Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mol Cancer Ther ; 15(7): 1757-67, 2016 07.
Article in English | MEDLINE | ID: mdl-27196777

ABSTRACT

UNLABELLED: Ionizing radiation (IR) is a key therapeutic regimen for many head and neck cancers (HNC). However, the 5-year overall survival rate for locally advanced HNCs is approximately 50% and better therapeutic efficacy is needed. NAD(P)H: quinone oxidoreductase 1 (NQO1) is overexpressed in many cancers, and ß-lapachone (ß-lap), a unique NQO1 bioactivatable drug, exploits this enzyme to release massive reactive oxygen species (ROS) that synergize with IR to kill by programmed necrosis. ß-Lap represents a novel therapeutic opportunity in HNC leading to tumor-selective lethality that will enhance the efficacy of IR. Immunohistochemical staining and Western blot assays were used to assess the expression levels of NQO1 in HNC cells and tumors. Forty-five percent of endogenous HNCs expressed elevated NQO1 levels. In addition, multiple HNC cell lines and tumors demonstrated elevated levels of NQO1 expression and activity and were tested for anticancer lethality and radiosensitization by ß-lap using long-term survival assays. The combination of nontoxic ß-lap doses and IR significantly enhanced NQO1-dependent tumor cell lethality, increased ROS, TUNEL-positive cells, DNA damage, NAD(+), and ATP consumption, and resulted in significant antitumor efficacy and prolonged survival in two xenograft murine HNC models, demonstrating ß-lap radiosensitization of HNCs through a NQO1-dependent mechanism. This translational study offers a potential biomarker-driven strategy using NQO1 expression to select tumors susceptible to ß-lap-induced radiosensitization. Mol Cancer Ther; 15(7); 1757-67. ©2016 AACR.


Subject(s)
Gene Expression , Head and Neck Neoplasms/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Radiation Tolerance/genetics , Radiation, Ionizing , Adenosine Triphosphate/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
2.
Cell Cycle ; 13(16): 2587-99, 2014.
Article in English | MEDLINE | ID: mdl-25486199

ABSTRACT

Indole-3-carbinol (I3C) is a natural anti-carcinogenic compound found at high concentrations in Brassica vegetables. I3C was recently reported to inhibit neutrophil elastase (NE) activity, while consequently limiting the proteolytic processing of full length cyclin E into pro-tumorigenic low molecular weight cyclin E (LMW-E). In this study, we hypothesized that inhibition of NE activity and resultant LMW-E generation is critical to the anti-tumor effects of I3C. LMW-E was predominately expressed by ERα-negative breast cancer cell lines. However, ERα-positive cell lines demonstrated the greatest sensitivity to the anti-tumor effects of I3C and its more potent N-alkoxy derivatives. We found that I3C was incapable of inhibiting NE activity or the generation of LMW-E. Therefore, this pathway did not contribute to the anti-tumor activity of I3C. Gene expression analyzes identified ligand-activated aryl hydrocarbon receptor (AhR), which mediated sensitivity to the anti-tumor effects of I3C in ERα-positive MCF-7 cells. In this model system, the reactive oxygen species (ROS)-induced upregulation of ATF-3 and pro-apoptotic BH3-only proteins (e.g. NOXA) contributed to the sensitivity of ERα-positive breast cancer cells to the anti-tumor effects of I3C. Overexpression of ERα in MDA-MB-231 cells, which normally lack ERα expression, increased sensitivity to the anti-tumor effects of I3C, demonstrating a direct role for ERα in mediating the sensitivity of breast cancer cell lines to I3C. Our results suggest that ERα signaling amplified the pro-apoptotic effect of I3C-induced AhR signaling in luminal breast cancer cell lines, which was mediated in part through oxidative stress induced upregulation of ATF-3 and downstream BH3-only proteins.


Subject(s)
Alcohols/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Indoles/pharmacology , Metabolic Networks and Pathways/drug effects , Alcohols/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin E/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/chemistry , Leukocyte Elastase/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects
3.
Tetrahedron ; 70(4): 984-990, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-25110363

ABSTRACT

A virtual library of 54 inositol analog mimics of In(1,4,5)P3 has been docked, scored, and ranked within the binding site of human inositol 1,4,5-trisphosphate 3-kinase A (IP3-3KA). Chemical synthesis of the best scoring structure that also met distance criteria for 3'-OH to -P in Phosphate has been attempted along with the synthesis of (1S,2R,3S,4S)-3-fluoro-2,4-dihydroxycyclohexanecarboxylic acid as an inositol analog, useful for non-invasive visualization and quantitation of IP3-3KA enzymatic activity.

4.
Antioxid Redox Signal ; 21(2): 237-50, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24512128

ABSTRACT

AIMS: ß-Lapachone (ß-lap), a novel radiosensitizer with potent antitumor efficacy alone, selectively kills solid cancers that over-express NAD(P)H: quinone oxidoreductase 1 (NQO1). Since breast or other solid cancers have heterogeneous NQO1 expression, therapies that reduce the resistance (e.g., NQO1(low)) of tumor cells will have significant clinical advantages. We tested whether NQO1-proficient (NQO1(+)) cells generated sufficient hydrogen peroxide (H2O2) after ß-lap treatment to elicit bystander effects, DNA damage, and cell death in neighboring NQO1(low) cells. RESULTS: ß-Lap showed NQO1-dependent efficacy against two triple-negative breast cancer (TNBC) xenografts. NQO1 expression variations in human breast cancer patient samples were noted, where ~60% cancers over-expressed NQO1, with little or no expression in associated normal tissue. Differential DNA damage and lethality were noted in NQO1(+) versus NQO1-deficient (NQO1(-)) TNBC cells and xenografts after ß-lap treatment. ß-Lap-treated NQO1(+) cells died by programmed necrosis, whereas co-cultured NQO1(-) TNBC cells exhibited DNA damage and caspase-dependent apoptosis. NQO1 inhibition (dicoumarol) or H2O2 scavenging (catalase [CAT]) blocked all responses. Only NQO1(-) cells neighboring NQO1(+) TNBC cells responded to ß-lap in vitro, and bystander effects correlated well with H2O2 diffusion. Bystander effects in NQO1(-) cells in vivo within mixed 50:50 co-cultured xenografts were dramatic and depended on NQO1(+) cells. However, normal human cells in vitro or in vivo did not show bystander effects, due to elevated endogenous CAT levels. Innovation and Conclusions: NQO1-dependent bystander effects elicited by NQO1 bioactivatable drugs (ß-lap or deoxynyboquinone [DNQ]) likely contribute to their efficacies, killing NQO1(+) solid cancer cells and eliminating surrounding heterogeneous NQO1(low) cancer cells. Normal cells/tissue are protected by low NQO1:CAT ratios.


Subject(s)
Bystander Effect/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Quinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Female , Humans , Mice , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/deficiency , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidation-Reduction/drug effects , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Bioorg Med Chem ; 22(4): 1450-8, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24457091

ABSTRACT

A series of degrasyn-like symmetrical compounds have been designed, synthesized, and screened against B cell malignancy (multiple myeloma, mantle cell lymphoma) cell lines. The lead compounds T5165804 and CP2005 showed higher nanomolar potency against these tumor cells in comparison to degrasyn and inhibited Usp9x activity in vitro and in intact cells. These observations suggest that this new class of compounds holds promise as cancer therapeutic agents.


Subject(s)
Antineoplastic Agents/chemistry , Nitriles/chemistry , Pyridines/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cyanoacrylates , Dimerization , Humans , Models, Molecular , Multiple Myeloma/drug therapy , Nitriles/pharmacology , Nitriles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism
6.
Bioorg Med Chem ; 22(1): 623-32, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24280068

ABSTRACT

We synthesized two series of imatinib mesylate (STI-571) analogs to develop a Bcr-Abl and c-KIT receptor-specific labeling agent for positron emission tomography (PET) imaging to measure Bcr-Abl and c-KIT expression levels in a mouse model. The methods of molecular modeling, synthesis of STI-571 and its analogs, in vitro kinase assays, and radiolabeling are described. Molecular modeling revealed that these analogs bind the same Bcr-Abl and c-KIT binding sites as those bound by STI-571. The analogs potently inhibit the tyrosine kinase activity of Bcr-Abl and c-KIT, similarly to STI-571. [(18)F]-labeled STI-571 was prepared with high specific activity (75 GBq/µmol) by nucleophilic displacement and an average radiochemical yield of 12%. [(131)I]-labeled STI-571 was prepared with high purity (>95%) and an average radiochemical yield of 23%. The uptake rates of [(18)F]-STI-571 in K562 cells expressing Abl and in U87WT cells overexpressing c-KIT were significantly higher than those in the U87 cell and could be inhibited by STI-71 (confirming the specificity of uptake). PET scans of K562 and U87WT tumor-bearing mice with [(18)F]-STI-571 as a contrast agent showed visible tumor uptake and tumor-to-non-target contrast.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Fusion Proteins, bcr-abl/metabolism , Piperazines/therapeutic use , Positron-Emission Tomography/methods , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents/chemistry , Benzamides/chemistry , Disease Models, Animal , Humans , Imatinib Mesylate , Mice , Models, Molecular , Piperazines/chemistry , Pyrimidines/chemistry
7.
Bioorg Med Chem ; 22(1): 435-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24280069

ABSTRACT

A gram scale synthesis of the glucuronide metabolites of curcumin were completed in four steps. The newly synthesized curcumin glucuronide compounds 2 and 3 along with curcumin 1 were tested and their anti-proliferative effects against KBM-5, Jurkat cell, U266, and A549 cell lines were reported. Biological data revealed that as much as 1 µM curcumin 1 exhibited anticancer activity and almost 100% cell kill was noted at 10 µM on two out of four cell lines; while curcumin mono-glucuronide 2 as well as di-glucuronide 3 displayed no suppression of cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Curcumin/pharmacology , Cell Line, Tumor , Curcumin/chemical synthesis , Humans , Jurkat Cells , Structure-Activity Relationship
8.
PLoS One ; 8(10): e77262, 2013.
Article in English | MEDLINE | ID: mdl-24204784

ABSTRACT

OBJECTIVE: The iboga alkaloids are a class of small molecules defined structurally on the basis of a common ibogamine skeleton, some of which modify opioid withdrawal and drug self-administration in humans and preclinical models. These compounds may represent an innovative approach to neurobiological investigation and development of addiction pharmacotherapy. In particular, the use of the prototypic iboga alkaloid ibogaine for opioid detoxification in humans raises the question of whether its effect is mediated by an opioid agonist action, or if it represents alternative and possibly novel mechanism of action. The aim of this study was to independently replicate and extend evidence regarding the activation of µ-opioid receptor (MOR)-related G proteins by iboga alkaloids. METHODS: Ibogaine, its major metabolite noribogaine, and 18-methoxycoronaridine (18-MC), a synthetic congener, were evaluated by agonist-stimulated guanosine-5´-O-(γ-thio)-triphosphate ([(35)S]GTPγS) binding in cells overexpressing the recombinant MOR, in rat thalamic membranes, and autoradiography in rat brain slices. RESULTS AND SIGNIFICANCE: In rat thalamic membranes ibogaine, noribogaine and 18-MC were MOR antagonists with functional Ke values ranging from 3 uM (ibogaine) to 13 uM (noribogaine and 18MC). Noribogaine and 18-MC did not stimulate [(35)S]GTPγS binding in Chinese hamster ovary cells expressing human or rat MORs, and had only limited partial agonist effects in human embryonic kidney cells expressing mouse MORs. Ibogaine did not did not stimulate [(35)S]GTPγS binding in any MOR expressing cells. Noribogaine did not stimulate [(35)S]GTPγS binding in brain slices using autoradiography. An MOR agonist action does not appear to account for the effect of these iboga alkaloids on opioid withdrawal. Taken together with existing evidence that their mechanism of action also differs from that of other non-opioids with clinical effects on opioid tolerance and withdrawal, these findings suggest a novel mechanism of action, and further justify the search for alternative targets of iboga alkaloids.


