Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374558

ABSTRACT

High levels of DNA methylation at CpG loci are associated with transcriptional repression of tumor suppressor genes and dysregulation of DNA repair genes. Human papilloma virus (HPV)-associated head and neck squamous cell carcinomas (HNSCC) have high levels of DNA methylation and methylation has been associated with dampening of an innate immune response in virally infected cells. We have been exploring demethylation as a potential treatment in HPV+ HNSCC and recently reported results of a window clinical trial showing that HNSCCs are particularly sensitive to demethylating agent 5-azacytidine (5-aza). Mechanistically, sensitivity is partially due to downregulation of HPV genes expression and restoration of tumor suppressors p53 and Rb. Here, for the first time, we show that 5-azaC treatment of HPV+ HNSCC induces replication and transcription-associated DNA double strand breaks (DSBs) that occur preferentially at demethylated genomic DNA. Blocking replication or transcription prevented formation of DNA DSBs and reduced sensitivity of HPV-positive head and neck cancer cells to 5-azaC, demonstrating that both replication and active transcription are required for formation of DSBs associated with 5-azaC.

2.
Clin Cancer Res ; 23(23): 7276-7287, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28916527

ABSTRACT

Purpose: DNA methylation in human papillomavirus-associated (HPV+) head and neck squamous cell carcinoma (HNSCC) may have importance for continuous expression of HPV oncogenes, tumor cell proliferation, and survival. Here, we determined activity of a global DNA-demethylating agent, 5-azacytidine (5-aza), against HPV+ HNSCC in preclinical models and explored it as a targeted therapy in a window trial enrolling patients with HPV+ HNSCC.Experimental Design: Sensitivity of HNSCC cells to 5-aza treatment was determined, and then 5-aza activity was tested in vivo using xenografted tumors in a mouse model. Finally, tumor samples from patients enrolled in a window clinical trial were analyzed to identify activity of 5-aza therapy in patients with HPV+ HNSCC.Results: Clinical trial and experimental data show that 5-aza induced growth inhibition and cell death in HPV+ HNSCC. 5-aza reduced expression of HPV genes, stabilized p53, and induced p53-dependent apoptosis in HNSCC cells and tumors. 5-aza repressed expression and activity of matrix metalloproteinases (MMP) in HPV+ HNSCC, activated IFN response in some HPV+ head and neck cancer cells, and inhibited the ability of HPV+ xenografted tumors to invade mouse blood vessels.Conclusions: 5-aza may provide effective therapy for HPV-associated HNSCC as an alternative or complement to standard cytotoxic therapy. Clin Cancer Res; 23(23); 7276-87. ©2017 AACR.


Subject(s)
Azacitidine/therapeutic use , Carcinoma, Squamous Cell/drug therapy , DNA Methylation/drug effects , Head and Neck Neoplasms/drug therapy , Papillomavirus Infections/drug therapy , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Humans , Mice, Nude , Papillomaviridae/drug effects , Papillomaviridae/physiology , Papillomavirus Infections/genetics , Papillomavirus Infections/virology
3.
Oncotarget ; 7(25): 38598-38611, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27233076

ABSTRACT

Radiation and chemotherapy that are commonly used to treat human cancers damage cellular DNA. DNA damage appears to be more toxic to cancer cells than normal cells, most likely due to deregulated checkpoint activation and/or deficiency in DNA repair pathways that are characteristics of many tumors. However, unwanted side effects arise as a result of DNA damage to normal cells during the treatment.Here, we show that roscovitine, a cyclin-dependent kinase (CDK) inhibitor that inhibits CDK-1, CDK-2, CDK-5, CDK-7, and CDK-9 due to competitive binding to the ATP site on the kinases, causes significant DNA damage followed by p53-dependent cell death in human papilloma virus (HPV)-positive, but not in HPV-negative, head and neck cancer cells. Since HPV positivity was a molecular marker for increased sensitivity of cells to roscovitine, we reasoned that systemic roscovitine administration would not be toxic to healthy HPV-negative tissue. Indeed, low roscovitine doses significantly inhibited the growth of HPV-associated xenografted tumors in mice without causing any detectable side effects.Given that inhibition of CDKs has been shown to inhibit replication of several viruses, we suggest that roscovitine treatment may represent a selective and safe targeted therapeutic option against HPV-positive head and neck cancer.


Subject(s)
Head and Neck Neoplasms/drug therapy , Purines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Female , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Mice , Mice, Nude , Papillomaviridae/isolation & purification , Papillomavirus Infections/pathology , Protein Kinase Inhibitors/pharmacology , Roscovitine , Xenograft Model Antitumor Assays
4.
Sci Transl Med ; 7(312): 312ra176, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26537256

ABSTRACT

Amplification of the MYCN oncogene predicts treatment resistance in childhood neuroblastoma. We used a MYC target gene signature that predicts poor neuroblastoma prognosis to identify the histone chaperone FACT (facilitates chromatin transcription) as a crucial mediator of the MYC signal and a therapeutic target in the disease. FACT and MYCN expression created a forward feedback loop in neuroblastoma cells that was essential for maintaining mutual high expression. FACT inhibition by the small-molecule curaxin compound CBL0137 markedly reduced tumor initiation and progression in vivo. CBL0137 exhibited strong synergy with standard chemotherapy by blocking repair of DNA damage caused by genotoxic drugs, thus creating a synthetic lethal environment in MYCN-amplified neuroblastoma cells and suggesting a treatment strategy for MYCN-driven neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , High Mobility Group Proteins/antagonists & inhibitors , Nervous System Neoplasms/drug therapy , Nervous System Neoplasms/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Elongation Factors/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Humans , Molecular Targeted Therapy , Signal Transduction/drug effects , Transcriptional Elongation Factors/metabolism
5.
Cancer Res ; 75(22): 4728-41, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26404003

ABSTRACT

Activation of Notch signaling in hematopoietic cells by tumors contributes to immune escape. T-cell defects in tumors can be reversed by treating tumor-bearing mice with multivalent forms of the Notch receptor ligand DLL-1, but the immunologic correlates of this effect have not been elucidated. Here, we report mechanistic insights along with the efficacy of combinational treatments of multivalent DLL-1 with oncoprotein targeting drugs in preclinical mouse models of lung cancer. Systemic DLL-1 administration increased T-cell infiltration into tumors and elevated numbers of CD44(+)CD62L(+)CD8(+) memory T cells while decreasing the number of regulatory T cells and limiting tumor vascularization. This treatment was associated with upregulation of Notch and its ligands in tumor-infiltrating T cells enhanced expression of T-bet and phosphorylation of Stat1/2. Adoptive transfer of T cells from DLL1-treated tumor-bearing immunocompetent hosts into tumor-bearing SCID-NOD immunocompromised mice attenuated tumor growth and extended tumor-free survival in the recipients. When combined with the EGFR-targeted drug erlotinib, DLL-1 significantly improved progression-free survival by inducing robust tumor-specific T-cell immunity. In tissue culture, DLL1 induced proliferation of human peripheral T cells, but lacked proliferative or clonogenic effects on lung cancer cells. Our findings offer preclinical mechanistic support for the development of multivalent DLL1 to stimulate antitumor immunity.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium-Binding Proteins , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Female , Flow Cytometry , Humans , Immunohistochemistry , Immunotherapy/methods , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/agonists , Recombinant Fusion Proteins/pharmacology
6.
Clin Cancer Res ; 20(16): 4274-88, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25125259

ABSTRACT

PURPOSE: To identify novel therapeutic drug targets for p53-mutant head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: RNAi kinome viability screens were performed on HNSCC cells, including autologous pairs from primary tumor and recurrent/metastatic lesions, and in parallel on murine squamous cell carcinoma (MSCC) cells derived from tumors of inbred mice bearing germline mutations in Trp53, and p53 regulatory genes: Atm, Prkdc, and p19(Arf). Cross-species analysis of cell lines stratified by p53 mutational status and metastatic phenotype was used to select 38 kinase targets. Both primary and secondary RNAi validation assays were performed on additional HNSCC cell lines to credential these kinase targets using multiple phenotypic endpoints. Kinase targets were also examined via chemical inhibition using a panel of kinase inhibitors. A preclinical study was conducted on the WEE1 kinase inhibitor, MK-1775. RESULTS: Our functional kinomics approach identified novel survival kinases in HNSCC involved in G2-M cell-cycle checkpoint, SFK, PI3K, and FAK pathways. RNAi-mediated knockdown and chemical inhibition of the WEE1 kinase with a specific inhibitor, MK-1775, had a significant effect on both viability and apoptosis. Sensitivity to the MK-1775 kinase inhibitor is in part determined by p53 mutational status, and due to unscheduled mitotic entry. MK-1775 displays single-agent activity and potentiates the efficacy of cisplatin in a p53-mutant HNSCC xenograft model. CONCLUSIONS: WEE1 kinase is a potential therapeutic drug target for HNSCC. This study supports the application of a functional kinomics strategy to identify novel therapeutic targets for cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA Interference , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
7.
Clin Cancer Res ; 20(9): 2300-11, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24599934

ABSTRACT

PURPOSE: Human papilloma virus (HPV)-associated (HPV+) oropharyngeal squamous cell carcinomas (OPSCC) have different molecular and biologic characteristics and clinical behavior compared with HPV-negative (HPV-) OPSCC. PIK3CA mutations are more common in HPV(+) OPSCC. To define molecular differences and tumor subsets, protein expression and phosphorylation were compared between HPV(+) and HPV(-) OPSCC and between tumors with and without PIK3CA mutations. EXPERIMENTAL DESIGN: Expression of 137 total and phosphorylated proteins was evaluated by reverse-phase protein array in 29 HPV(+) and 13 HPV(-) prospectively collected OPSCCs. Forty-seven OPSCCs were tested for hotspot-activating mutations in PIK3CA and AKT. Activation of PIK3CA downstream targets and sensitivity to pathway inhibitors were determined in HPV(+) head and neck cancer cells overexpressing wild-type or mutant PIK3CA. RESULTS: Analyses revealed 41 differentially expressed proteins between HPV(+) and HPV(-) OPSCC categorized into functional groups: DNA repair, cell cycle, apoptosis, phosphoinositide 3-kinase (PI3K)/AKT/mTOR, and receptor kinase pathways. All queried DNA repair proteins were significantly upregulated in HPV(+) samples. A total of 8 of 33 HPV(+) and 0 of 14 HPV(-) tumors contained activating PIK3CA mutations. Despite all activating PIK3CA mutations occurring in HPV(+) samples, HPV(+) tumors had lower mean levels of activated AKT and downstream AKT target phosphorylation. Ectopic expression of mutant PIK3CA in HPV(+) cells increased mTOR, but not AKT activity. HPV E6/E7 overexpression inhibited AKT phosphorylation in HPV-negative cells. Mutant PIK3CA overexpressing cells were more sensitive to a dual PI3K/mTOR inhibitor compared with an AKT inhibitor. CONCLUSIONS: Protein expression analyses suggest that HPV(+) and HPV(-) OPSCC differentially activate DNA repair, cell cycle, apoptosis, PI3K/AKT/mTOR, and receptor kinase pathways. PIK3CA mutations are more common in HPV(+) OPSCC and are associated with activation of mTOR, but not AKT. These data suggest that inhibitors for mTOR may have activity against HPV(+) PIK3CA mutant oropharyngeal cancers.


Subject(s)
Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Mutation , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/virology , Papillomaviridae , Papillomavirus Infections/complications , Phosphatidylinositol 3-Kinases/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/therapy , Humans , Male , Middle Aged , Neoplasm Staging , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/mortality , Oropharyngeal Neoplasms/therapy , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Protein Array Analysis , Risk Factors
8.
Mol Pharmacol ; 85(1): 62-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24136993

ABSTRACT

Interstitial adenosine stimulates neovascularization in part through A2B adenosine receptor-dependent upregulation of vascular endothelial growth factor (VEGF). In the current study, we tested the hypothesis that A2B receptors upregulate JunB, which can contribute to stimulation of VEGF production. Using the human microvascular endothelial cell line, human mast cell line, mouse cardiac Sca1-positive stromal cells, and mouse Lewis lung carcinoma (LLC) cells, we found that adenosine receptor-dependent upregulation of VEGF production was associated with an increase in VEGF transcription, activator protein-1 (AP-1) activity, and JunB accumulation in all cells investigated. Furthermore, the expression of JunB, but not the expression of other genes encoding transcription factors from the Jun family, was specifically upregulated. In LLC cells expressing A2A and A2B receptor transcripts, only the nonselective adenosine agonist NECA (5'-N-ethylcarboxamidoadenosine), but not the selective A2A receptor agonist CGS21680 [2-p-(2-carboxyethyl) phenylethylamino-5'-N-ethylcarboxamidoadenosine], significantly increased JunB reporter activity and JunB nuclear accumulation, which were inhibited by the A2B receptor antagonist PSB603 [(8-[4-[4-((4-chlorophenzyl)piperazide-1-sulfonyl)phenyl]]-1-propylxanthine]. Using activators and inhibitors of intracellular signaling, we demonstrated that A2B receptor-dependent accumulation of JunB protein and VEGF secretion share common intracellular pathways. NECA enhanced JunB binding to the murine VEGF promoter, whereas mutation of the high-affinity AP-1 site (-1093 to -1086) resulted in a loss of NECA-dependent VEGF reporter activity. Finally, NECA-dependent VEGF secretion and reporter activity were inhibited by the expression of a dominant negative JunB or by JunB knockdown. Thus, our data suggest an important role of the A2B receptor-dependent upregulation of JunB in VEGF production and possibly other AP-1-regulated events.


Subject(s)
Receptor, Adenosine A2B/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Adenosine/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Humans , Mice , Mutation , Promoter Regions, Genetic , RNA Interference , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
9.
Am J Physiol Renal Physiol ; 304(10): F1295-307, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23486012

ABSTRACT

Aquaporin 11 (AQP11) is a newly described member of the protein family of transport channels. AQP11 associates with the endoplasmic reticulum (ER) and is highly expressed in proximal tubular epithelial cells in the kidney. Previously, we identified and characterized a recessive mutation of the highly conserved Cys227 to Ser227 in mouse AQP11 that caused proximal tubule (PT) injury and kidney failure in mutant mice. The current study revealed induction of ER stress, unfolded protein response, and apoptosis as molecular mechanisms of this PT injury. Cys227Ser mutation interfered with maintenance of AQP11 oligomeric structure. AQP11 is abundantly expressed in the S1 PT segment, a site of major renal glucose flux, and Aqp11 mutant mice developed PT-specific mitochondrial injury. Glucose increased AQP11 protein expression in wild-type kidney and upregulation of AQP11 expression by glucose in vitro was prevented by phlorizin, an inhibitor of sodium-dependent glucose transport across PT. Total AQP11 levels in heterozygotes were higher than in wild-type mice but were not further increased in response to glucose. In Aqp11 insufficient PT cells, glucose potentiated increases in reactive oxygen species (ROS) production. ROS production was also elevated in Aqp11 mutation carriers. Phenotypically normal mice heterozygous for the Aqp11 mutation repeatedly treated with glucose showed increased blood urea nitrogen levels that were prevented by the antioxidant sulforaphane or by phlorizin. Our results indicate an important role for AQP11 to prevent glucose-induced oxidative stress in proximal tubules.


Subject(s)
Aquaporins/genetics , Endoplasmic Reticulum/metabolism , Kidney/metabolism , Oxidative Stress/genetics , Renal Insufficiency/genetics , Animals , Aquaporins/metabolism , Cell Line , Endoplasmic Reticulum Stress/physiology , Mice , Mutation , Reactive Oxygen Species/metabolism , Renal Insufficiency/metabolism , Up-Regulation
10.
J Immunol ; 187(11): 6120-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22039302

ABSTRACT

Extracellular adenosine and purine nucleotides are elevated in many pathological situations associated with the expansion of CD11b(+)Gr1(+) myeloid-derived suppressor cells (MDSCs). Therefore, we tested whether adenosinergic pathways play a role in MDSC expansion and functions. We found that A(2B) adenosine receptors on hematopoietic cells play an important role in accumulation of intratumoral CD11b(+)Gr1(high) cells in a mouse Lewis lung carcinoma model in vivo and demonstrated that these receptors promote preferential expansion of the granulocytic CD11b(+)Gr1(high) subset of MDSCs in vitro. Flow cytometry analysis of MDSCs generated from mouse hematopoietic progenitor cells revealed that the CD11b(+)Gr-1(high) subset had the highest levels of CD73 (ecto-5'-nucleotidase) expression (Δmean fluorescence intensity [MFI] of 118.5 ± 16.8), followed by CD11b(+)Gr-1(int) (ΔMFI of 57.9 ± 6.8) and CD11b(+)Gr-1(-/low) (ΔMFI of 12.4 ± 1.0) subsets. Even lower levels of CD73 expression were found on Lewis lung carcinoma tumor cells (ΔMFI of 3.2 ± 0.2). The high levels of CD73 expression in granulocytic CD11b(+)Gr-1(high) cells correlated with high levels of ecto-5'-nucleotidase enzymatic activity. We further demonstrated that the ability of granulocytic MDSCs to suppress CD3/CD28-induced T cell proliferation was significantly facilitated in the presence of the ecto-5'-nucleotidase substrate 5'-AMP. We propose that generation of adenosine by CD73 expressed at high levels on granulocytic MDSCs may promote their expansion and facilitate their immunosuppressive activity.


Subject(s)
Adenosine/metabolism , Granulocytes/metabolism , Myeloid Cells/metabolism , 5'-Nucleotidase/immunology , 5'-Nucleotidase/metabolism , Adenosine/immunology , Animals , CD11b Antigen/immunology , CD11b Antigen/metabolism , Carcinoma, Lewis Lung , Cell Differentiation/immunology , Cell Proliferation , Cell Separation , Female , Flow Cytometry , Granulocytes/cytology , Granulocytes/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P1/immunology , Receptors, Purinergic P1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...