Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
NPJ Breast Cancer ; 7(1): 38, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33824328

ABSTRACT

Triple-negative breast cancer (TNBC) remains the most lethal breast cancer subtype with poor response rates to the current chemotherapies and a lack of additional effective treatment options. We have identified deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) as a critical gatekeeper that protects tumour DNA from the genotoxic misincorporation of uracil during treatment with standard chemotherapeutic agents commonly used in the FEC regimen. dUTPase catalyses the hydrolytic dephosphorylation of deoxyuridine triphosphate (dUTP) to deoxyuridine monophosphate (dUMP), providing dUMP for thymidylate synthase as part of the thymidylate biosynthesis pathway and maintaining low intracellular dUTP concentrations. This is crucial as DNA polymerase cannot distinguish between dUTP and deoxythymidylate triphosphate (dTTP), leading to dUTP misincorporation into DNA. Targeting dUTPase and inducing uracil misincorporation during the repair of DNA damage induced by fluoropyrimidines or anthracyclines represents an effective strategy to induce cell lethality. dUTPase inhibition significantly sensitised TNBC cell lines to fluoropyrimidines and anthracyclines through imbalanced nucleotide pools and increased DNA damage leading to decreased proliferation and increased cell death. These results suggest that repair of treatment-mediated DNA damage requires dUTPase to prevent uracil misincorporation and that inhibition of dUTPase is a promising strategy to enhance the efficacy of TNBC chemotherapy.

3.
Nat Rev Clin Oncol ; 11(5): 282-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24732946

ABSTRACT

Over the past 60 years, chemotherapeutic agents that target thymidylate biosynthesis and the enzyme thymidylate synthase (TS) have remained among the most-successful drugs used in the treatment of cancer. Fluoropyrimidines, such as 5-fluorouracil and capecitabine, and antifolates, such as methotrexate and pemetrexed, induce a state of thymidylate deficiency and imbalances in the nucleotide pool that impair DNA replication and repair. TS-targeted agents are used to treat numerous solid and haematological malignancies, either alone or as foundational therapeutics in combination treatment regimens. We overview the pivotal discoveries that led to the rational development of thymidylate biosynthesis as a chemotherapeutic target, and highlight the crucial contribution of these advances to driving and accelerating drug development in the earliest era of cancer chemotherapy. The function of TS as well as the mechanisms and consequences of inhibition of this enzyme by structurally diverse classes of drugs with distinct mechanisms of action are also discussed. In addition, breakthroughs relating to TS-targeted therapies that transformed the clinical landscape in some of the most-difficult-to-treat cancers, such as pancreatic, colorectal and non-small-cell lung cancer, are highlighted. Finally, new therapeutic agents and novel mechanism-based strategies that promise to further exploit the vulnerabilities and target resistance mechanisms within the thymidylate biosynthesis pathway are reviewed.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Thymidine Monophosphate/biosynthesis , Thymidylate Synthase/antagonists & inhibitors , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , Drug Design , Drug Resistance, Neoplasm , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/therapeutic use , Humans , Models, Biological , Neoplasm Proteins/physiology , Neoplasms/enzymology , Prodrugs/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Thymidylate Synthase/physiology
4.
Invest New Drugs ; 31(4): 845-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23299388

ABSTRACT

Despite compelling preclinical data in colorectal cancer (CRC), the efficacy of HDACIs has been disappointing in the clinic. The goal of this study was to evaluate the effectiveness of vorinostat and panobinostat in a dose- and exposure-dependent manner in order to better understand the dynamics of drug action and antitumor efficacy. In a standard 72 h drug exposure MTS assay, notable concentration-dependent antiproliferative effects were observed in the IC50 range of 1.2-2.8 µmol/L for vorinostat and 5.1-17.5 nmol/L for panobinostat. However, shorter clinically relevant exposures of 3 or 6 h failed to elicit any significant growth inhibition and in most cases a >24 h exposure to vorinostat or panobinostat was required to induce a sigmoidal dose-response. Similar results were observed in colony formation assays where ≥ 24 h of exposure was required to effectively reduce colony formation. Induction of acetyl-H3, acetyl-H4 and p21 by vorinostat were transient and rapidly reversed within 12 h of drug removal. In contrast, panobinostat-induced acetyl-H3, acetyl-H4, and p21 persisted for 48 h after an initial 3 h exposure. Treatment of HCT116 xenografts with panobinostat induced significant increases in acetyl-H3 and downregulation of thymidylate synthase after treatment. Although HDACIs exert both potent growth inhibition and cytotoxic effects when CRC cells were exposed to drug for ≥ 24 h, these cells demonstrate an inherent ability to survive HDACI concentrations and exposure times that exceed those clinically achievable. Continued efforts to develop novel HDACIs with improved pharmacokinetics/phamacodynamics, enhanced intratumoral delivery and class/isoform-specificity are needed to improve the therapeutic potential of HDACIs and HDACI-based combination regimens in solid tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Acetylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Male , Mice , Neoplasm Proteins/genetics , Panobinostat , Thymidylate Synthase/metabolism , Time Factors , Tumor Stem Cell Assay , Vorinostat , Xenograft Model Antitumor Assays
5.
Angiogenesis ; 16(1): 29-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22945845

ABSTRACT

Lymphedema is mainly caused by lymphatic obstruction and manifested as tissue swelling, often in the arms and legs. Lymphedema is one of the most common post-surgical complications in breast cancer patients and presents a painful and disfiguring chronic illness that has few treatment options. Here, we evaluated the therapeutic potential of interleukin (IL)-8 in lymphatic regeneration independent of its pro-inflammatory activity. We found that IL-8 promoted proliferation, tube formation, and migration of lymphatic endothelial cells (LECs) without activating the VEGF signaling. Additionally, IL-8 suppressed the major cell cycle inhibitor CDKN1C/p57(KIP2) by downregulating its positive regulator PROX1, which is known as the master regulator of LEC-differentiation. Animal-based studies such as matrigel plug and cornea micropocket assays demonstrated potent efficacy of IL-8 in activating lymphangiogenesis in vivo. Moreover, we have generated a novel transgenic mouse model (K14-hIL8) that expresses human IL-8 in the skin and then crossed with lymphatic-specific fluorescent (Prox1-GFP) mouse. The resulting double transgenic mice showed that a stable expression of IL-8 could promote embryonic lymphangiogenesis. Moreover, an immunodeficient IL-8-expressing mouse line that was established by crossing K14-hIL8 mice with athymic nude mice displayed an enhanced tumor-associated lymphangiogenesis. Finally, when experimental lymphedema was introduced, K14-hIL8 mice showed an improved amelioration of lymphedema with an increased lymphatic regeneration. Together, we report that IL-8 can activate lymphangiogenesis in vitro and in vivo with a therapeutic efficacy in post-surgical lymphedema.


Subject(s)
Interleukin-8/therapeutic use , Lymphatic Vessels/physiopathology , Lymphedema/drug therapy , Lymphedema/etiology , Postoperative Complications/drug therapy , Postoperative Complications/etiology , Regeneration , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Down-Regulation/drug effects , Embryonic Development/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Interleukin-8/metabolism , Interleukin-8/pharmacology , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Lymphedema/pathology , Lymphedema/physiopathology , Mice , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Postoperative Complications/pathology , Postoperative Complications/physiopathology , Receptors, Interleukin-8/metabolism , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Regeneration/drug effects , Tretinoin/pharmacology , Tumor Microenvironment/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/metabolism
6.
Mol Cancer Ther ; 11(6): 1353-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22391039

ABSTRACT

Colorectal cancer is the second most common cause of cancer-related death in the United States. Recent studies showed that interleukin-8 (IL-8) and its receptors (CXCR1 and CXCR2) are significantly upregulated in both the tumor and its microenvironment, and act as key regulators of proliferation, angiogenesis, and metastasis. Our previous study showed that IL-8 overexpression in colorectal cancer cells triggers the upregulation of the CXCR2-mediated proliferative pathway. The aim of this study was to investigate whether the CXCR2 antagonist, SCH-527123, inhibits colorectal cancer proliferation and if it can sensitize colorectal cancer cells to oxaliplatin both in vitro and in vivo. SCH-527123 showed concentration-dependent antiproliferative effects in HCT116, Caco2, and their respective IL-8-overexpressing variants colorectal cancer cell lines. Moreover, SCH-527123 was able to suppress CXCR2-mediated signal transduction as shown through decreased phosphorylation of the NF-κB/mitogen-activated protein kinase (MAPK)/AKT pathway. These findings corresponded with decreased cell migration and invasion, while increased apoptosis in colorectal cancer cell lines. In vivo results verified that SCH-527123 treatment decreased tumor growth and microvessel density when compared with vehicle-treated tumors. Importantly, these preclinical studies showed that the combination of SCH-527123 and oxaliplatin resulted in a greater decrease in cell proliferation, tumor growth, apoptosis, and angiogenesis that was superior to single-agent treatment. Taken together, these findings suggest that targeting CXCR2 may block tumor proliferation, migration, invasion, and angiogenesis. In addition, CXCR2 blockade may further sensitize colorectal cancer to oxaliplatin treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Colonic Neoplasms/drug therapy , Cyclobutanes/pharmacology , Organoplatinum Compounds/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Cyclobutanes/therapeutic use , Drug Synergism , Drug Therapy, Combination , HCT116 Cells , Humans , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 11(3): 616-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22172489

ABSTRACT

Chemotherapies that target thymidylate synthase (TS) continue to see considerable clinical expansion in non-small cell lung cancer (NSCLC). One drawback to TS-targeted therapies is drug resistance and subsequent treatment failure. Novel therapeutic and biomarker-driven strategies are urgently needed. The enzyme deoxyuridine triphosphate nucleotidohydrolase (dUTPase) is reported to protect tumor cells from aberrant misincorporation of uracil during TS inhibition. The goal of this study was to investigate the expression and significance of dUTPase in mediating response to TS-targeted agents in NSCLC. The expression of dUTPase in NSCLC cell lines and clinical specimens was measured by quantitative real-time reverse transcriptase PCR and immunohistochemistry. Using a validated RNA interference approach, dUTPase was effectively silenced in a panel of NSCLC cell lines and response to the fluoropyrimidine fluorodeoxyuridine (FUdR) and the antifolate pemetrexed was analyzed using growth inhibition and clonogenic assays. Apoptosis was analyzed by flow cytometry. Significant variation in the quantity and cellular expression of dUTPase was observed, including clear evidence of overexpression in NSCLC cell line models and tumor specimens at the mRNA and protein level. RNA interference-mediated silencing of dUTPase significantly sensitized NSCLC cells to growth inhibition induced by FUdR and pemetrexed. This sensitization was accompanied by a significant expansion of intracellular dUTP pools and significant decreases in NSCLC cell viability evaluated by clonogenicity and apoptotic analyses. Together, these results strongly suggest that uracil misincorporation is a potent determinant of cytotoxicity to TS inhibition in NSCLC and that inhibition of dUTPase is a mechanism-based therapeutic approach to significantly enhance the efficacy of TS-targeted chemotherapeutic agents.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Pyrophosphatases/genetics , RNA Interference , Thymidylate Synthase/antagonists & inhibitors , Aged , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Deoxyuracil Nucleotides/metabolism , Floxuridine/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Immunoblotting , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Pemetrexed , Pyrophosphatases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism
8.
Nucleic Acids Res ; 39(17): e112, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21576234

ABSTRACT

Current methods for measuring deoxyribonucleoside triphosphates (dNTPs) employ reagent and labor-intensive assays utilizing radioisotopes in DNA polymerase-based assays and/or chromatography-based approaches. We have developed a rapid and sensitive 96-well fluorescence-based assay to quantify cellular dNTPs utilizing a standard real-time PCR thermocycler. This assay relies on the principle that incorporation of a limiting dNTP is required for primer-extension and Taq polymerase-mediated 5-3' exonuclease hydrolysis of a dual-quenched fluorophore-labeled probe resulting in fluorescence. The concentration of limiting dNTP is directly proportional to the fluorescence generated. The assay demonstrated excellent linearity (R(2) > 0.99) and can be modified to detect between ∼0.5 and 100 pmol of dNTP. The limits of detection (LOD) and quantification (LOQ) for all dNTPs were defined as <0.77 and <1.3 pmol, respectively. The intra-assay and inter-assay variation coefficients were determined to be <4.6% and <10%, respectively with an accuracy of 100 ± 15% for all dNTPs. The assay quantified intracellular dNTPs with similar results obtained from a validated LC-MS/MS approach and successfully measured quantitative differences in dNTP pools in human cancer cells treated with inhibitors of thymidylate metabolism. This assay has important application in research that investigates the influence of pathological conditions or pharmacological agents on dNTP biosynthesis and regulation.


Subject(s)
Deoxyribonucleotides/analysis , Nucleic Acid Hybridization/methods , DNA-Directed DNA Polymerase , Deoxyuracil Nucleotides/analysis , Fluorescence Resonance Energy Transfer , HCT116 Cells , Humans , Oligonucleotide Probes , Polymerase Chain Reaction , Ribonucleotides/chemistry , Templates, Genetic
9.
Mol Cancer Res ; 9(5): 660-70, 2011 May.
Article in English | MEDLINE | ID: mdl-21478268