Subject(s)
Bridged-Ring Compounds/pharmacology , Ibogaine/analogs & derivatives , Ibogaine/pharmacology , Receptors, Opioid, mu/metabolism , Thalamus/drug effects , Animals , Autoradiography , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Female , Gene Expression , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , HEK293 Cells , Humans , Organ Specificity , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/prevention & control , Thalamus/metabolism
9.
Mol Cancer Ther ; 12(10): 2110-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23883585

ABSTRACT

Improving patient outcome by personalized therapy involves a thorough understanding of an agent's mechanism of action. ß-Lapachone (clinical forms, Arq501/Arq761) has been developed to exploit dramatic cancer-specific elevations in the phase II detoxifying enzyme NAD(P)H:quinone oxidoreductase (NQO1). NQO1 is dramatically elevated in solid cancers, including primary and metastatic [e.g., triple-negative (ER-, PR-, Her2/Neu-)] breast cancers. To define cellular factors that influence the efficacy of ß-lapachone using knowledge of its mechanism of action, we confirmed that NQO1 was required for lethality and mediated a futile redox cycle where ∼120 moles of superoxide were formed per mole of ß-lapachone in 2 minutes. ß-Lapachone induced reactive oxygen species (ROS), stimulated DNA single-strand break-dependent poly(ADP-ribose) polymerase-1 (PARP1) hyperactivation, caused dramatic loss of essential nucleotides (NAD(+)/ATP), and elicited programmed necrosis in breast cancer cells. Although PARP1 hyperactivation and NQO1 expression were major determinants of ß-lapachone-induced lethality, alterations in catalase expression, including treatment with exogenous enzyme, caused marked cytoprotection. Thus, catalase is an important resistance factor and highlights H2O2 as an obligate ROS for cell death from this agent. Exogenous superoxide dismutase enhanced catalase-induced cytoprotection. ß-Lapachone-induced cell death included apoptosis-inducing factor (AIF) translocation from mitochondria to nuclei, TUNEL+ staining, atypical PARP1 cleavage, and glyceraldehyde 3-phosphate dehydrogenase S-nitrosylation, which were abrogated by catalase. We predict that the ratio of NQO1:catalase activities in breast cancer versus associated normal tissue are likely to be the major determinants affecting the therapeutic window of ß-lapachone and other NQO1 bioactivatable drugs.


Subject(s)
Breast Neoplasms/drug therapy , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/administration & dosage , Poly(ADP-ribose) Polymerases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catalase/genetics , Catalase/metabolism , DNA Breaks, Single-Stranded/drug effects , DNA Damage/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Necrosis/genetics , Necrosis/pathology , Poly (ADP-Ribose) Polymerase-1 , Reactive Oxygen Species/metabolism
10.
Bioorg Med Chem ; 21(17): 5182-7, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23859776

ABSTRACT

An improved method for the synthesis of 17ß-hydroxy-16α-iodo-wortmannin along with the first synthesis of 17ß-hydroxy-16α-iodoPX866 and [(131)I] radiolabeled 17ß-hydroxy-16α-[(131)I]iodo-wortmannin, as potential PET tracers for PI3K was also described. The differences between wortmannin and its iodo analogue were compared by covalently docking each structure to L833 in PI3K.


Subject(s)
Androstadienes/chemistry , Androstadienes/chemical synthesis , Gonanes/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Binding Sites , Gonanes/chemistry , Iodine Radioisotopes/chemistry , Isotope Labeling , Molecular Docking Simulation , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Positron-Emission Tomography , Protein Structure, Tertiary , Radiopharmaceuticals/chemistry , Wortmannin
11.
J Biol Chem ; 288(27): 19414-28, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23703610

ABSTRACT

Activation of the integrin family of cell adhesion receptors on progenitor cells may be a viable approach to enhance the effects of stem cell-based therapies by improving cell retention and engraftment. Here, we describe the synthesis and characterization of the first small molecule agonist identified for the integrin α4ß1 (also known as very late antigen-4 or VLA-4). The agonist, THI0019, was generated via two structural modifications to a previously identified α4ß1 antagonist. THI0019 greatly enhanced the adhesion of cultured cell lines and primary progenitor cells to α4ß1 ligands VCAM-1 and CS1 under both static and flow conditions. Furthermore, THI0019 facilitated the rolling and spreading of cells on VCAM-1 and the migration of cells toward SDF-1α. Molecular modeling predicted that the compound binds at the α/ß subunit interface overlapping the ligand-binding site thus indicating that the compound must be displaced upon ligand binding. In support of this model, an analog of THI0019 modified to contain a photoreactive group was used to demonstrate that when cross-linked to the integrin, the compound behaves as an antagonist instead of an agonist. In addition, THI0019 showed cross-reactivity with the related integrin α4ß7 as well as α5ß1 and αLß2. When cross-linked to αLß2, the photoreactive analog of THI0019 remained an agonist, consistent with it binding at the α/ß subunit interface and not at the ligand-binding site in the inserted ("I") domain of the αL subunit. Co-administering progenitor cells with a compound such as THI0019 may provide a mechanism for enhancing stem cell therapy.


