Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Neoplasia ; 20(1): 80-91, 2018 01.
Article in English | MEDLINE | ID: mdl-29216597

ABSTRACT

ONC201/TIC10 is a first-in-class small molecule inducer of TRAIL that causes early activation of the integrated stress response. Its promising safety profile and broad-spectrum efficacy in vitro have been confirmed in Phase I/II trials in several advanced malignancies. Binding and reporter assays have shown that ONC201 is a selective antagonist of the dopamine D2-like receptors, specifically, DRD2 and DRD3. We hypothesized that ONC201's interaction with DRD2 plays a role in ONC201's anticancer effects. Using cBioportal and quantitative reverse-transcription polymerase chain reaction analyses, we confirmed that DRD2 is expressed in different cancer cell types in a cell type-specific manner. On the other hand, DRD3 was generally not detectable. Overexpressing DRD2 in cells with low DRD2 levels increased ONC201-induced PARP cleavage, which was preceded and correlated with an increase in ONC201-induced CHOP mRNA expression. On the other hand, knocking out DRD2 using CRISPR/Cas9 in three cancer cell lines was not sufficient to abrogate ONC201's anticancer effects. Although ONC201's anticancer activity was not dependent on DRD2 expression in the cancer cell types tested, we assessed the cytotoxic potential of DRD2 blockade. Transient DRD2 knockdown in HCT116 cells activated the integrated stress response and reduced cell number. Pharmacological antagonism of DRD2 significantly reduced cell viability. Thus, we demonstrate in this study that disrupting dopamine receptor expression and activity can have cytotoxic effects that may at least be in part due to the activation of the integrated stress response. On the other hand, ONC201's anticancer activity goes beyond its ability to antagonize DRD2, potentially due to ONC201's ability to activate other pathways that are independent of DRD2. Nevertheless, blocking the dopamine D1-like receptor DRD5 via siRNA or the use of a pharmacological antagonist promoted ONC201-induced anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplasms/metabolism , Receptors, Dopamine/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Imidazoles , Neoplasms/genetics , Pyridines , Pyrimidines , RNA, Small Interfering/genetics , Receptors, Dopamine/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism
2.
PLoS One ; 12(8): e0180541, 2017.
Article in English | MEDLINE | ID: mdl-28767654

ABSTRACT

Cancer stem cells (CSCs) correlate with recurrence, metastasis and poor survival in clinical studies. Encouraging results from clinical trials of CSC inhibitors have further validated CSCs as therapeutic targets. ONC201 is a first-in-class small molecule imipridone in Phase I/II clinical trials for advanced cancer. We have previously shown that ONC201 targets self-renewing, chemotherapy-resistant colorectal CSCs via Akt/ERK inhibition and DR5/TRAIL induction. In this study, we demonstrate that the anti-CSC effects of ONC201 involve early changes in stem cell-related gene expression prior to tumor cell death induction. A targeted network analysis of gene expression profiles in colorectal cancer cells revealed that ONC201 downregulates stem cell pathways such as Wnt signaling and modulates genes (ID1, ID2, ID3 and ALDH7A1) known to regulate self-renewal in colorectal, prostate cancer and glioblastoma. ONC201-mediated changes in CSC-related gene expression were validated at the RNA and protein level for each tumor type. Accordingly, we observed inhibition of self-renewal and CSC markers in prostate cancer cell lines and patient-derived glioblastoma cells upon ONC201 treatment. Interestingly, ONC201-mediated CSC depletion does not occur in colorectal cancer cells with acquired resistance to ONC201. Finally, we observed that basal expression of CSC-related genes (ID1, CD44, HES7 and TCF3) significantly correlate with ONC201 efficacy in >1000 cancer cell lines and combining the expression of multiple genes leads to a stronger overall prediction. These proof-of-concept studies provide a rationale for testing CSC expression at the RNA and protein level as a predictive and pharmacodynamic biomarker of ONC201 response in ongoing clinical studies.


Subject(s)
Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/physiopathology , Colorectal Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/physiopathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Central Nervous System Neoplasms/genetics , Colorectal Neoplasms/genetics , Glioblastoma/genetics , HCT116 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Imidazoles , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Neoplastic Stem Cells/metabolism , Pyridines , Pyrimidines , Transcriptome , Wnt Signaling Pathway/drug effects
3.
Cancer Res ; 75(18): 3842-52, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26294215

ABSTRACT

The tumor-suppressor p53 prevents cancer development via initiating cell-cycle arrest, cell death, repair, or antiangiogenesis processes. Over 50% of human cancers harbor cancer-causing mutant p53. p53 mutations not only abrogate its tumor-suppressor function, but also endow mutant p53 with a gain of function (GOF), creating a proto-oncogene that contributes to tumorigenesis, tumor progression, and chemo- or radiotherapy resistance. Thus, targeting mutant p53 to restore a wild-type p53 signaling pathway provides an attractive strategy for cancer therapy. We demonstrate that small-molecule NSC59984 not only restores wild-type p53 signaling, but also depletes mutant p53 GOF. NSC59984 induces mutant p53 protein degradation via MDM2 and the ubiquitin-proteasome pathway. NSC59984 restores wild-type p53 signaling via p73 activation, specifically in mutant p53-expressing colorectal cancer cells. At therapeutic doses, NSC59984 induces p73-dependent cell death in cancer cells with minimal genotoxicity and without evident toxicity toward normal cells. NSC59984 synergizes with CPT11 to induce cell death in mutant p53-expressing colorectal cancer cells and inhibits mutant p53-associated colon tumor xenograft growth in a p73-dependent manner in vivo. We hypothesize that specific targeting of mutant p53 may be essential for anticancer strategies that involve the stimulation of p73 in order to efficiently restore tumor suppression. Taken together, our data identify NSC59984 as a promising lead compound for anticancer therapy that acts by targeting GOF-mutant p53 and stimulates p73 to restore the p53 pathway signaling.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Nitrofurans/pharmacology , Nuclear Proteins/physiology , Piperazines/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/physiology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Screening Assays, Antitumor , Drug Synergism , Gene Knockdown Techniques , Genes, p53 , Humans , Irinotecan , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitrofurans/chemistry , Nitrofurans/toxicity , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Piperazines/chemistry , Piperazines/toxicity , Proteolysis , Proto-Oncogene Mas , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism , Tumor Protein p73 , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
4.
Clin Colorectal Cancer ; 13(2): 119-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24461492

ABSTRACT

INTRODUCTION: Therapeutic plasma 5-fluorouracil (5-FU) levels are achieved in only 20% to 30% of patients with the current practice of administering 5-FU doses based on body surface area (BSA). Alternatively, 5-FU doses can be adjusted based on 5-FU pharmacokinetic (PK) monitoring. Although benefits of PK monitoring of 5-FU in metastatic colorectal cancer (CRC) have been reported, its utility among patients with early stage disease has not been reported. PATIENTS AND METHODS: We retrospectively examined the effect of 5-FU PK monitoring in 84 CRC patients (49 stage IV and 35 stage II/III) receiving mFOLFOX6 (modifiedFOLFOX6; modified 5-fluorouracil, leucovorin, oxaliplatin protocol) or mFOLFIRI (modified 5-fluorouracil, leucovorin, irinotecan protocol). Forty-six of the 84 patients received 5-FU doses based on BSA and 38 received doses that were adjusted with PK monitoring. 5-FU plasma levels were measured using a nanoparticle immunoassay method. RESULTS: 5-fluorouracil PK monitoring significantly improved disease-free survival in stage II/III patients (P = .0429). There was also a trend towards improved progression-free survival among stage IV patients who had their 5-FU levels PK-monitored (P = .16). Moreover, 5-FU PK monitoring significantly reduced (P = .0437) and delayed (P = .0144) adverse effects in stage II/III patients. Toxicity occurred after the second 5-FU dose in the BSA group and after the sixth to seventh dose in the PK monitoring group. In stage IV patients, the onset of toxicities was also delayed with PK monitoring (P = .0605). CONCLUSION: We provide evidence that PK monitoring of 5-FU is potentially beneficial for late stage and early stage CRC. These results contribute to the growing body of evidence regarding patient benefit when treatment decisions are based on the individual patient characteristics, in this case, a patients' 5-FU levels.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Drug Monitoring/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Colorectal Neoplasms/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Fluorouracil/therapeutic use , Humans , Leucovorin/administration & dosage , Leucovorin/pharmacokinetics , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/therapeutic use , Precision Medicine , Retrospective Studies
5.
Pharmaceuticals (Basel) ; 6(8): 988-1038, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-24276379

ABSTRACT

The use of pharmaceuticals for colon cancer treatment has been increasingly personalized, in part due to the development of new molecular tools. In this review, we discuss the old and new colon cancer chemotherapeutics, and the parameters that have been shown to be predictive of efficacy and safety of these chemotherapeutics. In addition, we discuss how alternate pharmaceuticals have been developed in light of a potential lack of response or resistance to a particular chemotherapeutic.

6.
Apoptosis ; 16(12): 1285-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21910008

ABSTRACT

The overexpression of the pro-apoptotic protein Prostate Apoptosis Response Protein-4 in colon cancer has been shown to increase response to the chemotherapeutic agent 5-fluorouracil (5-FU). Although colon cancer cells endogenously express Par-4, the presence or overexpression of Par-4 alone does not cause apoptosis. We hypothesize that Par-4 is inactivated in colon cancer. In colon cancer, the levels and the kinase activity of the nonreceptor tyrosine kinase c-Src increase with tumor progression. One of the downstream effectors of c-Src is Akt1. Akt1 has been shown to inhibit the pro-apoptotic activity of Par-4 in prostate cancer cells. We therefore investigated the potential of activating Par-4 by inhibiting c-Src. Colon carcinoma cell lines were treated with the Src kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2) in combination with the chemotherapeutic agent 5-FU. Treating cells with PP2 and 5-FU resulted in reduced interaction of Par-4 with Akt1 and with the scaffolding protein 14-3-3σ, and mobilization of Par-4 to the nucleus. Par-4 was shown to interact not only with Akt1 and 14-3-3σ, but also with c-Src. Overexpression of c-Src induced the phosphorylation of Par-4 at tyrosine site/s. Thus, in this study, we have shown that Par-4 can be activated by inhibiting Src with a pharmacological inhibitor and adding a chemotherapeutic agent. The activation of the pro-apoptotic protein Par-4 as reported in this study is a novel mechanism by which apoptosis occurs with a Src kinase inhibitor and 5-FU. In addition, we have demonstrated that the pro-apoptotic activity of endogenously expressed Par-4 can be increased in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Colonic Neoplasms/genetics , Fluorouracil/pharmacology , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , Exonucleases/genetics , Exonucleases/metabolism , Exoribonucleases , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
7.
Mol Cancer ; 9: 98, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20433755

ABSTRACT

BACKGROUND: Diminished expression or activity of prostate apoptosis response protein 4 (Par-4) has been demonstrated in a number of cancers, although reports on Par-4 expression during colon cancer progression are lacking. An understanding of the molecular events in conjunction with the genetic networks affected by Par-4 is warranted. RESULTS: Colon cancer specimens derived from patients have significantly diminished expression of Par-4 mRNA relative to paired normal colon. Hence, the functional consequences of reintroducing Par-4 into HT29 colon cancer cells were assessed. Overexpression augmented the interaction of Par-4 with NF kappaB in the cytosol but not nucleus, and facilitated apoptosis in the presence of 5-fluorouracil (5-FU). Analogous findings were obtained when AKT1 pro-survival signaling was inhibited. Transcriptome profiling identified approximately 700 genes differentially regulated by Par-4 overexpression in HT29 cells. Nearly all Par-4-regulated genes were shown by promoter analysis to contain cis-binding sequences for NF kappaB, and meta-analysis of patient expression data revealed that one-third of these genes exist as a recurrent co-regulated network in colon cancer specimens. Sets of genes involved in programmed cell death, cell cycle regulation and interestingly the microRNA pathway were found overrepresented in the network. Noteworthy, Par-4 overexpression decreased NF kappaB occupancy at the promoter of one particular network gene DROSHA, encoding a microRNA processing enzyme. The resulting down-regulation of DROSHA was associated with expression changes in a cohort of microRNAs. Many of these microRNAs are predicted to target mRNAs encoding proteins with apoptosis-related functions. Western and functional analyses were employed to validate several predictions. For instance, miR-34a up-regulation corresponded with a down-regulation of BCL2 protein. Treating Par-4-overexpressing HT29 cells with a miR-34a antagomir functionally reversed both BCL2 down-regulation and apoptosis by 5-FU. Conversely, bypassing Par-4 overexpression by direct knockdown of DROSHA expression in native HT29 cells increased miR-34a expression and 5-FU sensitivity. CONCLUSION: Our findings suggest that the initiation of apoptotic sensitivity in colon cancer cells can be mediated by Par-4 binding to NF kappaB in the cytoplasm with consequential changes in the expression of microRNA pathway components.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Chromatin Immunoprecipitation , Fluorouracil/pharmacology , Gene Expression , Gene Expression Profiling , HT29 Cells , Humans , Immunohistochemistry , Male , MicroRNAs/genetics , Microscopy, Confocal , NF-kappa B/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/biosynthesis , Ribonuclease III/genetics , Signal Transduction/genetics
8.
Clin Exp Metastasis ; 26(2): 77-87, 2009.
Article in English | MEDLINE | ID: mdl-18839319

ABSTRACT

Src kinase has been linked to increased motility in the progression and metastasis of human colon cancer, although the mechanisms are not fully understood. Integrins are involved in metastasis by mediating attachment and migration of cells, as well as through transducing signals. This study examines the link between Src and integrin activity in the metastatic process in colon cancer cells. To determine Src involvement in integrin expression, the human colon cancer cell line, HCT116, was transfected with an activated Src construct and assayed for its ability to attach to and migrate across collagen and laminin. These cells attached more readily and migrated less rapidly on the extracellular matrix (ECM) than did cells transfected with empty vector. Examination of integrin levels showed a decrease in the alpha3 subunit in Src transfected cells as well as decreased cell surface localization of alpha3 integrin. The downregulation of alpha3 integrin was reversed by inhibition of Src and by inhibition of MAP kinase. Inhibition of alpha3 integrin using shRNA resulted in decreased MMP7 secretion, a possible cause of decreased invasion with low alpha3 integrin expression. This study shows that Src overexpression downregulates alpha3 integrin total protein expression and localization to the cell surface of HCT116 colon cancer cells. This indicates that Src activity may enhance metastasis by altering alpha3 integrin expression.


Subject(s)
Integrin alpha3/biosynthesis , src-Family Kinases/physiology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Chromones/pharmacology , Colonic Neoplasms , Down-Regulation/physiology , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/physiology , Flavonoids/pharmacology , Humans , Matrix Metalloproteinase 7/metabolism , Morpholines/pharmacology , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridones/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
9.
Int J Cancer ; 122(12): 2665-73, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18351644

ABSTRACT

Src kinase has been linked as a causative agent in the progression of a number of cancers including colon, breast, lung and melanoma. Src protein and activity levels are increased in colorectal cancer and liver metastases arising secondary to colon cancer. However, although Src protein is increased in colon cancer as early as the adenomatous polyp stage, a role for Src in carcinogenesis has not been established. We developed the c-SRC transgenic mouse in the C57BL/6 strain to address the issue of carcinogenesis in cells with high levels of Src expression. The transgene was constructed with the human c-SRC gene downstream of the mouse metallothionein promoter to create zinc inducible gene expression. In these C57BL/6 mice, Src protein was increased in a number of tissues both with and without zinc induction. No additional carcinogenic agent was administered. After 20 months, mice were assessed for tumor development in the liver and GI tract, as well as other organs. Of the mice with the transgene, 15% developed tumors in the liver while no tumors were detected in wild type C57BL/6 mice. A further study was conducted by crossing c-SRC C57BL/6 mice with p21 nullizygous mice to determine the effect of oncogene expression combined with inactivation of the tumor suppressor gene, p21. Addition of the c-SRC transgene to the p21-/- background increased tumor formation almost 3-fold, while it increased metastasis 6-fold. The data from our study show, for the first time, that Src kinase may play a role in carcinogenesis.


Subject(s)
Neoplasms, Experimental/enzymology , src-Family Kinases/metabolism , Animals , Base Sequence , DNA Primers , Humans , Metallothionein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
10.
Int J Biochem Cell Biol ; 38(5-6): 1004-14, 2006.
Article in English | MEDLINE | ID: mdl-16324875

ABSTRACT

In diabetic animals, enhanced production of vascular endothelial growth factor is thought to be a major contributor to the development of diabetic retinopathy. In the present study, glucosamine-treated R28 retinal neuronal cells were used as an experimental model system to explore the possible involvement of the hexosamine biosynthetic pathway in the diabetes-induced changes in mRNA translation. Glucosamine treatment enhanced vascular endothelial growth factor production subsequent to changes in phosphorylation of the alpha-subunit of eukaryotic initiation factor 2, with no change in vascular endothelial growth factor mRNA content. Possible mechanisms through which glucosamine might act to increase eukaryotic initiation factor 2alpha phosphorylation include enhanced O-linked glycosylation of protein kinase or phosphatase regulatory proteins and/or induction of oxidative stress. However, increasing global protein O-glycosylation through inhibition of O-beta-N-acetylglucosaminidase did not mimic the effect of glucosamine on eukaryotic initiation factor 2alpha phosphorylation. Likewise, attenuating glucosamine-induced oxidative stress with two different antioxidants did not reduce glucosamine-induced eukaryotic initiation factor 2alpha phosphorylation. Glucosamine treatment was also found to promote eukaryotic initiation factor 2alpha phosphorylation in wild-type mouse embryonic fibroblasts, but not in mouse embryonic fibroblasts lacking the eukaryotic initiation factor 2alpha kinase referred to as RNA-dependent protein kinase-like endoplasmic-reticulum associated kinase, implicating the kinase in the glucosamine-induced increase in eukaryotic initiation factor 2alpha phosphorylation. Overall, the results are consistent with glucosamine causing activation of RNA-dependent protein kinase-like endoplasmic-reticulum associated kinase, which phosphorylates eukaryotic initiation factor 2alpha and consequently upregulates translation of mRNAs encoding specific proteins, such as vascular endothelial growth factor.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Glucosamine/pharmacology , eIF-2 Kinase/metabolism , Animals , Antioxidants/pharmacology , Cell Line , Hydrogen Peroxide/pharmacology , Mice , Phosphorylation/drug effects , Rats , Retina , Up-Regulation , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...