ad-ΙκΒ super-repressor by adding 1-50 μί of purified virus to the cells. After 3 days, the expression of the ΙκΒ super-repressor was ascertained by immunoblot. In a parallel experiment, the transduced cells were resuspended and allowed to readhere in BAY 11-7085 for 8 h. The percentage of surviving (nonapoptotic) cells was determined using crystal violet staining.
Immunoblotting:
[00171] The cells were washed twice in ice cold PBS and then lysed in 4°C lysis buffer (50 mM HEPES, 150 mM NaCl, 1.5 mM MgCl2, 1 mM EGTA, 100 mM NaF, 10 mM Na2P04, 1 mM Na3V04, 10% glycerol, 1% TRITON™ X-100, and ^g/mL each of aprotinin, leupeptin, chymostatin, pepstatin) and clarified at 12,000 rpm at 4°C for 15 minutes. The lysates were normalized for total protein concentration. Aliquots of the lysates were then mixed 1 : 1 in sample buffer (125 mM Tris-HCl (pH 6.8), 20% glycerol, 4%) sodium dodecyl sulfate, 2% β-mercaptoethanol, 10 μg/mL bromophenol blue) and boiled for 3 minutes. The samples were loaded onto SDS-PAGE gels (7.5 or 10%> acrylamide). The proteins were transferred to nitrocellulose from the gels and incubated in blocking buffer (1% bovine serum albumin (Bio-Rad), 100 mM Tris-Cl (pH 7.4), 0.9% NaCl, 0.1% Nonidet) overnight at 4°C.
[00172] For immunodetection, the blocked blots were probed with primary antibody for 2 hours at 4°C on a rocker. The blots were washed twice for 10 minutes each in blocking buffer, then incubated with a 1 :2000 dilution of a rabbit-anti-mouse secondary antibody (for monoclonal primary antibodies; for polyclonal antibodies, this step was skipped). After two washes in blocking buffer (150 mM NaCl, 20 mM Tris pH 7.4, 0.5% Tween 20, 5% bovine serum albumin, and 0.2gm/500 mL final concentration of sodium azide), the blots were finally incubated with a 1 :2000 dilution of alkaline phosphatase-conjugated rabbit-anti-mouse antibodies (Jackson) in blocking buffer. Alkaline phosphatase was detected by a colorimetric reaction using a BCIP (5-bromo-4-chloro-3-indolyl
phosphate)/NBT (nitroblue tetrazolium) kit (Zymed).
Athymic mouse colon cancer xenograft studies [00173] HT-29 and HCT-116 cells were harvested in 0.25% trypsin-PBS-EDTA, washed once in media and PBS, and resuspended in PBS at 1 million cells per 200 μΐ,. One million cells were injected either subcutaneous ly in the backs or intraperitoneally in 5 week old female nu/nu athymic mice (Charles River Labs). For the mice with
subcutaneous tumors, the tumors were allowed to establish themselves for 10 days after which they were randomized to BAY 11-7085 or DMSO. For the mice with
intraperitoneal tumors, the mice received DMSO or BAY 11-7085 (5 mg/kg) 24 hours before the cancer cells were injected intraperitoneally. All mice received BAY 11-7085 (5 mg/kg) or an equal volume of DMSO (about 200 μΐ,) twice weekly for 21-32 days.
Subcutaneous tumor sizes were determined by measuring the length and width with calipers. These studies were approved by the University of Utah Institutional Review Board and Institutional Animal Care and Use Committee and performed in the Animal Resource Center. Mice were euthanized when they experienced a greater than 10% loss in body weight or if they appeared ill. Post-mortem examinations included sectioning of kidney, lung, and liver tissues which were stained with hematoxylin and eosin followed by examination for tissue toxicity/damage by an experienced mouse pathologist (E.J.E.) who was blinded to therapy. Results
NF-KB inhibitors diminish colon cancer cell proliferation
[00174] Two compounds, BAY 11-7082 and BAY 11-7085, known to inhibit NF-KB and arthritis in rodent models (Pierce et al, "Novel inhibitors of cytokine-induced ΙκΒα phosphorylation and endothelial cell adhesion molecule expression show antiinflammatory effects in vivo " J Biol Chem 272:21096-103, 1997) were tested on colon cancer cell lines. BAY 11-7082 and BAY 11-7085 are soluble compounds that inhibit ΙκΒ phosphorylation and TNFa-induced NF-κΒ activation (Id.). They were also found to activate JNK/SAPK and p38, members of the MAP kinase family of signal transduction proteins (Id.). Incubation of the colon cancer cell lines, DLD-1, HCT-116, HT-29, with 10 μΜ of BAY 11-7085, but not BAY 11-7082 greatly inhibited cell proliferation (FIGs. 1A and B).
[00175] In order to determine whether the differences in the inhibitory effects of BAY 11-7082 and BAY 11-7085 on colon cancer cell proliferation were due to differences in the activities of the two compounds, NF-κΒ electrophoretic mobility shift assays (EMS A) were performed. Following 3 and 6 hour incubations of HT-29 cells with the two compounds at 10 μΜ concentrations, a more profound inhibition of NF-κΒ activity occurred with BAY 11-7085 than BAY 11-7082 (FIG. 1C). Consistent with previous reports in a variety of cancer cells, HT-29 colon cancer cells demonstrated constitutive activation of NF-KB. DLD-1 and HCT-116 cells demonstrated constitutive activation of NF-KB by EMS A as well (data not shown).
[00176] When HT-29 colon cancer cells were treated with the NF-κΒ inhibitors, BAY 11-7085 or MG-132 for 24 h, the percentage of apoptotic cells (annexin V-positive and propidium iodide-negative) was relatively greater for MG-132 than BAY 11-7085, and was proportional to the number of floating cells. It was confirmed in HT-29 cells that were treated with increasing concentrations of BAY 11-7085 for 24 h. After separating the HT-29 cells that remained adherent and those that became nonadherent after BAY 11- 7085 treatment, the percentage of apoptotic cells was determined for each group. Whereas the majority of nonadherent cells were apoptotic, only a minority of the adherent cells was apoptotic. The percentage of nonadherent apoptotic cells decreased after treatment with 50 and 100 μΜ BAY 11-7085 compared with lower concentrations of BAY 11-7085, because many of the cells were positive for both annexin V and propridium iodide indicating that they had completed apoptosis and were now necrotic. Shown another way, HT-29 cells were treated with 20 μΜ MG-132 for 8 h, after which the adherent and nonadherent cells were collected separately, and then lysed and immunoblotted for cleaved PARP, a product of caspase cleavage. Whereas there was only a small increase in cleaved PARP in the cells that remained adherent, there was a large increase in cleaved PARP in the nonadherent cells. It has been shown that the loss of colon cancer cell adhesion was associated with the induction of apoptosis by IFN and TNFa. The inhibition of colon cancer cell adhesion appears to be related to the apoptotic effects of the NF-KB inhibitors.
[00177] In order to determine whether BAY 11 -7082 or BAY 11 -7085 caused significant direct cell toxicity, an LDH-based cell toxicity assay was performed on DLD- 1, HCT-116, and HT-29 cell lines. One to ten μΜ of BAY 11-7082 or BAY 11-7085 for 24 hours resulted in a maximum of 20% cell toxicity. Thus, direct cell toxicity due to BAY 11-7082 or BAY 11-7085 could not alone explain the decreased colon cancer cell proliferation.
NF-KB inhibitors inhibit colon cancer cell tumorigenicity
[00178] Anchorage-independent cell growth is a hallmark of cancer cells and is a good correlation to tumorigencity in vivo. Therefore, the effects of BAY 11-7082 and BAY 11- 7085 were examined on colon cancer cell growth using the soft agar colony formation assay. At 10 μΜ concentrations of BAY 11-7082 or BAY 11-7085 for 6 days, anchorage- independent proliferation of the DLD-1 and HT-29, but not HCT-116, colon cancer cell lines was significantly inhibited (FIG. 2A).
[00179] A recent study showed that inhibition of NF-κΒ in colon cancer cell xenografts in athymic mice by direct injection of adenovirus containing the ΙκΒ super-repressor mutant into the tumors alone failed to reduce the tumor size (Wang et al., "Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-κΒ," Nat Med 5:412-7, 1999). However, BAY 11-7082 or BAY 11-7085 alone inhibit colon cancer xenograft growth in athymic mice. HCT-116 and HT-29 cell lines were used to generate subcutaneous tumors in athymic female mice. The mice were subsequently randomized to be treated with vehicle (DMSO) alone or BAY 11-7085 1 mg/kg twice weekly for 28 days.
[00180] While HT-29 cells readily formed tumors in the athymic mice, HCT-116 cells formed only very small tumors in most cases. There was a statistically significant overall reduction in the tumor volumes in the HT-29 xenografts in the BAY 11-7085 treatment group compared with the control group (FIG. 2B). There was no statistically significant overall difference in the tumor volumes in the HCT-116 xenografts in the treatment or control groups (FIG. 2C).
NF-KB inhibitor-induced apoptosis is associated with a loss and inhibition of cell adhesion
[00181] Treatment of HT-29 cells with BAY 11 -7082 or BAY 11 -7085 at doses up to 20 μΜ for 24 hours caused an increase in the level of apoptosis as measured by the annexin V-FITC assay (FIG. 3A), which is sensitive for cells in the early stages of apoptosis. However, 20 μΜ of MG-132, another NF-κΒ inhibitor, caused a greater increase in apoptosis (FIG. 3A). Disclosed herein, the level of apoptosis correlated with the degree of loss of cell adhesion manifested by floating cells. HT-29 cells were treated with increasing concentrations of BAY 11-7085 for 8 hours, after which the percentage of apoptotic cells was determined separately for the cells that remained adherent and those that became non-adherent. While the non-adherent cells were predominantly apoptotic, only a minority of the adherent cells was apoptotic (FIG. 3B). Note that the percentage of apoptosis in the non-adherent cell fraction decreased at 50 and 100 μΜ concentrations of BAY 11-7085. This was due to the fact that after 8 hours of treatment, many of the cells at higher doses of BAY 11-7085 were annexin V-positive and failed to exclude propidium iodide meaning that they had completed apoptosis and were now necrotic. Similar results were obtained using other colon cancer cell lines, HCT-116 and MG-132. Following treatment of HCT-116 cells with 20 μΜ MG-132 for 24 hours there was a readily apparent population of cells that had lost adhesion. While there was only a small increase in cleaved PARP, a product of caspase cleavage and activation, in the cells that remained adherent, there was a large increase in cleaved PARP in the non-adherent cells (FIG. 3C). [00182] Pretreatment of adherent HT-29 cells for three days with various
concentrations of BAY 11-7085 followed by a one-hour cell adhesion assay demonstrated a significant and rapid inhibition of cell adhesion at high concentrations of BAY 11-7085 (FIG. 3D), suggesting that NF-κΒ promoted cell adhesion. Even though BAY 11-7085 induced a loss of cell adhesion, it was important to show whether HT-29 cells were actually susceptible to anoikis. When HT-29 cells were cultured in suspension for 24 hours on dishes coated with poly-HEMA, a substrate that completely inhibited HT-29 cell adhesion, only a minority (17%) of the floating cells were apoptotic by the annexin V assay. The fact that only a small fraction of suspended HT-29 cells were apoptotic, suggested that anoikis was not the principal mechanism responsible for the apoptotic activity of the NF-κΒ inhibitors. Loss of colon cancer adhesion activates NF-KB
[00183] Various concentrations of BAY 1 1-7085 were added to HT-29 cells that were transiently suspended in trypsin-PBS for approximately 15 minutes before replating. The transiently suspended cells were quickly allowed to readhere in order to prevent anoikis. Alternatively, BAY 11-7085 was added to adherent HT-29 cell monolayers. BAY-11- 7085 at all concentrations tested caused a significant increase in apoptosis of the transiently suspended HT-29 cells (FIG. 4A). On the other hand, even the highest doses of BAY 11-7085 caused only a relatively small degree of apoptosis of adherent compared with transiently suspended HT-29 cells (FIG. 5). Similar results were obtained with BAY 11-7082 at the same doses. [00184] Since a transient loss of cell adhesion greatly enhanced the apoptotic effect of BAY 11-7085, we hypothesized that activation of NF-κΒ may be affected by a loss of cell adhesion. To test this, DLD-1, HCT-116, and HT-29 cells were transiently suspended in trypsin-PBS and then quickly allowed to readhere. Immunofluorescent studies on the localization of the p65 NF-κΒ subunit revealed nuclear p65 in cells one hour after being transiently in suspension (FIG. 4B). This was followed by a decrease in nuclear p65 localization from 3-24 hours after replating. In fact, by 24 hours, little nuclear p65 staining was seen although much cytoplasmic p65 staining was present. Similar results were obtained with DLD-1 (FIG. 4C) and HCT-116 (FIG. 4D) colon cancer cell lines. These results demonstrated that transient suspension of colon cancer cells resulted in activation of NF-KB. [00185] An NF-κΒ binding assay was used to demonstrate the activation of NF-κΒ by transient suspension. Adherent monolayers of DLD-1, HCT-116, and HT-29 cells were treated with increasing concentrations of BAY 11-7085 for 8 hours were scraped off plates and subjected to an NF-κΒ activation assay. The scraped cells were lysed and added to 96-well plates containing immobilized oligonucleotides corresponding to the NF-KB consensus binding sequence (TRANS-AM™ assay). NF-κΒ binding to the immobilized oligonucleotides was detected using a p65 monoclonal primary antibody and a horseradish peroxidase-conjugated anti-mouse secondary antibody. The assay showed that there was relatively little activation of NF-κΒ compared to the positive control (HeLa cells treated with TNF ) (FIG. 5 A). However, when the transiently suspended cells were allowed to readhere, a large induction of NF-κΒ activation occurred (FIG. 5A). Thus it was readhesion rather than just transient suspension that was the stimulus causing the large increase in NF-κΒ activation.
[00186] The same NF-κΒ binding assay was used to determine whether NF-KB inhibitors could diminish the activation of NF-κΒ by readhesion. HT-29 cells transiently suspended with trypsin were mixed with increasing concentrations of BAY 11-7085 then placed on culture dishes for 8 hours. Alternatively, adherent HT-29 cells were treated with increasing concentrations of BAY 11-7085 for 8 hours. The cells were scraped, and all the cells were collected and lysed. The NF-κΒ binding assay confirmed the strong activation of NF-κΒ caused by the readhesion of transiently suspended HT-29 cells and demonstrated a dose-related inhibition of this NF-κΒ activation by BAY 11-7085 (FIG. 5B, transiently suspended cells). On the other hand, there was a steady and low level of constitutive NF-κΒ activation in the adherent HT-29 cells that was relatively unaffected by treatment with increasing concentrations of BAY 11-7085 (FIG. 5B, adherent cells). [00187] Since trypsin can activate protease-activated receptor-2 in colon cancer cell lines, which then results in increased cell proliferation, (Darmoul et al., "Initiation of human colon cancer cell proliferation by trypsin acting at protease-activated receptor-2," Br J Cancer 85:772-9, 2001). NF-κΒ activation was determined in adherent and confluent DLD-1, HCT-116, Caco-2 and HT-29 cells that were removed from plastic dishes by trypsin or scraping. Scraping of the cells did not result in NF-κΒ activation (FIG. 5C). In addition, cells that were transiently suspended in trypsin-PBS but not allowed to readhere, did not show significantly more activation of NF-κΒ by the TRANS-AM™ assay (FIG. 5C). To determine whether the transient activation of NF-κΒ caused by cell readhesion would render all of the colon cancer cells tested more susceptible to BAY 11-7085- induced apoptosis, DLD-1, HCT-116, Caco-2 and HT-29 colon cancer cell lines were transiently suspended, treated with BAY 11-7085 and allowed to readhere. While DLD-1 and HT-29 cells showed the same propensity for BAY 11-7085-induced apoptosis, Caco-2 cells showed a moderate increase, while HCT-116 cells did not (FIG. 5C).
[00188] The colon cancer cell lines used were chosen for differences in their origins. While DLD-1 and HT-29 cells carry mutated APC alleles, HCT-116 cells do not, (Groden et al, "Response of colon cancer cell lines to the introduction of APC, a colon- specific tumor suppressor gene," Cancer Res 55: 1531-9, 1995); however, HCT-116 cells carry activating mutations in the β-catenin gene (CTNNB1) (Ilyas et al, "β-catenin mutations in cell lines established from human colorectal cancers," Proc Natl Acad Sci USA 94: 10330- 4, 1997) whose gene product is normally regulated by the APC gene product (Munemitsu et al, "Regulation of intracellular β-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein," Proc Natl Acad Sci USA 92:3046-50, 1995; Rubinfeld et al., "The APC protein and E-cadherin form similar but independent complexes with a- catenin, β-catenin, and plakoglobin," J Biol Chem 270:5549-55, 1995). In addition, HCT- 116 cells do not express COX-2 while DLD-1 and HT-29 cells do (His et al., "Lack of cyclooxygenase-2 activity in HT-29 human colorectal carcinoma cells," Exp Cell Res 256:563-70, 2000; Shao et al, "Regulation of constitutive cyclooxygenase-2 expression in colon carcinoma cells," J Biol Chem 275:33951-6, 2000; Tsuji et al, "Evidences for involvement of cyclooxygenase-2 in proliferation of two gastrointestinal cancer cell lines," Prostaglandins Leukot Ess ent Fatty Acids 55:179-83, 1996). COX-2 is commonly overexpressed in colorectal tumors and plays roles in survival and metastasis in colorectal cancers.
[00189] COX-2 overexpression plays a role in the susceptibility of the colon cancer cells to BAY 11-7085-induced apoptosis. 293 cells transfected with a ponasterone- inducible COX-2 construct (293-COX-2) were treated with BAY 11-7085. Treatment of 293-COX-2 cells with 1 μg/mL of ponasterone for 48 hours led to a large induction of COX-2 protein, while the uninduced cells showed no measurable COX-2 protein expression by western blot (FIG. 5E) Uninduced 293-COX-2 cells showed no increase in apoptosis following treatment with BAY 11-7085 (FIG. 5F). However, ponasterone- induced 293-COX-2 cells demonstrated a marked increase in BAY 11-7085-induced apoptosis (FIG. 5E). This data indicated that differences in COX-2 expression can influence the ability of BAY 11-7085 to cause apoptosis.
[00190] Two other soluble NF-κΒ inhibitors, MG-132 and PDTC, were used to test their activity in the induction of apoptosis of colon cancer cells during readhesion.
Compared with BAY 11-7085, MG-132 more potently induced apoptosis of HT-29 colon cancer cells during readhesion. PDTC caused apoptosis of HT-29 cells during readhesion as well.
[00191] The specific involvement of NF-κΒ in the apoptotic mechanism of BAY 11- 7085 was examined specifically. To accomplish this, DLD-1 and HT-29 cells were transduced with an adenovirus containing the 1KB super-repressor construct to specifically inhibit NF-κΒ. The ΙκΒ super-repressor was created previously by mutating two key serine residues within the IkB gene resulting in an encoded ΙκΒ protein that is incapable of being targeted for ubiquitination and, hence, degradation by the proteasome. The transduced DLD-1 and HT-29 cells were then transiently suspended and allowed to readhere at concentrations of BAY 11-7085 below the in vitro apoptotic threshold (>20 μΜ) for these cell lines. Expression of the IkB super-repressor in DLD-1 and HT-29 cells significantly lowered the apoptotic threshold of BAY 11-7085 compared with the controls. The effect of the IKB super-repressor was much greater for DLD-1 than HT-29 cells because the former expressed higher levels of IKB super-repressor protein than the latter cells. Furthermore, the IKB super-repressor protein was functional in both cell lines.
Therefore, inhibition of NF-κΒ is important to the apoptotic effect of BAY 11-7085 during colon cancer cell readhesion.
NF-KB inhibitor prevents intraabdominal metastasis in vivo [00192] Since BAY 11-7085 induced apoptosis of readherent HT-29 cells, an in vivo model was used to test the ability of the drug to prevent metastasis by seeding of intraabdominal tissue with colon cancer cells. During colorectal cancer surgery, seeding of the peritoneal cavity by tumor cells with tumor cell implantation of the peritoneal surfaces can occur. Seeding results from the transient suspension of cancer cells, through a loss of adhesion either naturally ro because of surgical displacement, followed by readhesion to other tissues. The HT-29 and HCT-116 colon cancer cell lines were injected into the abdominal cavities of athymic mice that had been pretreated 24 hours earlier with either intraperitoneal vehicle alone (DMSO) or BAY 11-7085 (1 mg/kg). The mice were then treated twice weekly with vehicle or BAY 11-7085 for a total of 21 days. Mice sacrificed 7 days after the introduction of colon cancer cells intraabdominally showed no evidence of tumoral implantation of the parietal or visceral peritoneal surfaces.
[00193] After 21 days, there was clear evidence of metastases involving the parietal and visceral peritoneum of mice injected with HCT-116 cells regardless of whether they had received the vehicle or BAY 11-7085 (FIG. 6A and B). However, of the mice injected intraabdominally with HT-29 cells and treated with BAY 1 1-7085, only 2 of 6 mice demonstrated parietal peritoneal metastases and none of the 6 showed any evidence of hepatic metastases (Table 3).
TABLE 3:
Peritoneal and liver metastases following intraperitoneal injections of HT-29 cells into athymic mice.
Figure imgf000061_0001
[00194] Of the mice that had been injected intraabdominally with HT-29 cells and treated with vehicle, 5 of 6 developed metastases of the parietal peritoneum and all 6 developed hepatic metastases (FIG. 6C and Table 3). Interestingly, the majority of the metastases involved ventral and dependent areas of parietal and visceral peritoneum, suggesting that the colon cancer cells took some time to implant.
NF-KB inhibitors decrease expression of anti-apoptotic proteins
[00195] Inhibition of NF-κΒ increased the susceptibility of cancer cells to TNFa- induced apoptosis in other studies (Han et al, "Activation of NF-κΒ determines the sensitivity of human colon cancer cells to TNFa-induced apoptosis," Biol Pharm Bull 23:420-6, 2000; Wang et al., "Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-κΒ," Nat Med 5:412-7, 1999). Furthermore, TNFa is expressed by a number of colon cancer cell lines including HT-29 cells (Jung et al., "A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion," J Clin Invest 95:55-65, 1995). In order to explore the possibility that BAY 11-7082 and BAY 11-7085 can be sensitizing colon cancer cells to TNFa-induced apoptosis, HT-29 cells were pretreated with a monoclonal antibody, cA2, which inhibits TNFa binding to TNFa receptors (D'Haens et al., "Endoscopic and histological healing with infliximab anti-tumor necrosis factor antibodies in Crohn's disease: A European multicenter trial," Gastroenterology 116: 1029- 34, 1999) followed by treatment with BAY 11-7085. A previous study demonstrated TNFa-induced activation of NF-κΒ in HT-29 cells (Yamamoto et al., "Sulindac inhibits activation of the NF-κΒ pathway," J Biol Chem 274:27307-14, 1999). In order to ascertain whether cA2 alone had any measurable effects on colon cancer cells, DLD-1, HCT-116, and HT-29 colon cancer cells were treated with various concentrations of cA2 in a cell proliferation assay. Interestingly, cA2, at 10 and 50 μg/mL, caused a significant inhibition of colon cancer cell proliferation after several days of treatment (FIG. 7A). However, pretreatment of transiently suspended HT-29 cells with 50 μg/mL of cA2 for 48 hours did not diminish BAY 11-7085-induced apoptosis of adherent (FIG. 7B). This indicated that endogenous TNFa was not necessary for BAY 11-7085-induced apoptosis.
[00196] NF-KB regulates the expression of a number of genes, including c-IAP-1, c- IAP-2, TRAF-1, and TRAF-2, that encode proteins that mediate cell survival (Stehlik et al., "Nuclear factor (NF)-KB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha- induced apoptosis," J Exp Med
188:211-6, 1998; Wang et al, "NF-κΒ antiapoptosis: induction of TRAF1 and TRAF2 and c-IAPl and c- IAP2 to suppress caspase-8 activation," Science 281 : 1680-3, 1998; Wu et al, "IEX-1L, an apoptosis inhibitor involved in NF-κΒ -mediated cell survival," Science 281 :998-1001, 1998). No decrease in c-IAP-1 protein expression occurred when transiently suspended or adherent HT-29 cells were treated with 20 μΜ of BAY 11-7085 for 8 hours (FIG. 7C). HT-29 cells did not express detectable levels of c-IAP-2 by Western blots. The same treatment of HT-29 cells did result in a decrease in the protein expression of both TRAF-1 and TRAF-2 proteins (FIG. 7C). [00197] Recently, the anti-apoptotic protein, FLIP, which inhibits TNFa-mediated apoptosis, was found to be regulated by NF-κΒ and overexpressed in a variety of cancers. (Elnemr et al., "Human pancreatic cancer cells disable function of Fas receptors at several levels in Fas signal transduction pathway," Int J Oncol 18:311-6, 2001 ; Irmler et al. , "Inhibition of death receptor signals by cellular FLIP," Nature 388: 190-5, 1997; Kreuz et al, "NF-KB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling," Mo I Cell Biol 21 :3964-73, 2001; Ryu et al., "Increased expression of cFLIP(L) in colonic adenocarcinoma," J Pathol 194: 15-9, 2001; Tepper et al.,
"Modulation of caspase-8 and FLICE-inhibitory protein expression as a potential mechanism of Epstein-Barr virus tumorigenesis in Burkitt's lymphoma," Blood 94: 1727- 37, 1999). Interestingly, FLIP expression is upregulated by cell adhesion in endothelial cells and plays a role in inhibiting anoikis (Aoudjit and Vuori, "Matrix attachment regulates Fas-induced apoptosis in endothelial cells: a role for c-flip and implications for anoikis," J Cell Biol 152:633-43, 2001). As disclosed herein, FLIP was expressed in the DLD-1 and HT-29, but not the HCT-116, cell lines. 20 μΜ of BAY 11-7085 caused only a slight diminution in the expression of the 54 kilodalton (long) isoform of FLIP in adherent, but not transiently suspended, HT-29 cells (FIG. 7C). Although decreased FLIP expression could be involved in BAY 11-7085-induced apoptosis of HT-29 cells, it would not explain BAY 11-7085-induced apoptosis in DLD-1 cells since they do not express FLIP (FIG. 7D).
[00198] Treatment of adherent colon cancer cells with BAY 11-7082, BAY 11-7085, or MG-132 inhibited colon cancer cell proliferation, tumorigenicity, and adhesion, and induced apoptosis. These effects were not uniform amongst the selected colon cancer cell lines used. While the cell proliferation of all three cell lines were readily inhibited by BAY 11-7085, reduced tumorigenicity and apoptosis induction by treatment with BAY 11-7082 and BAY 11-7085 was only achieved in the DLD-1 and HT-29 cell lines. Results obtained using ponasterone-inducible COX-2 transgene expression in 293 cells
demonstrated a likely role for COX-2 protein expression or overexpression in the apoptotic response to BAY 11-7085.
[00199] While cells that became non-adherent following treatment with NF-KB inhibitors were apoptotic, those cells that remained adherent were not. In addition, pretreatment of HT-29 cells with BAY 11-7085 caused a significant inhibition of cell adhesion at higher doses. The proapoptotic effect of BAY 11-7085 was greater when it was added to transiently suspended HT-29 and DLD-1 cells {i.e., just before plating) versus adherent cells. These results indicated that transient suspension of colon cancer cells increased the susceptibility of the cells to apoptosis due to NF-κΒ inhibition.
[00200] When adherent DLD-1, HCT-116, and HT-29 cells were scraped off of plastic dishes and assayed for NF-κΒ activation, the level of NF-κΒ DNA binding was low relative to the positive control (HeLa cells stimulated with TNFa). When transiently suspended DLD-1, HCT-116, and HT-29 were allowed to readhere, there was a rapid and large activation (similar to the positive control) and increased nuclear localization of NF- KB. Thus, it was the readhesion of the transiently suspended colon cancer cells that activated NF-κΒ. This large activation of NF-κΒ induced by the readhesion of transiently suspended colon cancer cells was rapid and almost totally inhibited by treatment with as little as 10 μΜ BAY 11-7085. On the other hand, adherent colon cancer cells
demonstrated a low but constitutive level of NF-κΒ activation, which could not be inhibited with doses of BAY 11-7085 up to 50 μΜ.
[00201] It appears that NF-κΒ inhibitors cause apoptosis of colon cancer cells in a two- step process. First, the NF-κΒ inhibitors inhibit cell adhesion of adherent colon cancer cells in vitro. The readhesion of these floating cells causes a large and transient activation of NF-KB. This renders the readherent cells exquisitely susceptible to NF-κΒ inhibitor- induced apoptosis. The fact that the vast majority of transiently suspended HT-29 and DLD-1 cells, which were allowed to readhere, became apoptotic following treatment with BAY 11 -7082 or BAY 11-7085, indicated that NF-κΒ is an important survival factor for certain cancer cells, such as colon cancer cells, during the process of readhesion, particularly cancer cells that express COX-2 and/or mutant APC genes.
[00202] As metastatic cancers must transiently suspend to pass through the circulatory system and then readhere to be invasive, these results indicate that the compositions disclosed herein can be used for the prevention of metastasis. The process of metastasis has been proposed to involve a number of sequential steps: invasion, dissociation, intavasation into the circulatory or lymphatic systems, dissemination, arrest in the microcirculation, extravasation, and invasion of distant tissues (Engers et ah,
"Mechanisms of tumor metastasis: cell biological aspects and clinical implications," J Cancer Res Clin Oncol 126:682-92, 2000). Each of these steps involves dynamic changes in cell adhesion including a complete absence of adhesion during dissemination. Although circulating cancer cells in humans can be abundantly found in venous blood samples of advanced cancer patients (Mehes et al, "Circulating breast cancer cells are frequently apoptotic," Am J Pathol 159: 17-20, 2001). Fortunately, the process of metastasis is surprisingly inefficient with as few as 0.05% of circulating tumor cells producing stable metastases (Liotta et al., "Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation," Cancer Res 34:997-1004, 1974; Nicolson, "Gene expression, cellular diversification and tumor progression to the metastatic phenotype," Bioessays 13:337-42, 1991; Weiss, "Metastatic inefficiency," In Liver Metastasis, L. Weiss, and H. Gilbert, eds. (Boston, Hall), pp. 126-157, 1985). Although many circulating tumor cells become arrested in the microcirculation, the majority of these cells remain viable and capable of extravasation in vivo (Chambers et al, "Steps in tumor metastasis: new concepts from intravital videomicroscopy," Cancer Metastasis Rev 14:279-301, 1995). Thus, the rate limiting step in metastasis is the colonization of distant tissues by extravasated tumor cells (Id.). [00203] While the five-year survival rate for early stage colorectal cancers (TNM I-II) without metastases is greater than 80%, for late-staged cancers (TNM III-IV) with metastases it is less than 50%.(Boland, "Malignant Tumors of the Colon." In Textbook of Gastroenterology, T. Yamada, D. H. Alpers, L. Laine, C. Owyang, and D. W. Powell, eds. Philadelphia, Lippincott Williams and Wilkins, pp. 2023-2082, 1999). Most newly diagnosed colorectal cancers are TNM stage III-IV, which means that most colorectal cancer patients already have metastasis of the primary tumor by the time of presentation. In fact, micrometastasis to lymph nodes was detected by RT-PCR of carcinoembryonic antigen in 54% of patients deemed to be TNM stage II by current staging methods (Liefers et al., "Micrometastases and survival in stage II colorectal cancer, N EnglJ Med 339:223- 8, 1998). In this study, the five-year survival of the patients with lymph node
micrometases was 50% versus 91% for those without micrometases. This means that the stage II patients with micrometastases behaved clinically more like TMN stage III patients with regards to survival. Thus, a greater proportion of newly diagnosed colorectal cancers already have metastases than previously thought. Indeed, breast cancer cells in the blood of patients with advanced breast cancer were found as frequently as 1 per 1000
mononuclear cells (Mehes et al, "Circulating breast cancer cells are frequently apoptotic," Am J Pathol 159: 17-20, 2001). Although the majority of these cells were apoptotic, a minority were not, demonstrating that a significant number of cancer cells invade into the circulatory system and survive.
Example 2:
Materials:
[00204] BAY 11-7085 (Biomol), anisomycin, ciglitazone (Cayman), pioglitazone (Cayman), troglitazone (Cayman), rosiglitazone (Cayman), GW9662 (Cayman), MC-555 (Cayman) were solubilized in DMSO. Cleaved PARP pAb (New England Biolabs), FLIP (Santa Cruz), and actin mAb (Lab Vision) were used for the Western blots. D-Luciferin was purchased from Xenogen. Ketamine was purchased from the University of Utah Pharmacy.
Cell Cultures and Transfection:
[00205] BxPc-3, Su.86.86, and PL-45 pancreatic and HT-29 colon cancer cell lines were purchased from ATCC and cultured in DMEM supplemented with 10% fetal bovine serum, glutamine, penicillin, and streptomycin. All cells were cultured at 37° C in a humidified incubator in a 5% C02 atmosphere.
[00206] The cancer cell lines were stably transfected with pLuc-puro, a constitutive luciferase reporter (a kind gift from Dr. Stephen Lessnick, University of Utah) and selected in puromycin-containing media.
Cell survival and readhesion assay:
[00207] Pancreatic cancer cell cultures (BxPc-3 and Su.86.86) and colon cancer (HT- 29) cells were transiently suspended and 105 cells were transferred to 96-well plates and treated in vitro with varying concentrations of BAY 11-7085, thiazolidinediones, or DMSO for eight hours. To gently remove nonadherent cells, the wells were carefully washed from the wells twice with 100 μΙ,ΛνεΙΙ of blocking buffer (1% BSA in PBS) and twice again with ice-cold PBS. It was previously shown that the floating cells are apoptotic (Scaife et ah, "Nuclear factor kappaB inhibitors induce adhesion-dependent colon cancer apoptosis: implications for metastasis," Cancer Res 62(23):6870-8, 2002). Attached surviving cells were fixed with 100 μΕΛνεΙΙ of 100% ice-cold methanol for 10 minutes, and then allowed to air dry. Cells were stained with 100 μΙ,ΛνεΙΙ of 0.1% crystal violet for 10 minutes then gently washed four times with 200 μίΛνεΙΙ distilled H20 and four times with PBS. The plates were then completely air-dried. The stained cells were then solubilized in 100 μίΛνεΙΙ of 1% sodium deoxycholate for 10-60 minutes at room temperature. Plates were read at 590 nm in a spectrophotometer with the background reading from blank wells subtracted. The absorption at 590 nm is proportional to the number of surviving treated cells {Id .
NF-KB Assay:
[00208] Pancreatic cancer (BxPc-3, Su.86.86, and PL-45) and colon cancer (HT-29) cell cultures were transiently suspended and treated in vitro with varying concentrations of BAY 11-7085 or rosiglitazone for 4 hours. Nuclear lysates were generated, normalized for total protein concentration, and used to determine NF-κΒ activity in an ELISA kit (Panomics) per the manufacturer's instructions.
Apoptosis assay: [00209] BxPc-3, PL-45, Su.86.86 and HT-29 cells were transiently suspended and treated in vitro with varying concentrations of BAY 11-7085 or DMSO for four to eight hours. Adherent and nonadherent treated cells were collected, lysates prepared, and protein concentrations normalized prior to Western Blot analysis using the Pierce BCA protein assay (Pierce, Rockford, 111.). The degree of apoptosis was determined by western blotting for cleaved PARP.
Athymic Mouse Pancreatic Cancer Xenograft Studies:
[00210] The animal studies were approved by and performed in accordance with the University of Utah Institutional Animal Care and Use Committee. Athymic 5-week-old female nu/nu mice were randomized to intraperitoneal (IP) treatment with either BAY 11- 7085 (5 mg/kg) or an equal volume of vehicle (DMSO) 24 hours prior to IP injection of 106 suspended Su.86.86, BxPc-3, or HT-29 cells. The mice continued to receive DMSO or BAY 11-7085 (5 mg/kg) IP every 3 days for 9 days, or, DMSO and rosiglitazone (5 mg/kg) IP every day for 9 days. A mixture of 10 of firefly D-luciferin (15 mg/mL)
(Xenogen), 200 μΐ, ketamine HC1, and 20 μΐ, xylazine (100 mg/mL) was injected IP just prior to bioluminescent imaging in order to anesthetize the mice and provide substrate for the luciferase expressing cancer cells. Prior to necropsy, the mice were euthanized by an overdose of inhaled enflurane followed by cervical dislocation. The abdominal cavity was exposed surgically and the intraabdominal tumor implants imaged with the bioluminescent imaging system. The tumor implants for each mouse was verified with a dissecting zoom microscope.
Results:
[00211] It was previously shown that readhesion of transiently suspended colon cancer cells in vitro resulted in transient (approximately 3-8 hours) and large increases (>10-fold) in NF-KB activity (Scaife et al., "Nuclear factor kappaB inhibitors induce adhesion- dependent colon cancer apoptosis: implications for metastasis," Cancer Res 62(23):6870- 8, 2002). The transient activation of NF-κΒ was also observed during the readhesion of multiple pancreatic cancer cell lines and was inhibited by the NF-κΒ inhibitor BAY 11- 7085 (FIG. 9).
[00212] Treatment of pancreatic and colon cancer cells with 10-20 μΜ BAY 11-7085 during cell readhesion increased apoptosis (cleaved PARP levels) (FIG. 10A). Since the induction of apoptosis of cancer cells during cell readhesion results in cell detachment (not shown), the number of readherent cells that survived treatment (FIG. 10B) was inversely proportional to the degree of apoptosis.
[00213] The detection of intraperitoneal cancer cells portends a very poor prognosis as they are typically resistant to standard therapy once they implant in the peritoneum (Vogel et al, "Detection and prognostic impact of disseminated tumor cells in pancreatic carcinoma," Pancreatology 2(2):79-88, 2002). Intra-abdominal cancer cells can seed the peritoneal cavity and do so by adhering to the peritoneum, a tough connective tissue lining the abdominal cavity and organs. Since BAY 11-7085 was able to induce apoptosis of readhering pancreatic cancer cells in vitro, the ability of BAY 11-7085 to inhibit intraabdominal seeding of human pancreatic cancer cell lines was tested in an athymic mouse model of peritoneal carcinomatosis. Su.86 and BxPc-3 pancreatic cancer cell lines were selected for these experiments on the basis of their differing sensitivities to BAY 11-7805 in vitro (FIG. 10B). [00214] Athymic mice were pretreated with vehicle (DMSO) or 5 mg/kg of BAY 11- 7085 (6 mice per arm). Twenty- four hours later, 106 transiently suspended Su.86 or BxPc- 3 pancreatic cancer cells were delivered into the intra-abdominal cavity by intraperitoneal (IP) injection. The mice continued to receive BAY 11-7085 or vehicle every 3 days for 9 days (4 doses total). The cell lines used were stably transfected with a firefly luciferase construct in order to allow visualization of small tumor implants in anesthetized mice using a bioluminescent imager. Small immoveable intra-abominal tumor implants were readily visualized as early as 3-4 days following intraperitoneal delivery of the cell lines (not shown). Nine days after the IP delivery of the pancreatic cancer cells, the mice were injected with luciferin and then euthanized. The abdominal cavities of the mice were rapidly exposed and the tumor implants visualized and imaged with the bioluminescent imager (FIGs. 11 A-D). Bioluminescent detection during necropsies aided the detection of tiny peritoneal tumor implants (2-3 mm) that were only otherwise visible with a zoom dissecting microscope. Tumor implants were observed on the parietal and visceral peritoneum with a predilection for the surface of the liver compared with the intestines or spleen (FIGs. 11 A-B). There was no evidence of metastases within the liver, lungs, or spleen of the mice (not shown) indicating that hematogenous spread had not occurred. For Su.86 pancreatic cancer cells, 100% of the control mice demonstrated tumor implants within the abdominal cavity and on the liver, while 33% and 0% (p<0.05) of BAY 11- 7085-treated mice showed intra-abdominal or hepatic tumor implants, respectively (Table 4). For BxPc-3 pancreatic cancer cells, 100% and 67% of the control mice developed intraabdominal and liver tumor implants, respectively, while 71% and 29% (p<0.05) of the BAY 11 -7085-treated mice developed intraabdominal and liver tumor implants, respectively (Table 4). None of the mice died or showed overt signs of toxicity. These results showed that BAY 11-7085 reduced peritoneal seeding by pancreatic cancer cells.
TABLE 4:
Figure imgf000069_0001
[00215] Other NF-κΒ inhibitors such as bortezomib and infliximab failed to induced apoptosis of colon and pancreatic cancer cells during readhesion (not shown) suggesting that BAY 11-7085 was not acting through NF-κΒ inhibition alone. Thus, the mechanism of action of BAY 11-7085 was examined in more detail. Although BAY 11-7085 and the closely related compound BAY 11-7082 are known to inhibit NF-κΒ, these compounds have been reported to activate TNK, a member of the Mitogen Activated Protein Kinase (MAPK) family as well. Exposure of cells to TNFa can result in either cell survival or apoptosis and JNK was recently shown to have a pivotal role in this decision. TNK activation results in the degradation of FLIP, an important inhibitor of caspase 8 and permits death receptor signaling-induced caspase 8 activation. Thus, the roles of TNK and FLIP in BAY 11-7085-induced apoptosis were examined.
[00216] BAY 11-7085 or anisomycin, a known activator of JNK, readily induced JNK activity (phosphorylation of c-JUN) in colon and pancreatic cancer cell lines (FIG. 12A). Furthermore, the drug-induced activation of TNK was inhibited by increasing
concentrations of the TNK- specific inhibitor SP600125, demonstrating that JNK specifically mediated the phosphorylation of c-JUN in these experiments (FIG. 12A).
[00217] Treatment of readhering HT-29 colon cancer cells, which express FLIP (FIG. 13 A), with BAY 11-7085 caused the down-regulation of FLIP after only 3 hours (FIG. 13B). This demonstrated that BAY 11-7085 can induce the down-regulation of FLIP and within the time frame that apoptosis occurs. Since HCT-116 colon cancer cells have relatively low levels of FLIP compared with other cancer cell lines (FIG. 13 A), they were transfected with human c-FLIP. HCT-116 cells that were transfected with human c-FLIP or empty vector were transiently suspended and allowed to readhere in the presence of BAY 11-7085 for 8 hours. While BAY 11-7085 induced apoptosis (Annexin V-FITC assay) in the control cells, the cells over-expressing FLIP were much more resistant to
BAY 11-7085 (FIG. 13B). Thus, FLIP appears to be an important target of BAY 11-7085 and is able to inhibit BAY 11-7085-mediated apoptosis during colon cancer cell readhesion.
[00218] In order to determine the role of TNK activity in FLIP downregulation, pancreatic and colon cancer cell lines were treated with anisomycin for 1.5 and 4 hours. Anisomycin caused a decrease in FLIP expression in pancreatic and colon cell lines tested (FIG. 14A). Although not as effective as BAY 11-7085, anisomycin induced apoptosis in colon and pancreatic cancer cell lines during cell readhesion (FIG. 14B). Anisomycin increased the pro-apoptotic activity of BAY 1 1-7085 on readhering colon and pancreatic cancer cells (FIG. 14C). These results showed that JNK activation is important and contributes to the pro-apoptotic activity of BAY 11-7085 during cancer cell readhesion.
[00219] However, it was previously shown that the TNFa receptor itself did not mediate NF-κΒ activation during colon cancer cell readhesion (Scaife et al, "Nuclear factor kappaB inhibitors induce adhesion-dependent colon cancer apoptosis: implications for metastasis," Cancer Res 62(23):6870-8, 2002). Of other members of the TNF receptor superfamily, Receptor Activator of NF-κΒ (RANK) is highly expressed in the digestive tract. Most of the colon cancer cell lines tested showed expression of RANK and its ligand, RANKL (FIG. 15 A). Incubation of colon cancer cell lines with soluble RANK, which inhibits ligand-dependent RANK activation, significantly reduced the activation of NF-KB (FIG. 15B). Thus, RANK appears to be important in the activation of NF-KB during colon cancer cell readhesion.
[00220] These studies indicate that the dual activity (inhibition of NF-κΒ and activation of JNK) of BAY 11-7085 was responsible for its pro-apoptotic activity in readhering cancer cells. Studies have shown that thiazolidinediones inhibit NF-κΒ and activate JNK in cancer cell lines (Bae et al, "Critical role of c-Jun N-terminal protein kinase activation in troglitazone -induced apoptosis of human HepG2 hepatoma cells," Mol Pharmacol 63(2):401-8, 2003; Yin et al, "Signaling pathways involved in induction of GADD45 gene expression and apoptosis by troglitazone in human MCF-7 breast carcinoma cells," Oncogene 23(26):4614-23, 2004) and inhibit NF-κΒ (Ghanim et al, "Suppression of nuclear factor-kappaB and stimulation of inhibitor kappaB by troglitazone: evidence for an anti-inflammatory effect and a potential antiatherosclerotic effect in the obese," J Clin Endocrinol Metab 86(3): 1306-12, 2001 ; Mohanty et al, "Evidence for a potent antiinflammatory effect of rosiglitazone," J Clin Endocrinol Metab 89(6):2728-35, 2004) and sensitize cells to death receptor induced apoptosis by down-regulating c-FLIP expression (Schultze et al, "Troglitazone sensitizes tumor cells to TRAIL-induced apoptosis via down-regulation of FLIP and Survivin," Apoptosis 11(9): 1503-12, 2006;
Akasaki et al, "A peroxisome proliferator-activated receptor-gamma agonist, troglitazone, facilitates caspase-8 and -9 activities by increasing the enzymatic activity of protein- tyrosine phosphatase- IB on human glioma cells," J Biol Chem 281(10):6165-74, 2006) and troglitazone was shown to sensitize tumor cells to TRAIL-induced apoptosis via down-regulation of FLIP. Thus, the ability of thiazolidinediones to induce the apoptosis of readhering colon and pancreatic cancer cells was tested. HT-29 cells were allowed to readhere in the presence of vehicle (DMSO) or increasing concentrations of rosiglitazone. Transiently suspended colon and pancreatic cancer cells that were allowed to readhere to plates for 3 hours showed a marked increase in NF-κΒ activity (FIG. 15 A). Rosiglitazone at 30 μΜ significantly inhibited readhesion-induced NF-κΒ activation within 3 hours (FIG. 15 A) albeit to a lesser degree than BAY 11-7085 (FIG. 9). HT-29 cells that were allowed to readhere in the presence of 30 μΜ rosiglitazone, ciglitazone, or troglitazone for 8 hours exhibited increased apoptosis, demonstrated by increased cleaved PARP levels (FIG. 15B).
[00221] Thirty μΜ rosiglitazone caused decreased FLIP expression in readhering BxPc3 and HT-29 cancer cells, but not Su.86 pancreatic cancer cells (FIG. 15C). BxPc3, Su.86, and HT-29 cells were transiently suspended and allowed to readhere in the presence of various concentrations of rosiglitazone, pioglitazone, troglitazone, ciglitazone, and MCC-555 (a potent PPARy agonist). All three cell lines showed decreased survival in the presence of various thiazolidinediones, but not to MCC-555 (FIGs. 15D-G), suggesting the lack of involvement of PPARy. While rosiglitazone decreased the survival of readhering BxPc3 and HT-29 cells in a concentration dependent fashion, Su.86 cells were resistant to rosiglitazone (FIGs. 15D-G). Thus, the ability of rosiglitazone to cause downregulation of FLIP correlated with its ability to diminish the survival of readhering colon and pancreatic cancer cell lines.
[00222] In order to determine if the cell-type specific effects of the thiazolidinediones were due to differing PPARy agonist activity, HT-29 and Su.86 cells, which overexpress and lack PPARy expression, respectively (Yang and Frucht, "Activation of the PPAR pathway induces apoptosis and COX-2 inhibition in HT-29 human colon cancer cells," Carcinogenesis 22(9): 1379-83, 2001; Galli et al., "Antidiabetic thiazolidinediones inhibit invasiveness of pancreatic cancer cells via PPARgamma independent mechanisms," Gut 53(11): 1688-97, 2004), were treated with ciglitazone in the presence or absence of the irreversible PPARy inhibitor GW9662 (Weber et al., "PPARy is not required for the inhibitory actions of PGJ2 on cytokine signaling in pancreatic beta-cells," Am J Physiol Endocrinol Metab 286(3):E329-36, 2004). Ciglitazone decreased the survival of both cell lines in a dose-dependent fashion (FIGs. 16A-B). However, the addition of 30 μΜ GW 9662 did not significantly affect the efficacy of ciglitazone in these assays (FIGs. 16A-B). These data suggest that PPARy agonist activity is not required for the pro-apoptotic activity of the thiazolidinediones during cancer cell readhesion.
[00223] The ability of rosiglitazone to inhibit intra-abdominal seeding of BxPc-3, HT- 29, and Su.86 cancer cells was tested. Athymic mice (6 mice in each arm) received 5 mg/kg of rosiglitazone or vehicle (DMSO) IP. Twenty-four hours later, 106 BxPc-3, HT- 29, and Su.86 cancer cells were delivered IP. Rosiglitazone (5 mg/kg) was given IP daily thereafter for 8 more days. For BxPC-3 pancreatic cancer cells, 83% and 50% of the control mice, but only 33% and 17% of rosiglitazone -treated mice developed
intraabdominal and hepatic tumor implants, respectively (Table 5). Following IP injection of athymic mice with HT-29 colon cancer cells, treatment with rosiglitazone resulted in a 50% reduction in intraabdominal tumor implants compared with vehicle (Table 5). For Su.86 pancreatic cancer cells, 83% and 67% of control mice, and 83% and 83% of rosiglitazone-treated mice developed intraabdominal and hepatic tumor implants, respectively (Table 5). None of the mice died or showed signs of overt toxicity.
TABLE 5:
Figure imgf000073_0001
[00224] Peritoneal carcinomatosis is a fatal disorder with limited treatment options (Fujiwara, "Intraperitoneal chemotherapy and intraperitoneal washing cytology in management of ovarian cancer," Gan To Kagaku Ryoho 27 Suppl 2:354-8, 2000; Kanellos et al., "Incidence and prognostic value of positive peritoneal cytology in colorectal cancer," Dis Colon Rectum 46(4):535-9, 2003; Nakatsuka et al., "Positive washing cytology in patients with pancreatic cancer indicates a contraindication of
pancreatectomy," Int J Surg Investig 1(4):311-7, 1999; Santala et al., "Peritoneal cytology and preoperative serum CA 125 level are important prognostic indicators of overall survival in advanced endometrial cancer," Anticancer Res 23(3C):3097-103, 2003;
Terauchi et al., "Combination chemotherapy with paclitaxel and intraperitoneal cisplatin for ovarian cancer with disseminated lesions in the peritoneum and the diaphragm," IntJ Clin Oncol 7(6):356-60, 2002; Yachida et al, "Implications of peritoneal washing cytology in patients with potentially resectable pancreatic cancer," Br J Surg 89(5):573-8, 2002). In this study, it is demonstrated that BAY 11-7085 and thiazolidinediones induce adhesion-dependent pancreatic and colon cancer cell apoptosis in vitro and that this has a biological consequence on intra-abdominal tumor seeding in vivo. Both of these drugs inhibit NFKB (Scaife et al., "Nuclear factor kappaB inhibitors induce adhesion-dependent colon cancer apoptosis: implications for metastasis," Cancer Res 62(23):6870-8, 2002; Su et al., "A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response," J Clin Invest 104(4):383-9, 1999) and the strong dependence of digestive tract cancer cells on NF-κΒ activity for survival during readhesion provides a unique therapeutic target for inhibiting intra-abdominal seeding and metastasis. [00225] It has been shown that transiently suspended colon cancer cells exhibit a strong and transient (3-8 hours) activation of NF-κΒ during cell readhesion. It is described herein that now treatment of readherent pancreatic as well as colon cancer cells with the BAY 11-7085 and thiazolidinediones inhibit the adhesion-induced NF-κΒ activity and cause apoptosis. The ability to induce apoptosis in readhering cancer cells is not universal to all NF-KB inhibitors that were tested. For example infliximab, an TNFr antagonistic antibody, and, bortezomib, a 26S proteasome inhibitor that indirectly inhibits NF-κΒ by preventing the degradation of ΙκΒ (Voorhees and Orlowski, "The proteasome and proteasome inhibitors in cancer therapy," Annu Rev Pharmacol Toxicol 46: 189-213, 2006), failed to induce apoptosis of readhering HT-29 colon cancer cells. [00226] JNK antagonizes NF-KB-mediated cell survival signaling by causing the down- regulation of FLIP, an important inhibitor of caspase 8 activity. BAY 11-7085 and thiazolidinediones activate JNK and cause the rapid downregulation of FLIP protein in colon and pancreatic cancer cells. This is not likely due to their inhibitory activity of the compounds on NF-κΒ, which regulates the transcription of CFLAR, the gene encoding FLIP. The ability of anisomycin, a JNK agonist, to cause down-regulation of FLIP supports the role of JNK in mediating the down-regulation of FLIP. It is shown herein that the pro-apoptotic effect of BAY 11-7085 on readhering pancreatic cancer cells was enhanced by JNK activation by anisomycin in a dose-dependent fashion. Furthermore, BAY 11-7085 caused the down-regulation of FLIP in readhering colon cancer cells, and, over-expression of FLIP in colon cancer cells caused greatly increased resistance to BAY 11-7085-induced apoptosis during cell readhesion. Finally, is is shown herein that BAY 11-7085 and to a lesser extent thiazolidinediones inhibited NF-κΒ during colon and pancreatic cancer readhesion. On a molar basis, BAY 11-7085 was a more potent NF-KB inhibitor than rosiglitazone, thus potentially explaining the greater reduction in peritoneal implants generated with pancreatic cancer cell lines. These studies show the relevance of targeting both NF-κΒ and JNK during cancer cell readhesion in order to down-regulate FLIP.
[00227] The identification of the thiazolidinedione rosiglitazone as an inhibitor of peritoneal seeding is important since this compound is in clinical use and has a relatively low toxicity profile. Thiazolidinediones have been shown to inhibit NF-κΒ and this may or may not be related to their PPARy agonist activity (Su et al., "A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response," J Clin Invest 104(4):383-9, 1999). We demonstrated that multiple thiazolidinediones were effective in inhibiting NF-κΒ and inducing apoptosis of colon and pancreatic cancer cells during readhesion. Furthermore, rosiglitazone inhibited the intraabdominal seeding of colon and pancreatic cancer cell lines as well. [00228] The proapoptotic activity of the thiazolidinediones may be independent of PPARy agonist activity as they showed pro-apoptotic activity in Su.86 and PL45 pancreatic cancer cells, neither of which expresses PPARy (Galli et al., "Antidiabetic thiazolidinediones inhibit invasiveness of pancreatic cancer cells via PPARgamma independent mechanisms," Gut 53(11): 1688-97, 2004). In addition, the irreversible PPARy inhibitor GW9662 failed to inhibit the pro-apoptotic activity of pioglitazone during cancer cell readhesion. Finally, others showed recently that rosiglitazone and pioglitazone inhibited pancreatic cancer cell invasion in a PPARy-independent fashion {Id showing that these compounds possess PPARy-independent antitumorigenic activities. [00229] The appearance of discrete and immoveable bio luminescent peritoneal implants only a few days after seeding and the efficacy of the compounds with only 10 days of treatment indicates that the effect of these drugs occurs in the initial steps of tumor implantation of the peritoneum. Pharmacokinetic studies showed Cmax blood levels of 2.5 μΜ of pioglitazone were measured in humans after a single or multiple oral doses of the drug. [00230] The importance of the current studies is that they provide evidence for the efficacy of compounds that inhibit NF-κΒ, and simultaneously activate JNK in reducing peritoneal carcinomatosis. Furthermore, FLIP appears to be an important target of these drugs and promotes cancer cell survival during readhesion. Once established, peritoneal carcinomatosis from abdominal malignancies is extremely difficult to treat and ultimately fatal. The possibility of preventing pancreatic or colon cancer seeding by treating patients with agents that induce apoptosis of readhering cancer cells in the perioperative setting is an unexplored area and an unmet clinical need.