Subject(s)
Cell Movement/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Integrin alpha4beta1/agonists , Models, Molecular , Stem Cells/metabolism , CD11a Antigen/genetics , CD11a Antigen/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/physiology , Cell- and Tissue-Based Therapy/methods , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Heterocyclic Compounds, 4 or More Rings/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Jurkat Cells , Stem Cells/cytology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
12.
Mol Cancer Ther ; 12(5): 654-62, 2013 May.
Article in English | MEDLINE | ID: mdl-23303403

ABSTRACT

We have previously shown that the antiallergic drug cromolyn blocks S100P interaction with its receptor receptor for advanced glycation end product (RAGE) and improves gemcitabine effectiveness in pancreatic ductal adenocarcinoma (PDAC). However, the concentration required to achieve its effectiveness was high (100 µmol/L). In this study, we designed and synthesized analogs of cromolyn and analyzed their effectiveness compared with the parent molecule. An ELISA was used to confirm the binding of S100P with RAGE and to test the effectiveness of the different analogs. Analog 5-methyl cromolyn (C5OH) blocked S100P binding as well as the increases in NF-κB activity, cell growth, and apoptosis normally caused by S100P. In vivo C5OH systemic delivery reduced NF-κB activity to a greater extent than cromolyn and at 10 times lesser dose (50 mg vs. 5 mg). Treatment of mice-bearing syngeneic PDAC tumors showed that C5OH treatment reduced both tumor growth and metastasis. C5OH treatment of nude mice bearing orthotopic highly aggressive pancreatic Mpanc96 cells increased the overall animal survival. Therefore, the cromolyn analog, C5OH, was found to be more efficient and potent than cromolyn as a therapeutic for PDAC.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/metabolism , Cromolyn Sodium/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Calcium-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cromolyn Sodium/analogs & derivatives , Cromolyn Sodium/chemistry , Drug Design , Drug Evaluation, Preclinical , Humans , Male , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Binding/drug effects , Receptor for Advanced Glycation End Products/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem ; 21(4): 932-9, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23294827

ABSTRACT

Curcumin (diferuloylmethane) is a potent anti-inflammatory and anti-tumorigenic agent that has shown preclinical activity in diverse cancers. Curcumin up-regulates heat shock protein 70 (hsp70) mRNA in several different cancer cell lines. Hsp70 contributes to an escape from the apoptotic effects of curcumin by several different mechanisms including prevention of the release of apoptosis inducing factor from the mitochondria and inhibition of caspases 3 and 9. Previously we showed that the combination of curcumin plus a heat shock protein inhibitor was synergistic in its down-regulation of the proliferation of a human schwannoma cell line (HEI-193) harboring an NF2 mutation, possibly because curcumin up-regulated hsp70, which also binds merlin, the NF2 gene product. In order to determine if curcumin also interacts directly with hsp70 and to discover other binding partners of curcumin, we synthesized biotinylated curcumin (bio-curcumin) and treated HEI-193 schwannoma cells. Cell lysates were prepared and incubated with avidin-coated beads. Peptides pulled down from this reaction were sequenced and it was determined that biotinylated curcumin bound hsp70, hsp90, 3-phosphoglycerate dehydrogenase, and a ß-actin variant. These binding partners may serve to further elucidate the underlying mechanisms of curcumin's actions.


Subject(s)
Curcumin/chemistry , HSP70 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/chemistry , Phosphoglycerate Dehydrogenase/chemistry , Binding Sites , Biotin/chemistry , Cell Line, Tumor , Curcumin/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Docking Simulation , Neurilemmoma/metabolism , Neurilemmoma/pathology , Phosphoglycerate Dehydrogenase/metabolism , Protein Binding , Protein Structure, Tertiary
14.
Tetrahedron Lett ; 54(41): 5555-5567, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-25110364

ABSTRACT

The C1-C6 region of the potent cytotoxic agent psymberin has been synthesized. The key transformations of the synthesis are an auxiliary-controlled addition of a Sn(II)-glycolate enolate to an aldehyde to yield the anti aldol product and transforming the primary alcohol into a terminal olefin utilizing organoselenium chemistry.

15.
Tetrahedron Lett ; 54(43): 5799-5801, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-25110365

ABSTRACT

We report the synthesis of a macrocycle utilizing a novel framework of standard amino acids in combination with subunits that we have named as Linked Amino Acid Mimetics (LAAM's). Macrocycles based on the LAAM concept provide both a peptide targeting region and two independently variable functional regions. In the prototype structure, the commonly known Arg-Gly-Asp (RGD) sequence was used for the targeting region. The functional regions contain a phenyl group, and the linkage was formed via a Ring-Closing Metathesis (RCM) reaction.

16.
Lett Org Chem ; 9(10): 762-766, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-24883052

ABSTRACT

An updated and practical approach to the synthesis of dithymoquinone via one-step photoirradiation of thymoquinone (2-methyl-5-isopropyl-1,4-benzoquinone) is described. Synthesis resulted in a 55% yield of one structural isomer (trans-anti derivative), as confirmed by HPLC, NMR spectroscopy and first ever single-crystal X-ray diffraction analyses.

17.
Cell Signal ; 23(12): 2076-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21855629

ABSTRACT

AG490 is a tyrosine kinase inhibitor with activity against Jak2 and apoptotic activity in specific leukemias. Due to its weak kinase inhibitory activity and poor pharmacology, we conducted a cell-based screen for derivatives with improved Jak2 inhibition and activity in animals. Two hits emerged from an initial small chemical library screen, and more detailed structure-activity relationship studies led to the development of WP1130 with 50-fold greater activity in suppressing Jak2-dependent cytokine signaling than AG490. However, WP1130 did not directly suppress Jak2 kinase activity, but mediated Jak2 ubiquitination resulting in its trafficking through HDAC6 to perinuclear aggresomes without cytokine stimulation or SOCS-1 induction. Jak2 primarily contained K63-linked ubiquitin polymers, and mutation of this lysine blocked Jak2 ubiquitination and mobilization in WP1130-treated cells. Further analysis demonstrated that WP1130, but not AG490, acts as a deubiquitinating enzyme (DUB) inhibitor, possibly through a Michael addition reaction. We conclude that chemical modification of AG490 resulted in development of a DUB inhibitor with activity against a DUB capable of modulating Jak2 ubiquitination, trafficking and signal transduction.


Subject(s)
Endopeptidases/metabolism , Janus Kinase 2/metabolism , Protease Inhibitors/pharmacology , Signal Transduction/drug effects , Tyrphostins/pharmacology , Ubiquitination , Amino Acid Substitution , Cell Line, Tumor , Cell Proliferation/drug effects , Cyanoacrylates , Drug Evaluation, Preclinical , Endopeptidases/chemistry , Enzyme Assays , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-6/pharmacology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nitriles/pharmacology , Phosphorylation , Proteasome Inhibitors , Protein Transport/drug effects , Proteolysis , Pyridines/pharmacology , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
18.
Cancer ; 117(19): 4424-38, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21491416

ABSTRACT

BACKGROUND: Epigenetic therapy has had a significant impact on the management of hematologic malignancies, but its role in the treatment of ovarian cancer remains to be defined. The authors previously demonstrated that treatment of ovarian and breast cancer cells with DNA methyltransferase and histone deacetylase (HDAC) inhibitors can up-regulate the expression of imprinted tumor suppressors. In this study, demethylating agents and HDAC inhibitors were tested for their ability to induce re-expression of tumor suppressor genes, inhibiting growth of ovarian cancer cells in culture and in xenografts. METHODS: Ovarian cancer cells (Hey and SKOv3) were treated with demethylating agents (5-aza-20-deoxycytidine [DAC] or 5-azacitidine [AZA]) or with HDAC inhibitors (suberoylanilide hydroxamicacid [SAHA] or trichostatin A [TSA]) to determine their impact on cellular proliferation, cell cycle regulation, apoptosis, autophagy, and re-expression of 2 growth inhibitory imprinted tumor suppressor genes: guanosine triphosphate-binding Di-RAS-like 3 (ARHI) and paternally expressed 3 (PEG3). The in vivo activities of DAC and SAHA were assessed in a Hey xenograft model. RESULTS: The combination of DAC and SAHA produced synergistic inhibition of Hey and SKOv3 cell growth by apoptosis and cell cycle arrest. DAC induced autophagy in Hey cells that was enhanced by SAHA. Treatment with both agents induced re-expression of ARHI and PEG3 in cultured cells and in xenografts, correlating with growth inhibition. Knockdown of ARHI decreased DAC-induced autophagy. DAC and SAHA inhibited the growth of Hey xenografts and induced autophagy in vivo. CONCLUSIONS: A combination of DAC and SAHA inhibited ovarian cancer growth while inducing apoptosis, G2/M arrest, autophagy, and re-expression of imprinted tumor suppressor genes.


Subject(s)
Apoptosis/drug effects , Autophagy , Azacitidine/analogs & derivatives , Genes, Tumor Suppressor/drug effects , Genomic Imprinting , Hydroxamic Acids/pharmacology , Ovarian Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine , Drug Synergism , Drug Therapy, Combination , Epigenomics , Female , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transplantation, Heterologous , Vorinostat
19.
Cancer Res ; 71(10): 3658-68, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21444672

ABSTRACT

AZD6244 is a small molecule inhibitor of the MEK (MAP/ERK kinase) pathway currently in clinical trials. However, the mechanisms mediating intrinsic resistance to MEK inhibition are not fully characterized. To define molecular mechanisms of MEK inhibitor resistance, we analyzed responses of 38 lung cancer cell lines following AZD6244 treatment and their genome-wide gene expression profiles and identified a panel of genes correlated with sensitivity or resistance to AZD6244 treatment. In particular, ingenuity pathway analysis revealed that activation of the STAT3 pathway was associated with MEK inhibitor resistance. Inhibition of this pathway by JSI-124, a STAT3-specific small molecule inhibitor, or with STAT3-specific siRNA sensitized lung cancer cells to AZD6244 and induced apoptosis. Moreover, combining a STAT3 inhibitor with AZD6244 induced expression of BIM and PARP cleavage, whereas activation of the STAT3 pathway inhibited BIM expression and elicited resistance to MEK inhibitors. We found that the STAT3-regulated microRNA miR-17 played a critical role in MEK inhibitor resistance, such that miR-17 inhibition sensitized resistant cells to AZD6244 by inducing BIM and PARP cleavage. Together, these results indicated that STAT3-mediated overexpression of miR-17 blocked BIM expression and caused resistance to AZD6244. Our findings suggest novel approaches to overcome resistance to MEK inhibitors by combining AZD6244 with STAT3 or miR-17 inhibitors.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , RNA, Small Interfering/metabolism
20.
Blood ; 117(11): 3151-62, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21248063

ABSTRACT

Although chronic myelogenous leukemia (CML) is effectively controlled by Bcr-Abl kinase inhibitors, resistance to inhibitors, progressive disease, and incomplete eradication of Bcr-Abl-expressing cells are concerns for the long-term control and suppression of this disease. We describe a novel approach to targeting key proteins in CML cells with a ubiquitin-cycle inhibitor, WP1130. Bcr-Abl is rapidly modified with K63-linked ubiquitin polymers in WP1130-treated CML cells, resulting in its accumulation in aggresomes, where is it unable to conduct signal transduction. Induction of apoptosis because of aggresomal compartmentalization of Bcr-Abl was observed in both imatinib-sensitive and -resistant cells. WP1130, but not Bcr-Abl kinase inhibitors, directly inhibits Usp9x deubiquitinase activity, resulting in the down-regulation of the prosurvival protein Mcl-1 and facilitating apoptosis. These results demonstrate that ubiquitin-cycle inhibition represents a novel and effective approach to blocking Bcr-Abl kinase signaling and reducing Mcl-1 levels to engage CML cell apoptosis. This approach may be a therapeutic option for kinase inhibitor-resistant CML patients.


Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Signal Transduction , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Cyanoacrylates , Drug Resistance, Neoplasm/drug effects , Endopeptidases/metabolism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , Models, Biological , Nitriles/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Transport/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Substrate Specificity/drug effects , Ubiquitin Thiolesterase/metabolism , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...