ABSTRACT

Aberrant activation of Wnt/ß-catenin signaling, resulting in the expression of Wnt-regulated oncogenes, is recognized as a critical factor in the etiology of colorectal cancer. Occupancy of ß-catenin at promoters of Wnt target genes drives transcription, but the mechanism of ß-catenin action remains poorly understood. Here, we show that CARM1 (coactivator-associated arginine methyltransferase 1) interacts with ß-catenin and positively modulates ß-catenin-mediated gene expression. In colorectal cancer cells with constitutively high Wnt/ß-catenin activity, depletion of CARM1 inhibits expression of endogenous Wnt/ß-catenin target genes and suppresses clonal survival and anchorage-independent growth. We also identified a colorectal cancer cell line (RKO) with a low basal level of ß-catenin, which is dramatically elevated by treatment with Wnt3a. Wnt3a also increased the expression of a subset of endogenous Wnt target genes, and CARM1 was required for the Wnt-induced expression of these target genes and the accompanying dimethylation of arginine 17 of histone H3. Depletion of ß-catenin from RKO cells diminished the Wnt-induced occupancy of CARM1 on a Wnt target gene, indicating that CARM1 is recruited to Wnt target genes through its interaction with ß-catenin and contributes to transcriptional activation by mediating events (including histone H3 methylation) that are downstream from the actions of ß-catenin. Therefore, CARM1 is an important positive modulator of Wnt/ß-catenin transcription and neoplastic transformation, and may thereby represent a novel target for therapeutic intervention in cancers involving aberrantly activated Wnt/ß-catenin signaling.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Histones/genetics , Protein-Arginine N-Methyltransferases/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Histones/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
10.
Cancer Res ; 71(10): 3635-48, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21464044

ABSTRACT

As key molecules that drive progression and chemoresistance in gastrointestinal cancers, epidermal growth factor receptor (EGFR) and HER2 have become efficacious drug targets in this setting. Lapatinib is an EGFR/HER2 kinase inhibitor suppressing signaling through the RAS/RAF/MEK (MAP/ERK kinase)/MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase)/AKT pathways. Histone deacetylase inhibitors (HDACi) are a novel class of agents that induce cell cycle arrest and apoptosis following the acetylation of histone and nonhistone proteins modulating gene expression and disrupting HSP90 function inducing the degradation of EGFR-pathway client proteins. This study sought to evaluate the therapeutic potential of combining lapatinib with the HDACi panobinostat in colorectal cancer (CRC) cell lines with varying EGFR/HER2 expression and KRAS/BRAF/PIK3CA mutations. Lapatinib and panobinostat exerted concentration-dependent antiproliferative effects in vitro (panobinostat range 7.2-30 nmol/L; lapatinib range 7.6-25.8 µmol/L). Combined lapatinib and panobinostat treatment interacted synergistically to inhibit the proliferation and colony formation in all CRC cell lines tested. Combination treatment resulted in rapid induction of apoptosis that coincided with increased DNA double-strand breaks, caspase-8 activation, and PARP cleavage. This was paralleled by decreased signaling through both the PI3K and MAPK pathways and increased downregulation of transcriptional targets including NF-κB1, IRAK1, and CCND1. Panobinostat treatment induced downregulation of EGFR, HER2, and HER3 mRNA and protein through transcriptional and posttranslational mechanisms. In the LoVo KRAS mutant CRC xenograft model, the combination showed greater antitumor activity than either agent alone, with no apparent increase in toxicity. Our results offer preclinical rationale warranting further clinical investigation combining HDACi with EGFR and HER2-targeted therapies for CRC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Histone Deacetylase Inhibitors/pharmacology , Humans , Indoles , Lapatinib , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Neoplasm Transplantation , Panobinostat , Signal Transduction
11.
Pharmacogenomics ; 12(1): 27-39, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21174620

ABSTRACT

AIMS: Survival and response rates in metastatic colorectal cancer remain poor, despite advances in drug development. There is increasing evidence to suggest that gender-specific differences may contribute to poor clinical outcome. We tested the hypothesis that genomic profiling of metastatic colorectal cancer is dependent on gender. MATERIALS & METHODS: A total of 152 patients with metastatic colorectal cancer who were treated with oxaliplatin and continuous infusion 5-fluorouracil were genotyped for 21 polymorphisms in 13 cancer-related genes by PCR. Classification and regression tree analysis tested for gender-related association of polymorphisms with overall survival, progression-free survival and tumor response. RESULTS: Classification and regression tree analysis of all polymorphisms, age and race resulted in gender-specific predictors of overall survival, progression-free survival and tumor response. Polymorphisms in the following genes were associated with gender-specific clinical outcome: estrogen receptor ß, EGF receptor, xeroderma pigmentosum group D, voltage-gated sodium channel and phospholipase A2. CONCLUSION: Genetic profiling to predict the clinical outcome of patients with metastatic colorectal cancer may depend on gender.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/genetics , Gene Expression Profiling , Sex Characteristics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Genotype , Humans , Male , Middle Aged , Neoplasm Metastasis , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Polymerase Chain Reaction , Polymorphism, Genetic
12.
Int J Cancer ; 128(9): 2038-49, 2011 May 01.
Article in English | MEDLINE | ID: mdl-20648559