[00231] As shown in Table 6, BAY 11-7085 inhibits intraabdominal implantation of readhering pancreatic cancer cells in vivo. Adult female athymic mice were randomized to receive either vehicle (DMSO) or drug (BAY 11-7085 or rosiglitazone) IP. Twenty-four hours later, 106 Su.86 or BxPC-3 pancreatic cancer cells, that were stably transfected with a firefly lucif erase construct, were delivered IP. The mice continued to receive the BAY 11-7085 every 3 days or rosiglitazone daily for 9 days after the cells were injected into the mice. At the end of treatment, the mice received luciferin IP and sacrificed. The abdominal cavities were exposed and the mice placed immediately in a bioluminescent imager and photographed. The individual intraabdominal peritoneal tumors were counted. The subset of tumor implants limited to the liver surface was determined as well. The results are expressed as the percentage of mice that demonstrated multiple intraabdominal tumor implants. The proportion of mice with peritoneal and/or hepatic tumor implants were compared using a test of proportions (*p<0.05).
TABLE 6:
Figure imgf000076_0001
DMSO Rosiglitazone
Su.86 83% peritoneal; 67% hepatic 83%o peritoneal; 83% hepatic
BxPC-3 83%o peritoneal; 50%> hepatic 33%o* peritoneal; 17%>* hepatic
Example 3
[00232] SU.86 pancreatic cancer cells were sorted for CD 133 expression cancer stem cells (CSC) by FACS and cocultured with monolayers of mesothelial cells, which line the peritoneum, and, express FASL and TRAIL. 4xl04CD133+ SU.86 cells stably expressing firefly luciferase were injected intraperitoneally (IP) into nude mice. Mice received BAY 11-7085 or vehicle (DMSO) IP. Peritoneal implants were quantified in situ by bioluminescent scans.
[00233] In a screen, BAY 11-7085 rapidly (~ 1 hr) induced ΓΝΚ-dependent FLIP proteasomal degradation in SU.86 cells. Pre-treatment of the DR-resistant CSC with 10μΜ BAY 11-7085 (BAY) or of SU.86 unsorted cells with FLIP siRNA restored DR- mediated apoptosis during cell adhesion to mesothelial cells. Inhibition of JNK, TRAIL, or FAS reversed BAY-mediated cell apoptosis during adhesion to mesothelial cells. BAY targeted FLIP for proteasomal degradation via INK activation. In vivo, BAY 11-7085 (5mg/kg IP TIW) greatly inhibited CSC peritoneal implants over 3 months (FIG. 14) without overt toxicity. Even a single dose of BAY 11-7085 administered IP together with unsorted SU.86 cells dramatically inhibited peritoneal implants after 1 month with 6 control mice having a two fold greater light output than the 6 BAY treated mice (p<0.05). Bortezomib or gemcitabine failed to restore DR-mediated apoptosis or inhibit peritoneal implants.
[00234] Inhibition of FLIP in pancreatic CSC and the bulk tumor cells allowed normal cells to kill them through juxtracrine DR activation.
References:
(1) Battu, S., Chable-Rabinovitch, FL, Rigaud, M., and Beneytout, J. L. (1998).
Cyclooxygenase-2 expression in human adenocarcinoma cell line HT29 cl. l9A,
Anticancer Res 18, 2397-403. (2) Cao, S., Troutt, A. B., and Rustum, Y. M. (1998). Interleukin 15 protects against toxicity and potentiates antitumor activity of 5-fluorouracil alone and in combination with leucovorin in rats bearing colorectal cancer, Cancer Res 58, 1695-9.
(3) DuBois, R. N., Giardiello, F. M., and Smalley, W. E. (1996). Nonsteroidal anti- inflammatory drugs, eicosanoids, and colorectal cancer prevention, Gastroenterol Clin
North Am 25, 773-91.
(4) Eberhart, C. E., Coffey, R. J., Radhika, A., Giardiello, F. M., Ferrenbach, S., and DuBois, R. N. (1994). Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas, Gastroenterology 107, 1183-8. (5) Elder, D. J., Halton, D. E., Hague, A., and Paraskeva, C. (1997). Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti- inflammatory drug: independence from COX-2 protein expression, Clin Cancer Res 3, 1679-83.
(6) Giri, D. K., and Aggarwal, B. B. (1998). Constitutive activation of NF- B causes resistance to apoptosis in human cutaneous T cell lymphoma HuT-78 cells. Autocrine role of tumor necrosis factor and reactive oxygen intermediates, J Biol Chem 273, 14008-14.
(7) Golab, J., Kozar, K., Kaminski, R., Czajka, A., Marczak, M., Switaj, T., Giermasz, A., Stoklosa, T., Lasek, W., Zagozdzon, R., et al. (2000). Interleukin 12 and indomethacin exert a synergistic, angiogenesis- dependent antitumor activity in mice, Life Sci 66, 1223- 30.
(8) He, T. C, Chan, T. A., Vogelstein, B., and Kinzler, K. W. (1999). PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs, Cell 99, 335-45.
(9) Higgins, K. A., Perez, J. R., Coleman, T. A., Dorshkind, K., McComas, W. A., Sarmiento, U. M., Rosen, C. A., and Narayanan, R. (1993). Antisense inhibition of the p65 subunit of NF-κΒ blocks tumorigenicity and causes tumor regression, Proc Natl Acad Sci U S A 90, 9901-5. (10) Ichijo, H., Nishida, E., Irie, K., ten Dijke, P., Saitoh, M., Moriguchi, T., Takagi, M., Matsumoto, K., Miyazono, K., and Gotoh, Y. (1997). Induction of apoptosis by ASK1, a mammalian MAPKK that activates SAPK/JNK and p38 signaling pathways, Science 275, 90-4. (11) Kutchera, W., Jones, D. A., Matsunami, N., Groden, J., Mclntyre, T. M.,
Zimmerman, G. A., White, R. L., and Prescott, S. M. (1996). Prostaglandin H synthase 2 is expressed abnormally in human colon cancer: evidence for a transcriptional effect, Proc Natl Acad Sci U S A 93, 4816-20.
(12) Lee, J. W., and Juliano, R. L. (2000). α5β1 integrin protects intestinal epithelial cells from apoptosis through a phosphatidylinositol 3 -kinase and protein kinase Independent pathway, Mol Biol Cell 11, 1973-87.
(13) Meredith, J. E., Jr., Fazeli, B., and Schwartz, M. A. (1993). The extracellular matrix as a cell survival factor, Mol Biol Cell 4, 953-61.
(14) Palombella, V. J., Rando, O. J., Goldberg, A. L., and Maniatis, T. (1994). The ubiquitin-proteasome pathway is required for processing the NF- B 1 precursor protein and the activation of NF- B, Cell 78, 773-85.
(15) Peleg, II, Lubin, M. F., Cotsonis, G. A., Clark, W. S., and Wilcox, C. M. (1996). Long-term use of nonsteroidal antiinflammatory drugs and other chemopreventors and risk of subsequent colorectal neoplasia, Dig Dis Sci 41, 1319-26. (16) Remacle-Bonnet, M. M., Garrouste, F. L., Heller, S., Andre, F., Marvaldi, J. L., and Pommier, G. J. (2000). Insulin-like growth factor-I protects colon cancer cells from death factor-induced apoptosis by potentiating tumor necrosis factor alpha- induced mitogen- activated protein kinase and nuclear factor kappaB signaling pathways, Cancer Res 60, 2007-17. (17) Rosette, C, and Karin, M. (1996). Ultraviolet light and osmotic stress: activation of the JNK cascade through multiple growth factor and cytokine receptors, Science 274, 1194-7. (18) Shanmugathasan, M., and Jothy, S. (2000). Apoptosis, anoikis and their relevance to the pathobiology of colon cancer, Pathol Int 50, 273-9.
(19) Sheng, H., Shao, J., Kirkland, S. C, Isakson, P., Coffey, R. J., Morrow, J.,
Beauchamp, R. D., and DuBois, R. N. (1997). Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2, J Clin Invest 99, 2254-9.
(20) Shiff, S. J., Qiao, L., Tsai, L. L., and Rigas, B. (1995). Sulindac sulfide, an aspirinlike compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells, J Clin Invest 96, 491-503.
(21) Shinohara, H., Killion, J. J., Kuniyasu, H., Kumar, R., and Fidler, I. J. (1998). Prevention of intestinal toxic effects and intensification of irinotecan's therapeutic efficacy against murine colon cancer liver metastases by oral administration of the lipopeptide JBT 3002, Clin Cancer Res 4, 2053-63.
(22) Shureiqi, I., Chen, D., Lee, J. J., Yang, P., Newman, R. A., Brenner, D. E., Lotan, R., Fischer, S. M., and Lippman, S. M. (2000). 15-LOX-l : a novel molecular target of nonsteroidal anti-inflammatory drug-induced apoptosis in colorectal cancer cells, J Natl Cancer Inst 92, 1136-42.
(23) Shureiqi, I., Wojno, K. J., Poore, J. A., Reddy, R. G., Moussalli, M. J., Spindler, S. A., Greenson, J. K., Normolle, D., Hasan, A. A., Lawrence, T. S., and Brenner, D. E. (1999). Decreased 13-S-hydroxyoctadecadienoic acid levels and 15 -lipoxygenase- 1 expression in human colon cancers, Carcinogenesis 20, 1985-95.
(24) Singh, A. K., and Trotman, B. W. (1998). Use and safety of aspirin in the chemoprevention of colorectal cancer, J Assoc Acad Minor Phys 9, 40-4.
(25) Sinicrope, F. A., Pazdur, R., and Levin, B. (1996). Phase I trial of sulindac plus 5- fluorouracil and levamisole: potential adjuvant therapy for colon carcinoma, Clin Cancer Res 2, 37-41.
(26) Smalley, W. E., and DuBois, R. N. (1997). Colorectal cancer and nonsteroidal antiinflammatory drugs, Adv Pharmacol 39, 1-20. (27) Sokoloski, J. A., Sartorelli, A. C, Rosen, C. A., and Narayanan, R. (1993).
Antisense oligonucleotides to the p65 subunit of NF- B block CD1 lb expression and alter adhesion properties of differentiated HL-60 granulocytes, Blood 82, 625-32.
(28) Tsujii, M., and DuBois, R. N. (1995). Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2, Cell 83, 493-
501.
(29) Yin, M. J., Yamamoto, Y., and Gaynor, R. B. (1998). The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-β, Nature 396, 77-80.
(30) Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS, Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAPl and c- IAP2 to suppress caspase-8 activation. Science 1998;281(5383): 1680-3.
(31) Wu MX, Ao Z, Prasad KV, Wu R, Schlossman SF. IEX-1L, an apoptosis inhibitor involved in NF-kappaB-mediated cell survival. Science 1998;281(5379):998-1001.
(32) Stehlik C, de Martin R, Kumabashiri I, Schmid JA, Binder BR, Lipp J. Nuclear factor (NF)-kappaB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha- induced apoptosis. J Exp Med
1998;188(l):211-6.
(33) Newton R, Stevens DA, Hart LA, Lindsay M, Adcock IM, Barnes PJ.
Superinduction of COX-2 mRNA by cycloheximide and interleukin-lbeta involves increased transcription and correlates with increased NF- kappaB and JNK activation. FEBS Lett 1997;418(l-2): 135-8.
(34) Newton R, Kuitert LM, Bergmann M, Adcock IM, Barnes PJ. Evidence for involvement of NF-kappaB in the transcriptional control of COX-2 gene expression by IL- lbeta. Biochem Biophys Res Commun 1997;237(l):28-32. (35) Schmedtje JF, Jr., Ji YS, Liu WL, DuBois RN, Runge MS. Hypoxia induces cyclooxygenase-2 via the NF-kappaB p65 transcription factor in human vascular endothelial cells. J Biol Chem 1997;272(l):601-8. (36) Crofford LJ, Tan B, McCarthy CJ, Hla T. Involvement of nuclear factor kappa B in the regulation of cyclooxygenase-2 expression by interleukin-1 in rheumatoid
synoviocytes. Arthritis Rheum 1997;40(2):226-36.
(37) Huang S, Pettaway CA, Uehara H, Bucana CD, Fidler IJ. Blockade of NF-kappaB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene 2001;20(31):4188-97.
(38) Dubuquoy L, Rousseaux C, Thuru X, et al. PPARgamma as a new therapeutic target in inflammatory bowel diseases. Gut 2006;55(9): 1341-9.
[00235] It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other examples of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims

CLAIMS What is claimed is:
1. A method for inhibiting pancreatic cancer stem cell proliferation: comprising administering to a subject diagnosed with pancreatic cancer, a compound having the structure
Figure imgf000083_0001
wherein R2, R3, and R4 are, independently, hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubstituted aromatic, wherein the compound is the E- or Z-isomer.
2. The method of claim 1, wherein the compound causes a decrease in the expression of anti-apoptotic proteins.
3. The method of claim 1, wherein the compound inhibits ΙκΒ NF-κΒ
phosphorylation.
4. The method of claim 1, wherein the compound inhibits TNFa induced NF-KB activation.
5. The method of claim 1, wherein the compound inhibits FLIP.
6. The method of claim 1, wherein R3 and R4 are hydrogen.
7. The method of claim 1, wherein R2 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, substituted or unsubstituted phenyl, or benzyl.
8. The method of claim 1, wherein R2 is a phenyl group having at least one alkyl group.
9. The method of claim 1, wherein the compound is the E-isomer.
10. The method of claim 1, wherein the compound has the structure
Figure imgf000084_0001
11. The method of claim 1 , wherein compound has the structure
Figure imgf000084_0002
12. The method of claim 1, wherein the compound is administered prior to a resection.
13. The method of claim 1, wherein the compound is administered after a resection.
14. The method of claim 1, wherein the compound is administered within 5 days of a resection.
15. The method of claim 1, wherein the compound is administered within 1 day of a resection.
16. The method of claim 1, wherein the compound is administered within 10 hours of a resection.
17. The method of claim 1, wherein the compound is administered within 1 hour of a resection.
18. The method of claim 1, wherein the compound is administered within 0.5 hours of the resection.
19. The method of claim 1, wherein the composition is administered by intraveneous injection or drip.
20. The method of claim 1, wherein the composition is administered intraperitoneally.
21. The method of claim 1 , wherein the composition is administered peri-operatively.
22. A method of screening compounds for anticancer activity, comprising:
(a) providing a compound or a library of compounds;
(b) assaying the compound or the library of compounds for NF-κΒ inhibitory activity;
(c) assaying the compound or library of compounds for FLIP inhibitory
activity;
(d) identifying compounds with both NF-κΒ inhibitory activity and FLIP
inhibitory activity, the identified compounds having anticancer activity.
23. The method of claim 22, further comprising assaying the compound or the library of compounds for JNK activation activity.
24. The method of claim 23, wherein the NF-κΒ assay, the FLIP assay, or the JNK assay, or all of the NF-κΒ, FLIP, and JNK assays use a DLD-1 cancer cell line, a HT-29 cancer cell line, a Su.86 cell line or a BxPC-3 cell line.
25. The method of claim 23, wherein the the assays are performed simultaneously.
26. The method of claim 1, wherein the compound or the library of compounds
comprisies an olefin having at least one electron- withdrawing group.
27. The method of claim 22, wherein the compound or the library of compounds
comprises an olefin having at least two electron-withdrawing groups.
28. The method of claim 27, wherein the electron- withdrawing group comprises a cyano group, a sulfo-oxy group, a phospho-oxy group, a carboxyl group, a nitro group, a halogen, a halogenated alkyl group, an unsubstituted aromatic ring, or a substituted aromatic ring having at least one cyano group, sulfo-oxy group, phospho-oxy group, carboxyl group, hydroxy 1 group, amino group, ether group, halogenated alkyl group, halogen, or nitro group.
29. The method of claim 28, wherein the phospho-oxy group has the structure
Figure imgf000086_0001
wherein R1 is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, substituted or unsubsituted aromatic.
The method of claim 28, wherein the sulfo-oxy group has the structure
Figure imgf000086_0002
wherein R is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubsituted aromatic.
31. The method of claim 22, wherein the compound or the library of compounds comprises an olefin having a cyano group and a sulfo-oxy group having the structure
O
-S Rz
O wherein R is hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubsituted aromatic.
32. The method of claim 22, wherein the compound or the library of compounds comprises the structure
Figure imgf000087_0001
wherein R2, R3 and R4 are, independently, hydrogen, alkyl, halogenated alkyl, alkenyl, alkynyl, aralkyl, or substituted or unsubsituted aromatic, wherein the compound is the E- or Z-isomer.
33. The method of claim 32, wherein R3 and R4 are hydrogen.
34. The method of claim 32, wherein R2 is methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, substituted or unsubstituted phenyl, or benzyl.
35. The method of claim 32, wherein R2 is a phenyl group having at least one alkyl group.
36. The method of claim 32, wherein the compound is the E-isomer.
The method of claim 22, wherein the compound or the library of compounds comprises the structure
Figure imgf000087_0002
The method of claim 22, wherein the compound or the library of compounds compises the structure
Figure imgf000088_0001
39. The method of claim 22, wherein the compound or the library of compounds comprises a thiazolidinedione.
40. The method of claim 39, wherein the compound or the library of compounds comprises rosiglitazone, pioglitazone, troglitazone, or ciglitazone.
41. The method of claim 22, wherein the compound or the library of compounds comprises anisomycin.
PCT/US2011/032299 2010-04-13 2011-04-13 Inhibitors of flip to treat cancer WO2011130395A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US27413210P 2010-04-13 2010-04-13
US61/274,132 2010-04-13

Publications (1)

Publication Number Publication Date
WO2011130395A1 true WO2011130395A1 (en) 2011-10-20

Family

ID=44799010

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/032299 WO2011130395A1 (en) 2010-04-13 2011-04-13 Inhibitors of flip to treat cancer

Country Status (1)

Country Link
WO (1) WO2011130395A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014043144A1 (en) * 2012-09-11 2014-03-20 Olatec Industries Llc Methods for treating inflammation and pain
US9763909B2 (en) 2012-09-11 2017-09-19 Olatec Therapeutics Llc Methods for treating inflammation and pain
WO2018130679A1 (en) * 2017-01-16 2018-07-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for reducing cd95- mediated cell motility
US10071087B2 (en) 2014-07-22 2018-09-11 Bioventures, Llc Compositions and methods for selectively depleting senescent cells
US10807977B2 (en) 2016-04-21 2020-10-20 Bioventures, Llc Compounds that induce degradation of anti-apoptotic Bcl-2 family proteins and the uses thereof
US11331328B2 (en) 2014-05-05 2022-05-17 Bioventures, Llc Compositions and methods for inhibiting antiapoptotic Bcl-2 proteins as anti-aging agents

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050124590A1 (en) * 2002-02-21 2005-06-09 Kuwada Scott K. Compositions and methods for inhibiting nf-kappab mediated tumorigenicity and adhesion dependent survival of cancer cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050124590A1 (en) * 2002-02-21 2005-06-09 Kuwada Scott K. Compositions and methods for inhibiting nf-kappab mediated tumorigenicity and adhesion dependent survival of cancer cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KERBAUY ET AL.: "NF-kB and FLIP in arsenic trioxide (ATO)-induced apoptosis in myelodyplastic syndromes (MDSs).", BLOOD, vol. 106, 2005, pages 3917 - 3925 *
MIMEAULT ET AL.: "Novel therapies against aggressive and recurrent epithelial cancers by molecular targeting tumor- and metastasis- initiating cells and their progenies.", ANTICANCER AGENTS MED CHEM, vol. 10, February 2010 (2010-02-01), pages 137 - 151 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014043144A1 (en) * 2012-09-11 2014-03-20 Olatec Industries Llc Methods for treating inflammation and pain
US9763909B2 (en) 2012-09-11 2017-09-19 Olatec Therapeutics Llc Methods for treating inflammation and pain
US11331328B2 (en) 2014-05-05 2022-05-17 Bioventures, Llc Compositions and methods for inhibiting antiapoptotic Bcl-2 proteins as anti-aging agents
US10071087B2 (en) 2014-07-22 2018-09-11 Bioventures, Llc Compositions and methods for selectively depleting senescent cells
US10758524B2 (en) 2014-07-22 2020-09-01 Bioventures, Llc Compositions and methods for selectively depleting senescent cells
US10807977B2 (en) 2016-04-21 2020-10-20 Bioventures, Llc Compounds that induce degradation of anti-apoptotic Bcl-2 family proteins and the uses thereof
US11319316B2 (en) 2016-04-21 2022-05-03 Bioventures, Llc Compounds that induce degradation of anti-apoptotic Bcl-2 family proteins and the uses thereof
WO2018130679A1 (en) * 2017-01-16 2018-07-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for reducing cd95- mediated cell motility

Similar Documents

Publication Publication Date Title
Avery et al. GLI1: a therapeutic target for cancer
Kamdje et al. Signaling pathways in breast cancer: therapeutic targeting of the microenvironment
Asati et al. K-Ras and its inhibitors towards personalized cancer treatment: Pharmacological and structural perspectives
Patsos et al. The endogenous cannabinoid, anandamide, induces COX-2-dependent cell death in apoptosis-resistant colon cancer cells
Fang et al. NFYB-induced high expression of E2F1 contributes to oxaliplatin resistance in colorectal cancer via the enhancement of CHK1 signaling
US8969313B2 (en) Methods and compounds for preventing and treating a tumour
WO2011130395A1 (en) Inhibitors of flip to treat cancer
US20110306675A1 (en) Compositions and methods for inhibiting nf-kb mediated tumorigenicity and adhesion-dependent survival of cancer cells
Massari et al. Antitumor activity of histamine and clozapine in a mouse experimental model of human melanoma
AU2009330492A1 (en) Enzastaurin for the treatment of cancer
Cheng et al. A novel inhibitor targets both Wnt signaling and ATM/p53 in colorectal cancer
Zhang et al. GLI1 inhibitor SRI-38832 attenuates chemotherapeutic resistance by downregulating NBS1 transcription in BRAFV600E colorectal cancer
AU2014207272A1 (en) Estrogen receptor inhibitors
US20110251135A1 (en) Inhibitors of peritoneal seeding of cancer cells
US20160030367A1 (en) Methods and Compositions for Preventing Metastasis and for Improving the Survival Time
Lee et al. Inhibitory effect of FSLLRY-NH 2 on inflammatory responses induced by hydrogen peroxide in HepG2 cells
US20200101070A1 (en) Methods of treating cancer having an active wnt/beta-catenin pathway
US9457016B2 (en) Methods for treating polycystic kidney disease
Matsuda et al. Therapeutic Efficacy of Vitamin D in Experimental c-MET–β-Catenin-Driven Hepatocellular Cancer
Khazak et al. A Two-Hybrid Approach to Identify Inhibitors of the RAS–RAF Interaction
US20160101076A1 (en) Use of Sumoylation Inhibitors for Treating Cancer
JP2007511471A (en) SHC proteins as therapeutic targets in proliferative diseases
Kumar et al. Impact of Hybrid-polar Histone Deacetylase Inhibitor m-Carboxycinnamic Acid bis-Hydroxyamide on Human Pancreatic Adenocarcinoma Cells
Sharma Inhibition of Tumor Cell Growth by Mefloquine via Multimechanistic Effects Involving Increased Cellular Stress, Inhibition of Autophagy, and Impairment of Cellular Energy Metabolism
Robinson Combinatorial Effects of 2, 3-Dichloro-5, 8-Dimethoxy-1, 4-Naphthoquinone and 4-Hydroxytamoxifen on Triple-Negative Breast Cancer Cell Lines

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: 11769518

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: 11769518

Country of ref document: EP

Kind code of ref document: A1