Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Bioorg Med Chem Lett ; 27(24): 5473-5480, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29150397

ABSTRACT

In this work, we describe the use of the rule of 3 fragment-based strategies from biochemical screening data of 1100 in-house, small, low molecular weight fragments. The sequential combination of in silico fragment hopping and fragment linking based on S160/Y161/A162 hinge residues hydrogen bonding interactions leads to the identification of novel 1H-benzo[d]imidazol-2-yl)-1H-indazol class of Phosphoinositide-Dependent Kinase-1 (PDK1) inhibitors. Consequent SAR and follow-up screening data led to the discovery of two potent PDK1 inhibitors: compound 32 and 35, with an IC50 of 80 nM and 94 nM, respectively. Further biological evaluation showed that, at the low nanomolar concentration, the drug had potent ability to inhibit phosphorylation of AKT and p70S6, and selectively kill the cancer cells with mutations in both PTEN and PI3K. The microarray data showed that DUSP6, DUSP4, and FOSL1 were down-regulated in the sensitive cell lines with the compound treatment. The in vivo test showed that 35 can significantly inhibit tumor growth without influencing body weight growth. Our results suggest that these compounds, especially 35, merit further pre-clinical evaluation.


Subject(s)
Drug Design , Indazoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Down-Regulation/drug effects , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Humans , Imidazoles/chemistry , Indazoles/chemical synthesis , Indazoles/pharmacology , Inhibitory Concentration 50 , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Molecular Docking Simulation , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Structure-Activity Relationship
2.
J Biol Chem ; 288(4): 2261-70, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23212921

ABSTRACT

During the process of branching morphogenesis, the mammary gland undergoes distinct phases of remodeling to form an elaborate ductal network that ultimately produces and delivers milk to newborn animals. These developmental events rely on tight regulation of critical cellular pathways, many of which are probably disrupted during initiation and progression of breast cancer. Transgenic mouse and in vitro organoid models previously identified growth factor signaling as a key regulator of mammary branching, but the functional downstream targets of these pathways remain unclear. Here, we used purified primary mammary epithelial cells stimulated with fibroblast growth factor-2 (FGF2) to model mammary branching morphogenesis in vitro. We employed a forward chemical genetic approach to identify modulators of this process and describe a potent compound, 1023, that blocks FGF2-induced branching. In primary mammary epithelial cells, we used lentivirus-mediated knockdown of the aryl hydrocarbon receptor (AHR) to demonstrate that 1023 acts through AHR to block branching. Using 1023 as a tool, we identified desmosomal adhesion as a novel target of AHR signaling and show that desmosomes are critical for AHR agonists to block branching. Our findings support a functional role for desmosomes during mammary morphogenesis and also in blocking FGF-induced invasion.


Subject(s)
Desmosomes/metabolism , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Animals , Cell Adhesion , Cells, Cultured , Collagen/chemistry , Down-Regulation , Drug Combinations , Epithelial Cells/cytology , Female , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factors/metabolism , Genetic Techniques , Laminin/chemistry , Mammary Glands, Animal/physiology , Mice , Morphogenesis , Proteoglycans/chemistry , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction
3.
Exp Cell Res ; 317(3): 307-18, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21075102

ABSTRACT

HGF signaling induces epithelial cells to disassemble cadherin-based adhesion and increase cell motility and invasion, a process termed epithelial-mesenchymal transition (EMT). EMT plays a major role in cancer metastasis, allowing individual cells to detach from the primary tumor, invade local tissue, and colonize distant tissues with new tumors. While invasion of vascular and lymphatic networks is the predominant route of metastasis, nerves also can act as networks for dissemination of cancer cell to distant sites in a process termed perineual invasion (PNI). Signaling between nerves and invasive cancer cells remains poorly understood, as does cellular decision making that selects the specific route of invasion. Here we examine how HGF signaling contributes to PNI using reductionist culture model systems. We find that TGFß, produced by PC12 cells, enhances scattering in response to HGF stimulation, increasing both cell-cell junction disassembly and cell migration. Further, gradients of TGFß induce migratory mesenchymal cells to undergo chemotaxis towards the source of TGFß. Interestingly, VEGF suppresses TGFß-induced enhancement of scattering. These results have broad implications for how combinatorial growth factor signaling contributes to cancer metastasis, suggesting that VEGF and TGFß might modulate HGF signaling to influence route selection during cancer progression.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hepatocyte Growth Factor/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Cell Movement , Neoplasm Metastasis , PC12 Cells , Rats , Signal Transduction , Transforming Growth Factor beta/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
4.
Nat Commun ; 12(1): 5337, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504101

ABSTRACT

TNK1 is a non-receptor tyrosine kinase with poorly understood biological function and regulation. Here, we identify TNK1 dependencies in primary human cancers. We also discover a MARK-mediated phosphorylation on TNK1 at S502 that promotes an interaction between TNK1 and 14-3-3, which sequesters TNK1 and inhibits its kinase activity. Conversely, the release of TNK1 from 14-3-3 allows TNK1 to cluster in ubiquitin-rich puncta and become active. Active TNK1 induces growth factor-independent proliferation of lymphoid cells in cell culture and mouse models. One unusual feature of TNK1 is a ubiquitin-association domain (UBA) on its C-terminus. Here, we characterize the TNK1 UBA, which has high affinity for poly-ubiquitin. Point mutations that disrupt ubiquitin binding inhibit TNK1 activity. These data suggest a mechanism in which TNK1 toggles between 14-3-3-bound (inactive) and ubiquitin-bound (active) states. Finally, we identify a TNK1 inhibitor, TP-5801, which shows nanomolar potency against TNK1-transformed cells and suppresses tumor growth in vivo.


Subject(s)
14-3-3 Proteins/genetics , Fetal Proteins/genetics , Lymphocytes/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein-Tyrosine Kinases/genetics , Ubiquitin/genetics , 14-3-3 Proteins/metabolism , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Fetal Proteins/antagonists & inhibitors , Fetal Proteins/metabolism , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lymphocytes/drug effects , Lymphocytes/pathology , Mice , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction , Survival Analysis , Tumor Burden/drug effects , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
5.
Cancer Res ; 80(7): 1551-1563, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31992541

ABSTRACT

Cytometry by time-of-flight (CyTOF) simultaneously measures multiple cellular proteins at the single-cell level and is used to assess intertumor and intratumor heterogeneity. This approach may be used to investigate the variability of individual tumor responses to treatments. Herein, we stratified lung tumor subpopulations based on AXL signaling as a potential targeting strategy. Integrative transcriptome analyses were used to investigate how TP-0903, an AXL kinase inhibitor, influences redundant oncogenic pathways in metastatic lung cancer cells. CyTOF profiling revealed that AXL inhibition suppressed SMAD4/TGFß signaling and induced JAK1-STAT3 signaling to compensate for the loss of AXL. Interestingly, high JAK1-STAT3 was associated with increased levels of AXL in treatment-naïve tumors. Tumors with high AXL, TGFß, and JAK1 signaling concomitantly displayed CD133-mediated cancer stemness and hybrid epithelial-to-mesenchymal transition features in advanced-stage patients, suggesting greater potential for distant dissemination. Diffusion pseudotime analysis revealed cell-fate trajectories among four different categories that were linked to clinicopathologic features for each patient. Patient-derived organoids (PDO) obtained from tumors with high AXL and JAK1 were sensitive to TP-0903 and ruxolitinib (JAK inhibitor) treatments, supporting the CyTOF findings. This study shows that single-cell proteomic profiling of treatment-naïve lung tumors, coupled with ex vivo testing of PDOs, identifies continuous AXL, TGFß, and JAK1-STAT3 signal activation in select tumors that may be targeted by combined AXL-JAK1 inhibition. SIGNIFICANCE: Single-cell proteomic profiling of clinical samples may facilitate the optimal selection of novel drug targets, interpretation of early-phase clinical trial data, and development of predictive biomarkers valuable for patient stratification.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Janus Kinase 1/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Feasibility Studies , Female , Flow Cytometry/methods , Humans , Janus Kinase 1/metabolism , Lung/pathology , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Nitriles , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA-Seq , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Single-Cell Analysis/methods , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tissue Array Analysis , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
6.
SLAS Discov ; 24(1): 77-85, 2019 01.
Article in English | MEDLINE | ID: mdl-30204534

ABSTRACT

ETS transcription factors from the ERG and ETV1/4/5 subfamilies are overexpressed in the majority of prostate cancer patients and contribute to disease progression. Here, we have developed two in vitro assays for the interaction of ETS transcription factors with DNA that are amenable to high-throughput screening. Using ETS1 as a model, we applied these assays to screen 110 compounds derived from a high-throughput virtual screen. We found that the use of lower-affinity DNA binding sequences, similar to those that ERG and ETV1 bind to in prostate cells, allowed for higher inhibition from many of these test compounds. Further pilot experiments demonstrated that the in vitro assays are robust for ERG, ETV1, and ETV5, three of the ETS transcription factors that are overexpressed in prostate cancer.


Subject(s)
High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-ets/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/genetics , Transcriptional Regulator ERG/genetics
7.
Oncotarget ; 9(98): 37173-37184, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30647852

ABSTRACT

Earlier we have shown the expression of a constitutively active receptor tyrosine kinase Axl in CLL B-cells from previously untreated CLL patients, and that Axl inhibitor TP-0903 induces robust leukemic B-cell death. To explore whether Axl is an effective target in relapsed/refractory CLL patients, we analyzed CLL B-cells obtained from CLL patients on ibrutinib therapy. Ibrutinib-exposed CLL B-cells were treated with increasing doses (0.01- 0.50µM) of a new formulation of high-affinity Axl inhibitor, TP-0903 (tartrate salt), for 24 hours and LD50 doses were determined. Sensitivity of CLL B-cells was compared with known prognostic factors and effect of TP-0903 was also evaluated on Axl signaling pathway in CLL B-cells from this cohort. We detected sustained overexpression of Axl in CLL B-cells from CLL patients on ibrutinib treatment, suggests targeting Axl could be a promising strategy to overcome drug resistance and killing of CLL B-cells in these patients. We found that CLL B-cells from sixty-nine percent of relapsed CLL patients actively on ibrutinib therapy were found to be highly sensitive to TP-0903 with induction of apoptosis at nanomolar doses (≤0.50 µM). TP-0903 treatment effectively inhibited Axl phosphorylation and reduced expression levels of anti-apoptotic proteins (Mcl-1, XIAP) in ibrutinib exposed CLL B-cells. In total, our in vitro preclinical studies showing that TP-0903 is very effective at inducing apoptosis in CLL B-cells obtained from ibrutinib-exposed patients supports further testing of this drug in relapsed/refractory CLL.

8.
Mol Cancer Ther ; 5(7): 1764-73, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891462

ABSTRACT

Aurora A and Aurora B are potential targets for anticancer drug development due to their roles in tumorigenesis and disease progression. To identify small-molecule inhibitors of the Aurora kinases, we undertook a structure-based design approach that used three-dimensional structural models of the Aurora A kinase and molecular docking simulations of chemical entities. Based on these computational methods, a new generation of inhibitors derived from quinazoline and pyrimidine-based tricyclic scaffolds were synthesized and evaluated for Aurora A kinase inhibitory activity, which led to the identification of 4-(6,7-dimethoxy-9H-1,3,9-triaza-fluoren-4-yl)-piperazine-1-carbothioic acid [4-(pyrimidin-2-ylsulfamoyl)-phenyl]-amide. The lead compound showed selectivity for the Aurora kinases when it was evaluated against a panel of diverse kinases. Additionally, the compound was evaluated in cell-based assays, showing a dose-dependent decrease in phospho-histone H3 levels and an arrest of the cell cycle in the G(2)-M fraction. Although biological effects were observed only at relatively high concentrations, this chemical series provides an excellent starting point for drug optimization and further development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Lead/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sulfonamides/pharmacology , Thiones/pharmacology , Antineoplastic Agents/chemistry , Aurora Kinase B , Aurora Kinases , Drug Screening Assays, Antitumor , Histones/metabolism , Humans , Lead/chemistry , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Thiones/chemistry
9.
Cancer Res ; 62(7): 2077-84, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11929828

ABSTRACT

p21(WAF1/CIP1) (p21) functions as a cyclin-dependent kinase (CDK) inhibitor and is a key mediator of p53-dependent growth arrest. However, its role in cell cycle regulation is complex, because it also appears to promote CDK activity in certain experimental contexts. Its potential role in tumor suppression was evaluated in MMTV-ras and MMTV-myc transgenic mice that were interbred to p21(WAF1/CIP1) knockout mice (p21-/-). p21 deficiency had differential effects on tumor incidence and age of onset, proliferation, and apoptosis in the presence of these two oncogenes. Tumors arising in MMTV-ras/p21-/- mice displayed higher S-phase fractions and correspondingly increased cyclin D1 and E/CDK activity than MMTV-ras tumors. In contrast, MMTV-myc/p21-/- tumors had lower S-phase fractions and levels of cyclin D1 and E/CDK activity than MMTV-myc tumors. In both tumor types, changes in cyclin D1 and E/CDK activity were paralleled by changes in the corresponding cyclin protein levels. Tumor cell apoptosis was also differentially influenced by p21 deficiency in the two models. MMTV-ras/p21-/- tumors exhibited a significant increase in spontaneous apoptosis as compared with MMTV-ras tumors, whereas p21 deficiency had minimal effect on apoptosis in MMTV-myc tumors. These results indicate that the effects of p21 expression on cellular proliferation are differentially affected by the expression of different oncogenes, and that p21 may play a role in promoting either growth arrest or proliferation, depending on the specific cellular context.


Subject(s)
Cyclins/deficiency , Genes, myc/physiology , Genes, ras/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Division/genetics , Cell Division/physiology , Cell Survival/genetics , Cell Survival/physiology , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/physiology , Female , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , S Phase/genetics , S Phase/physiology
10.
Cancer Res ; 62(10): 2890-6, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019169

ABSTRACT

To identify new diagnostic markers and drug targets for pancreatic cancer, we compared the gene expression patterns of pancreatic cancer cell lines growing in tissue culture with those of normal pancreas using cDNA microarray analysis. Fluorescently (cyanine 5) labeled cDNA probes, made individually from mRNA samples of nine pancreatic cell lines, were each combined with fluorescently (cyanine 3) labeled universal reference mRNA. The mixed probes of each sample were then hybridized with 5760 cDNA arrays (5289 unique cDNA sequences) printed on individual microscope slides. Fluorescently (cyanine 5) labeled normal pancreas mRNA was also compared with the same universal reference mRNA reference pool. The expression ratios of neoplastic versus normal pancreas cells were then calculated by multiplying the ratio of cancer versus the universal reference mRNA and the ratio of the universal reference mRNA cell versus normal pancreas. For 5289 different genes interrogated by the arrays, 30 of them showed an expression ratio 2 SD from the mean in at least three of the nine pancreatic cell lines studied. To confirm the expression profiles of these genes, quantitative reverse transcription-PCR and Northern blot were carried out for 25 of the overexpressed genes. To verify the overexpression in patient samples, two of the overexpressed genes, c-Myc and Rad51, were selected to undergo analysis by reverse transcription-PCR in frozen tumor tissues and by immunostaining in paraffin-embedded tissue section microarrays. The results of these experiments are in agreement with the microarray data. Potential up-regulated targets of note from this study include urokinase-type plasminogen activator receptor, serine/threonine kinase 15, thioredoxin reductase, and CDC28 protein kinase 2, as well as several others.


Subject(s)
Pancreatic Neoplasms/genetics , Biomarkers, Tumor/genetics , Blotting, Northern , DNA, Complementary/genetics , DNA, Complementary/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Profiling , HeLa Cells , Humans , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Sci Transl Med ; 8(325): 325fs4, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26865564

ABSTRACT

A nanoparticle formulation of an Aurora B inhibitor increases antitumor efficacy and reduces toxicity, which may be a precedent for the use of this technology with other small molecules (Ashton et al., this issue).


Subject(s)
Aurora Kinase B , Aurora Kinases , Aurora Kinase A , Humans , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors
13.
Curr Opin Pharmacol ; 2(4): 415-23, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12127874

ABSTRACT

Recent advances in telomerase inhibition have been achieved by using antisense oligonucleotides and ribozymes to target the telomerase mRNA or the telomerase RNA template. Also, small molecules are potent catalytic inhibitors of telomerase. However, therapeutic regimes incorporating these agents will be challenging to implement in the clinic because of their delayed effectiveness. Drugs that directly bind to the telomeres and stabilize secondary DNA structures such as G-quadruplexes are also potent inhibitors of telomerase and disrupt telomere structure. These G-quadruplex-interactive drugs could feasibly be used in synergy with more conventional cytotoxic agents to bring about more immediate responses in cancer cells that are less dependent upon telomere length. Recently, an emerging possible novel use of G-quadruplex-interactive drugs employs their ability to target G-quadruplexes in promoter regions of genes (such as c-MYC), which then serves to repress the production of the human telomerase reverse transcriptase protein.


Subject(s)
Drug Delivery Systems/methods , Telomerase/antagonists & inhibitors , Telomere/drug effects , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Telomerase/metabolism , Telomere/metabolism
14.
Mol Cancer Ther ; 2(2): 165-71, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589033

ABSTRACT

A transgenic mouse tumor model was used to investigate the role of p53 in tumor response to two different platinum-based chemotherapeutic agents: (a) cisplatin and (b) oxaliplatin, a diaminocyclohexane platine recently introduced into the clinic. MMTV-v-Ha-ras transgenic mice were interbred to p53-deficient mice to generate mice that develop salivary tumors either possessing or lacking p53. Tumor-bearing mice were then treated on either a 9-day schedule to assess overall tumor growth response or on a short-term treatment schedule to assess effects on cell cycle parameters and apoptosis. Both agents induced significant apoptosis and promoted overall tumor regression, regardless of the p53 status of the tumor. This is in contrast to previous studies using this model in which treatment with paclitaxel or doxorubicin promoted tumor growth arrest but not apoptosis. These findings indicate that even in the context of an activated ras gene that potentially mediates suppression of apoptosis, both cisplatin and oxaliplatin are capable of promoting an efficient p53-independent tumor response.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/therapeutic use , Genes, ras , Mammary Tumor Virus, Mouse/genetics , Organoplatinum Compounds/therapeutic use , Salivary Gland Neoplasms/drug therapy , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Female , Gene Expression Regulation, Neoplastic , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxaliplatin , Salivary Gland Neoplasms/genetics
15.
Mol Cancer Ther ; 2(6): 589-95, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12813139

ABSTRACT

Aurora-2 kinase has been shown to contribute to oncogenic transformation and is frequently overexpressed and amplified in many human tumor types. Aurora-2 belongs to a small family of mitotic serine/threonine kinases that regulate centrosome maturation, chromosome segregation, and cytokinesis. The mechanism behind the transforming activity of aurora-2 is not fully understood; however, the role of aurora-2 in regulating the centrosome cycle is likely responsible for its ability to transform cells. Aurora-2 overexpression has been correlated with centrosome amplification, which can be a driving cause of genomic instability in tumor cells. In addition, recent work has demonstrated that aurora-2 plays an active function in promoting entry into mitosis by regulating local translation of centrosomal stored mRNA, such as cyclin B1. These recent findings implicate aurora-2 as an important regulator of both genomic integrity and cell cycle progression in cancer cells and suggest that aurora-2 is an attractive target for anticancer drug development.


Subject(s)
Neoplasms/enzymology , Protein Serine-Threonine Kinases/physiology , Antineoplastic Agents/pharmacology , Aurora Kinases , Centrosome/ultrastructure , Cyclin B/metabolism , Cyclin B1 , Gene Expression Regulation, Neoplastic , Mitosis , Models, Biological , Protein Biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism
16.
Mol Cancer Ther ; 1(8): 565-73, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12479216

ABSTRACT

Cationic porphyrins are being studied as possible anticancer agents because of their ability to bind to and stabilize DNA guanine quadruplexes (G-quadruplexes). We have shown previously that the cationic porphyrin TMPyP4 is able to bind to and stabilize G-quadruplexes in human telomere sequences, resulting in inhibition of telomerase activity. To better understand the mechanism of action behind telomerase inhibition by TMPyP4, we performed a cDNA microarray analysis on cells treated with TMPyP4 and TMPyP2, a positional isomer of TMPyP4 that has low affinity for G-quadruplexes. Analysis of time course data from the microarray experiments revealed that TMPyP4 and TMPyP2 treatment altered the expression of several gene clusters. We found that c-MYC, an oncogene nearly ubiquitous in human tumors that bears the potential in its promoter to form a G-quadruplex, was among the genes specifically down-regulated by TMPyP4, but not by TMPyP2. The hTERT gene, which encodes the catalytic subunit of telomerase, is transcriptionally regulated by c-MYC, and we have found that TMPyP4 also causes a decrease in human telomerase reverse transcriptase transcripts, suggesting two possible mechanisms for the effect of TMPyP4 on telomerase activity. We also show that TMPyP4, but not TMPyP2, is able to prolong survival and decrease tumor growth rates in two xenograft tumor models. We believe that, because of the actions of TMPyP4 in decreasing both c-MYC protein levels and telomerase activity, as well as its anticancer effects in vivo, it is a worthwhile agent to pursue and develop further.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Down-Regulation , Porphyrins/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Telomerase/metabolism , Animals , Blotting, Northern , Blotting, Western , DNA/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins , HeLa Cells , Humans , Mice , Models, Chemical , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Mol Cancer Ther ; 2(3): 283-94, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12657723

ABSTRACT

The aurora kinases are a novel oncogenic family of mitotic serine/threonine kinases (S/T kinases) that are overexpressed in a number of solid tumors, including pancreas and colorectal cancer. A PSI-BLAST search [National Center for Biotechnology Information (NCBI)] with the sequence of the S/T kinase domain of human aurora1 kinase [also known as AUR1, ARK2, AIk2, AIM-1, and STK12] and human aurora2 kinase (also known as AUR2, ARK1, AIK, BTAK, and STK15) showed a high sequence similarity to the three-dimensional structures of bovine cAMP-dependent kinase [Brookhaven Protein Data Bank code 1CDK], murine cAMP-dependent kinase (1APM), and Caenorhabditis elegans twitchin kinase (1KOA). When the aurora1 or aurora2 sequence was input into the tertiary structure prediction programs THREADER and 3D-PSSM (three-dimensional position-sensitive scoring matrix), the top structural matches were 1CDK, 1APM, and 1KOA, confirming that these domains are structurally conserved. The structural models of aurora1 and aurora2 were built using 1CDK as the template structure. Molecular dynamics and docking simulations, targeting the ATP binding site of aurora2 with adenylyl imidodiphosphate (AMP-PNP), staurosporine, and six small molecular S/T kinase inhibitors, identified active-site residues that interact with these inhibitors differentially. The docked structures of the aurora2-AMP-PNP and aurora2-staurosporine complexes indicated that the adenine ring of AMP-PNP and the indolocarbazole moiety of staurosporine have similar positions and orientations and provided the basis for the docking of the other S/T kinase inhibitors. Inhibitors with isoquinoline and quinazoline moieties were recognized by aurora2 in which H-89 and 6,7-dimethoxyquinazoline compounds exhibited high binding energies compared with that of staurosporine. The calculated binding energies for the docked small-molecule inhibitors were qualitatively consistent with the IC(50) values generated using an in vitro kinase assay. The aurora2 structural model provides a rational basis for site-directed mutagenesis of the active site; design of novel H-89, staurosporine, and quinazoline analogues; and the screening of the available chemical database for the identification of other novel, small-molecular entities.


Subject(s)
Antineoplastic Agents/pharmacology , Caenorhabditis elegans Proteins , Computational Biology , Drug Design , Pancreatic Neoplasms/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Sequence , Animals , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Caenorhabditis elegans/enzymology , Calmodulin-Binding Proteins/chemistry , Cattle , Cyclic AMP-Dependent Protein Kinases/chemistry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Muscle Proteins/chemistry , Pancreatic Neoplasms/drug therapy , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Sequence Homology, Amino Acid , Staurosporine/chemistry , Staurosporine/metabolism , Structure-Activity Relationship
18.
Mol Cancer Ther ; 3(5): 641-6, 2004 May.
Article in English | MEDLINE | ID: mdl-15141022

ABSTRACT

Pancreas cancer is the fourth leading cause of cancer-related death in adults in the United States. New molecular targets for diagnosis and therapy of this disease are desperately needed. In this study, we report on the mitotic serine-threonine kinase polo-like kinase 1 (Plk1) in pancreatic cancer. Plk1 mRNA was found to be overexpressed in 9 of 10 tested pancreatic cancer cell lines and in 4 of 4 tested human tumors. Immunohistochemical staining of a pancreatic tissue microarray showed that 26 of the 35 tumors taken directly from patients overexpressed Plk1. We also examined the effects of depleting Plk1 in pancreatic cancer cells by the use of antisense oligonucleotides. Antisense-treated pancreatic cancer cells showed cell cycle arrest in G(2)-M as well as a drastic reduction in proliferation rates. These data suggest that Plk1 is a potential therapeutic target in devising a treatment for patients with pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Protein Kinases/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Gene Dosage , Gene Expression , Humans , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Pancreatic Neoplasms/genetics , Protein Kinases/deficiency , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Polo-Like Kinase 1
19.
Mol Cancer Ther ; 3(4): 451-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15078988

ABSTRACT

Aurora-2 is a serine threonine kinase that associates with the centrosome. Overexpression or ectopic expression of Aurora-2 appears to alter centrosome number and function and has been implicated in a variety of human cancers. In this work, we demonstrate that Aurora-2 is both amplified and overexpressed in human pancreatic cancer cell lines, with a 2-5-fold increase in gene copy number and a 3-4-fold increase in protein levels compared with controls. Aurora-2 is also amplified and overexpressed in pancreatic cancers taken directly from patients. An immunohistochemistry of tissues taken directly from patients demonstrated an overexpression of Aurora-2 in 26 of 28 pancreatic cancers compared with 18 normal pancreas samples. Antisense nucleotides specifically targeted at Aurora-2 arrest the cell cycle in pancreatic cancer cells, indicating the potential of Aurora-2 as a therapeutic target in pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Aurora Kinases , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , G2 Phase/drug effects , Gene Expression , Humans , Immunohistochemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Pancreatic Neoplasms/genetics
20.
Appl Immunohistochem Mol Morphol ; 23(10): 717-23, 2015.
Article in English | MEDLINE | ID: mdl-26551340

ABSTRACT

BACKGROUND: The Provirus integrating site Moloney murine leukemia virus (Pim) family are proteins with serine/threonine kinase activity. Studies have demonstrated overexpression of Pims in cancer. To our knowledge, only a single study has examined Pim-1 in urothelial carcinoma. The aim of this investigation was to evaluate Pim-1, Pim-2, and Pim-3 in urothelial carcinoma and assess for expression that may contribute to disease progression and serve as a site for targeted therapy. METHODS: This retrospective study included 137 cases taken from specimens from the University of Utah, Department of Pathology (2008 to 2011). Tissue was stained with antibodies against Pim-1, Pim-2, and Pim-3. Cases were classified into 3 groups, based upon current World Health Organization criteria (invasive high-grade urothelial carcinoma [IHG] [n=84], noninvasive high-grade urothelial carcinoma/carcinoma in situ [n=32], and noninvasive low-grade urothelial carcinoma [NILG] [n=21]). Cases were scored and recorded as positive or negative on the basis of the percentage of cells with cytoplasmic and/or nuclear staining. RESULTS: NILG showed higher expression of Pim-1 (relative expression rate [RER]=2.28; 95% confidence interval [CI], 0.183-0.764) and Pim-3 (RER=3.06; 95% CI, 0.423-0.816) compared with other lesions. IHG had lower expression of Pim-1 (RER=0.31; 95% CI, 0.401-0.844) and Pim-3 (RER=0.354; 95% CI, 0.322-0.816) and noninvasive high-grade urothelial carcinoma (NIHG) demonstrated increased expression of Pim-1 and (RER=2.09; 95% CI, 0.124-0.739) and Pim-2 (RER=1.70; 95% CI, 0.151-0.591). At least 1 Pim kinase protein was expressed at the following rates: 49% in IHG, 66% in NIHG, and 76% in NILG. CONCLUSION: A high percentage of urothelial carcinomas express Pim kinases. Pim expression differs in NILG, NIHG, and IHG lesions.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-pim-1/biosynthesis , Urologic Neoplasms , Urothelium , Female , Humans , Male , Urologic Neoplasms/enzymology , Urologic Neoplasms/pathology , Urothelium/enzymology , Urothelium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL