35 results on '"Park JW"'
Search Results
2. Abstract P3-01-02: Detection of circulating tumor cells (CTC) in blood and disseminated tumor cells (DTC) in bone marrow at surgery identifies breast cancer patients (pts) with long-term risk of distant recurrence and breast cancer-specific death
- Author
-
Magbanua, MJM, primary, Yau, C, additional, Wolf, D, additional, Lee, JS, additional, Chattopadhyay, A, additional, Scott, JH, additional, Yoder, E, additional, Hwang, S, additional, Alvarado, M, additional, Ewing, CA, additional, Delson, AL, additional, van't Veer, L, additional, Esserman, L, additional, and Park, JW, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P2-01-01: Trajectory patterns of circulating tumor cells (CTC) in chemotherapy-treated metastatic breast cancer (MBC) patients predict poor clinical outcomes: CALGB 40502 (Alliance)/NCCTG N063H study
- Author
-
Magbanua, MJ, primary, Hendrix, L, additional, Hyslop, T, additional, Barry, WT, additional, Winer, EP, additional, Hudis, C, additional, Toppmeyer, D, additional, Burnstein, H, additional, Qadir, M, additional, Ma, C, additional, Scott, JH, additional, Park, JW, additional, and Rugo, HS, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract S3-01: IMENEO: International MEta-analysis of circulating tumor cell detection in early breast cancer patients treated by NEOadjuvant chemotherapy
- Author
-
Bidard, F-C, primary, Michiels, S, additional, Mueller, V, additional, Riethdorf, S, additional, Esserman, LJ, additional, Lucci, A, additional, Naume, B, additional, Horiguchi, J, additional, Gisbert-Criado, R, additional, Sleijfer, S, additional, Toi, M, additional, Garcia-Saenz, JA, additional, Hartkopf, A, additional, Generali, D, additional, Rothe, F, additional, Smerage, J, additional, Muinelo, L, additional, Stebbing, J, additional, Viens, P, additional, Magbanua, M, additional, Hall, CS, additional, Engebråtenm, O, additional, Takata, D, additional, Vidal-Martínez, J, additional, Onstenk, W, additional, Fujisawa, N, additional, Diaz-Rubio, E, additional, Taran, F-A, additional, Cappelletti, MR, additional, Ignatiadis, M, additional, Name, N, additional, Proudhon, C, additional, Wolf, D, additional, Bowman Bauldry, J, additional, Borgen, E, additional, Nagaoka, R, additional, Carañana, V, additional, Kraan, J, additional, Maestro, M, additional, Brucker, SY, additional, Weber, K, additional, Reyal, F, additional, Amara, D, additional, Gopalkrishna Karhade, M, additional, Ruud Mathiesen, R, additional, Tokiniwa, H, additional, Llombart-Cussac, A, additional, D'Hollander, K, additional, Cottu, P, additional, Park, JW, additional, Loibl, S, additional, Pierga, J-Y, additional, and Pantel, K, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract PD6-8: Prognostic value of disseminated tumor cells according to ER/PR and HER2 status in non-metastatic breast cancer
- Author
-
Lee, JS, primary, Magbanua, MJM, additional, Moore, AP, additional, Esserman, LJ, additional, and Park, JW, additional
- Published
- 2013
- Full Text
- View/download PDF
6. Abstract P3-11-01: Prospective study of cognitive function in women with early stage breast cancer: Predictors of cognitive decline and relationship to cognitive complaints
- Author
-
Heflin, LH, primary, Fang, S, additional, DeLuca, A, additional, Melisko, MM, additional, Moasser, M, additional, Park, JW, additional, Chien, AJ, additional, Munster, P, additional, Landau, SM, additional, Kramer, JH, additional, Jagust, WJ, additional, and Rugo, HS, additional
- Published
- 2013
- Full Text
- View/download PDF
7. Abstract P2-01-05: Parallel DNA and RNA profiling of EpCAM-positive cells in blood of metastatic breast cancer (MBC) patients confirm their malignant nature
- Author
-
Magbanua, MJM, primary, Hauranieh, L, additional, Sosa, EV, additional, Pendyala, P, additional, Scott, JH, additional, Rugo, HS, additional, and Park, JW, additional
- Published
- 2012
- Full Text
- View/download PDF
8. Abstract P4-16-07: Selective use of post-mastectomy radiation therapy in the neoadjuvant setting
- Author
-
Warren, Peled A, primary, Wang, F, additional, Stover, AC, additional, Rugo, HS, additional, Melisko, ME, additional, Park, JW, additional, Alvarado, M, additional, Ewing, CA, additional, Esserman, LJ, additional, Fowble, B, additional, and Hwang, ES, additional
- Published
- 2012
- Full Text
- View/download PDF
9. Abstract P6-01-05: Enhancement of18F-FDG uptake and glycolysis by epidermal growth factor via PI3K activation in T47D breast cancer cells
- Author
-
Lee, EJ, primary, Park, JW, additional, Cung, Q, additional, Jung, K-H, additional, Lee, JH, additional, Paik, J-Y, additional, and Lee, K-H, additional
- Published
- 2012
- Full Text
- View/download PDF
10. Abstract PD04-10: Genome-Wide Copy Number Analysis of Circulating Tumor Cells (CTCs) from Metastatic Breast Cancer Patients
- Author
-
Magbanua, MJM, primary, Sosa, EV, additional, Scott, JH, additional, Pinkel, D, additional, Rugo, HS, additional, and Park, JW., additional
- Published
- 2010
- Full Text
- View/download PDF
11. Simultaneous quantitative assessment of tumor cells in blood and bone marrow in primary breast cancer patients.
- Author
-
Moore, AP, primary, Magbanua, MM, additional, Scott, J, additional, Moore, D, additional, Alvarado, MD, additional, Esserman, LJ, additional, and Park, JW, additional
- Published
- 2009
- Full Text
- View/download PDF
12. Phase I study of sunitinib in combination with metronomic dosing of cyclophosphamide and methotrexate in patients with metastatic breast cancer.
- Author
-
Lyandres, J, primary, Melisko, M, additional, Moasser, M, additional, Goga, A, additional, Park, JW, additional, and Rugo, HS, additional
- Published
- 2009
- Full Text
- View/download PDF
13. Tumor-Associated Macrophages Enhance Tumor Hypoxia and Aerobic Glycolysis.
- Author
-
Jeong H, Kim S, Hong BJ, Lee CJ, Kim YE, Bok S, Oh JM, Gwak SH, Yoo MY, Lee MS, Chung SJ, Defrêne J, Tessier P, Pelletier M, Jeon H, Roh TY, Kim B, Kim KH, Ju JH, Kim S, Lee YJ, Kim DW, Kim IH, Kim HJ, Park JW, Lee YS, Lee JS, Cheon GJ, Weissman IL, Chung DH, Jeon YK, and Ahn GO
- Subjects
- Animals, B7-H1 Antigen immunology, Carcinoma, Non-Small-Cell Lung etiology, Carcinoma, Non-Small-Cell Lung metabolism, Female, Humans, Lung Neoplasms etiology, Lung Neoplasms metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Prognosis, T-Lymphocytes immunology, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha metabolism, B7-H1 Antigen metabolism, Carcinoma, Non-Small-Cell Lung pathology, Glycolysis, Lung Neoplasms pathology, Macrophages immunology, Tumor Hypoxia immunology
- Abstract
Tumor hypoxia and aerobic glycolysis are well-known resistance factors for anticancer therapies. Here, we demonstrate that tumor-associated macrophages (TAM) enhance tumor hypoxia and aerobic glycolysis in mice subcutaneous tumors and in patients with non-small cell lung cancer (NSCLC). We found a strong correlation between CD68 TAM immunostaining and PET
18 fluoro-deoxyglucose (FDG) uptake in 98 matched tumors of patients with NSCLC. We also observed a significant correlation between CD68 and glycolytic gene signatures in 513 patients with NSCLC from The Cancer Genome Atlas database. TAM secreted TNFα to promote tumor cell glycolysis, whereas increased AMP-activated protein kinase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha in TAM facilitated tumor hypoxia. Depletion of TAM by clodronate was sufficient to abrogate aerobic glycolysis and tumor hypoxia, thereby improving tumor response to anticancer therapies. TAM depletion led to a significant increase in programmed death-ligand 1 (PD-L1) expression in aerobic cancer cells as well as T-cell infiltration in tumors, resulting in antitumor efficacy by PD-L1 antibodies, which were otherwise completely ineffective. These data suggest that TAM can significantly alter tumor metabolism, further complicating tumor response to anticancer therapies, including immunotherapy. SIGNIFICANCE: These findings show that tumor-associated macrophages can significantly modulate tumor metabolism, hindering the efficacy of anticancer therapies, including anti-PD-L1 immunotherapy., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
14. FIH Is an Oxygen Sensor in Ovarian Cancer for G9a/GLP-Driven Epigenetic Regulation of Metastasis-Related Genes.
- Author
-
Kang J, Shin SH, Yoon H, Huh J, Shin HW, Chun YS, and Park JW
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Epigenesis, Genetic, Female, Follow-Up Studies, Gene Expression Regulation, Neoplastic, Histocompatibility Antigens genetics, Histone-Lysine N-Methyltransferase genetics, Humans, Hydroxylation, Hypoxia, Mice, Mixed Function Oxygenases genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Peritoneal Neoplasms genetics, Peritoneal Neoplasms metabolism, Prognosis, Repressor Proteins genetics, Transcription, Genetic, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers, Tumor metabolism, Histocompatibility Antigens metabolism, Histone-Lysine N-Methyltransferase metabolism, Mixed Function Oxygenases metabolism, Ovarian Neoplasms pathology, Oxygen metabolism, Peritoneal Neoplasms secondary, Repressor Proteins metabolism
- Abstract
The prolyl hydroxylase domain-containing proteins (PHD1-3) and the asparaginyl hydroxlyase factor inhibiting HIF (FIH) are oxygen sensors for hypoxia-inducible factor-driven transcription of hypoxia-induced genes, but whether these sensors affect oxygen-dependent epigenetic regulation more broadly is not known. Here, we show that FIH exerts an additional role as an oxygen sensor in epigenetic control by the histone lysine methyltransferases G9a and GLP. FIH hydroxylated and inhibited G9a and GLP under normoxia. When the FIH reaction was limited under hypoxia, G9a and GLP were activated and repressed metastasis suppressor genes, thereby triggering cancer cell migration and peritoneal dissemination of ovarian cancer xenografts. In clinical specimens of ovarian cancer, expression of FIH and G9a were reciprocally associated with patient outcomes. We also identified mutations of FIH target motifs in G9a and GLP, which exhibited excessive H3K9 methylation and facilitated cell invasion. This study provides insight into a new function of FIH as an upstream regulator of oxygen-dependent chromatin remodeling. It also implies that the FIH-G9a/GLP pathway could be a potential target for inhibiting hypoxia-induced cancer metastasis. Significance: These findings deepen understanding of oxygen-dependent gene regulation and cancer metastasis in response to hypoxia. Cancer Res; 78(5); 1184-99. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
15. Quantitative Whole Genome Sequencing of Circulating Tumor Cells Enables Personalized Combination Therapy of Metastatic Cancer.
- Author
-
Gulbahce N, Magbanua MJM, Chin R, Agarwal MR, Luo X, Liu J, Hayden DM, Mao Q, Ciotlos S, Li Z, Chen Y, Chen X, Li Y, Zhang RY, Lee K, Tearle R, Park E, Drmanac S, Rugo HS, Park JW, Drmanac R, and Peters BA
- Subjects
- Female, Humans, Middle Aged, Neoplasm Metastasis, Neoplastic Cells, Circulating pathology, Genomics methods, Neoplasms drug therapy, Neoplastic Cells, Circulating metabolism
- Abstract
Much effort has been dedicated to developing circulating tumor cells (CTC) as a noninvasive cancer biopsy, but with limited success as yet. In this study, we combine a method for isolation of highly pure CTCs using immunomagnetic enrichment/fluorescence-activated cell sorting with advanced whole genome sequencing (WGS), based on long fragment read technology, to illustrate the utility of an accurate, comprehensive, phased, and quantitative genomic analysis platform for CTCs. Whole genomes of 34 CTCs from a patient with metastatic breast cancer were analyzed as 3,072 barcoded subgenomic compartments of long DNA. WGS resulted in a read coverage of 23× per cell and an ensemble call rate of >95%. These barcoded reads enabled accurate detection of somatic mutations present in as few as 12% of CTCs. We found in CTCs a total of 2,766 somatic single-nucleotide variants and 543 indels and multi-base substitutions, 23 of which altered amino acid sequences. Another 16,961 somatic single nucleotide variant and 8,408 indels and multi-base substitutions, 77 of which were nonsynonymous, were detected with varying degrees of prevalence across the 34 CTCs. On the basis of our whole genome data of mutations found in all CTCs, we identified driver mutations and the tissue of origin of these cells, suggesting personalized combination therapies beyond the scope of most gene panels. Taken together, our results show how advanced WGS of CTCs can lead to high-resolution analyses of cancers that can reliably guide personalized therapy. Cancer Res; 77(16); 4530-41. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
16. SIRT1 and AMPK mediate hypoxia-induced resistance of non-small cell lung cancers to cisplatin and doxorubicin.
- Author
-
Shin DH, Choi YJ, and Park JW
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, Apoptosis drug effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Drug Resistance, Neoplasm, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Mice, Nude, Signal Transduction, Sirtuin 1 genetics, Xenograft Model Antitumor Assays, AMP-Activated Protein Kinases metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Cell Hypoxia physiology, Cisplatin pharmacology, Doxorubicin pharmacology, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Sirtuin 1 metabolism
- Abstract
SIRT1 is an NAD(+)-dependent protein deacetylase induced by metabolic stresses, such as nutrition or oxygen deprivation. Although SIRT1 contributes to aging and metabolic disorders, its role in cancer progression and therapeutic responses remains controversial. Because hypoxia occurs widely in solid tumors, where it provokes drug resistance, we investigated the involvement of SIRT1 in hypoxia-induced chemoresistance. SIRT1 was downregulated in a panel of non-small cell lung carcinoma (NSCLC) cells exposed to hypoxia for 48 hours. The master metabolic kinase AMP-activated protein kinase (AMPK) was inactivated under the same conditions, likely due to attenuation of the SIRT1/LKB1-mediated AMPK activation process. Notably, hypoxic inactivation of this SIRT1-AMPK pathway led to cisplatin and doxorubicin resistance. Mechanistic investigations suggested that this pathway supported the cytotoxic response to cisplatin and doxorubicin by licensing an apoptotic process controlled by mitochondria. We confirmed the involvement of this pathway in a mouse xenograft model of human NSCLC. Furthermore, we demonstrated that a SIRT1 activator SRT1720 augmented the antitumor effects of cisplatin, and these effects could be blocked by administration of an AMPK inhibitor compound C. Taken together, our results offer preclinical proof-of-concept to target the SIRT1-AMPK pathway as a strategy to overcome hypoxia-induced chemoresistance in NSCLC.
- Published
- 2014
- Full Text
- View/download PDF
17. Molecular profiling of tumor cells in cerebrospinal fluid and matched primary tumors from metastatic breast cancer patients with leptomeningeal carcinomatosis.
- Author
-
Magbanua MJ, Melisko M, Roy R, Sosa EV, Hauranieh L, Kablanian A, Eisenbud LE, Ryazantsev A, Au A, Scott JH, and Park JW
- Subjects
- Biomarkers, Tumor genetics, Breast Neoplasms pathology, Comparative Genomic Hybridization, Female, Flow Cytometry, Humans, Meningeal Carcinomatosis secondary, Neoplastic Cells, Circulating pathology, Breast Neoplasms cerebrospinal fluid, Breast Neoplasms genetics, Cerebrospinal Fluid metabolism, Gene Expression Profiling, Meningeal Carcinomatosis cerebrospinal fluid, Meningeal Carcinomatosis genetics, Neoplastic Cells, Circulating metabolism
- Abstract
Although leptomeningeal carcinomatosis is a well-established clinical syndrome, virtually nothing is known about the tumor cells responsible for this particularly aggressive metastatic process. To isolate cerebrospinal fluid-derived tumor cells (CSFTC) from 15 patients with metastatic breast cancer diagnosed with leptomeningeal carcinomatosis, CSF samples were subjected to a two-step method involving immunomagnetic enrichment and fluorescence-activated cell sorting (IE/FACS), a technique previously used for isolating circulating tumor cells (CTC) from blood. CSFTCs were subjected to genome-wide copy number analysis by array comparative genomic hybridization. Genomic profiling was successfully performed for 13 of 15 patients (87%). Copy number analysis in CSFTCs revealed genomic alterations commonly observed in primary breast cancer and CTCs, indicating their malignant origin. Interestingly, 12 (92%) harbored high-level gains on the 8q24 locus, which includes the MYC oncogene. Comparison of CSFTCs against corresponding archival primary tumors in six patients revealed clonal relationships with some divergence. Good concordance among serial samples attested to the reproducibility of the assay. Our approach for isolation and molecular analysis of CSFTCs yielded new insights into the molecular nature of these cells. Further genomic and functional analyses may help elucidate mechanisms by which tumor cells metastasize to the central nervous system.
- Published
- 2013
- Full Text
- View/download PDF
18. Genomic profiling of isolated circulating tumor cells from metastatic breast cancer patients.
- Author
-
Magbanua MJ, Sosa EV, Roy R, Eisenbud LE, Scott JH, Olshen A, Pinkel D, Rugo HS, and Park JW
- Subjects
- Breast Neoplasms blood, Comparative Genomic Hybridization, Female, Gene Dosage, Humans, Neoplasm Metastasis genetics, Neoplasm Metastasis pathology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Separation methods, Flow Cytometry methods, Gene Expression Profiling methods, Neoplastic Cells, Circulating metabolism
- Abstract
Molecular characterization of circulating tumor cells (CTC) from blood is technically challenging because cells are rare and difficult to isolate. We developed a novel approach to isolate CTCs from blood via immunomagnetic enrichment followed by fluorescence-activated cell sorting (IE-FACS). Isolated CTCs were subjected to genome-wide copy number analysis via array comparative genomic hybridization (aCGH). In clinical studies, CTCs were isolated from 181 patients with metastatic breast cancer, 102 of which were successfully profiled, including matched archival primary tumor from five patients. CTCs revealed a wide range of copy number alterations including those previously reported in breast cancer. Comparison with two published aCGH datasets of primary breast tumors revealed similar frequencies of recurrent genomic copy number aberrations. In addition, serial testing of CTCs confirmed reproducibility and indicated genomic change over time. Comparison of CTCs with matched archival primary tumors confirmed shared lineage as well as some divergence. We showed that it is feasible to isolate CTCs away from hematopoietic cells with high purity through IE-FACS and profile them via aCGH analysis. Our approach may be used to explore genomic events involved in cancer progression and to monitor therapeutic efficacy of targeted therapies in clinical trials in a relatively noninvasive manner.
- Published
- 2013
- Full Text
- View/download PDF
19. HIF-2alpha enhances beta-catenin/TCF-driven transcription by interacting with beta-catenin.
- Author
-
Choi H, Chun YS, Kim TY, and Park JW
- Subjects
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Cell Growth Processes physiology, Cell Hypoxia physiology, Cell Line, Tumor, E1A-Associated p300 Protein genetics, E1A-Associated p300 Protein metabolism, HEK293 Cells, Humans, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Protein Structure, Tertiary, Signal Transduction, Transcription Factor 4, Transcription Factors genetics, Transcription, Genetic, Transcriptional Activation, beta Catenin genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Carcinoma, Renal Cell metabolism, Kidney Neoplasms metabolism, Transcription Factors metabolism, beta Catenin metabolism
- Abstract
The tumor-promoting factors β-catenin and hypoxia-inducible factor (HIF) are often found to be coactivated in rapidly growing tumors. Recently, it was shown that HIF-1α negatively regulates Wnt/β-catenin signaling by sequestering β-catenin from β-catenin/T-cell factor (TCF). However, no investigation has been undertaken on the involvement of HIF-2α in β-catenin regulation. In this study, it was found that, like HIF-1α, HIF-2α interacts with β-catenin, but at a different site. Furthermore, HIF-2α was found to assemble with β-catenin/TCF and facilitate gene transcription. Mutational analyses revealed that transactivation domains of HIF-2α promote p300 coactivator recruitment by β-catenin. Furthermore, HIF-2α and β-catenin were found to associate in the nuclei of 786-0 renal cell carcinoma cells, and HIF-2α was found to be required for β-catenin activation in these cells and for their proliferation. These results suggest that this interaction contributes to the unrestrained growth of tumor cells containing coactivated HIF-2α and β-catenin. Interestingly, these actions of HIF-2α oppose those of HIF-1α on β-catenin and cell growth, and this suggests that HIF-1α/HIF-2α balance may importantly determine cell growth when hypoxia and Wnt stimulation coexist., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
20. Deficiencies in the Fanconi anemia DNA damage response pathway increase sensitivity to HPV-associated head and neck cancer.
- Author
-
Park JW, Pitot HC, Strati K, Spardy N, Duensing S, Grompe M, and Lambert PF
- Subjects
- 4-Nitroquinoline-1-oxide, Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, DNA Damage, Fanconi Anemia genetics, Fanconi Anemia metabolism, Fanconi Anemia Complementation Group D2 Protein metabolism, Female, Fluorescent Antibody Technique, Head and Neck Neoplasms chemically induced, Head and Neck Neoplasms metabolism, Humans, Male, Mice, Mice, 129 Strain, Mice, Knockout, Mice, Transgenic, Papillomavirus E7 Proteins metabolism, Quinolones, Signal Transduction, Fanconi Anemia Complementation Group D2 Protein genetics, Genetic Predisposition to Disease genetics, Head and Neck Neoplasms genetics, Papillomavirus E7 Proteins genetics
- Abstract
Patients with the rare genetic disease, Fanconi anemia (FA), are highly susceptible to squamous cell carcinomas arising at multiple anatomic sites including the head and neck region. Human papillomaviruses (HPVs), particularly HPV16, are associated with ∼20% of head and neck squamous cell carcinomas (HNSCCs) in the general population. Some but not other investigators have reported that HNSCCs in FA patients are much more frequently positive for HPV. In addition, studies have demonstrated an interaction between the HPV16 E7 oncoprotein and the FA pathway, a DNA damage response pathway deficient in FA patients. On the basis of these studies, it was hypothesized that the FA pathway contributes to repair of DNA damage induced by HPV16 E7, providing one explanation for why FA patients are predisposed to HPV-associated HNSCCs. To determine the importance of the FA pathway in modulating the oncogenic abilities of E7, we crossed K14E7 transgenic (K14E7) and fancD2 knockout mice (FancD2(-/-)) to establish K14E7/FancD2(-/-) and K14E7/FancD2(+/+) mice and monitored their susceptibility to HNSCC when treated with a chemical carcinogen. K14E7/FancD2(-/-) mice had a significantly higher incidence of HNSCC compared with K14E7/FancD2(+/+) mice. This difference correlated with an increased proliferative index and the increase in expression of biomarkers that are used to assess levels of DNA damage. These animal studies support the hypotheses that FA patients have increased susceptibility to HPV-associated cancer and that the FA DNA damage response pathway normally attenuates the oncogenic potential of HPV16 E7.
- Published
- 2010
- Full Text
- View/download PDF
21. Depletion of embryonic stem cell signature by histone deacetylase inhibitor in NCCIT cells: involvement of Nanog suppression.
- Author
-
You JS, Kang JK, Seo DW, Park JH, Park JW, Lee JC, Jeon YJ, Cho EJ, and Han JW
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Binding Sites genetics, Blotting, Western, Cell Differentiation drug effects, Cell Differentiation genetics, Cell Line, Tumor, Cell Proliferation drug effects, Chromatin Immunoprecipitation, DNA Methylation drug effects, Embryonal Carcinoma Stem Cells pathology, Histone Deacetylase Inhibitors, Humans, Nanog Homeobox Protein, Promoter Regions, Genetic genetics, Protein Binding, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Transcription Factors metabolism, Embryonal Carcinoma Stem Cells metabolism, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Homeodomain Proteins genetics, Peptides, Cyclic pharmacology
- Abstract
The embryonic stem cell-like gene expression signature has been shown to be associated with poorly differentiated aggressive human tumors and has attracted great attention as a potential target for future cancer therapies. Here, we investigate the potential of the embryonic stem cell signature as molecular target for the therapy and the strategy to suppress the embryonic stem cell signature. The core stemness gene Nanog is abnormally overexpressed in human embryonic carcinoma NCCIT cells showing gene expression profiles similar to embryonic stem cells. Down-regulation of the gene by either small interfering RNAs targeting Nanog or histone deacetylase inhibitor apicidin causes reversion of expression pattern of embryonic stem cell signature including Oct4, Sox2, and their target genes, leading to cell cycle arrest, inhibition of colony formation in soft agar, and induction of differentiation into all three germ layers. These effects are antagonized by reintroduction of Nanog. Interestingly, embryonic carcinoma cells (NCCIT, NTERA2, and P19) exhibit a higher sensitivity to apicidin in down-regulation of Nanog compared with embryonic stem cells. Furthermore, the down-regulation of Nanog expression by apicidin is mediated by a coordinated change in recruitment of epigenetic modulators and transcription factors to the promoter region. These findings indicate that overexpression of stemness gene Nanog in NCCIT cells is associated with maintaining stem cell-like phenotype and suggest that targeting Nanog might be an approach for improved therapy of poorly differentiated tumors.
- Published
- 2009
- Full Text
- View/download PDF
22. Hypoxia-inducible factor-1alpha obstructs a Wnt signaling pathway by inhibiting the hARD1-mediated activation of beta-catenin.
- Author
-
Lim JH, Chun YS, and Park JW
- Subjects
- Acetylation, Acetyltransferases metabolism, Cell Hypoxia physiology, Cell Proliferation, Cells, Cultured, Gene Expression Regulation, HCT116 Cells, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Models, Biological, N-Terminal Acetyltransferase A, N-Terminal Acetyltransferase E, Protein Binding, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Signal Transduction, TCF Transcription Factors antagonists & inhibitors, TCF Transcription Factors metabolism, Transcription Factor 7-Like 2 Protein, Acetyltransferases physiology, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Wnt Proteins antagonists & inhibitors, beta Catenin metabolism
- Abstract
Although a splice variant of mouse mARD1s was found to acetylate and destabilize hypoxia-inducible factor-1alpha (HIF-1alpha), human hARD1 has no such activities. Nonetheless, hARD1 has been reported to bind directly with HIF-1alpha. Here, we addressed the functional significance of the hARD1-HIF-1alpha interaction. Because hARD1 acetylates and activates beta-catenin, we examined whether HIF-1alpha regulates the hARD1-mediated activation of Wnt signaling. It was found that HIF-1alpha binds hARD1 through the oxygen-dependent degradation domain and, in so doing, dissociates hARD1 from beta-catenin, which prevents beta-catenin acetylation. In LiCl-stimulated HEK293 or cancer cell lines with active Wnt signaling, beta-catenin acetylation and activity were suppressed in hypoxia, and these suppressions were mediated by HIF-1alpha. Moreover, HIF-1alpha disruption of hARD1/beta-catenin repressed TCF4 activity, resulting in c-Myc suppression and p21(cip1) induction. In addition, we confirmed that the HIF-1alpha NH(2) terminal inactivates TCF4 by directly binding beta-catenin. In conclusion, HIF-1alpha was found to inactivate the Wnt signaling by binding to hARD1 or beta-catenin, which may contribute to the hypoxia-induced growth arrest of tumor cells.
- Published
- 2008
- Full Text
- View/download PDF
23. Human arrest defective 1 acetylates and activates beta-catenin, promoting lung cancer cell proliferation.
- Author
-
Lim JH, Park JW, and Chun YS
- Subjects
- Acetylation, Acetyltransferases genetics, Carcinoma, Non-Small-Cell Lung enzymology, Carcinoma, Non-Small-Cell Lung pathology, Cell Growth Processes physiology, Cell Line, Tumor, Cyclin D1 genetics, Cyclin D1 metabolism, G1 Phase physiology, Humans, Lung Neoplasms enzymology, Lung Neoplasms pathology, N-Terminal Acetyltransferase A, N-Terminal Acetyltransferase E, Promoter Regions, Genetic, Protein Binding, RNA, Small Interfering genetics, TCF Transcription Factors metabolism, Transcription Factor 7-Like 2 Protein, beta Catenin antagonists & inhibitors, beta Catenin genetics, Acetyltransferases metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Lung Neoplasms metabolism, beta Catenin metabolism
- Abstract
Arrest defective 1 (ARD1), an acetyltransferase, is essential for the yeast life cycle. Although its human homologue (hARD1) has been identified, its biological functions in human cells remain unclear. In the present study, we examined the biological function of hARD1. In H1299 and A549 lung cancer cells, hARD1-silencing RNA inhibited cell proliferation and induced G(1) arrest. Cyclin D1 was also found to be down-regulated in these growth-arrested cells, and the ectopic expression of cyclin D1 rescued cell growth. hARD1 knockdown repressed the promoter activity of the cyclin D1 gene, which inhibited the transcription of cyclin D1. Moreover, hARD1 knockdown reduced the binding of beta-catenin/TCF4 transcription factor to cyclin D1 promoter and repressed its transcriptional activity. Inversely, hARD1 expression increased the transcriptional activity of beta-catenin. Both endogenous and ectopically expressed hARD1 was coimmunoprecipitated with beta-catenin. hARD1 knockdown did not affect beta-catenin expression or degradation but noticeably reduced acetylated beta-catenin. The beta-catenin binding and acetylation by hARD1 were observed in vitro. Therefore, it is suggested that hARD1 participates in proliferation of lung cancer cells via the activation of beta-catenin.
- Published
- 2006
- Full Text
- View/download PDF
24. Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models.
- Author
-
Kirpotin DB, Drummond DC, Shao Y, Shalaby MR, Hong K, Nielsen UB, Marks JD, Benz CC, and Park JW
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacokinetics, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Female, Humans, Liposomes chemistry, Mice, Receptor, ErbB-2 biosynthesis, Transplantation, Heterologous, Breast Neoplasms immunology, Liposomes immunology, Liposomes pharmacokinetics, Nanostructures, Receptor, ErbB-2 immunology
- Abstract
We describe evidence for a novel mechanism of monoclonal antibody (MAb)-directed nanoparticle (immunoliposome) targeting to solid tumors in vivo. Long-circulating immunoliposomes targeted to HER2 (ErbB2, Neu) were prepared by the conjugation of anti-HER2 MAb fragments (Fab' or single chain Fv) to liposome-grafted polyethylene glycol chains. MAb fragment conjugation did not affect the biodistribution or long-circulating properties of i.v.-administered liposomes. However, antibody-directed targeting also did not increase the tumor localization of immunoliposomes, as both targeted and nontargeted liposomes achieved similarly high levels (7-8% injected dose/g tumor tissue) of tumor tissue accumulation in HER2-overexpressing breast cancer xenografts (BT-474). Studies using colloidal gold-labeled liposomes showed the accumulation of anti-HER2 immunoliposomes within cancer cells, whereas matched nontargeted liposomes were located predominantly in extracellular stroma or within macrophages. A similar pattern of stromal accumulation without cancer cell internalization was observed for anti-HER2 immunoliposomes in non-HER2-overexpressing breast cancer xenografts (MCF-7). Flow cytometry of disaggregated tumors posttreatment with either liposomes or immunoliposomes showed up to 6-fold greater intracellular uptake in cancer cells due to targeting. Thus, in contrast to nontargeted liposomes, anti-HER2 immunoliposomes achieved intracellular drug delivery via MAb-mediated endocytosis, and this, rather than increased uptake in tumor tissue, was correlated with superior antitumor activity. Immunoliposomes capable of selective internalization in cancer cells in vivo may provide new opportunities for drug delivery.
- Published
- 2006
- Full Text
- View/download PDF
25. YC-1 induces S cell cycle arrest and apoptosis by activating checkpoint kinases.
- Author
-
Yeo EJ, Ryu JH, Chun YS, Cho YS, Jang IJ, Cho H, Kim J, Kim MS, and Park JW
- Subjects
- Apoptosis physiology, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular enzymology, Carcinoma, Hepatocellular pathology, Cell Growth Processes drug effects, Cell Line, Tumor, Checkpoint Kinase 1, Checkpoint Kinase 2, Enzyme Activation drug effects, Humans, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors, Indazoles pharmacokinetics, Liver Neoplasms drug therapy, Liver Neoplasms enzymology, Liver Neoplasms pathology, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors pharmacology, S Phase physiology, Apoptosis drug effects, Indazoles pharmacology, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, S Phase drug effects
- Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha) seems central to tumor growth and progression because it up-regulates genes essential for angiogenesis and the hypoxic adaptation of cancer cells, which is why HIF-1alpha inhibition is viewed as a cancer therapy strategy. Paradoxically, HIF-1alpha also leads to cell cycle arrest or the apoptosis of cancer cells. Thus, the possibility cannot be ruled out that HIF-1alpha inhibitors unlock cell cycle arrest under hypoxic conditions and prevent cell death, which would limit the anticancer effect of HIF-1alpha inhibitors. Previously, we reported on the development of YC-1 as an anticancer agent that inhibits HIF-1alpha. In the present study, we evaluated the effects of YC-1 on hypoxia-induced cell cycle arrest and cell death. It was found that YC-1 does not reverse the antiproliferative effect of hypoxia, but rather that it induces S-phase arrest and apoptosis at therapeutic concentrations that inhibit HIF-1alpha and tumor growth; however, YC-1 did not stimulate cyclic guanosine 3',5'-monophosphate production in this concentration range. It was also found that YC-1 activates the checkpoint kinase-mediated intra-S-phase checkpoint, independently of ataxia-telangiectasia mutated kinase or ataxia-telangiectasia mutated and Rad3-related kinase. These results imply that YC-1 does not promote the regrowth of hypoxic tumors because of its cell cycle arrest effect. Furthermore, YC-1 may induce the combined anticancer effects of HIF-1alpha inhibition and cell growth inhibition.
- Published
- 2006
- Full Text
- View/download PDF
26. Development of a highly active nanoliposomal irinotecan using a novel intraliposomal stabilization strategy.
- Author
-
Drummond DC, Noble CO, Guo Z, Hong K, Park JW, and Kirpotin DB
- Subjects
- Animals, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Camptothecin administration & dosage, Camptothecin chemistry, Camptothecin pharmacokinetics, Camptothecin toxicity, Colonic Neoplasms drug therapy, Colonic Neoplasms metabolism, Drug Stability, Female, HT29 Cells, Humans, Irinotecan, Liposomes administration & dosage, Liposomes pharmacokinetics, Liposomes toxicity, Mice, Mice, Nude, Nanostructures toxicity, Rats, Rats, Sprague-Dawley, Xenograft Model Antitumor Assays, Camptothecin analogs & derivatives, Drug Delivery Systems methods, Liposomes chemistry, Nanostructures chemistry
- Abstract
Liposome formulations of camptothecins have been actively pursued because of the potential for significant pharmacologic advantages from successful drug delivery of this important class of anticancer drugs. We describe nanoliposomal CPT-11, a novel nanoparticle/liposome construct containing CPT-11 (irinotecan) with unprecedented drug loading efficiency and in vivo drug retention. Using a modified gradient loading method featuring a sterically hindered amine with highly charged, multivalent anionic trapping agents, either polymeric (polyphosphate) or nonpolymeric (sucrose octasulfate), liposomes were capable of entrapping CPT-11 at extremely high drug-to-lipid ratios (>800 g CPT-11/mol phospholipid) and retaining encapsulated drug in vivo with a half-life of drug release in the circulation of 56.8 hours. CPT-11 was also protected from hydrolysis to the inactive carboxylate form and from metabolic conversion to SN-38 while circulating. The maximum tolerated dose in normal mice was determined to be 80 mg/kg for free CPT-11 and >320 mg/kg for nanoliposomal CPT-11. Nanoliposomal CPT-11 showed markedly superior efficacy when compared with free CPT-11 in human breast (BT474) and colon (HT29) cancer xenograft models. This study shows that intraliposomal stabilization of CPT-11 using a polymeric or highly charged, nonpolymeric polyanionic trapping agent results in a markedly active antitumor agent with low toxicity.
- Published
- 2006
- Full Text
- View/download PDF
27. Novel nanoliposomal CPT-11 infused by convection-enhanced delivery in intracranial tumors: pharmacology and efficacy.
- Author
-
Noble CO, Krauze MT, Drummond DC, Yamashita Y, Saito R, Berger MS, Kirpotin DB, Bankiewicz KS, and Park JW
- Subjects
- Animals, Brain Neoplasms metabolism, Camptothecin administration & dosage, Camptothecin chemistry, Camptothecin pharmacokinetics, Camptothecin toxicity, Cell Line, Tumor, Convection, Humans, Irinotecan, Liposomes administration & dosage, Liposomes chemistry, Liposomes pharmacokinetics, Liposomes toxicity, Male, Nanostructures chemistry, Nanostructures toxicity, Phospholipids administration & dosage, Phospholipids chemistry, Phospholipids pharmacokinetics, Phospholipids toxicity, Rats, Rats, Sprague-Dawley, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Camptothecin analogs & derivatives, Drug Delivery Systems methods
- Abstract
We hypothesized that combining convection-enhanced delivery (CED) with a novel, highly stable nanoparticle/liposome containing CPT-11 (nanoliposomal CPT-11) would provide a dual drug delivery strategy for brain tumor treatment. Following CED in rat brains, tissue retention of nanoliposomal CPT-11 was greatly prolonged, with >20% injected dose remaining at 12 days for all doses. Tissue residence was dose dependent, with doses of 60 microg (3 mg/mL), 0.8 mg (40 mg/mL), and 1.6 mg (80 mg/mL) resulting in tissue half-life (t(1/2)) of 6.7, 10.7, and 19.7 days, respectively. In contrast, CED of free CPT-11 resulted in rapid drug clearance (tissue t(1/2) = 0.3 day). At equivalent CED doses, nanoliposomal CPT-11 increased area under the time-concentration curve by 25-fold and tissue t(1/2) by 22-fold over free CPT-11; CED in intracranial U87 glioma xenografts showed even longer tumor retention (tissue t(1/2) = 43 days). Plasma levels were undetectable following CED of nanoliposomal CPT-11. Importantly, prolonged exposure to nanoliposomal CPT-11 resulted in no measurable central nervous system (CNS) toxicity at any dose tested (0.06-1.6 mg/rat), whereas CED of free CPT-11 induced severe CNS toxicity at 0.4 mg/rat. In the intracranial U87 glioma xenograft model, a single CED infusion of nanoliposomal CPT-11 at 1.6 mg resulted in significantly improved median survival (>100 days) compared with CED of control liposomes (19.5 days; P = 4.9 x 10(-5)) or free drug (28.5 days; P = 0.011). We conclude that CED of nanoliposomal CPT-11 greatly prolonged tissue residence while also substantially reducing toxicity, resulting in a highly effective treatment strategy in preclinical brain tumor models.
- Published
- 2006
- Full Text
- View/download PDF
28. Epidermal growth factor receptor-targeted immunoliposomes significantly enhance the efficacy of multiple anticancer drugs in vivo.
- Author
-
Mamot C, Drummond DC, Noble CO, Kallab V, Guo Z, Hong K, Kirpotin DB, and Park JW
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Brain Neoplasms drug therapy, Brain Neoplasms immunology, Brain Neoplasms metabolism, Breast Neoplasms immunology, Breast Neoplasms metabolism, Cetuximab, Doxorubicin administration & dosage, Drug Delivery Systems, Epirubicin administration & dosage, ErbB Receptors genetics, ErbB Receptors immunology, Female, Glioblastoma immunology, Glioblastoma metabolism, Humans, Immunoglobulin Fab Fragments immunology, Mice, Mice, Nude, Rats, Rats, Sprague-Dawley, Transfection, Transplantation, Heterologous, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Vinblastine administration & dosage, Vinblastine analogs & derivatives, Vinorelbine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, ErbB Receptors drug effects, Glioblastoma drug therapy, Immunoconjugates therapeutic use, Liposomes administration & dosage
- Abstract
We previously reported the development of epidermal growth factor receptor (EGFR)-targeted immunoliposomes that bind and internalize in tumor cells which overexpress EGFR and/or mutant EGFR variant III (EGFRvIII), enabling intracellular delivery of potent anticancer agents in vitro. We now describe in vivo proof-of-concept for this approach for the delivery of multiple anticancer drugs in EGFR-overexpressing tumor models. Anti-EGFR immunoliposomes were constructed modularly with Fab' fragments of cetuximab (IMC-C225), covalently linked to liposomes containing probes and/or anticancer drugs. Pharmacokinetic and biodistribution studies confirmed long circulation times (t(1/2) = 21 hours) and efficient accumulation in tumors (up to 15% ID/g) irrespective of the presence of the targeting ligand. Although total accumulations of anti-EGFR immunoliposomes and nontargeted liposomes in EGFR-overexpressing tumors were comparable, only immunoliposomes internalized extensively within tumor cells (92% of analyzed cells versus <5% for nontargeted liposomes), indicating different mechanisms of delivery at the cellular level. In vivo therapy studies in a series of xenograft models featuring overexpression of EGFR and/or EGFRvIII showed the superiority of immunoliposomal delivery of encapsulated drugs, which included doxorubicin, epirubicin, and vinorelbine. For each of these drugs, anti-EGFR immunoliposome delivery showed significant antitumor effects and was significantly superior to all other treatments, including the corresponding free or liposomal drug (P < 0.001-0.003). We conclude that anti-EGFR immunoliposomes provide efficient and targeted drug delivery of anticancer compounds and may represent a useful new treatment approach for tumors that overexpress the EGFR.
- Published
- 2005
- Full Text
- View/download PDF
29. Distribution of liposomes into brain and rat brain tumor models by convection-enhanced delivery monitored with magnetic resonance imaging.
- Author
-
Saito R, Bringas JR, McKnight TR, Wendland MF, Mamot C, Drummond DC, Kirpotin DB, Park JW, Berger MS, and Bankiewicz KS
- Subjects
- Animals, Antibiotics, Antineoplastic administration & dosage, Antibiotics, Antineoplastic pharmacokinetics, Carbocyanines administration & dosage, Carbocyanines pharmacology, Convection, Doxorubicin administration & dosage, Doxorubicin pharmacokinetics, Fluorescent Dyes administration & dosage, Fluorescent Dyes pharmacokinetics, Gadolinium administration & dosage, Gadolinium pharmacokinetics, Glioma metabolism, Gliosarcoma metabolism, Liposomes administration & dosage, Liposomes toxicity, Magnetic Resonance Imaging, Male, Rats, Rats, Sprague-Dawley, Tissue and Organ Procurement, Brain metabolism, Brain Neoplasms metabolism, Liposomes pharmacokinetics
- Abstract
Although liposomes have been used as a vehicle for delivery of therapeutic agents in oncology, their efficacy in targeting brain tumors has been limited due to poor penetration through the blood-brain barrier. Because convection-enhanced delivery (CED) of liposomes may improve the therapeutic index for targeting brain tumors, we conducted a three-stage study: stage 1 established the feasibility of using in vivo magnetic resonance imaging (MRI) to confirm adequate liposomal distribution within targeted regions in normal rat brain. Liposomes colabeled with gadolinium (Gd) and a fluorescent indicator, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine-5,5'-disulfonic acid [DiI-DS; formally DiIC(18)(3)-DS], were administered by CED into striatal regions. The minimum concentration of Gd needed for monitoring, correlation of infused volume with distribution volume, clearance of infused liposome containing Gd and DiI-DS (Lip/Gd/DiI-DS), and potential local toxicity were evaluated. After determination of adequate conditions for MRI detection in normal brain, stage 2 evaluated the feasibility of in vivo MRI monitoring of liposomal distribution in C6 and 9L-2 rat glioma models. In both models, the distribution of Lip/Gd/DiI-DS covering the tumor mass was well defined and monitored with MRI. Stage 3 was designed to develop a clinically relevant treatment strategy in the 9L-2 model by infusing liposome containing Gd (Lip/Gd), prepared in the same size as Lip/Gd/DiI-DS, with Doxil, a liposomal drug of similar size used to treat several cancers. MRI detection of Lip/Gd coadministered with Doxil provided optimum CED parameters for complete coverage of 9L-2 tumors. By permitting in vivo monitoring of therapeutic distribution in brain tumors, this technique optimizes local drug delivery and may provide a basis for clinical applications in the treatment of malignant glioma.
- Published
- 2004
- Full Text
- View/download PDF
30. Phorbol ester stimulates the nonhypoxic induction of a novel hypoxia-inducible factor 1alpha isoform: implications for tumor promotion.
- Author
-
Chun YS, Lee KH, Choi E, Bae SY, Yeo EJ, Huang LE, Kim MS, and Park JW
- Subjects
- Alternative Splicing, Animals, Cell Division physiology, Cell Hypoxia physiology, Cell Line, Tumor, DNA Methylation, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Mice, Mice, Nude, Protein Isoforms, RNA, Messenger genetics, Signal Transduction drug effects, Transcription Factors genetics, Transcription Factors physiology, Transcriptional Activation, Transfection, Up-Regulation drug effects, Carcinogens pharmacology, Tetradecanoylphorbol Acetate pharmacology, Transcription Factors biosynthesis
- Abstract
Hypoxia-inducible factor-1 (HIF-1), which is present at higher levels in human tumors, plays important roles in tumor promotion. It is composed of HIF-1alpha and HIF-1beta subunits and its activity depends on the amount of HIF-1alpha, which is tightly controlled by cellular oxygen tension. In addition to hypoxia, various nonhypoxic stimuli can stabilize HIF-1alpha in tumor cells, implying that both hypoxic and nonhypoxic stimuli contribute to the overexpression of HIF-1alpha in tumors. On the other hand, phorbol esters such as phorbol-12-myristate-13-acetate (PMA) are known to be potent tumor promoters. Here, we identified a novel HIF-1alpha isoform, which is regulated primarily by PMA. The variant mRNA lacks exon 11 and produces a 785-amino acid isoform (HIF-1alpha(785)) without altering the reading frame and therefore the COOH-terminal transcriptional activity. HIF-1alpha(785) is induced markedly by PMA and heat shock, the latter of which is also known to induce HIF-1alpha. HIF-1alpha(785) escapes from lysine acetylation because of the loss of Lys(532) and was stabilized under normoxic conditions. Its expression was blocked by reducing agents and by a mitogen-activated protein/extracellular signal-regulated kinase-1 inhibitor and enhanced by hydrogen peroxide. In addition, HIF-1alpha(785) overexpression strikingly enhanced tumor growth in vivo. These results suggest that HIF-1alpha(785) is induced by PMA under normoxic conditions via a redox-dependent mitogen-activated protein/extracellular signal-regulated kinase-1 pathway and that it plays an important role in tumor promotion.
- Published
- 2003
31. Protective role of alpha-phenyl-N-t-butylnitrone against ionizing radiation in U937 cells and mice.
- Author
-
Lee JH and Park JW
- Subjects
- Animals, Cell Death radiation effects, Cyclic N-Oxides, DNA Damage, Humans, Liver metabolism, Liver radiation effects, Male, Mice, Mice, Inbred ICR, Mitochondria radiation effects, Oxidation-Reduction, Oxidative Stress, U937 Cells, Nitrogen Oxides pharmacology, Radiation-Protective Agents pharmacology
- Abstract
Ionizing radiation (IR) induces the production of reactive oxygen species (ROS), which play an important causative role in radiation damage. alpha-Phenyl-N-t-butylnitrone (PBN) is one of the most widely used spin-trapping compounds for investigating the existence of free radicals in biological systems. We investigated the protective role of PBN against IR in U937 cells and mice. On exposure to IR, there was a distinct difference between the control cells and the cells pretreated with PBN in regard to viability, cellular redox status, and oxidative damage to cells. Lipid peroxidation, oxidative DNA damage, and protein oxidation were significantly lower in the cells treated with PBN when the cells were exposed to IR. Although the activities of antioxidant enzymes were comparable in PBN-treated and control cells, the [GSSG]:[GSH + GSSG] ratio and the generation of intracellular ROS were higher and the [NADPH]:[NADP(+) + NADPH] ratio was lower in control cells compared with PBN-treated cells. The IR-induced mitochondrial damage reflected by the altered mitochondrial permeability transition, the increase in the accumulation of ROS, the reduction of ATP production, and the morphological change were significantly higher in control cells compared with PBN-treated cells. PBN administration for 14 days with a daily dosage of 30 mg/kg provided substantial protection against killing and oxidative damage to mice exposed to whole body irradiation. These data indicate that PBN may have great application potential as a new class of in vivo, nonsulfur-containing radiation protector.
- Published
- 2003
32. The oncogene phosphatidylinositol 3'-kinase catalytic subunit alpha promotes angiogenesis via vascular endothelial growth factor in ovarian carcinoma.
- Author
-
Zhang L, Yang N, Katsaros D, Huang W, Park JW, Fracchioli S, Vezzani C, Rigault de la Longrais IA, Yao W, Rubin SC, and Coukos G
- Subjects
- Apoptosis physiology, Catalytic Domain, Cell Division physiology, Chromones pharmacology, Endothelial Growth Factors genetics, Enzyme Inhibitors pharmacology, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Intercellular Signaling Peptides and Proteins genetics, Lymphokines genetics, Morpholines pharmacology, Neovascularization, Pathologic enzymology, Ovarian Neoplasms enzymology, Ovarian Neoplasms genetics, Phosphatidylinositol 3-Kinases biosynthesis, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, RNA, Messenger biosynthesis, RNA, Messenger genetics, Transcription Factors metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors biosynthesis, Intercellular Signaling Peptides and Proteins biosynthesis, Lymphokines biosynthesis, Neovascularization, Pathologic genetics, Ovarian Neoplasms blood supply, Phosphatidylinositol 3-Kinases genetics
- Abstract
The gene of phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) has been implicated as an oncogene in ovarian cancer [L. Shayesteh et al., Nat. Genet., 21: 99-102, 1999]. In this study, we examined the expression of PIK3CA mRNA and its p110alpha protein product in human ovarian carcinoma and investigated its role in regulating angiogenesis via vascular endothelial growth factor (VEGF). PIK3CA mRNA was detected in 66.6% of stage I and 93.9% of advanced stage ovarian cancer specimens and in all 17 ovarian cancer cell lines. PIK3CA mRNA levels were significantly higher in invasive carcinomas compared with benign and low malignant potential neoplasms (P = 0.007), but no significant difference was seen between early and advanced stage carcinomas (P = 0.812). Strong expression of immunoreactive p110alpha was detected in tumor cells and/or stroma endothelium. PIK3CA expression in vivo positively correlated, both at the mRNA and the protein level, with the expression of VEGF as well as with the extent of microvascular development. Furthermore, PIK3CA mRNA overexpression positively correlated with increased proliferation and decreased apoptosis of tumor cells in vivo. In vitro, PIK3CA expression positively correlated with the expression of VEGF in ovarian cancer cells, whereas the phosphatidylinositol 3'-kinase inhibitor Ly294002 reduced both the constitutive and inducible expression of hypoxia-inducible factor-1alpha at the mRNA and protein levels and abrogated VEGF up-regulation by glucose starvation. Furthermore, Ly294002 suppressed cell proliferation and, at higher doses, induced marked apoptosis in ovarian cancer cells. Collectively, these data strongly indicate that PIK3CA supports ovarian cancer growth through multiple and independent pathways affecting cell proliferation, apoptosis and angiogenesis, and plays an important role in ovarian cancer progression.
- Published
- 2003
33. Tumor-derived vascular endothelial growth factor up-regulates angiopoietin-2 in host endothelium and destabilizes host vasculature, supporting angiogenesis in ovarian cancer.
- Author
-
Zhang L, Yang N, Park JW, Katsaros D, Fracchioli S, Cao G, O'Brien-Jenkins A, Randall TC, Rubin SC, and Coukos G
- Subjects
- Angiogenesis Inducing Agents genetics, Angiopoietin-1, Angiopoietin-2, Animals, Carcinoma blood supply, Carcinoma metabolism, Carcinoma pathology, Cell Line, Transformed drug effects, Cell Line, Transformed metabolism, Dactinomycin pharmacology, Endothelial Growth Factors pharmacology, Endothelium, Vascular metabolism, Female, Genes, Reporter, Humans, Intercellular Signaling Peptides and Proteins pharmacology, Lymphokines pharmacology, Membrane Glycoproteins biosynthesis, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, Models, Biological, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Neoplasm Proteins pharmacology, Nucleic Acid Synthesis Inhibitors pharmacology, Ovarian Neoplasms blood supply, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Pericytes pathology, RNA, Messenger biosynthesis, RNA, Neoplasm biosynthesis, Receptor Protein-Tyrosine Kinases biosynthesis, Receptor Protein-Tyrosine Kinases genetics, Receptor, TIE-2, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins physiology, Stromal Cells drug effects, Stromal Cells metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Angiogenesis Inducing Agents biosynthesis, Carcinoma physiopathology, Endothelial Growth Factors physiology, Endothelium, Vascular drug effects, Gene Expression Regulation, Neoplastic, Intercellular Signaling Peptides and Proteins physiology, Lymphokines physiology, Neoplasm Proteins physiology, Neovascularization, Pathologic physiopathology, Ovarian Neoplasms physiopathology, Paracrine Communication
- Abstract
Vascular remodeling in host tissues surrounding growing tumors is implicated in the successful development of tumor neovasculature. Cooperation between vascular endothelial growth factor (VEGF) and angiopoietins (Angs) is considered to be critical in this context. However, the mechanisms regulating the coordinated expression of these molecules remain, to date, elusive. In this study, we used a murine ovarian cancer angiogenesis model induced by overexpression of VEGF, as well as 52 human ovarian cancer specimens and 36 established cancer cell lines to characterize the expression and regulation of Ang-2 in the context of tumor angiogenesis. Using a combination of immunohistochemistry, laser capture microdissection and real-time quantitative reverse transcription-PCR, we showed that tumor-derived VEGF significantly up-regulated the expression of Ang-2 in host stroma endothelial cells, resulting in markedly increased Ang-2/Tie-2 mRNA copy number ratio in vivo. In vitro experiments showed that VEGF directly up-regulated Ang-2, which is mediated via VEGF receptor-2/flk-1/KDR pathway, in cultured endothelial cells through transcriptional activation rather than the enhanced mRNA stability. In human ovarian cancer, Ang-2 was primarily expressed in stroma endothelial cells and detectable in tumor cells of only 12% tumor specimens; however, it was not detected in the majority of established ovarian cancer cell lines. In addition, a significant correlation was observed between VEGF and Ang-2 mRNA expression (P < 0.01) but not between VEGF and Ang-1 or Tie-2 in human ovarian cancer specimens. In the mouse ovarian cancer model, up-regulation of Ang-2 in host stroma endothelial cells was significantly associated with pericyte loss and instability of the host vasculature surrounding the tumor. Our study suggests a novel mechanism by which tumor-derived VEGF interacts with Angs/Tie-2 system in host stroma endothelial cells and induces in a paracrine manner the remodeling of host vasculature to support angiogenesis during tumor growth.
- Published
- 2003
34. Epidermal growth factor receptor (EGFR)-targeted immunoliposomes mediate specific and efficient drug delivery to EGFR- and EGFRvIII-overexpressing tumor cells.
- Author
-
Mamot C, Drummond DC, Greiser U, Hong K, Kirpotin DB, Marks JD, and Park JW
- Subjects
- Adenocarcinoma pathology, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal therapeutic use, Antibodies, Neoplasm administration & dosage, Antineoplastic Agents blood, Brain Neoplasms pathology, Breast Neoplasms pathology, Carcinoma, Squamous Cell pathology, Doxorubicin blood, Drug Delivery Systems, Drug Design, ErbB Receptors genetics, ErbB Receptors immunology, Female, Glioblastoma pathology, Humans, Immunoconjugates blood, Immunoglobulin Fab Fragments immunology, Liposomes administration & dosage, Liposomes blood, Neoplasm Proteins immunology, Transfection, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Vinorelbine, Vulvar Neoplasms pathology, Antineoplastic Agents administration & dosage, Doxorubicin administration & dosage, ErbB Receptors drug effects, Immunoconjugates administration & dosage, Methotrexate administration & dosage, Neoplasm Proteins drug effects, Vinblastine administration & dosage, Vinblastine analogs & derivatives
- Abstract
We hypothesized that immunoliposomes (ILs) that target epidermal growth factor receptor (EGFR) and/or its truncated variant EGFRvIII can be constructed to provide efficient intracellular drug delivery in tumor cells overexpressing these receptors. Monoclonal antibody fragments included Fab' fragments derived from C225, which binds both EGFR and EGFRvIII, or novel anti-EGFR scFv C10, which binds EGFR only. Monoclonal antibody fragments were covalently linked to liposomes containing various reporters or drugs. ILs were evaluated for specific binding, internalization, and cytotoxicity in EGFR/EGFRvIII-overexpressing cell lines in vitro. Flow cytometry and fluorescence microscopy showed that EGFR-targeted ILs, but not nontargeted liposomes or irrelevant ILs, were efficiently bound and internalized by EGFR-overexpressing cells, including glioma cells (U-87), carcinoma cells (A-431 and MDA-MB-468), and EGFRvIII stable transfectants (NR-6M). Furthermore, EGFR-targeted ILs did not bind to non-EGFR-overexpressing cells (MCF-7 and parental NR-6). ILs showed 3 orders of magnitude greater accumulation in NR-6-EGFRvIII stable transfectants versus parental NR-6 cells. Quantitative internalization studies indicated binding of EGFR-targeted ILs to target cells within 5 min, followed by intracellular accumulation beginning at 15 min; total uptake reached approximately 13,000 ILs/cell. ILs were used to deliver cytotoxic drugs doxorubicin, vinorelbine, or methotrexate to EGFR/EGFRvIII-overexpressing target cells in vitro. In each case, the IL agent was significantly more cytotoxic than the corresponding nontargeted liposomal drug in target cells, whereas it was equivalent in cells lacking EGFR/EGFRvIII overexpression. We conclude that EGFR-targeted ILs provide efficient and targeted delivery of anticancer drugs in cells overexpressing EGFR or EGFRvIII.
- Published
- 2003
35. Dykellic acid inhibits phorbol myristate acetate-induced matrix metalloproteinase-9 expression by inhibiting nuclear factor kappa B transcriptional activity.
- Author
-
Woo JH, Park JW, Lee SH, Kim YH, Lee IK, Gabrielson E, Lee SH, Lee HJ, Kho YH, and Kwon TK
- Subjects
- Enzyme Induction drug effects, Gene Expression Regulation, Neoplastic drug effects, Genes, Reporter, Humans, Luciferases analysis, Luciferases genetics, Matrix Metalloproteinase 2 biosynthesis, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 9 genetics, NF-kappa B chemistry, NF-kappa B physiology, Neoplasm Invasiveness, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins chemistry, Neoplasm Proteins genetics, Proline pharmacology, Promoter Regions, Genetic drug effects, Protein Structure, Tertiary, RNA, Messenger biosynthesis, RNA, Messenger genetics, Tetradecanoylphorbol Acetate pharmacology, Thiocarbamates pharmacology, Thioctic Acid pharmacology, Transcription Factor AP-1 metabolism, Transfection, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured enzymology, Matrix Metalloproteinase 9 biosynthesis, NF-kappa B antagonists & inhibitors, Neoplasm Proteins biosynthesis, Proline analogs & derivatives, Propionates pharmacology, Pyrones pharmacology, Tetradecanoylphorbol Acetate antagonists & inhibitors, Transcriptional Activation drug effects
- Abstract
Proteolytic degradation of the extracellular matrix and tumor metastasis correlate with expression of endopeptidases known as matrix metalloproteinases (MMPs). Expression of MMPs is regulated by cytokines and signal transduction pathways, including those activated by phorbol myristate acetate. We found that dykellic acid, a fungal metabolite, significantly inhibits the phorbol myristate acetate-induced increase in MMP-9 expression and activity. These effects of dykellic acid are time- and dose-dependent, and correlate with decreased MMP-9 promoter activity and mRNA expression. Whereas this compound does not affect DNA binding activity of nuclear factor kappa B (NF kappa B), dykellic acid does inhibit transactivation of NF kappa B. These data demonstrate a role for NF kappa B in the regulation of MMP-9 expression and the ability of dykellic acid to suppress this action of NF kappa B.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.