Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(5): 3400-3418, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38387069

ABSTRACT

The use of ß-lactam (BL) and ß-lactamase inhibitor combination to overcome BL antibiotic resistance has been validated through clinically approved drug products. However, unmet medical needs still exist for the treatment of infections caused by Gram-negative (GN) bacteria expressing metallo-ß-lactamases. Previously, we reported our effort to discover pan inhibitors of three main families in this class: IMP, VIM, and NDM. Herein, we describe our work to improve the GN coverage spectrum in combination with imipenem and relebactam. This was achieved through structure- and property-based optimization to tackle the GN cell penetration and efflux challenges. A significant discovery was made that inhibition of both VIM alleles, VIM-1 and VIM-2, is essential for broad GN coverage, especially against VIM-producing P. aeruginosa. In addition, pharmacokinetics and nonclinical safety profiles were investigated for select compounds. Key findings from this drug discovery campaign laid the foundation for further lead optimization toward identification of preclinical candidates.


Subject(s)
Anti-Bacterial Agents , beta-Lactamase Inhibitors , Humans , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamase Inhibitors/chemistry , Anti-Bacterial Agents/chemistry , Imipenem/pharmacology , beta-Lactamases , Gram-Negative Bacteria , Microbial Sensitivity Tests
2.
J Med Chem ; 65(24): 16234-16251, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36475645

ABSTRACT

With the emergence and rapid spreading of NDM-1 and existence of clinically relevant VIM-1 and IMP-1, discovery of pan inhibitors targeting metallo-beta-lactamases (MBLs) became critical in our battle against bacterial infection. Concurrent with our fragment and high-throughput screenings, we performed a knowledge-based search of known metallo-beta-lactamase inhibitors (MBLIs) to identify starting points for early engagement of medicinal chemistry. A class of compounds exemplified by 11, discovered earlier as B. fragilis metallo-beta-lactamase inhibitors, was selected for in silico virtual screening. From these efforts, compound 12 was identified with activity against NDM-1 only. Initial exploration on metal binding design followed by structure-guided optimization led to the discovery of a series of compounds represented by 23 with a pan MBL inhibition profile. In in vivo studies, compound 23 in combination with imipenem (IPM) robustly lowered the bacterial burden in a murine infection model and became the lead for the invention of MBLI clinical candidates.


Subject(s)
Bacterial Infections , beta-Lactamase Inhibitors , Animals , Mice , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamase Inhibitors/chemistry , Imipenem/pharmacology , Imipenem/therapeutic use , beta-Lactamases/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests
3.
PLoS Pathog ; 12(5): e1005585, 2016 05.
Article in English | MEDLINE | ID: mdl-27144276

ABSTRACT

Here we describe a chemical biology strategy performed in Staphylococcus aureus and Staphylococcus epidermidis to identify MnaA, a 2-epimerase that we demonstrate interconverts UDP-GlcNAc and UDP-ManNAc to modulate substrate levels of TarO and TarA wall teichoic acid (WTA) biosynthesis enzymes. Genetic inactivation of mnaA results in complete loss of WTA and dramatic in vitro ß-lactam hypersensitivity in methicillin-resistant S. aureus (MRSA) and S. epidermidis (MRSE). Likewise, the ß-lactam antibiotic imipenem exhibits restored bactericidal activity against mnaA mutants in vitro and concomitant efficacy against 2-epimerase defective strains in a mouse thigh model of MRSA and MRSE infection. Interestingly, whereas MnaA serves as the sole 2-epimerase required for WTA biosynthesis in S. epidermidis, MnaA and Cap5P provide compensatory WTA functional roles in S. aureus. We also demonstrate that MnaA and other enzymes of WTA biosynthesis are required for biofilm formation in MRSA and MRSE. We further determine the 1.9Å crystal structure of S. aureus MnaA and identify critical residues for enzymatic dimerization, stability, and substrate binding. Finally, the natural product antibiotic tunicamycin is shown to physically bind MnaA and Cap5P and inhibit 2-epimerase activity, demonstrating that it inhibits a previously unanticipated step in WTA biosynthesis. In summary, MnaA serves as a new Staphylococcal antibiotic target with cognate inhibitors predicted to possess dual therapeutic benefit: as combination agents to restore ß-lactam efficacy against MRSA and MRSE and as non-bioactive prophylactic agents to prevent Staphylococcal biofilm formation.


Subject(s)
Bacterial Proteins/metabolism , Racemases and Epimerases/metabolism , Staphylococcus aureus/metabolism , Staphylococcus epidermidis/metabolism , Teichoic Acids/biosynthesis , Animals , Bacterial Proteins/chemistry , Biofilms/growth & development , Cell Wall/metabolism , Crystallography, X-Ray , Disease Models, Animal , Methicillin-Resistant Staphylococcus aureus , Mice , Microbial Sensitivity Tests , Microscopy, Fluorescence , Nuclear Magnetic Resonance, Biomolecular , Racemases and Epimerases/chemistry , Staphylococcal Infections/metabolism
4.
ACS Med Chem Lett ; 7(3): 324-9, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985323

ABSTRACT

A new subseries of substituted piperidines as p53-HDM2 inhibitors exemplified by 21 has been developed from the initial lead 1. Research focused on optimization of a crucial HDM2 Trp23-ligand interaction led to the identification of 2-(trifluoromethyl)thiophene as the preferred moiety. Further investigation of the Leu26 pocket resulted in potent, novel substituted piperidine inhibitors of the HDM2-p53 interaction that demonstrated tumor regression in several human cancer xenograft models in mice. The structure of HDM2 in complex with inhibitors 3, 10, and 21 is described.

5.
Cancer Biol Ther ; 4(12): 1355-60, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16294023

ABSTRACT

To determine if A1 adenosine receptors mediate breast tumorigenesis, we evaluated A1 receptor expression in human tumor cell lines and human primary breast tumor tissues using both quantitative RT-PCR and Western blot analysis. A1 receptor mRNA expression is upregulated in all breast tumor cell lines examined (n=7) compared to normal mammary epithelial cells/cell lines (n=3) as determined by quantitative RT-PCR analysis. Western blot analysis indicates that protein expression of A1 adenosine receptor is higher in 15 (62.5%) of 24 human primary breast tumor tissues than in matched normal breast tissue. To explore its cellular function, the A1 adenosine receptor was depleted by small interfering RNA (siRNA) in MDA-MB-468 human breast tumor cells. Depletion of A1 receptors in MDA-MB-468 breast tumor cells attenuated both cell growth and cell proliferation as measured by cell number counts and [(14)C]-thymidine incorporation, respectively. Cell cycle analysis indicated that depletion of A1 receptors by siRNA impairs G(1) checkpoint, leading to marked accumulation of cells in G(2)/M phase, in agreement with the inhibitory effect on cell proliferation. Further supporting this finding, synchronization studies of Hela cells in various cell cycle phases suggest that A1 receptor expression is suppressed in G(2)/M cells and depletion of A1 receptor expression by siRNA produced differential expression of several key cell cycle regulators, i.e., accumulation of the cyclin-dependent kinase inhibitor p27 with concomitant reduction of CDK4 and cyclin E proteins. In addition to the impact on cell cycle progression, depletion of A1 receptors by siRNA results in substantial cell death and apoptosis as determined by FACS analysis and annexin V staining method. Together these findings suggest that the A1 adenosine receptor may contribute to tumor cell growth and survival in breast tumor cells.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Proliferation , RNA Interference , Receptor, Adenosine A1/metabolism , Breast/cytology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/metabolism
6.
J Biol Chem ; 280(35): 31101-8, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16006564

ABSTRACT

Lonafarnib (SCH66336) is a farnesyl transferase inhibitor (FTI) that inhibits the post-translational lipid modification of H-Ras and other farnesylated proteins. K- and N-Ras are also substrates of farnesyl transferase; however, upon treatment with FTIs, they are alternatively prenylated by geranylgeranyl transferase-1. Despite the failure to abrogate prenylation of K- and N-Ras, growth of many tumors in preclinical models is inhibited by FTIs. This suggests that the anti-proliferative action of FTIs is dependent on blocking the farnesylation of other proteins. Rheb (Ras homologue enriched in brain) is a farnesylated small GTPase that positively regulates mTOR (mammalian target of rapamycin) signaling. We found that Rheb and Rheb2 mRNA were elevated in various tumor cell lines relative to normal cells. Peptides derived from the carboxyl termini of human Rheb and Rheb2 are in vitro substrates for farnesyl transferase but not geranylgeranyl transferase-1. Rheb prenylation in cell culture was completely inhibited by SCH66336, indicating a lack of alternative prenylation. SCH66336 treatment also inhibited the phosphorylation of S6 ribosomal protein, a downstream target of Rheb and mTOR signaling. SCH66336 did not inhibit S6 phosphorylation in cells expressing Rheb-CSVL, a mutant construct of Rheb designed to be geranylgeranylated. Importantly, expression of Rheb-CSVL also abrogated SCH66336 enhancement of tamoxifen- and docetaxel-induced apoptosis in MCF-7 breast cancer cells and ES-2 ovarian cancer cells, respectively. Further, inhibition of Rheb signaling by rapamycin treatment, small interfering RNA, or dominant negative Rheb enhanced tamoxifen- and docetaxel-induced apoptosis, similar to FTI treatment. These studies demonstrated that Rheb is modified by farnesylation, is not a substrate for alternative prenylation, and plays a role in SCH66336 enhancement of the anti-tumor response to other chemotherapeutics.


Subject(s)
Alkyl and Aryl Transferases , Bridged-Ring Compounds/therapeutic use , Monomeric GTP-Binding Proteins/metabolism , Neoplasms/drug therapy , Neuropeptides/metabolism , Piperidines/metabolism , Protein Kinases/metabolism , Pyridines/metabolism , Tamoxifen/therapeutic use , Taxoids/therapeutic use , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Caspases/metabolism , Cell Line, Tumor , Farnesyltranstransferase , Humans , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Phosphorylation , Protein Kinases/genetics , Protein Prenylation , RNA, Messenger/metabolism , Ras Homolog Enriched in Brain Protein , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases
7.
J Biol Chem ; 280(13): 12935-43, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15659406

ABSTRACT

In this study, differential gene expression between normal human mammary epithelial cells and their malignant counterparts (eight well established breast cancer cell lines) was studied using Incyte GeneAlbum 1-6, which contains 65,873 cDNA clones representing 33,515 individual genes. 3,152 cDNAs showed a > or =3.0-fold expression level change in at least one of the human breast cancer cell lines as compared with normal human mammary epithelial cells. Integration of breast tumor gene expression data with the genes in the tumor suppressor p53 signaling pathway yielded 128 genes whose expression is altered in breast tumor cell lines and in response to p53 expression. A hierarchical cluster analysis of the 128 genes revealed that a significant portion of genes demonstrate an opposing expression pattern, i.e. p53-activated genes are down-regulated in the breast tumor lines, whereas p53-repressed genes are up-regulated. Most of these genes are involved in cell cycle regulation and/or apoptosis, consistent with the tumor suppressor function of p53. Follow-up studies on one gene, RAI3, suggested that p53 interacts with the promoter of RAI3 and repressed its expression at the onset of apoptosis. The expression of RAI3 is elevated in most tumor cell lines expressing mutant p53, whereas RAI3 mRNA is relatively repressed in the tumor cell lines expressing wild-type p53. Furthermore, ectopic expression of RAI3 in 293 cells promotes anchorage-independent growth and small interfering RNA-mediated depletion of RAI3 in AsPc-1 pancreatic tumor cells induces cell morphological change. Taken together, these data suggest a role for RAI3 in tumor growth and demonstrate the predictive power of integrative genomics.


Subject(s)
Gene Expression Regulation, Neoplastic , Genome, Human , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Agar/chemistry , Apoptosis , Breast Neoplasms/metabolism , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cluster Analysis , DNA, Complementary/metabolism , Down-Regulation , Epithelial Cells/metabolism , Genomics/methods , HeLa Cells , Humans , Mammary Glands, Human/metabolism , Models, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
8.
Methods Mol Biol ; 281: 33-54, 2004.
Article in English | MEDLINE | ID: mdl-15220520

ABSTRACT

The completion of the human genome sequence and availability of cDNA microarray technology provide new approaches to explore global cellular regulatory mechanisms. Here we present a strategy to identify genes regulated by specific transcription factors in the human genome, and apply it to p53. We first collected promoters or introns of all genes available using two methods: GenBank annotation and a computationally derived transcript map. The "FindPatterns" program is then used to search sequences in regulatory regions that match the p53 DNA-binding consensus sequence, resulting in the p53 Target Database. This database collects human genes that have at least one p53 DNA-binding sequence in their regulatory region. cDNA microarray was also used to identify genes that respond to p53 at a genomic scale. Integration of the microarray data and the p53 Target Database should greatly enrich direct p53 target genes. Taqman analysis and quantitative chromatin immunoprecipitation analysis are used to validate the in silico prediction and microarray data. Enrichment factor analysis is used to demonstrate that in silico prediction greatly enriches for genes that are transcriptionally regulated by p53 and assists us to identify other signaling pathways that are potentially connected to p53. The approaches can be extended to other transcription factors. The methods shown here illustrate a novel approach to the analysis of global gene regulatory networks through the integration of human genomic sequence information and genome-wide gene expression analysis.


Subject(s)
Computational Biology/methods , DNA/analysis , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Tumor Suppressor Protein p53/genetics , Binding Sites , Chromatin , Female , Gene Expression Regulation , Genome, Human , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Precipitin Tests/methods , RNA, Messenger/genetics , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Oncogene ; 22(23): 3645-54, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12789273

ABSTRACT

The temporal gene expression profile during the entire process of apoptosis and cell cycle progression in response to p53 in human ovarian cancer cells was explored with cDNA microarrays representing 33 615 individual human genes. A total of 1501 genes (4.4%) were found to respond to p53 (approximately 80% of these were repressed by p53) using 2.5-fold change as a cutoff. It was anticipated that most of p53 responsive genes resulted from the secondary effect of p53 expression at late stage of apoptosis. To delineate potential p53 direct and indirect target genes during the process of apoptosis and cell cycle progression, microarray data were combined with global p53 DNA-binding site analysis. Here we showed that 361 out of 1501 p53 responsive genes contained p53 consensus DNA-binding sequence(s) in their regulatory region, approximately 80% of which were repressed by p53. This is the first time that a large number of p53-repressed genes have been identified to contain p53 consensus DNA-binding sequence(s) in their regulatory region. Hierarchical cluster analysis of these genes revealed distinct temporal expression patterns of transcriptional activation and repression by p53. More genes were activated at early time points, while more repressed genes were found after the onset of apoptosis. A small-scale quantitative chromatin immunoprecipitation analysis indicated that in vivo p53-DNA interaction was detected in eight out of 10 genes, most of which were repressed by p53 at the early onset of apoptosis, suggesting that a portion of p53 target genes in the human genome could be negatively regulated by p53 via sequence-specific DNA binding. The approaches and genes described here should aid the understanding of global gene regulatory network of p53.


Subject(s)
Apoptosis/genetics , Cell Cycle/genetics , Ovarian Neoplasms/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Amino Acid Sequence , Binding Sites , Chromatin/metabolism , Cluster Analysis , Conserved Sequence , Female , Gene Expression Profiling , Genome, Human , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Precipitin Tests , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection/methods , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
10.
J Biol Chem ; 277(39): 36329-37, 2002 Sep 27.
Article in English | MEDLINE | ID: mdl-12138103

ABSTRACT

In this study we used adenovirus vector-mediated transduction of either the p53 gene (rAd-p53) or the p21(WAF1/CIP1) gene (rAd-p21) to mimic both p53-dependent and -independent up-regulation of p21(WAF1/CIP1) within a human ovarian cancer cell line, 2774, and the derivative cell lines, 2774qw1 and 2774qw2. We observed that rAd-p53 can induce apoptosis in both 2774 and 2774qw1 cells but not in 2774qw2 cells. Surprisingly, overexpression of p21(WAF1/CIP1) also triggered apoptosis within these two cell lines. Quantitative reverse transcription-PCR analysis revealed that the differential expression of BAX, BCL2, and caspase 3 genes, specific in rAd-p53-induced apoptotic cells, was not altered in rAd-p21-induced apoptotic cells, suggesting p21(WAF1/CIP1)-induced apoptosis through a pathway distinguishable from p53-induced apoptosis. Expression analysis of 2774qw1 cells infected with rAd-p21 on 60,000 cDNA microarrays identified 159 genes in response to p21(WAF1/CIP1) expression in at least one time point with 2.5-fold change as a cutoff. Integration of the data with the parallel microarray experiments with rAd-p53 infection allowed us to extract 66 genes downstream of both p53 and p21(WAF1/CIP1) and 93 genes in response to p21(WAF1/CIP1) expression in a p53-independent pathway. The genes in the former set may play a dual role in both p53-dependent and p53-independent pathways, and the genes in the latter set gave a mechanistic molecular explanation for p53-independent p21(WAF1/CIP1)-induced apoptosis. Furthermore, promoter sequence analysis suggested that transcription factor E2F family is partially responsible for the differential expression of genes following p21(WAF1/CIP1). This study has profound significance toward understanding the role of p21(WAF1/CIP1) in p53-independent apoptosis.


Subject(s)
Apoptosis , Cyclins/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , Transcription, Genetic , Adenoviridae/genetics , Caspase 3 , Caspases/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , DNA, Complementary/metabolism , Female , Genetic Vectors , Genome, Human , Humans , In Situ Nick-End Labeling , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Up-Regulation , bcl-2-Associated X Protein
11.
J Biol Chem ; 277(31): 28118-26, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12011061

ABSTRACT

Tumors evade cell death by constitutively activating cell survival pathways and suppressing intrinsic death machinery. Activation of cell survival pathways leads to transcriptional repression of genes associated with cell death and activation of ones promoting anti-apoptosis. Akt/protein kinase B phosphorylates forkhead transcription factors and prevents their nuclear localization, leading to repression of genes involved in apoptosis, such as Fas ligand (FasL). Using bioinformatic approaches, we have identified three consensus sequences for forkhead transcription factor binding in transforming growth factor beta2 (TGF-beta2) promoter. TGF-beta inhibits cell proliferation and induces apoptosis in many cell types, and acquisition of TGF-beta resistance is linked to tumorigenesis. In this study, we show that activated Akt down-regulates TGF-beta2 promoter, and sequences within the promoter that are related to consensus forkhead binding sites are necessary for repression. Forkhead factor FKHRL1 binds in vitro to the three consensus sequences and can activate TGF-beta2 promoter in normal and Akt-transformed cell lines. In human breast and pancreatic tumors, activated Akt expression correlated with down-regulation of TGF-beta 2 mRNA levels. A number of tumor cells expressing activated Akt were responsive to TGF-beta addition, indicating the presence of an intact TGF-beta-signaling pathway. These results suggest that repression of TGF-beta 2 promoter activity in cells expressing activated Akt may play a role in promoting tumorigenesis and escape from the growth-inhibitory and/or apoptotic effects of TGF-beta.


Subject(s)
Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/genetics , Base Sequence , Binding Sites , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Consensus Sequence , Forkhead Transcription Factors , Gene Expression Regulation/immunology , Genes, Reporter , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , Transforming Growth Factor beta2 , Tumor Cells, Cultured
12.
Oncogene ; 21(17): 2613-22, 2002 Apr 18.
Article in English | MEDLINE | ID: mdl-11965534

ABSTRACT

Survivin is an inhibitor of apoptosis protein, which is over-expressed in most tumors. Aberrant expression of survivin and loss of wild-type p53 in many tumors prompted us to investigate a possible link between these two events. Here we show that wild-type p53 represses survivin expression at both mRNA and protein levels. Transient transfection analyses revealed that the expression of wild-type p53, but not mutant p53, was associated with strong repression of the survivin promoter in various cell types. The over-expression of exogenous survivin protein rescues cells from p53-induced apoptosis in a dose-dependent manner, suggesting that loss of survivin mediates, at least, in part the p53-dependent apoptotic pathway. In spite of the presence of two putative p53-binding sites in the survivin promoter, deletion and mutation analyses suggested that neither site is required for transcriptional repression of survivin expression. This was confirmed by chromatin immunoprecipitation assays. Further analyses suggested that the modification of chromatin within the survivin promoter could be a molecular explanation for silencing of survivin gene transcription by p53.


Subject(s)
Apoptosis , Chromosomal Proteins, Non-Histone/genetics , Cysteine Proteinase Inhibitors/genetics , Microtubule-Associated Proteins , Tumor Suppressor Protein p53/genetics , Adenoviridae , Antibiotics, Antineoplastic/pharmacology , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cysteine Proteinase Inhibitors/metabolism , DNA Primers/chemistry , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins , Luciferases/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Molecular Sequence Data , Neoplasm Proteins , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Precipitin Tests , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...