Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 20(1): 65-80, 2021 01.
Article in English | MEDLINE | ID: mdl-33356791

ABSTRACT

Palbociclib, a selective CDK4/6 kinase inhibitor, is approved in combination with endocrine therapies for the treatment of advanced estrogen receptor positive (ER+) breast cancer. In pre-clinical cancer models, CDK4/6 inhibitors act primarily as cytostatic agents. In two commonly studied ER+ breast cancer cell lines (MCF7 and T47D), CDK4/6 inhibition drives G1-phase arrest and the acquisition of a senescent-like phenotype, both of which are reversible upon palbociclib withdrawal (incomplete senescence). Here we identify an ER+ breast cancer cell line, CAMA1, in which palbociclib treatment induces irreversible cell cycle arrest and senescence (complete senescence). In stark contrast to T47D and MCF7 cells, mTORC1 activity is not stably suppressed in CAMA1 cells during palbociclib treatment. Importantly, inhibition of mTORC1 signaling either by the mTORC1 inhibitor rapamycin or by knockdown of Raptor, a unique component of mTORC1, during palbociclib treatment of CAMA1 cells blocks the induction of complete senescence. These results indicate that sustained mTORC1 activity promotes complete senescence in ER+ breast cancer cells during CDK4/6 inhibitor-induced cell cycle arrest. Consistent with this mechanism, genetic depletion of TSC2, a negative regulator of mTORC1, in MCF7 cells resulted in sustained mTORC1 activity during palbociclib treatment and evoked a complete senescence response. These findings demonstrate that persistent mTORC1 signaling during palbociclib-induced G1 arrest is a potential liability for ER+ breast cancer cells, and suggest a strategy for novel drug combinations with palbociclib.


Subject(s)
Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cellular Senescence/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Humans , MCF-7 Cells , Mice , Signal Transduction/drug effects
2.
Nucleic Acids Res ; 48(17): 9462-9477, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32821942

ABSTRACT

CRISPR/Cas9 functional genomic screens have emerged as essential tools in drug target discovery. However, the sensitivity of available genome-wide CRISPR libraries is impaired by guides which inefficiently abrogate gene function. While Cas9 cleavage efficiency optimization and essential domain targeting have been developed as independent guide design rationales, no library has yet combined these into a single cohesive strategy to knock out gene function. Here, in a massive reanalysis of CRISPR tiling data using the most comprehensive feature database assembled, we determine which features of guides and their targets best predict activity and how to best combine them into a single guide design algorithm. We present the ProteIN ConsERvation (PINCER) genome-wide CRISPR library, which for the first time combines enzymatic efficiency optimization with conserved length protein region targeting, and also incorporates domains, coding sequence position, U6 termination (TTT), restriction sites, polymorphisms and specificity. Finally, we demonstrate superior performance of the PINCER library compared to alternative genome-wide CRISPR libraries in head-to-head validation. PINCER is available for individual gene knockout and genome-wide screening for both the human and mouse genomes.


Subject(s)
Algorithms , CRISPR-Cas Systems , Databases, Genetic , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Amino Acids/genetics , Animals , Cell Line , Conserved Sequence , Enzymes/genetics , Enzymes/metabolism , Genome , Genomic Library , Humans , Mice , RNA, Guide, Kinetoplastida/genetics , Reproducibility of Results , Thymidine/genetics
3.
Clin Cancer Res ; 21(22): 5121-5130, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-25991817

ABSTRACT

PURPOSE: Endocrine therapy, using tamoxifen or an aromatase inhibitor, remains first-line therapy for the management of estrogen receptor (ESR1)-positive breast cancer. However, ESR1 mutations or other ligand-independent ESR1 activation mechanisms limit the duration of response. The clinical efficacy of fulvestrant, a selective estrogen receptor downregulator (SERD) that competitively inhibits agonist binding to ESR1 and triggers receptor downregulation, has confirmed that ESR1 frequently remains engaged in endocrine therapy-resistant cancers. We evaluated the activity of a new class of selective estrogen receptor modulators (SERM)/SERD hybrids (SSH) that downregulate ESR1 in relevant models of endocrine-resistant breast cancer. Building on the observation that concurrent inhibition of ESR1 and the cyclin-dependent kinases 4 and 6 (CDK4/6) significantly increased progression-free survival in advanced patients, we explored the activity of different SERD- or SSH-CDK4/6 inhibitor combinations in models of endocrine therapy-resistant ESR1(+) breast cancer. EXPERIMENTAL DESIGN: SERDs, SSHs, and the CDK4/6 inhibitor palbociclib were evaluated as single agents or in combination in established cellular and animal models of endocrine therapy-resistant ESR1(+) breast cancer. RESULTS: The combination of palbociclib with a SERD or an SSH was shown to effectively inhibit the growth of MCF7 cell or ESR1-mutant patient-derived tumor xenografts. In tamoxifen-resistant MCF7 xenografts, the palbociclib/SERD or SSH combination resulted in an increased duration of response as compared with either drug alone. CONCLUSIONS: A SERD- or SSH-palbociclib combination has therapeutic potential in breast tumors resistant to endocrine therapies or those expressing ESR1 mutations. See related commentary by DeMichele and Chodosh, p. 4999.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/genetics , Selective Estrogen Receptor Modulators/administration & dosage , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Mutation , Piperazines/administration & dosage , Pyridines/administration & dosage , Tamoxifen/administration & dosage , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 21(13): 2905-10, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25941111

ABSTRACT

Cancer cells bypass normal controls over mitotic cell-cycle progression to achieve a deregulated state of proliferation. The retinoblastoma tumor suppressor protein (pRb) governs a key cell-cycle checkpoint that normally prevents G1-phase cells from entering S-phase in the absence of appropriate mitogenic signals. Cancer cells frequently overcome pRb-dependent growth suppression via constitutive phosphorylation and inactivation of pRb function by cyclin-dependent kinase (CDK) 4 or CDK6 partnered with D-type cyclins. Three selective CDK4/6 inhibitors, palbociclib (Ibrance; Pfizer), ribociclib (Novartis), and abemaciclib (Lilly), are in various stages of development in a variety of pRb-positive tumor types, including breast cancer, melanoma, liposarcoma, and non-small cell lung cancer. The emerging, positive clinical data obtained to date finally validate the two decades-old hypothesis that the cyclin D-CDK4/6 pathway is a rational target for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cyclin D/antagonists & inhibitors , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
5.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646013

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab-maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non-cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab-maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952-63. ©2015 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Immunoconjugates/pharmacology , Trastuzumab/pharmacology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Inhibitory Concentration 50 , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Protein Transport , Proteome , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction , Transcriptome , Trastuzumab/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Oncotarget ; 6(5): 2928-38, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25671303

ABSTRACT

In addition to genetic alterations, cancer cells are characterized by myriad epigenetic changes. EZH2 is a histone methyltransferase that is over-expressed and mutated in cancer. The EZH2 gain-of-function (GOF) mutations first identified in lymphomas have recently been reported in melanoma (~2%) but remain uncharacterized. We expressed multiple EZH2 GOF mutations in the A375 metastatic skin melanoma cell line and observed both increased H3K27me3 and dramatic changes in 3D culture morphology. In these cells, prominent morphological changes were accompanied by a decrease in cell contractility and an increase in collective cell migration. At the molecular level, we observed significant alteration of the axonal guidance pathway, a pathway intricately involved in the regulation of cell shape and motility. Furthermore, the aggressive 3D morphology of EZH2 GOF-expressing melanoma cells (both endogenous and ectopic) was attenuated by EZH2 catalytic inhibition. Finally, A375 cells expressing exogenous EZH2 GOF mutants formed larger tumors than control cells in mouse xenograft studies. This study not only demonstrates the first functional characterization of EZH2 GOF mutants in non-hematopoietic cells, but also provides a rationale for EZH2 catalytic inhibition in melanoma.


Subject(s)
Cell Movement , Cell Proliferation , Cell Shape , Epigenesis, Genetic , Melanoma/genetics , Mutation , Polycomb Repressive Complex 2/genetics , Skin Neoplasms/genetics , Animals , Cell Line , Cell Movement/drug effects , Cell Shape/drug effects , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/pathology , Mice, Nude , Molecular Targeted Therapy , Neoplasm Invasiveness , Polycomb Repressive Complex 2/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Tumor Burden , Xenograft Model Antitumor Assays
7.
Pac Symp Biocomput ; : 10-9, 2015.
Article in English | MEDLINE | ID: mdl-25592564

ABSTRACT

The Cell Index Database, (CELLX) (http://cellx.sourceforge.net) provides a computational framework for integrating expression, copy number variation, mutation, compound activity, and meta data from cancer cells. CELLX provides the computational biologist a quick way to perform routine analyses as well as the means to rapidly integrate data for offline analysis. Data is accessible through a web interface which utilizes R to generate plots and perform clustering, correlations, and statistical tests for associations within and between data types for ~20,000 samples from TCGA, CCLE, Sanger, GSK, GEO, GTEx, and other public sources. We show how CELLX supports precision oncology through indications discovery, biomarker evaluation, and cell line screening analysis.


Subject(s)
Databases, Genetic , Neoplasms/genetics , Software , Biomarkers, Tumor/genetics , Cell Line, Tumor , Computational Biology , Humans , Internet , Neoplasms/therapy , Precision Medicine
8.
Cell Stress Chaperones ; 15(6): 913-27, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20717760

ABSTRACT

Cancer cells are exposed to external and internal stresses by virtue of their unrestrained growth, hostile microenvironment, and increased mutation rate. These stresses impose a burden on protein folding and degradation pathways and suggest a route for therapeutic intervention in cancer. Proteasome and Hsp90 inhibitors are in clinical trials and a 20S proteasome inhibitor, Velcade, is an approved drug. Other points of intervention in the folding and degradation pathway may therefore be of interest. We describe a simple screen for inhibitors of protein synthesis, folding, and proteasomal degradation pathways in this paper. The molecular chaperone-dependent client v-Src was fused to firefly luciferase and expressed in HCT-116 colorectal tumor cells. Both luciferase and protein tyrosine kinase activity were preserved in cells expressing this fusion construct. Exposing these cells to the Hsp90 inhibitor geldanamycin caused a rapid reduction of luciferase and kinase activities and depletion of detergent-soluble v-Src::luciferase fusion protein. Hsp70 knockdown reduced v-Src::luciferase activity and, when combined with geldanamycin, caused a buildup of v-Src::luciferase and ubiquitinated proteins in a detergent-insoluble fraction. Proteasome inhibitors also decreased luciferase activity and caused a buildup of phosphotyrosine-containing proteins in a detergent-insoluble fraction. Protein synthesis inhibitors also reduced luciferase activity, but had less of an effect on phosphotyrosine levels. In contrast, certain histone deacetylase inhibitors increased luciferase and phosphotyrosine activity. A mass screen led to the identification of Hsp90 inhibitors, ubiquitin pathway inhibitors, inhibitors of Hsp70/Hsp40-mediated refolding, and protein synthesis inhibitors. The largest group of compounds identified in the screen increased luciferase activity, and some of these increase v-Src levels and activity. When used in conjunction with appropriate secondary assays, this screen is a powerful cell-based tool for studying compounds that affect protein synthesis, folding, and degradation.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Molecular Chaperones/antagonists & inhibitors , Proteasome Inhibitors , Protein Folding/drug effects , Protein Synthesis Inhibitors/pharmacology , Benzoquinones/pharmacology , Cell Line, Tumor , Drug Evaluation, Preclinical , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/pharmacology , Humans , Lactams, Macrocyclic/pharmacology , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Oncogene Protein pp60(v-src)/chemistry , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Small Interfering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitin/antagonists & inhibitors , Ubiquitin/metabolism
9.
Bioorg Med Chem Lett ; 20(12): 3821-5, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20471256

ABSTRACT

A series of tricyclic anilinopyrimidines were synthesized and evaluated as IKKbeta inhibitors. Several analogues, including tricyclic phenyl (10, 18a, 18c, 18d, and 18j) and thienyl (26 and 28) derivatives were shown to have good in vitro enzyme potency and excellent cellular activity. Pharmaceutical profiling of a select group of tricyclic compounds compared to the non-tricyclic analogues suggested that in some cases, the improved cellular activity may be due to increased clog P and permeability.


Subject(s)
I-kappa B Kinase/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Animals , Cell Line , Cell Membrane Permeability , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Structure-Activity Relationship
10.
Eur J Cancer ; 46(10): 1781-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20399641

ABSTRACT

Bosutinib (SKI-606) is an orally active Src and Abl kinase inhibitor presently in Phase III trials for treatment of chronic myelogenous leukaemia (CML), and in Phase II trials for treatment of breast cancer. Bosutinib is a potent antiproliferative and proapoptotic agent in CML cells and inhibits Bcr-Abl mediated signalling at nanomolar concentrations. Short-term administration of bosutinib causes regression of K562 and KU812 CML tumour xenografts. BaF3 murine myeloid cells expressing wild-type Bcr-Abl are sensitive to bosutinib treatment, as are BaF3 cells expressing many imatinib-resistant forms of Bcr-Abl. Recent studies indicate that bosutinib is active against a broader spectrum of kinases than originally believed. These additional inhibitory activities have interesting possibilities for further clinical development. This review will focus on preclinical studies supporting the clinical development of bosutinib for treatment of CML, with a discussion on the broader potential of this agent in other oncology indications.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Nitriles/therapeutic use , Quinolines/therapeutic use , src-Family Kinases/therapeutic use , Aniline Compounds/chemistry , Animals , Cell Communication , Cell Line, Tumor , Clinical Trials as Topic , Cyclic N-Oxides/pharmacology , Drug Resistance, Neoplasm , Humans , Mice , Neoplasm Transplantation , Nitriles/chemistry , Protein Kinase Inhibitors/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Transplantation, Heterologous , src-Family Kinases/chemistry
11.
Eur J Med Chem ; 45(4): 1379-86, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20074837

ABSTRACT

A series of 8,9-dimethoxy-5-(2-aminoalkoxy-pyridin-3-yl)-benzo[c][2,7]naphthyridin-4-ylamine-based inhibitors of 3-phosphoinositide-dependent kinase-1 (PDK-1) has been identified. Several examples appear to be potent and relatively selective inhibitors of PDK-1 over the related AGC kinases PKA, PKB/AKT, and p70S6K. The introduction of a stereochemical center beside the amino substituent on the aminoalkoxy-side chain had little effect upon the inhibitory activity against these enzymes, and X-ray crystallographic analyses of a representative pair of enantiomeric inhibitors bound to the active site of PDK-1 revealed comparable binding modes for each enantiomer.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Static Electricity , Structure-Activity Relationship
12.
J Med Chem ; 53(2): 897-910, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20025292

ABSTRACT

We are introducing a novel series of 2,4-diaminoquinazolines as beta-catenin/Tcf4 inhibitors which were identified by ligand-based design. Here we elucidate the SAR of this series and explain how we were able to improve key molecular properties such as solubility and cLogP leading to compound 9. Analogue 9 exhibited better biological activity and improved physical and pharmacological properties relative to the HTS hit 49. Furthermore, 9 demonstrated good cell growth inhibition against several human colorectal cancer lines such as LoVo and HT29. In addition, treatment with compound 9 led to gene expression changes that overlapped significantly with the transcriptional profile resulting from the pathway inhibition by siRNA knockdown of beta-catenin or Tcf4. Subsequently, 9 was tested for efficacy in a beta-catenin/RKE-mouse xenograft, where it led to more then 50% decrease in tumor volume.


Subject(s)
Colorectal Neoplasms/drug therapy , Quinazolines/chemical synthesis , TCF Transcription Factors/drug effects , beta Catenin/drug effects , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/drug effects , Cell Line, Tumor , Colorectal Neoplasms/pathology , Drug Design , Humans , Mice , Quinazolines/pharmacology , Quinazolines/therapeutic use , Structure-Activity Relationship , Transcription Factor 4 , Transcription Factors/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem Lett ; 19(17): 4980-3, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19640711

ABSTRACT

The synthesis and SAR of a series of 2,4-diamino-quinazoline derivatives as beta-catenin/Tcf-4 inhibitors are described. This series was developed by modifying the initial lead 1, which was identified by screening of our compound library and found to inhibit the beta-catenin/Tcf-4 pathway. Replacement of the biphenyl moiety in compound 1 with the N-phenylpiperidine-4-carboxamide chain as in 2, resulted in a number of new analogues, which are potent inhibitors of the beta-catenin/Tcf-4 pathway. Compound such as 16k exhibited good cellular potency, solubility, metabolic stability and oral bioavailability.


Subject(s)
Anilides/chemistry , Antineoplastic Agents/chemistry , Colorectal Neoplasms/drug therapy , Quinazolines/chemistry , TCF Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Anilides/chemical synthesis , Anilides/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Quinazolines/chemical synthesis , Quinazolines/pharmacokinetics , Structure-Activity Relationship , TCF Transcription Factors/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
15.
Mol Carcinog ; 48(8): 678-84, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19444813

ABSTRACT

Many anticancer agents activate NF-kappaB, which plays an important role in the survival of cancer cells. Inhibition of NF-kappaB activity may therefore potentiate the efficacy of anticancer agents. We found that a previously used anticancer agent Streptonigrin (SN) was also a potent NF-kappaB inducer. Using a specific IKKbeta inhibitor IV (Podolin et al., J Pharmacol Exp Ther 2005; 312: 373-381), we revealed that the activation of NF-kappaB was mediated through DNA damage-induced activation of IKK complex. Furthermore, we demonstrated that SN-induced DNA damage was unrelated to reactive oxygen species but to the hydroquinone form of SN converted by the NAD(P)H:quinine oxidoreductase (NQO1). The study suggests that the combination of SN with IKK inhibitor may improve efficacy over the use of single agent.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Melanoma/drug therapy , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Streptonigrin/pharmacology , Blotting, Western , Caspases/metabolism , Cell Proliferation/drug effects , DNA Damage , Histones/genetics , Histones/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
16.
Cancer Res ; 68(22): 9519-24, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010928

ABSTRACT

Tumor necrosis factor alpha (TNFalpha) has been used to treat patients with certain tumor types. However, its antitumor activity has been undermined by the activation of IkappaBalpha kinase (IKK), which in turn activates nuclear factor-kappaB (NF-kappaB) to help cancer cells survive. Therefore, inhibition of TNFalpha-induced IKK activity with specific IKK inhibitor represents an attractive strategy to treat cancer patients. This study reveals IKI-1 as a potent small molecule inhibitor of IKKalpha and IKKbeta, which effectively blocked TNFalpha-mediated IKK activation and subsequent NF-kappaB activity. Using gene profiling analysis, we show that IKI-1 blocked most of the TNFalpha-mediated mRNA expression, including many genes that play important roles in cell survival. We further show that in vitro and in vivo combination of TNFalpha with IKI-1 had superior potency than either agent alone. This increased potency was due primarily to the increased apoptosis in the presence of both TNFalpha and IKI-1. Additionally, IKKbeta small interfering RNA transfected cells were more sensitive to the treatment of TNFalpha. The study suggests that the limited efficacy of TNFalpha in cancer treatment was due in part to the activation of NF-kappaB, allowing tumor cells to escape apoptosis. Therefore, the combination of IKI-1 with TNFalpha may improve the efficacy of TNFalpha for certain tumor types.


Subject(s)
I-kappa B Kinase/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , I-kappa B Kinase/metabolism , Mice , Pancreatic Neoplasms/pathology , Phosphorylation , Protein Transport/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Xenograft Model Antitumor Assays
17.
J Med Chem ; 51(3): 373-5, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18197612

ABSTRACT

Heat shock protein 90 (Hsp90) is a molecular chaperone that is responsible for activating many signaling proteins and is a promising target in tumor biology. We have identified small-molecule benzisoxazole derivatives as Hsp90 inhibitors. Crystallographic studies show that these compounds bind in the ATP binding pocket interacting with the Asp93. Structure based optimization led to the identification of potent analogues, such as 13, with good biochemical profiles.


Subject(s)
Antineoplastic Agents/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/chemical synthesis , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/metabolism , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , K562 Cells , Models, Molecular , Protein Conformation , Structure-Activity Relationship
18.
J Med Chem ; 50(23): 5547-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17941624

ABSTRACT

With high-throughput screening, substituted dibenzo[c,f][2,7]naphthyridine 1 was identified as a novel potent and selective phosphoinositide-dependent kinase-1 (PDK-1) inhibitor. Various regions of the lead molecule were explored to understand the SAR requirement for this scaffold. The crystal structure of 1 with kinase domain of PDK-1 confirmed the binding in the active site. The key interaction of the molecule with the active site residues, observed SAR, and the biological profile are discussed in detail.


Subject(s)
Naphthyridines/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Naphthyridines/chemistry , Protein Serine-Threonine Kinases/chemistry , Structure-Activity Relationship
19.
Cancer Res ; 65(12): 5358-64, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958584

ABSTRACT

Src up-regulation is a common event in human cancers. In colorectal cancer, increased Src levels are an indicator of poor prognosis, and progression to metastatic disease is associated with substantial increases in Src activity. Therefore, we examined the activity of SKI-606, a potent inhibitor of Src and Abl kinases, against colon tumor lines in vitro and in s.c. tumor xenograft models. SKI-606 inhibited Src autophosphorylation with an IC(50) of approximately 0.25 micromol/L in HT29 cells. Phosphorylation of Tyr(925) of focal adhesion kinase, a Src substrate, was reduced by similar concentrations of inhibitor. Antiproliferative activity on plastic did not correlate with Src inhibition in either HT29 or Colo205 cells (IC(50)s, 1.5 and 2.5 micromol/L, respectively), although submicromolar concentrations of SKI-606 inhibited HT29 cell colony formation in soft agar. SKI-606 also caused loosely aggregated Colo205 spheroids to condense into compact spheroids. On oral administration to nude mice at the lowest efficacious dose, peak plasma concentrations of approximately 3 micromol/L, an oral bioavailability of 18%, and a t(1/2) of 8.6 hours were observed. SKI-606 was orally active in s.c. colon tumor xenograft models and caused substantial reductions in Src autophosphorylation on Tyr(418) in HT29 and Colo205 tumors. SKI-606 inhibited HT29 tumor growth on once daily administration, whereas twice daily administration was necessary to inhibit Colo205, HCT116, and DLD1 tumor growth. These results support development of SKI-606 as a therapeutic agent for treatment of colorectal cancer.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Administration, Oral , Aniline Compounds/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Female , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Nitriles/pharmacokinetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Quinolines/pharmacokinetics , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
20.
Yeast ; 22(3): 219-39, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15704212

ABSTRACT

Eukaryotic chaperonins, the Cct complexes, are assembled into two rings, each of which is composed of a stoichiometric array of eight different subunits, which are denoted Cct1p-Cct8p. Overexpression of a single CCT gene in Saccharomyces cerevisiae causes an increase of the corresponding Cct subunit, but not of the Cct complex. Nevertheless, overexpression of certain Cct subunits, especially CCT6, suppresses a wide range of abnormal phenotypes, including those caused by the diverse types of conditional mutations tor2-21, lst8-2 and rsp5-9 and those caused by the concomitant overexpression of Sit4p and Sap155p. The examination of 73 altered forms of Cct6p revealed that the cct6-24 mutation, containing GDGTT --> AAAAA replacements of the conserved ATP-binding motif, was unable to suppress any of these traits, although the cct6-24 allele was completely functional for growth. These results provide evidence for functional differences among Cct subunits and for physiological properties of unassembled subunits. We suggest that the suppression is due to the competition of specific Cct subunits for activities that normally modify various cellular components. Furthermore, we also suggest that the Cct subunits can act as suppressors only in certain states, such as when associated with ATP.


Subject(s)
Chaperonins/physiology , Saccharomyces cerevisiae/physiology , Chaperonin Containing TCP-1 , Chaperonins/biosynthesis , Chaperonins/genetics , DNA, Fungal/chemistry , DNA, Fungal/genetics , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Fungal , Mass Spectrometry , Models, Biological , Plasmids , Polymerase Chain Reaction , Protein Multimerization , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...