ABSTRACT

Interleukin-8 (IL-8), a chemokine with a defining CXC amino acid motif, is known to possess tumorigenic and proangiogenic properties. Overexpression of IL-8 has been detected in many human tumors, including colorectal cancer (CRC), and is associated with poor prognosis. The goal of our study was to determine the role of IL-8 overexpression in CRC cells in vitro and in vivo. We stably transfected the IL-8 cDNA into two human colon cancer cell lines, HCT116 and Caco2, and selected IL-8-secreting transfectants. Real-time RT-PCR confirmed that IL-8 mRNA was overexpressed in IL-8 transfectants with 45- to 85-fold higher than parental cells. The IL-8-transfected clones secreted 19- to 28-fold more IL-8 protein than control and parental cells as detected by ELISA. The IL-8 transfectants demonstrated increased cellular proliferation, cell migration and invasion based on functional assays. Growth inhibition studies showed that IL-8 overexpression lead to a significant resistance to oxaliplatin (p < 0.0001). Inhibition of IL-8 overexpression with small interfering RNA reversed the observed increases in tumorigenic functions and oxaliplatin resistance, suggesting that IL-8 not only provides a proliferative advantage but also promotes the metastatic potential of colon cancer cells. Using a tumor xenograft model, IL-8-expressing cells formed significantly larger tumors than the control cells with increased microvessel density. Together, these findings indicate that overexpression of IL-8 promotes tumor growth, metastasis, chemoresistance and angiogenesis, implying IL-8 to be an important therapeutic target in CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Interleukin-8/metabolism , Neovascularization, Pathologic/metabolism , Animals , Blotting, Western , Caco-2 Cells , Colonic Neoplasms/blood supply , Drug Resistance, Neoplasm/physiology , Enzyme-Linked Immunosorbent Assay , HCT116 Cells , Humans , Immunohistochemistry , Mice , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
13.
Cancer Chemother Pharmacol ; 65(5): 979-88, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20062993

ABSTRACT

PURPOSE: We conducted a phase I/II clinical trial to determine the safety and feasibility of combining vorinostat with 5-fluorouracil (5-FU) in patients with metastatic colorectal cancer (mCRC) and elevated intratumoral thymidylate synthase (TS). METHODS: Patients with mCRC who had failed all standard therapeutic options were eligible. Intratumoral TS mRNA expression and peripheral blood mononuclear cell (PBMC) histone acetylation were measured before and after 6 consecutive days of vorinostat treatment at 400 mg PO daily. 5-FU/LV were given on days 6 and 7 and repeated every 2 weeks, along with continuous daily vorinostat. Dose escalation occurred in cohorts of three to six patients. RESULTS: Ten patients were enrolled. Three dose levels were explored in the phase I portion of the study. Two dose-limiting toxicities (DLTs) were observed at the starting dose level, which resulted in dose de-escalation to levels -1 and -2. Given the occurrence of two DLTs at each of the dose levels, we were unable to establish a maximum tolerated dose (MTD). Two patients achieved significant disease stabilization for 4 and 6 months. Grade 3 and 4 toxicities included fatigue, thrombocytopenia and mucositis. Intratumoral TS downregulation > or = 50% was observed in one patient only. Acetylation of histone 3 was observed in PBMCs following vorinostat treatment. CONCLUSIONS: The study failed to establish a MTD and was terminated. The presence of PBMC histone acetylation indicates biological activity of vorinostat, however, consistent reductions in intratumoral TS mRNA were not observed. Alternate vorinostat dose-scheduling may alleviate the toxicity and achieve optimal TS downregulation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Hydroxamic Acids/therapeutic use , Adult , Aged , Colorectal Neoplasms/enzymology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Fluorouracil/administration & dosage , Histone Acetyltransferases/metabolism , Humans , Hydroxamic Acids/administration & dosage , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Leukocytes, Mononuclear/enzymology , Male , Maximum Tolerated Dose , Middle Aged , RNA, Messenger/metabolism , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Treatment Failure , Vorinostat
14.
BMC Med Genomics ; 2: 67, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19948057

ABSTRACT

BACKGROUND: Despite the significant progress made in colon cancer chemotherapy, advanced disease remains largely incurable and novel efficacious chemotherapies are urgently needed. Histone deacetylase inhibitors (HDACi) represent a novel class of agents which have demonstrated promising preclinical activity and are undergoing clinical evaluation in colon cancer. The goal of this study was to identify genes in colon cancer cells that are differentially regulated by two clinically advanced hydroxamic acid HDACi, vorinostat and LBH589 to provide rationale for novel drug combination partners and identify a core set of HDACi-regulated genes. METHODS: HCT116 and HT29 colon cancer cells were treated with LBH589 or vorinostat and growth inhibition, acetylation status and apoptosis were analyzed in response to treatment using MTS, Western blotting and flow cytometric analyses. In addition, gene expression was analyzed using the Illumina Human-6 V2 BeadChip array and Ingenuity Pathway Analysis. RESULTS: Treatment with either vorinostat or LBH589 rapidly induced histone acetylation, cell cycle arrest and inhibited the growth of both HCT116 and HT29 cells. Bioinformatic analysis of the microarray profiling revealed significant similarity in the genes altered in expression following treatment with the two HDACi tested within each cell line. However, analysis of genes that were altered in expression in the HCT116 and HT29 cells revealed cell-line-specific responses to HDACi treatment. In addition a core cassette of 11 genes modulated by both vorinostat and LBH589 were identified in both colon cancer cell lines analyzed. CONCLUSION: This study identified HDACi-induced alterations in critical genes involved in nucleotide metabolism, angiogenesis, mitosis and cell survival which may represent potential intervention points for novel therapeutic combinations in colon cancer. This information will assist in the identification of novel pathways and targets that are modulated by HDACi, providing much-needed information on HDACi mechanism of action and providing rationale for novel drug combination partners. We identified a core signature of 11 genes which were modulated by both vorinostat and LBH589 in a similar manner in both cell lines. These core genes will assist in the development and validation of a common gene set which may represent a molecular signature of HDAC inhibition in colon cancer.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Acetylation , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Indoles , Oligonucleotide Array Sequence Analysis/methods , Panobinostat , Vorinostat
15.
Int J Cancer ; 125(12): 2957-69, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19536776

ABSTRACT

Members of the human epidermal receptor (HER) family are frequently associated with aggressive disease and poor prognosis in multiple malignancies. Lapatinib is a dual tyrosine kinase inhibitor targeting the epidermal growth factor receptor (EGFR) and HER-2. This study evaluated the therapeutic potential of lapatinib, alone and in combination with SN-38, the active metabolite of irinotecan (CPT-11), in colon and gastric cancer cell lines. Concentration-dependent antiproliferative effects of both lapatinib and SN-38 were observed in all colon and gastric cancer cell lines tested but varied significantly between individual cell lines (lapatinib range 0.08-11.7 muM; SN-38 range 3.6-256 nM). Lapatinib potently inhibited the growth of a HER-2 overexpressing gastric cancer cell line and demonstrated moderate activity in gastric and colon cancer cells with detectable HER-2 expression. The combination of lapatinib and SN-38 interacted synergistically to inhibit cell proliferation in all colon and gastric cancer cell lines tested. Cotreatment with lapatinib and SN-38 also resulted in enhanced cell cycle arrest and the induction of apoptosis with subsequent cellular pharmacokinetic analysis demonstrating that lapatinib promoted the increased intracellular accumulation and retention of SN-38 when compared to SN-38 treatment alone. Finally, the combination of lapatinib and CPT-11 demonstrated synergistic antitumor efficacy in the LoVo colon cancer mouse xenograft model with no apparent increase in toxicity compared to CPT-11 monotherapy. These results provide compelling preclinical rationale indicating lapatinib to be a potentially efficacious chemotherapeutic combination partner for irinotecan in the treatment of gastrointestinal carcinomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chromatography, Liquid , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colony-Forming Units Assay , ErbB Receptors/metabolism , Flow Cytometry , Humans , Irinotecan , Lapatinib , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Int J Cancer ; 125(2): 463-73, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19384949

ABSTRACT

Despite recent therapeutic advances, the response rates to chemotherapy for patients with metastatic colon cancer remain at approximately 50% with the fluoropyrimidine, 5-fluorouracil (5-FU), continuing to serve as the foundation chemotherapeutic agent for the treatment of this disease. Previous studies have demonstrated that overexpression of thymidylate synthase (TS) is a key determinant of resistance to 5-FU-based chemotherapy. Therefore, there is a significant need to develop alternative therapeutic strategies to overcome TS-mediated resistance. In this study, we demonstrate that the histone deacetylase inhibitors (HDACi) vorinostat and LBH589 significantly downregulate TS gene expression in a panel of colon cancer cell lines. Downregulation of TS was independent of p53, p21 and HDAC2 expression and was achievable in vivo as demonstrated by mouse xenograft models. We provide evidence that HDACi treatment leads to a potent transcriptional repression of the TS gene. Combination of the fluoropyrimidines 5-FU or FUdR with both vorinostat and LBH589 enhanced cell cycle arrest and growth inhibition. Importantly, the downstream effects of TS inhibition were significantly enhanced by this combination including the inhibition of acute TS induction and the enhanced accumulation of the cytotoxic nucleotide intermediate dUTP. These data demonstrate that HDACi repress TS expression at the level of transcription and provides the first evidence suggesting a direct mechanistic link between TS downregulation and the synergistic interaction observed between HDACi and 5-FU. This study provides rationale for the continued clinical evaluation of HDACi in combination with 5-FU-based therapies as a strategy to overcome TS-mediated resistance.


Subject(s)
Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylase Inhibitors , Pyrimidines/pharmacology , Thymidylate Synthase/genetics , Base Sequence , Cell Division/drug effects , Cell Line, Tumor , Colonic Neoplasms/enzymology , DNA Primers , Drug Synergism , Flow Cytometry , Humans , Polymerase Chain Reaction
17.
Cancer Res ; 69(9): 3736-45, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366806

ABSTRACT

The receptor tyrosine kinase EphB2 is expressed by colon progenitor cells; however, only 39% of colorectal tumors express EphB2 and expression levels decline with disease progression. Conversely, EphB4 is absent in normal colon but is expressed in all 102 colorectal cancer specimens analyzed, and its expression level correlates with higher tumor stage and grade. Both EphB4 and EphB2 are regulated by the Wnt pathway, the activation of which is critically required for the progression of colorectal cancer. Differential usage of transcriptional coactivator cyclic AMP-responsive element binding protein-binding protein (CBP) over p300 by the Wnt/beta-catenin pathway is known to suppress differentiation and increase proliferation. We show that the beta-catenin-CBP complex induces EphB4 and represses EphB2, in contrast to the beta-catenin-p300 complex. Gain of EphB4 provides survival advantage to tumor cells and resistance to innate tumor necrosis factor-related apoptosis-inducing ligand-mediated cell death. Knockdown of EphB4 inhibits tumor growth and metastases. Our work is the first to show that EphB4 is preferentially induced in colorectal cancer, in contrast to EphB2, whereby tumor cells acquire a survival advantage.


Subject(s)
Colorectal Neoplasms/enzymology , Receptor, EphB2/biosynthesis , Receptor, EphB4/biosynthesis , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Fluorescent Antibody Technique , HT29 Cells , Humans , Immunoblotting , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Receptor, EphB4/genetics , Transfection , Transplantation, Heterologous , beta Catenin/metabolism
18.
Nucleic Acids Res ; 37(1): 78-95, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19015155

ABSTRACT

Deoxyuridine triphosphate nucleotidohydrolase (dUTPase) catalyzes the hydrolysis of dUTP to dUMP and PPi. Although dUTP is a normal intermediate in DNA synthesis, its accumulation and misincorporation into DNA is lethal. Importantly, uracil misincorporation is a mechanism of cytotoxicity induced by fluoropyrimidine chemotherapeutic agents including 5-fluorouracil (5-FU) and elevated expression of dUTPase is negatively correlated with clinical response to 5-FU-therapy. In this study we performed the first functional characterization of the dUTPase promoter and demonstrate a role for E2F-1 and Sp1 in driving dUTPase expression. We establish a direct role for both mutant and wild-type forms of p53 in modulating dUTPase promoter activity. Treatment of HCT116 p53(+/+) cells with the DNA-damaging agent oxaliplatin induced a p53-dependent transcriptional downregulation of dUTPase not observed in the isogenic null cell line. Oxaliplatin treatment induced enrichment of p53 at the dUTPase promoter with a concomitant reduction in Sp1. The suppression of dUTPase by oxaliplatin promoted increased levels of dUTP that was enhanced by subsequent addition of fluoropyrimidines. The novel observation that oxaliplatin downregulates dUTPase expression may provide a mechanistic basis contributing to the synergy observed between 5-FU and oxaliplatin in the clinic. Furthermore, these studies provide the first evidence of a direct transcriptional link between the essential enzyme dUTPase and the tumor suppressor p53.


Subject(s)
Antineoplastic Agents/toxicity , Gene Expression Regulation, Neoplastic , Organoplatinum Compounds/toxicity , Pyrophosphatases/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , Deoxyuracil Nucleotides/biosynthesis , Down-Regulation , Gene Expression Regulation, Enzymologic , Humans , Oxaliplatin , Promoter Regions, Genetic , Pyrophosphatases/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sp1 Transcription Factor/physiology , Thymidylate Synthase/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
19.
Clin Cancer Res ; 14(23): 7884-95, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047118

ABSTRACT

PURPOSE: Recently, an objective response rate of 12% was reported in a phase II study of cetuximab in patients with epidermal growth factor receptor (EGFR)-expressing metastatic colorectal cancer (mCRC) refractory to fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy (IMC-0144). In this large molecular correlates study, we tested whether K-ras mutation status and polymorphisms in genes involved in the EGFR-signaling pathway were associated with clinical outcome in IMC-0144. EXPERIMENTAL DESIGN: We analyzed all available tissue samples from 130 of 346 mCRC patients enrolled in the IMC-0144 phase II clinical trial of cetuximab. Genomic DNA was extracted from formalin-fixed paraffin-embedded tumor tissues, and K-ras mutation status and the genotypes were analyzed using PCR-RFLP, direct DNA-sequencing, and 5'-end [gamma-33P] ATP-labeled PCR-protocols. RESULTS: The PFS of patients with cyclooxygenase-2 (COX-2) -765 G>C [C/C; risk ratio (RR), 0.31; 95% confidence interval (95% CI), 0.12-0.84; P = 0.032], COX-2 +8473 T>C (C/C; RR, 0.67; 95% CI, 0.40-1.13; P = 0.003), EGF +61 A>G (G/G; RR, 0.57; 95% CI, 0.34-0.95; P = 0.042), and EGFR +497 G>A (A/G; RR, 0.82; 95% CI, 0.56-1.20; P = 0.017) genotypes was significantly longer compared with those with other genotypes. In addition, patients whose tumors did not have K-ras mutations showed better RR, PFS, and overall survival than patients with K-ras mutations. In multivariable analysis, COX-2 +8473 T>C (adjusted P = 0.013) and EGFR +497 G>A (adjusted P = 0.010) remained significantly associated with progression-free survival, independent of skin rash toxicity, K-ras mutation status, and Eastern Cooperative Group performance status. CONCLUSIONS: Polymorphisms in COX-2 and EGFR may be useful independent molecular markers to predict clinical outcome in patients with mCRC treated with single-agent cetuximab, independent of skin rash toxicity, K-ras mutation, and Eastern Cooperative Oncology Group performance status.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , ErbB Receptors/genetics , Antibodies, Monoclonal, Humanized , Cetuximab , Clinical Trials, Phase II as Topic , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease-Free Survival , Epidermal Growth Factor/genetics , Genes, ras/genetics , Humans , Kaplan-Meier Estimate , Multicenter Studies as Topic , Mutation , Polymorphism, Restriction Fragment Length
20.
Mol Cancer Ther ; 7(9): 3029-37, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790783

ABSTRACT

For over 40 years, the fluoropyrimidine 5-fluorouracil (5-FU) has remained the central agent in therapeutic regimens employed in the treatment of colorectal cancer and is frequently combined with the DNA-damaging agents oxaliplatin and irinotecan, increasing response rates and improving overall survival. However, many patients will derive little or no benefit from treatment, highlighting the need to identify novel therapeutic targets to improve the efficacy of current 5-FU-based chemotherapeutic strategies. dUTP nucleotidohydrolase (dUTPase) catalyzes the hydrolysis of dUTP to dUMP and PPi, providing substrate for thymidylate synthase (TS) and DNA synthesis and repair. Although dUTP is a normal intermediate in DNA synthesis, its accumulation and misincorporation into DNA as uracil is lethal. Importantly, uracil misincorporation represents an important mechanism of cytotoxicity induced by the TS-targeted class of chemotherapeutic agents including 5-FU. A growing body of evidence suggests that dUTPase is an important mediator of response to TS-targeted agents. In this article, we present further evidence showing that elevated expression of dUTPase can protect breast cancer cells from the expansion of the intracellular uracil pool, translating to reduced growth inhibition following treatment with 5-FU. We therefore report the implementation of in silico drug development techniques to identify and develop small-molecule inhibitors of dUTPase. As 5-FU and the oral 5-FU prodrug capecitabine remain central agents in the treatment of a variety of malignancies, the clinical utility of a small-molecule inhibitor to dUTPase represents a viable strategy to improve the clinical efficacy of these mainstay chemotherapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Pyrophosphatases/antagonists & inhibitors , Thymidine/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology , DNA Damage , Drug Design , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Humans , Immunohistochemistry , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...