Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Neurooncol ; 152(2): 233-243, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33517558

ABSTRACT

PURPOSE: Depatux-m is an antibody drug conjugate (ADC) that targets and inhibits growth of cancer cells overexpressing the epidermal growth factor receptor (EGFR) or the 2-7 deletion mutant (EGFRvIII) in tumor models in vitro and in vivo. Treatment of patients suffering from relapsed/refractory glioblastoma (GBM) with a combination of depatux-m and temozolomide (TMZ) tended to increase overall survival. As a first step to understand the nature of the interaction between the two drugs, we investigated whether the interaction was synergistic, additive or antagonistic. METHODS: The efficacy of ADCs, antibodies, TMZ and radiation was tested in xenograft models of GBM, U-87MG and U-87MG EGFRvIII. Both models express EGFR. U-87MG EGFRvIII was transduced to express EGFRvIII. Changes in tumor volume, biomarkers of cell death and apoptosis after treatment were used to measure efficacy of the various treatments. Synergism of depatux-m and TMZ was verified in three-dimensional cultures of U-87MG and U-87MG EGFRvIII by the method of Chou and Talalay. RESULTS: Combined with TMZ and radiotherapy (RT), depatux-m inhibited xenograft growth of U-87MG and U-87MG EGFRvIII more than either treatment with depatux-m or TMZ + RT. Durability of the response to depatux-m + TMZ + RT or depatux-m + TMZ was more pronounced in U-87MG EGFRvIII than in U-87MG. Efficacy of depatux-m + TMZ was synergistic in U-87MG EGFRvIII and additive in U-87MG. CONCLUSION: Adding depatux-m enhances the efficacy of standard of care therapy in preclinical models of GBM. Durability of response to depatux-m + TMZ in vivo and synergy of the drug-drug interaction correlates with the amount of antigen expressed by the tumor cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms , Glioblastoma , Temozolomide/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Xenograft Model Antitumor Assays
2.
J Org Chem ; 86(3): 2499-2521, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33417458

ABSTRACT

Thailanstatin A and spliceostatin D, two naturally occurring molecules endowed with potent antitumor activities by virtue of their ability to bind and inhibit the function of the spliceosome, and their natural siblings and designed analogues, constitute an appealing family of compounds for further evaluation and optimization as potential drug candidates for cancer therapies. In this article, the design, synthesis, and biological investigation of a number of novel thailanstatin A analogues, including some accommodating 1,1-difluorocyclopropyl and tetrahydrooxazine structural motifs within their structures, are described. Important findings from these studies paving the way for further investigations include the identification of several highly potent compounds for advancement as payloads for antibody-drug conjugates (ADCs) as potential targeted cancer therapies and/or small molecule drugs, either alone or in combination with other anticancer agents.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Antineoplastic Agents/pharmacology , Pyrans/pharmacology
3.
Nucleic Acids Res ; 42(15): 9771-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25100870

ABSTRACT

NONO, SFPQ and PSPC1 make up a family of proteins with diverse roles in transcription, RNA processing and DNA double-strand break (DSB) repair. To understand long-term effects of loss of NONO, we characterized murine embryonic fibroblasts (MEFs) from knockout mice. In the absence of genotoxic stress, wild-type and mutant MEFs showed similar growth rates and cell cycle distributions, and the mutants were only mildly radiosensitive. Further investigation showed that NONO deficiency led to upregulation of PSPC1, which replaced NONO in a stable complex with SFPQ. Knockdown of PSPC1 in a NONO-deficient background led to severe radiosensitivity and delayed resolution of DSB repair foci. The DNA-dependent protein kinase (DNA-PK) inhibitor, NU7741, sensitized wild-type and singly deficient MEFs, but had no additional effect on doubly deficient cells, suggesting that NONO/PSPC1 and DNA-PK function in the same pathway. We tested whether NONO and PSPC1 might also affect repair indirectly by influencing mRNA levels for other DSB repair genes. Of 12 genes tested, none were downregulated, and several were upregulated. Thus, NONO or related proteins are critical for DSB repair, NONO and PSPC1 are functional homologs with partially interchangeable functions and a compensatory response involving PSPC1 blunts the effect of NONO deficiency.


Subject(s)
DNA Repair , DNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , DNA Breaks, Double-Stranded , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Radiation Tolerance , Up-Regulation
4.
Cancer Cell ; 7(6): 547-59, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950904

ABSTRACT

The p53 tumor suppressor protein is regulated by its interaction with HDM2, which serves as a ubiquitin ligase (E3) to target p53 for degradation. We have identified a family of small molecules (HLI98) that inhibits HDM2's E3 activity. These compounds show some specificity for HDM2 in vitro, although at higher concentrations effects on unrelated RING and HECT domain E3s are detectable, which could be due, at least in part, to effects on E2-ubiquitin thiol-ester levels. In cells, the compounds allow the stabilization of p53 and HDM2 and activation of p53-dependent transcription and apoptosis, although other p53-independent toxicity was also observed.


Subject(s)
Enzyme Inhibitors/pharmacology , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Endosomal Sorting Complexes Required for Transport , Enzyme Inhibitors/chemistry , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavins/chemistry , Gene Expression/drug effects , Humans , Mice , Molecular Structure , Nedd4 Ubiquitin Protein Ligases , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding/drug effects , Proteins/antagonists & inhibitors , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Transfection , Tumor Suppressor Protein p53/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
5.
Nucleic Acids Res ; 35(6): 1919-32, 2007.
Article in English | MEDLINE | ID: mdl-17337438

ABSTRACT

Alternative splicing produces functionally distinct proteins participating in cellular processes including differentiation and development. CoAA is a coactivator that regulates transcription-coupled splicing and its own pre-mRNA transcript is alternatively spliced. We show here that the CoAA gene is embryonically expressed and alternatively spliced in multiple tissues to three splice variants, CoAA, CoAM and CoAR. During retinoic-acid-induced P19 stem cell differentiation, the expression of CoAA undergoes a rapid switch to its dominant negative splice variant CoAM in the cavity of the embryoid body. CoAM functionally inhibits CoAA, and their switched expression up-regulates differentiation marker Sox6. Using a CoAA minigene cassette, we find that the switched alternative splicing of CoAA and CoAM is regulated by the cis-regulating sequence upstream of the CoAA basal promoter. Consistent to this, we show that p54(nrb) and PSF induce CoAM splice variant through the cis-regulating sequence. We have previously shown that the CoAA gene is amplified in human cancers with a recurrent loss of this cis-regulating sequence. These results together suggest that the upstream regulatory sequence contributes to alternative splicing of the CoAA gene during stem cell differentiation, and its selective loss in human cancers potentially deregulates CoAA alternative splicing and alters stem cell differentiation.


Subject(s)
Alternative Splicing , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Oncogene Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Cell Differentiation , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Developmental , High Mobility Group Proteins/biosynthesis , Mice , Molecular Sequence Data , Nuclear Matrix-Associated Proteins/metabolism , Oncogene Proteins/metabolism , PTB-Associated Splicing Factor , RNA-Binding Proteins/metabolism , Regulatory Elements, Transcriptional , SOXD Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/metabolism
6.
ACS Appl Mater Interfaces ; 10(48): 41678-41689, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30418738

ABSTRACT

Atomic Layer Deposition (ALD) is very attractive for producing optical quality thin films, including transparent barrier films on metal-coated astronomical mirrors. To date, ALD of mirror coatings has been limited to relatively small-sized substrates. A new ALD tool has been designed, constructed, and tested to apply uniform protective coatings over a 0.9 m diameter substrate in a 1 m diameter scale deposition plane. The new tool, which we have named the meter scale ALD system (MSAS), employs a unique chamber design that isolates a large substrate surface to be coated by utilizing the substrate as a wall of the reaction chamber. The MSAS is mechanically designed to be rapidly reconfigurable for selective area coating of custom substrates with arbitrary shape, size, and permanent backside hardware attachments. The design, implementation, results, and future applications of this new tool are discussed for coating large-area optical substrates, specifically protective coatings for silver mirrors, and other future large astronomical optics. To demonstrate the potential of this new design, aluminum oxide was deposited by thermal ALD using trimethylaluminum and water at a low reaction temperature of 60 °C. Growth rate and uniformity, which are dependent on precursor pulse times and chamber purge times, show that the two half-reactions occur in a saturated regime, matching typical characteristics of ideal ALD behavior. Aluminum oxide deposition process parameters of the MSAS are compared with those of a conventional 100 mm wafer-scale ALD tool, and saturated ALD growth over the 0.9 m substrate is realized with a simple scaling factor applied to precursor pulse and purge times. This initial test shows that lateral thickness uniformity across a 0.9 m substrate is within 2.5% of the average film thickness, and simple steps to realize 1% uniformity have been identified for next growths. Results show promising application of transparent robust dielectric films as uniform coatings across large optical components scaled to meter-sized substrates.

7.
Mol Cancer Ther ; 17(4): 795-805, 2018 04.
Article in English | MEDLINE | ID: mdl-29483208

ABSTRACT

Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795-805. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Glioblastoma/drug therapy , Immunoconjugates/pharmacology , Oligopeptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/chemistry , Apoptosis , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoconjugates/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
ACS Appl Mater Interfaces ; 9(18): 15841-15847, 2017 May 10.
Article in English | MEDLINE | ID: mdl-28418638

ABSTRACT

Silver thin films covered with dielectric films serving as protective coatings are desired for telescope mirrors, but durable coatings have proved elusive. As part of an effort to develop long-lived protected-silver mirrors, silver thin films were deposited by electron beam evaporation using a physical vapor deposition system at the University of California Observatories Astronomical Coatings Lab. The silver films were later covered with a stack of dielectric films utilizing silicon nitride and titanium dioxide deposited by ion-assisted electron beam evaporation to fabricate protected mirrors. In-situ argon ion bombardment was introduced after silver deposition and prior to the deposition of dielectric films to assess its effects on the performance of the mirrors. We found that ion bombardment of the silver influenced surface morphology and reflectivity, and these effects correlated with time between silver deposition and ion bombardment. The overall reflectivity at wavelengths in the range of 350-800 nm was found to improve due to ion bombardment, which was qualitatively interpreted as a result of decreased surface plasmon resonance coupling. We suggest that the observed decrease in coupling is caused by silver grain boundary pinning due to ion bombardment suppressing silver surface diffusion, forming smoother silver-dielectric interfaces.

9.
Mol Cancer Ther ; 15(4): 661-9, 2016 04.
Article in English | MEDLINE | ID: mdl-26846818

ABSTRACT

Targeting tumor-overexpressed EGFR with an antibody-drug conjugate (ADC) is an attractive therapeutic strategy; however, normal tissue expression represents a significant toxicity risk. The anti-EGFR antibody ABT-806 targets a unique tumor-specific epitope and exhibits minimal reactivity to EGFR in normal tissue, suggesting its suitability for the development of an ADC. We describe the binding properties and preclinical activity of ABT-414, an ABT-806 monomethyl auristatin F conjugate. In vitro, ABT-414 selectively kills tumor cells overexpressing wild-type or mutant forms of EGFR. ABT-414 inhibits the growth of xenograft tumors with high EGFR expression and causes complete regressions and cures in the most sensitive models. Tumor growth inhibition is also observed in tumor models with EGFR mutations, including activating mutations and those with the exon 2-7 deletion [EGFR variant III (EGFRvIII)], commonly found in glioblastoma multiforme. ABT-414 exhibits potent cytotoxicity against glioblastoma multiforme patient-derived xenograft models expressing either wild-type EGFR or EGFRvIII, with sustained regressions and cures observed at clinically relevant doses. ABT-414 also combines with standard-of-care treatment of radiation and temozolomide, providing significant therapeutic benefit in a glioblastoma multiforme xenograft model. On the basis of these results, ABT-414 has advanced to phase I/II clinical trials, and objective responses have been observed in patients with both amplified wild-type and EGFRvIII-expressing tumors. Mol Cancer Ther; 15(4); 661-9. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Epitopes , ErbB Receptors/antagonists & inhibitors , Immunoconjugates/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Antibody Affinity , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Disease Models, Animal , Epitopes/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Molecular Targeted Therapy , Mutation , Protein Binding , Xenograft Model Antitumor Assays
10.
Essays Biochem ; 39: 89-104, 2003.
Article in English | MEDLINE | ID: mdl-14585076

ABSTRACT

Although cancer is a disease that will afflict one out of three people in the Western world, when considered at a cellular level, it is a rare clonal event. Long-lived organisms, such as humans, have evolved strategies to restrict the development of potentially malignant cells, and one such mechanism is the coupling of proliferative and apoptotic pathways. Multiple oncogenes have the ability to trigger apoptosis when expressed in an inappropriate fashion, and this is thought to restrict tumour formation by eliminating potentially malignant cells that have acquired a mutation stimulating proliferation. Hence for a tumour to arise, in addition to mutations that drive proliferation, mutations that prevent apoptosis are also a prerequisite.


Subject(s)
Apoptosis , Gene Expression Regulation , Oncogenes/physiology , Animals , Cell Division , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , Genes, myc/physiology , Genes, p53/physiology , Humans , Mice , Mitochondria/metabolism , Models, Biological , Mutation , Nuclear Proteins/metabolism , Signal Transduction , Transcriptional Activation , Tumor Protein p73 , Tumor Suppressor Proteins
11.
Mol Cancer Ther ; 14(5): 1141-51, 2015 May.
Article in English | MEDLINE | ID: mdl-25731184

ABSTRACT

Despite clinical efficacy, current approved agents targeting EGFR are associated with on-target toxicities as a consequence of disrupting normal EGFR function. MAb 806 is a novel EGFR antibody that selectively targets a tumor-selective epitope suggesting that a mAb 806-based therapeutic would retain antitumor activity without the on-target toxicities associated with EGFR inhibition. To enable clinical development, a humanized variant of mAb 806 designated ABT-806 was generated and is currently in phase 1 trials. We describe the characterization of binding and functional properties of ABT-806 compared with the clinically validated anti-EGFR antibody cetuximab. ABT-806 binds the mutant EGFRvIII with high affinity and, relative to cetuximab, exhibits increased potency against glioblastoma multiforme cell line and patient-derived xenografts expressing this form of the receptor. ABT-806 also inhibits the growth of squamous cell carcinoma xenograft models expressing high levels of wild-type EGFR, associated with inhibition of EGFR signaling, although higher doses of ABT-806 than cetuximab are required for similar activity. ABT-806 enhances in vivo potency of standard-of-care therapies used to treat glioblastoma multiforme and head and neck squamous cell carcinoma. An indium-labeled version of ABT-806, [(111)In]-ABT-806, used to investigate the relationship between dose and receptor occupancy, revealed greater receptor occupancy at lowers doses in an EGFRvIII-expressing model and significant uptake in an orthotopic model. Collectively, these results suggest that ABT-806 may have antitumor activity superior to cetuximab in EGFRvIII-expressing tumors, and similar activity to cetuximab in tumors highly overexpressing wild-type EGFR with reduced toxicity.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Cetuximab/administration & dosage , ErbB Receptors/immunology , ErbB Receptors/metabolism , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Standard of Care , Xenograft Model Antitumor Assays
12.
Mol Cell Biochem ; 286(1-2): 67-76, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16432762

ABSTRACT

XIAP-associated factor 1 (Xaf1) binds XIAP and re-localizes it to the nucleus, thus inhibiting XIAP activity and enhancing apoptosis [1]. Xaf1 expression is reduced or absent in tumor samples and cell lines suggesting it may function as a tumor suppressor [2-5]. To further study Xaf1 function we generated Xaf1 inducible cells in the osteosarcoma cell line Saos-2. Despite Xaf1 inducing apoptosis that is dramatically enhanced by TNFalpha we find no evidence for an interaction between Xaf1 and XIAP. Furthermore, Xaf1 expression sensitized XIAP-/- fibroblasts to TNFalpha, demonstrating the existence of a novel mechanism of Xaf1 induced apoptosis distinct from antagonizing XIAP. Xaf1 expression promotes cytochrome c release that cannot be blocked by inhibition of caspase activity. This implicates a role for the mitochondrial apoptotic pathway, consistent with the ability of Bcl2 to block Xaf1 induced apoptosis. The data indicate that in Saos2 cells Xaf1 activates the mitochondrial apoptotic pathway to facilitate cytochrome c release, thus amplifying apoptotic signals from death receptors.


Subject(s)
Apoptosis/drug effects , Neoplasm Proteins/physiology , Tumor Necrosis Factor-alpha/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caspase Inhibitors , Cell Line, Tumor , Cells, Cultured , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mitochondria/drug effects , Mitochondria/physiology , Mutation/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
13.
Proc Natl Acad Sci U S A ; 99(6): 3529-34, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11891335

ABSTRACT

The p53-related protein p73 has many functions similar to that of p53 including the ability to induce cell-cycle arrest and apoptosis. Both p53 and p73 function as transcription factors, and p73 activates expression of many genes that also are regulated by p53. Despite their similarities, it is evident that p53 and p73 are not interchangeable functionally, with p73 playing a role in normal growth and development that is not shared by p53. In this paper we describe the ability of p73beta but not p53 to activate expression of the cyclin-dependent kinase inhibitor p57(KIP) and KvLQT1, two genes that are coregulated in an imprinted region of the genome. Our results suggest that p73 may regulate expression of genes through mechanisms that are not shared by p53, potentially explaining the different contributions of p53 and p73 to normal development.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Potassium Channels, Voltage-Gated , Up-Regulation , Animals , Blotting, Western , Cell Line , Cyclin-Dependent Kinase Inhibitor p57 , E2F Transcription Factors , Genes, Tumor Suppressor , Genomic Imprinting/genetics , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Mice , Nuclear Proteins/genetics , Potassium Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL