Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Onco Targets Ther ; 12: 6515-6523, 2019.
Article in English | MEDLINE | ID: mdl-31616161

ABSTRACT

PURPOSE: The efficacy of traditional therapies for oral carcinoma (OC) is limited. Oncolytic adenovirus, a novel strategy of cancer therapy, shows potential use in OC treatment. However, its clinical application is limited by pre-existing neutralizing antibodies. Thus, this study aimed to examine the efficacy of a new modified adenovirus against OC in vitro and in vivo. MATERIALS AND METHODS: A multiple modified adenovirus (MMAD) armed with IL-13 (MMAD-IL-13) was constructed, and its effect on Cal-27 cells was examined. The potency of MMAD-IL-13 was examined in vitro and in vivo. For in vitro experiment, CCK-8 kit was used to determine the IC50 of MMAD-IL-3 in OC cell lines. For in vivo experiment, Cal-27 xenograft models were used to determine the antitumor effect of MMAD-IL-13. Apoptosis was measured in Cal-27 cells by Western blotting assay. Immunity response was detected in Cal-27 xenograft models 7 days after intratumoral injection with MMAD-IL-13. The potency of MMAD and MMAD-IL-13 was compared in Cal-27 peripheral blood mononuclear cells (PBMCs) models. RESULTS: MMAD-IL-13 was successfully constructed; the harvested virus could be replicated and they overexpressed human IL-13 in Cal-27 cells. Compared with MMAD, MMAD-IL-13 showed enhanced antitumor effect in vitro by inducing apoptosis and reducing percentage of M2 macrophages in tumor environment in vivo. MMAD-IL-13 also showed potent antitumor effect in Cal-27, SCC-4, and Tca8113 cells in vitro and in Cal-27 xenograft models in vivo. However, MMAD-IL13 did not harm normal human oral epithelial cells in vitro and exhibited no effect on body weight in Cal-27 xenograft models. In Cal-27 PBMC models, MMAD-IL-13 showed stronger antitumor effect than MMAD. CONCLUSION: A new oncolytic adenovirus carrying the human IL-13 gene was constructed. This virus effectively led to remission of tumor development and death of OC cells in vivo and in vitro, showing its potential as a clinical cancer therapy.

2.
Theranostics ; 9(5): 1247-1263, 2019.
Article in English | MEDLINE | ID: mdl-30867828

ABSTRACT

Background and Aims: Prostate specific membrane antigen (PSMA) is specifically expressed on prostate epithelial cells and markedly overexpressed in almost all prostate cancers. TRIM24 is also up-regulated from localized prostate cancer to metastatic castration-resistant prostate cancer (CRPC). Because of the high relevance of TRIM24 for cancer development and the universal expression of PSMA in CPRC, we investigated the efficacy of human monoclonal PSMA antibody (PSMAb)-based platform for the targeted TRIM24 siRNA delivery and its therapeutic efficacy in CRPC in vivo and in vitro. Methods: The therapeutic complexes were constructed by conjugating PSMAb and sulfo-SMCC-protamine, and encapsulating TRIM24 siRNA. Flow cytometry, immunofluorescence, and fluorescence imaging were performed to detect the receptor-binding, internalization, and targeted delivery of PSMAb-sulfo-SMCC-protamine (PSP)-FAM-siRNA complex (PSPS) in vitro and in vivo. CCK-8, plate-colony formation, apoptosis, cell cycle, and Transwell assays were performed to evaluate the therapeutic potential of the PSP-TRIM24 siRNA complex in vitro, whereas the in vivo therapeutic efficacy was monitored by small animal imaging, radiography, and micro CT. Results: We confirmed that PSP could efficiently protect siRNA from enzymatic digestion, enable targeted delivery of siRNA, and internalize and release siRNA into PSMA-positive (PSMA+) prostate cancer cells in vitro and in vivo. Silencing TRIM24 expression by the PSP-TRIM24 siRNA complex could dramatically suppress proliferation, colony-formation, and invasion of PSMA+ CRPC cells in vitro, and inhibit tumor growth of PSMA+ CRPC xenografts and bone loss in PSMA+ CRPC bone metastasis model without obvious toxicity at therapeutic doses in vivo. Conclusion: PSMAb mediated TRIM24 siRNA delivery platform could significantly inhibit cell proliferation, colony-formation, and invasion in PSMA+ CRPC in vitro and suppressed tumor growth and bone loss in PSMA+ CRPC xenograft and bone metastasis model.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, Surface/immunology , Carrier Proteins/antagonists & inhibitors , Glutamate Carboxypeptidase II/immunology , Molecular Targeted Therapy/methods , Prostatic Neoplasms, Castration-Resistant/drug therapy , RNA, Small Interfering/administration & dosage , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Male , Mice, Nude , Models, Theoretical , Therapeutic Uses , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
3.
Cancer Lett ; 403: 119-127, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28634045

ABSTRACT

EGFR amplification and mutations are the most common oncogenic events in GBM. EGFR overexpression correlates with GBM invasion, but the underlying mechanisms are poorly understood. In a previous study, we showed that AJAP1 is involved in regulating F-actin to inhibit the invasive ability of GBM. In addition, in a GBM cell line, the AJAP1 promoter was highly bound by H3K27me3 and, through bioinformatics analysis, we found that AJAP1 expression was negatively correlated with EGFR. In this study, we found that the pathway downstream of EGFR had a higher activation level in GBM cell lines, which led to excessive tumor suppressor silencing. Therefore, we deduced that in glioma cells, the pathway downstream of EGFR remodels the cytoskeleton via AJAP1 epigenetic silencing to enhance invasion. Furthermore, MK2206 reversed AJAP1 downregulation by inhibiting the EGFR pathway. In vivo, MK2206 also inhibited the proliferation and local invasion of 87-EGFRvIII. These data suggest that activation of the EGFR signal transduction pathway genetically silences anti-oncogenes to enhance GBM malignancy. MK2206 might be a promising therapeutic for EGFR/EGFRvIII-positive GBMs.


Subject(s)
Actin Cytoskeleton/metabolism , Brain Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , DNA Methylation , Epigenesis, Genetic , ErbB Receptors/metabolism , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Computational Biology , Databases, Genetic , Enhancer of Zeste Homolog 2 Protein/metabolism , ErbB Receptors/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinase/metabolism , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Transfection , Xenograft Model Antitumor Assays
4.
Cancer Lett ; 388: 269-280, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27998759

ABSTRACT

Worldwide, glioblastoma (GBM) is the most lethal and frequent intracranial tumor. Despite decades of study, the overall survival of GBM patients remains unchanged. epidermal growth factor receptor (EGFR) amplification and gene mutation are thought to be negatively correlated with prognosis. In this study, we used proteomics to determine that UBXN1 is a negative downstream regulator of the EGFR mutation vIII (EGFRvIII). Via bioinformatics analysis, we found that UBXN1 is a factor that can improve glioma patients' overall survival time. We also determined that the down-regulation of UBXN1 is mediated by the upregulation of H3K27me3 in the presence of EGFRvIII. Because NF-κB can be negatively regulated by UBXN1, we believe that EGFRwt/vIII activates NF-κB by suppressing UBXN1 expression. Importantly, we used the latest genomic editing tool, CRISPR/Cas9, to knockout EGFRwt/vIII on exon 17 and further proved that UBXN1 is negatively regulated by EGFRwt/vIII. Furthermore, knockout of EGFR/EGFRvIII could benefit GBM in vitro and in vivo, indicating that CRISPR/Cas9 is a promising therapeutic strategy for both EGFR amplification and EGFR mutation-bearing patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Glioma/genetics , Glioma/metabolism , NF-kappa B/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Epigenomics , ErbB Receptors/metabolism , Exons , Female , Humans , Mice , Mice, Nude , Signal Transduction , Transfection
5.
Nanoscale ; 7(25): 11063-74, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26050964

ABSTRACT

Electronic bipolar resistance switching (eBRS) in an Al/TiOx/Al structure, where the TiOx layer was reactively sputter-deposited, was examined in conjunction with a structural analysis using transmission electron microscopy. A thin (3-5 nm) insulating Al(Ti)Ox layer was formed at the bottom Al electrode interface, which provided the necessary asymmetric potential barrier for the eBRS to emerge, whereas the top Al electrode interface appeared to have provided the fluent carrier (electron) injection. The set and reset switching were related to the trapping and detrapping of the carriers at the trap centers, the characteristic energy of which was ∼0.86 eV, across the entire electrode area. The general features of this material system as the feasible RS memory were insufficient: endurance cycle, <∼8000, and retention time at 85 °C, 10(6) s. However, the detailed analysis of the switching behavior based on the space-charge limited current conduction mechanism, and its variation with the switching cycles, provided useful information on the general features of the eBRS, which could also be applicable to other binary (or even ternary) metal-oxide RS systems based on the electronic switching mechanism.

6.
Oncotarget ; 6(21): 18631-40, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26087181

ABSTRACT

In order to determine the diagnostic and prognostic value of miR-26a in Cholangiocarcinoma (CCA), we compared miR-26a levels in serum from 66 CCA patients and 66 healthy controls, which was followed by serum analysis between the pre-operative serum and post-operative serum of these CCA patients. We found the concentration levels of miR-26a in serum of CCA patients were significantly higher than that from healthy controls (P < 0.01). Furthermore, the concentration levels of miR-26a in the post-operative serum were significantly reduced when compared to the pre-operative serum (P < 0.001). High miR-26a in serum was correlated significantly with clinical stage, distant metastasis, differentiation status, and poor survival of CCA patients. More importantly, serum miR-26a was an independent prognostic marker for CCA. In conclusion, our results suggested that miR-26a in serum might be a potential and useful noninvasive biomarker for the early detection of CCA.


Subject(s)
Bile Duct Neoplasms/genetics , Biomarkers, Tumor/genetics , Cholangiocarcinoma/genetics , MicroRNAs/genetics , Bile Duct Neoplasms/blood , Bile Duct Neoplasms/diagnosis , Biomarkers, Tumor/blood , Case-Control Studies , Cell Line, Tumor , Cholangiocarcinoma/blood , Cholangiocarcinoma/diagnosis , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/blood , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Neoplasm Staging , Postoperative Period , Preoperative Period , Prognosis , Reverse Transcriptase Polymerase Chain Reaction
7.
CNS Neurosci Ther ; 20(9): 830-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24810017

ABSTRACT

BACKGROUND AND AIMS: The nuclear localization of ß-catenin, a mediator of canonical Wnt signaling, has been indicated in a variety of cancers and is frequently related to tumor progression and metastasis. Therefore, targeting ß-catenin is an attractive therapeutic strategy for cancers. METHODS: Herein, we identified a natural, small molecule inhibitor of ß-catenin signaling, BASI, and evaluated its therapeutic efficacy both in vitro and in orthotopic mouse models of glioma. RESULTS: BASI significantly suppressed proliferation and invasion and induced apoptosis in glioblastoma cells and resulted in the remarkable attenuation of orthotopic tumor growth in vivo. Furthermore, we found that BASI altered the expression of several microRNAs, which mediated the posttranscriptional silencing of ß-catenin expression either directly or indirectly through a von Hippel-Lindau (VHL)-mediated ß-catenin degradation pattern. CONCLUSIONS: Taken together, our findings offer preclinical validation of BASI as a promising new type of ß-catenin inhibitor with a mechanism of inhibition that has broad potential for the improved treatment of glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , MicroRNAs/metabolism , Neuroblastoma/drug therapy , Signal Transduction/drug effects , Trypsin Inhibitor, Kunitz Soybean/pharmacology , beta Catenin/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/pathology , CREB-Binding Protein/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colony-Forming Units Assay , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice, Nude , MicroRNAs/genetics , Neuroblastoma/pathology , Protein Binding/drug effects , beta Catenin/genetics
8.
Mol Cancer ; 13: 63, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24650032

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is amplified in 40% of human glioblastomas. However, most glioblastoma patients respond poorly to anti-EGFR therapy. MicroRNAs can function as either oncogenes or tumor suppressor genes, and have been shown to play an important role in cancer cell proliferation, invasion and apoptosis. Whether microRNAs can impact the therapeutic effects of EGFR inhibitors in glioblastoma is unknown. METHODS: miR-566 expression levels were detected in glioma cell lines, using real-time quantitative RT-PCR (qRT-PCR). Luciferase reporter assays and Western blots were used to validate VHL as a direct target gene of miR-566. Cell proliferation, invasion, cell cycle distribution and apoptosis were also examined to confirm whether miR-566 inhibition could sensitize anti-EGFR therapy. RESULTS: In this study, we demonstrated that miR-566 is up-regulated in human glioma cell lines and inhibition of miR-566 decreased the activity of the EGFR pathway. Lentiviral mediated inhibition of miR-566 in glioblastoma cell lines significantly inhibited cell proliferation and invasion and led to cell cycle arrest in the G0/G1 phase. In addition, we identified von Hippel-Lindau (VHL) as a novel functional target of miR-566. VHL regulates the formation of the ß-catenin/hypoxia-inducible factors-1α complex under miR-566 regulation. CONCLUSIONS: miR-566 activated EGFR signaling and its inhibition sensitized glioblastoma cells to anti-EGFR therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Signal Transduction , Animals , Blotting, Western , Cell Line, Tumor , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Glioblastoma/metabolism , Heterografts , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Transfection , Von Hippel-Lindau Tumor Suppressor Protein/genetics
9.
CNS Neurosci Ther ; 20(5): 429-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24483339

ABSTRACT

AIMS: Down-regulation of AJAP1 in glioblastoma multiforme (GBM) has been reported. However, the expression profiles of AJAP1 in gliomas and the underlying mechanisms of AJAP1 function on invasion are still poorly understood. METHODS: The gene profiles of AJAP1 in glioma patients were studied among four independent cohorts. Confocal imaging was used to analyze the AJAP1 localization. After AJAP1 overexpression in GBM cell lines, cellular polarity, cytoskeleton distribution, and antitumor effect were investigated in vitro and in vivo. RESULTS: AJAP1 expression was significantly decreased in gliomas compared with normal brain in REMBRANDT and CGCA cohorts. Additionally, low AJAP1 expression was associated with worse survival in GBMs in REMBRANDT and TCGA U133A cohorts and was significantly associated with classical and mesenchymal subtypes of GBMs among four cohorts. Confocal imaging indicated AJAP1 localized in cell membranes in low-grade gliomas and AJAP1-overexpressing GBM cells, but difficult to assess in high-grade gliomas due to its absence. AJAP1 overexpression altered the cytoskeleton and cellular polarity in vitro and inhibited the tumor growth in vivo. CONCLUSIONS: AJAP1 is dysregulated at an early stage of gliomagenesis and may suppress glioma cell invasion and proliferation, which suggests that AJAP1 may be a potential diagnostic and prognostic marker for gliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Cell Adhesion Molecules/metabolism , Cytoskeleton/metabolism , Glioma/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cohort Studies , Cytoskeleton/ultrastructure , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , Kaplan-Meier Estimate , Mice, Nude , Microscopy, Confocal , Neoplasm Staging , Neoplasm Transplantation
10.
Cancer Lett ; 342(1): 139-49, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24012640

ABSTRACT

Epidermal growth factor receptors (EGFR) expression is frequently amplified in human glioblastoma cells. Nimotuzumab, a monoclonal antibody (mAb) against EGFR, has been used globally in clinics as an anti-cancer agent. It is largely unknown whether the blockade of miR-21, a microRNA that is upregulated in glioma cells, could amplify the effects of nimotuzumab. Herein, we have demonstrated that miR-21 directly targets von Hippel-Lindau (VHL) and peroxisome-proliferator-activated receptor α (PPARα) and that miR-21 regulates EGFR/AKT signaling through VHL/ß-catenin and the PPARα/AP-1 axis. Further, the expression of miR-21 is regulated by EGFR via the activation of ß-catenin and AP-1. These data indicate that a feedback loop exists between miR-21 and EGFR. We also show that the combination of nimotuzumab and an inhibitor of miR-21 is superior to single-agent therapy. These results clarify a novel association between miR-21 and EGFR in the regulation of cancer cell progression.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Base Sequence , Binding Sites , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , PPAR alpha/genetics , PPAR alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Transcription Factor AP-1/metabolism , Tumor Burden , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
11.
CNS Neurosci Ther ; 19(2): 98-108, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230963

ABSTRACT

BACKGROUND AND AIMS: Currently temozolomide (TMZ) as a potent agent is widely used to treat the glioblastoma multiforme (GBM), whereas recurrence due to intrinsic or acquired therapeutic resistance often occurs. Combination chemotherapy with TMZ may be a promising therapeutic strategy to improve treatment efficacy. METHODS: Aspirin, TMZ, and aspirin-/TMZ-coloaded poly (L-lactide-co-glycolide) (PLGA) microspheres were prepared by spray drying, and cytotoxicities of glioblastoma cells were measured. RESULTS: Aspirin microsphere treatment induced slight apoptosis and modestly inhibited proliferation of LN229 and U87 cells in vitro and in vivo through inhibition of ß-catenin transactivation. However, aspirin-/TMZ-coloaded microspheres presented synergistic antitumor efficacy compared with single TMZ-loaded microspheres. Aspirin/TMZ microspheres induced more apoptosis and repressed proliferation of LN229 and U87 cells. Corresponding to inhibition of ß-catenin signaling, ß-catenin/TCF4 transcriptional activity and STAT3 luciferase activity were strongly suppressed, and downstream targets expression was decreased. Furthermore, aspirin/TMZ microsphere intratumoral injection downregulated the expression of ß-catenin, TCF4, pAKT, pSTAT3, and PCNA and delayed tumor growth in nude mice harboring subcutaneous LN229 xenografts. CONCLUSIONS: Aspirin sensitized TMZ chemotherapy efficacy through inhibition of ß-catenin transactivation; furthermore, the coloaded microspheres achieved a sustained release action to reduce the TMZ dosage, offering the potential for improved treatment of glioblastomas.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Aspirin/administration & dosage , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Transcriptional Activation/drug effects , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Animals , Cell Line, Tumor , Dacarbazine/administration & dosage , Drug Carriers/administration & dosage , Drug Synergism , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microspheres , Random Allocation , Temozolomide , Transcriptional Activation/genetics , Treatment Outcome , Xenograft Model Antitumor Assays/methods , beta Catenin/genetics
12.
CNS Neurosci Ther ; 18(7): 573-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22630347

ABSTRACT

AIMS: MicroRNA-21 (miR-21) expression is increased in many types of human malignancy, including glioma. Recent studies report that miR-21 regulates cell invasion by targeting RECK, however, the underlying transcriptional regulation of miR-21 in glioma cells remains elusive. RESULTS: Here, we identify a positive correlation between miR-21 expression and pathological grade in glioma tissues. We demonstrate that ß-catenin pathway regulates miR-21 expression in human umbilical vein endothelial cell and glioma cells, and that this regulation is signal transducer and activator of transcription 3 (STAT3)-dependent. Further, chromatin immunoprecipitation and luciferase reporter analysis demonstrate that miR-21 is controlled by an upstream promoter containing a conserved STAT3 binding site. Notably, knockdown of miR-21-inhibited cell invasion by increasing RECK expression and decreased tumor growth in a xenograft model. CONCLUSION: These data provide compelling evidence that ß-catenin regulation of miR-21 via STAT3 plays a role in glioma cell invasion and proliferation and indicate that STAT3 is a potential therapeutic target for glioma intervention.


Subject(s)
GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/physiology , beta Catenin/physiology , Animals , Cell Line, Tumor , Down-Regulation/genetics , GPI-Linked Proteins/genetics , Gene Knockdown Techniques/methods , Gene Targeting/methods , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/biosynthesis , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Umbilical Veins/cytology , Umbilical Veins/metabolism , Up-Regulation/genetics , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL