Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(8): 1400-1411, 2021 08.
Article in English | MEDLINE | ID: mdl-34088831

ABSTRACT

Venetoclax is a small molecule inhibitor of the prosurvival protein BCL-2 that has gained market approval in BCL-2-dependent hematologic cancers including chronic lymphocytic leukemia and acute myeloid leukemia. Neuroblastoma is a heterogenous pediatric cancer with a 5-year survival rate of less than 50% for high-risk patients, which includes nearly all cases with amplified MYCN We previously demonstrated that venetoclax is active in MYCN-amplified neuroblastoma but has limited single-agent activity in most models, presumably the result of other pro-survival BCL-2 family protein expression or insufficient prodeath protein mobilization. As the relative tolerability of venetoclax makes it amenable to combining with other therapies, we evaluated the sensitivity of MYCN-amplified neuroblastoma models to rational combinations of venetoclax with agents that have both mechanistic complementarity and active clinical programs. First, the MDM2 inhibitor NVP-CGM097 increases the prodeath BH3-only protein NOXA to sensitize p53-wild-type, MYCN-amplified neuroblastomas to venetoclax. Second, the MCL-1 inhibitor S63845 sensitizes MYCN-amplified neuroblastoma through neutralization of MCL-1, inducing synergistic cell killing when combined with venetoclax. Finally, the standard-of-care drug cocktail cyclophosphamide and topotecan reduces the apoptotic threshold of neuroblastoma, thus setting the stage for robust combination efficacy with venetoclax. In all cases, these rational combinations translated to in vivo tumor regressions in MYCN-amplified patient-derived xenograft models. Venetoclax is currently being evaluated in pediatric patients in the clinic, including neuroblastoma (NCT03236857). Although establishment of safety is still ongoing, the data disclosed herein indicate rational and clinically actionable combination strategies that could potentiate the activity of venetoclax in patients with amplified MYCN with neuroblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Amplification , Gene Expression Regulation, Neoplastic/drug effects , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Animals , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Proliferation , Cyclophosphamide/administration & dosage , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Sulfonamides/administration & dosage , Topotecan/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
PLoS One ; 13(7): e0199699, 2018.
Article in English | MEDLINE | ID: mdl-29979702

ABSTRACT

ALDH1L1 is a folate-metabolizing enzyme abundant in liver and several other tissues. In human cancers and cell lines derived from malignant tumors, the ALDH1L1 gene is commonly silenced through the promoter methylation. It was suggested that ALDH1L1 limits proliferation capacity of the cell and thus functions as putative tumor suppressor. In contrast to cancer cells, mouse cell lines NIH3T3 and AML12 do express the ALDH1L1 protein. In the present study, we show that the levels of ALDH1L1 in these cell lines fluctuate throughout the cell cycle. During S-phase, ALDH1L1 is markedly down regulated at the protein level. As the cell cultures become confluent and cells experience increased contact inhibition, ALDH1L1 accumulates in the cells. In agreement with this finding, NIH3T3 cells arrested in G1/S-phase by a thymidine block completely lose the ALDH1L1 protein. Treatment with the proteasome inhibitor MG-132 prevents such loss in proliferating NIH3T3 cells, suggesting the proteasomal degradation of the ALDH1L1 protein. The co-localization of ALDH1L1 with proteasomes, demonstrated by confocal microscopy, supports this mechanism. We further show that ALDH1L1 interacts with the chaperone-dependent E3 ligase CHIP, which plays a key role in the ALDH1L1 ubiquitination and degradation. In NIH3T3 cells, silencing of CHIP by siRNA halts, while transient expression of CHIP promotes, the ALDH1L1 loss. The downregulation of ALDH1L1 is associated with the accumulation of the ALDH1L1 substrate 10-formyltetrahydrofolate, which is required for de novo purine biosynthesis, a key pathway activated in S-phase. Overall, our data indicate that CHIP-mediated proteasomal degradation of ALDH1L1 facilitates cellular proliferation.


Subject(s)
G1 Phase , Isoenzymes/metabolism , Proteasome Endopeptidase Complex/metabolism , Resting Phase, Cell Cycle , Retinal Dehydrogenase/metabolism , S Phase , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Cell Proliferation , Mice , NIH 3T3 Cells , Proteolysis
3.
J Nat Prod ; 72(3): 438-42, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19203291

ABSTRACT

Calothrixins A (1) and B (2) were converted to their O- and N-methylated derivatives, respectively. All four compounds were found to act as poisons of DNA topoisomerase I and to do so reversibly. Three of the calothrixins (1-3) were tested for their cytotoxicity toward cultured (p53 proficient) CEM leukemia cells and found to exhibit IC(50) values ranging from 0.20 to 5.13 muM. The cell cycle effects of calothrixins 1-3 were also studied. Calothrixin B (2) produced G(1) arrest at 0.1 muM concentration, while higher concentrations of calothrixins 1 and 3 resulted in cell accumulation in both the S and G(2)/M phases of the cell cycle. The cell cycle effects produced by the calothrixins were more readily reversible upon removal of the compounds than those produced by camptothecin.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Indole Alkaloids/chemistry , Indole Alkaloids/pharmacology , Topoisomerase I Inhibitors , Antineoplastic Agents/chemical synthesis , DNA Replication/drug effects , Drug Screening Assays, Antitumor , Humans , Indole Alkaloids/chemical synthesis , Molecular Structure
4.
J Am Chem Soc ; 130(39): 12888-9, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18774813

ABSTRACT

The topopyrones are fungal natural products shown previously to act as poisons of human DNA topoisomerase I. Recent total syntheses of the four known naturally occurring members of this class of compounds have enabled more detailed biochemical characterization. Presently it is shown that in addition to topoisomerase I, topopyrones A-D also act as poisons of human DNA topoisomerase II. The topopyrones thus represent a rare example of molecules capable of interacting effectively with more than one DNA topoisomerase.


Subject(s)
Anthraquinones/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyrones/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Anthraquinones/chemistry , Antigens, Neoplasm/metabolism , Base Sequence , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/chemistry , Humans , Pyrones/chemistry
5.
J Mol Biol ; 365(3): 561-9, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17095016

ABSTRACT

Many agents (e.g. camptothecins, indolocarbazoles, indenoisoquinolines, and dibenzonaphthyridines) stimulate topoisomerase I (TOP1)-mediated DNA cleavage (a behavior termed topoisomerase I poisoning) by interacting with both the DNA and the enzyme at the site of cleavage (typically by intercalation between the -1 and +1 base-pairs). The bibenzimidazoles, which include Hoechst 33258 and 33342, are a family of DNA minor groove-directed agents that also stimulate topoisomerase I-mediated DNA cleavage. However, the molecular mechanism by which these ligands poison TOP1 is poorly understood. Toward this goal, we have used a combination of mutational, footprinting, and DNA binding affinity analyses to define the DNA binding site for Hoechst 33258 and a related derivative that results in optimal induction of TOP1-mediated DNA cleavage. We show that this DNA binding site is located downstream from the site of DNA cleavage, encompassing the base-pairs from position +4 to +8. The distal nature of this binding site relative to the site of DNA cleavage suggests that minor groove-directed agents like the bibenzimidazoles poison TOP1 via a mechanism distinct from compounds like the camptothecins, which interact at the site of cleavage.


Subject(s)
Benzimidazoles/pharmacology , Bisbenzimidazole/pharmacology , DNA Cleavage , DNA Topoisomerases, Type I/metabolism , Indoles/pharmacology , Nucleic Acid Conformation/drug effects , Base Pairing/drug effects , Base Sequence , Benzimidazoles/chemistry , Binding Sites/drug effects , Bisbenzimidazole/chemistry , Camptothecin/pharmacology , DNA/chemistry , DNA/genetics , DNA Footprinting , Deoxyribonuclease I/metabolism , Fluorescence , Humans , Indoles/chemistry , Ligands , Molecular Sequence Data , Poly dA-dT/chemistry , Temperature
6.
J Med Chem ; 49(17): 5245-51, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16913713

ABSTRACT

The terbenzimidazoles are a class of anticancer agents that bind in the DNA minor groove. These compounds also exhibit a propensity for self-association, which can potentially impact their cellular bioavailabilities and activities. We have explored this possibility by using a broad range of biophysical and cytological techniques to characterize the self-association and cellular uptake properties of two terbenzimidazole analogues, 5-phenylterbenzimidazole (5PTB) and 5-phenyl-2'-(indolo-6-yl)bibenzimidazole (5P2'IBB). Concentration- and temperature-dependent fluorescence spectroscopy, dynamic light scattering, and transmission electron microscopy studies reveal that 5PTB and 5P2'IBB exhibit differing self-association properties. In this connection, 5PTB exhibits an enhanced propensity for self-association and forms larger and more stable aggregates than 5P2'IBB. In addition, the net uptake of 5PTB into human lymphoblast cells is diminished relative to that of 5P2'IBB. These observations suggest that the self-association properties of terbenzimidazoles modulate the cellular bioavailabilities of the compounds, with enhanced self-association propensity and aggregate size leading to reduced cellular bioavailability.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , DNA/drug effects , Indoles/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Molecular Structure , Structure-Activity Relationship , Temperature , Time Factors
7.
J Biol Chem ; 279(48): 50499-504, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15448130

ABSTRACT

Topoisomerase I (Top1), an abundant nuclear enzyme expressed throughout the cell cycle, relaxes DNA supercoiling by forming transient covalent DNA cleavage complexes. We show here that staurosporine, a ubiquitous inducer of apoptosis in mammalian cells, stabilizes cellular Top1 cleavage complexes. These complexes are formed indirectly as staurosporine cannot induce Top1 cleavage complexes in normal DNA with recombinant Top1 or nuclear extract from normal cells. In treated cells, staurosporine produces oxidative DNA lesions and generates reactive oxygen species (ROS). Quenching of these ROS by the antioxidant N-acetyl-l-cysteine or inhibition of the mitochondrial dependent production of ROS by the caspase inhibitor benzyloxycarbonyl-VAD prevents staurosporine-induced Top1 cleavage complexes. Down-regulation of Top1 by small interfering RNA decreases staurosporine-induced apoptotic DNA fragmentation. We propose that Top1 cleavage complexes resulting from oxidative DNA lesions generated by ROS in staurosporine-treated cells contribute to the full apoptotic response.


Subject(s)
Apoptosis/physiology , DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Enzyme Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Staurosporine/pharmacology , Apoptosis/drug effects , DNA/drug effects , DNA Fragmentation/drug effects , Humans , Tumor Cells, Cultured
8.
Cell Cycle ; 3(9): 1095-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15326388

ABSTRACT

Topoisomerase I (Top1) is expressed throughout the cell cycle. Top1 forms reversible and transient DNA cleavage complexes as it relaxes DNA supercoiling generated by transcription and replication. Recent findings indicate that mechanistically different inducers of apoptosis, arsenic trioxide, staurosporine and etoposide, which are inactive on purified Top1, induce Top1 cleavage complexes. These apoptotic Top1 cleavage complexes result from oxidative DNA lesions generated by reactive oxygen species during apoptosis. Their functional role could be to directly fragment chromatin and to further activate (amplify) apoptotic pathways.


Subject(s)
Apoptosis/physiology , DNA Replication/physiology , DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Oxidative Stress/physiology , Animals , DNA/genetics , DNA Fragmentation/physiology , Humans , Macromolecular Substances/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
9.
Biochem Pharmacol ; 68(5): 857-66, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15294448

ABSTRACT

Cisplatin, cis-[PtCl2(NH3)2], is commonly utilized in various combination chemotherapy protocols for the treatment of both ovarian and breast cancer while the corresponding trans isomer is therapeutically inactive. This work describes efforts to elucidate the cellular mechanism of action of a novel trans-platinum compound, trans-(dichloroamminethiazole)platinum(II) (ATZ), which demonstrates antiproliferative and cytotoxic effects against both MCF-7 human breast and A2780 human ovarian carcinoma cells in culture. A2780 cells were approximately twofold more sensitive to ATZ than MCF-7 cells in both cell growth and clonogenic survival assays. Dye exclusion studies revealed a 50-70% loss in cell viability within the first 12 h of drug treatment in both cell lines. This initial wave of cell death was succeeded by a prolonged interval of growth arrest during which a small fraction of apoptotic cells was detected. Binding of ATZ to DNA, as estimated by atomic absorption spectroscopy, was similar for the two cell lines and was almost completely reversed 24 h after drug removal. ATZ also induced DNA strand breakage as well as DNA-protein crosslinking during the initial 12 h period when the bulk of cell death was evident. However, neither the extent of DNA strand breakage nor that of DNA protein crosslinking was sufficient to explain the different drug sensitivity in the two cell lines. At 24 and 48 h after exposure of MCF-7 cells to high concentrations of ATZ, the formation of DNA-topoisomerase I complexes is detected, coincident with a high degree of apoptosis. These studies suggest that ATZ has the capacity to interfere with topoisomerase I in the tumor cell, a function not evident in cis-platinum-based drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , DNA Damage/drug effects , DNA, Neoplasm/drug effects , Organoplatinum Compounds/pharmacology , Animals , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Cross-Linking Reagents/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Female , Humans , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
10.
Int J Cancer ; 109(6): 848-54, 2004 May 10.
Article in English | MEDLINE | ID: mdl-15027118

ABSTRACT

Overcoming drug resistance has become an important issue in cancer chemotherapy. Among all known mechanisms that confer resistance, active efflux of chemotherapeutic agents by proteins from the ATP-binding cassette family has been extensively reported. The aim of the present study was to determine the involvement of ABCG2 in resistance to SN38 (the active metabolite of irinotecan) in colorectal cancer. By progressive exposure to increasing concentrations of SN38, we isolated 2 resistant clones from the human colon carcinoma cell line HCT116. These clones were 6- and 53-fold more resistant to SN38 than the HCT116-derived sensitive clone. Topoisomerase I expression was unchanged in our resistant variants. The highest resistance level correlated with an ABCG2 amplification. This overexpression was associated with a marked decrease in the intracellular accumulation of SN38. The inhibition of ABCG2 function by Ko143 demonstrated that enhanced drug efflux from resistant cells was mediated by the activity of ABCG2 protein and confirmed that ABCG2 is directly involved in acquired resistance to SN38. Furthermore, we show, for the first time in clinical samples, that the ABCG2 mRNA content in hepatic metastases is higher after an irinotecan-based chemotherapy than in irinotecan-naive metastases. In conclusion, this study supports the potential involvement of ABCG2 in the development of irinotecan resistance in vivo.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/drug therapy , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Antineoplastic Agents, Phytogenic/therapeutic use , Colon/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Topoisomerases, Type I/metabolism , Gene Expression Regulation, Neoplastic , Humans , Irinotecan , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Messenger/metabolism , RNA, Neoplasm , Reverse Transcriptase Polymerase Chain Reaction
11.
Proc Natl Acad Sci U S A ; 100(21): 12498-503, 2003 Oct 14.
Article in English | MEDLINE | ID: mdl-14523238

ABSTRACT

DNA topoisomerase II (Top2) is the target of some of the most effective anticancer DNA intercalators. To determine the effect of intercalating ligands at defined positions relative to a known DNA cleavage site for human Top2alpha, we synthesized oligodeoxynucleotides containing single trans-opened benzo[a]pyrene 7,8-diol 9,10-epoxide (DE) deoxyadenosine (dA) adducts of known absolute configuration, placed at specific positions in a duplex sequence containing staggered Top2 cleavage sites on both strands. Because the orientations of the intercalated hydrocarbon are known from NMR solution structures of duplex oligonucleotides containing these dA adducts, a detailed analysis of the relationship between the position of intercalation and trapping of Top2 is possible. Our findings demonstrate that (i) Top2 cleavage complexes are trapped by intercalation of the hydrocarbon at either of the staggered cleavage sites or immediately adjacent to the base pairs flanking the cleavage sites within the stagger; (ii) both concerted and nonconcerted cleavage by both subunits of a Top2 homodimer were detected depending on the position of the benzo[a]pyrene DE dA adduct; and (iii) intercalation immediately outside of the staggered Top2 cleavage site, and to a lesser extent in the middle of the stagger, prevents Top2 from cleaving DNA at this site, consistent with the effect of some intercalators as suppressors of Top2-mediated DNA cleavage. These results identify specific binding sites for intercalators that result in trapping of Top2. Such poisoning of Top2 by bulky polycyclic aromatic hydrocarbon DE adducts constitutes a potential mechanism for their carcinogenic activity.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/analogs & derivatives , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/drug effects , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/chemistry , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/pharmacology , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Base Sequence , Binding Sites , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Adducts/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins , Humans , In Vitro Techniques , Intercalating Agents/pharmacology , Models, Molecular , Molecular Structure , Poly-ADP-Ribose Binding Proteins , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Substrate Specificity
12.
Curr Med Chem Anticancer Agents ; 3(4): 271-90, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12769773

ABSTRACT

Topoisomerase inhibitors are among the most efficient inducers of apoptosis. The main pathways leading from topoisomerase-mediated DNA damage to cell death involve activation of caspases in the cytoplasm by proapoptotic molecules released from mitochondria. In some cells, apoptotic response also involves the death receptor Fas (APO-1/CD95). The engagement of these apoptotic effector pathways is tightly controlled by upstream regulatory pathways that respond to DNA lesions-induced by topoisomerase inhibitors in cells undergoing apoptosis. These include the proapoptotic Chk2, c-Abl and SAPK/JNK pathways, the survival PI(3)kinase-Akt-dependent pathway and the transcription factors p53 and NF-kappaB. Initiation of cellular responses to DNA lesions-induced by topoisomerase inhibitors is ensured by the protein kinases DNA-PK, ATM and ATR, which bind to DNA breaks. These kinases commonly called "DNA sensors" mediate their effects (DNA repair, cell cycle arrest and/or apoptosis) by phosphorylating a large number of substrates, including several downstream kinases such as c-Abl and the checkpoint protein Chk2. c-Abl induces apoptosis by activating cell death pathways (e.g., SAPK, p53 and p73) and inhibiting cell survival pathways [e.g., PI(3)kinase]. The DNA-damage regulating kinase Chk2, in addition to its role in cell cycle arrest and/or DNA repair, can induce apoptosis by phosphorylation/activation of the promyelocytic leukemia (PML) protein and p53. Finally, we will review the recent observations that support a role for topoisomerases in chromatin fragmentation during the execution phase of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Topoisomerase Inhibitors , DNA Damage , DNA Topoisomerases/genetics , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Models, Biological
13.
Mol Carcinog ; 33(1): 1-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11807952

ABSTRACT

In previous studies, we have shown that human breast and lung carcinoma cells and mouse nontransformed type II lung cells fail to undergo cell-cycle arrest in G(1) phase in response to treatment with hydrocarbon carcinogens but rather accumulate in the S phase with damaged DNA. This situation may lead to replication of DNA on a damaged template and enhance frequency of mutations. The mechanism of this G(1) arrest failure was examined. Western immunoblot analyses of MCF7 human mammary cancer cells exposed to actinomycin D (used as a positive control for G(1) cell-cycle arrest) or hydrocarbon carcinogens revealed that while all of these chemicals caused an increase in p53, only trace levels of p21(waf1/cip1) protein were observed in the hydrocarbon carcinogen-treated samples. Similarly, in murine lung E10 type II cells, p53 but not p21(waf1/cip1) protein increased in response to benzo[a]pyrene dihydrodiol epoxide. Treatment of either MCF7 mammary or E10 lung cells with the protease inhibitor calpain I resulted in increased levels of p21(waf1/cip1) protein and enhancement of arrest of the cells in early phases of the cell cycle (G(1) and early S phase). The results suggest that failure of cell-cycle arrest in carcinogen-treated mammary and lung cells is related to increased protease-mediated degradation of p21(waf1/cip1) and/or related regulatory proteins.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cyclins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Epithelial Cells/metabolism , Lung/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinogens/pharmacology , Chrysenes/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Dactinomycin/pharmacology , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/pharmacology , Humans , Mice , Nocodazole/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...