1
|
Hassan MS, Cwidak N, Johnson C, Däster S, Eppenberger-Castori S, Awasthi N, Li J, Schwarz MA, von Holzen U. Therapeutic Potential of the Cyclin-Dependent Kinase Inhibitor Flavopiridol on c-Myc Overexpressing Esophageal Cancer. Front Pharmacol 2021; 12:746385. [PMID: 34621175 PMCID: PMC8490822 DOI: 10.3389/fphar.2021.746385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Tumors with elevated c-Myc expression often exhibit a highly aggressive phenotype, and c-Myc amplification has been shown to be frequent in esophageal cancer. Emerging data suggests that synthetic lethal interactions between c-Myc pathway activation and small molecules inhibition involved in cell cycle signaling can be therapeutically exploited to preferentially kill tumor cells. We therefore investigated whether exploiting elevated c-Myc expression is effective in treating esophageal cancer with the CDK inhibitor flavopiridol. We found frequent overexpression of c-Myc in human esophageal cancer cell lines and tissues. c-Myc overexpression correlated with accelerated esophageal cancer subcutaneous xenograft tumor growth. Esophageal cancer cells with elevated c-Myc expression were found preferentially more sensitive to induction of apoptosis by the CDK inhibition flavopiridol compared to esophageal cancer cells with lower c-Myc expression. In addition, we observed that flavopiridol alone or in combination with the chemotherapeutic agent nanoparticle albumin-bound paclitaxel (NPT) or in combinations with the targeted agent BMS-754807 significantly inhibited esophageal cancer cell proliferation and subcutaneous xenograft tumor growth while significantly enhancing overall mice survival. These results indicate that aggressive esophageal cancer cells with elevated c-Myc expression are sensitive to the CDK inhibitor flavopiridol, and that flavopiridol alone or in combination can be a potential therapy for c-Myc overexpressing esophageal cancer.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States
| | - Nicholas Cwidak
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States
| | - Chloe Johnson
- University of Notre Dame, Notre Dame, IN, United States
| | | | | | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States
| | - Jun Li
- Harper Cancer Research Institute, South Bend, IN, United States.,University of Notre Dame, Notre Dame, IN, United States
| | - Margaret A Schwarz
- Harper Cancer Research Institute, South Bend, IN, United States.,Department of Pediatrics, Indiana University School of Medicine, South Bend, IN, United States
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States.,University of Basel, Basel, Switzerland.,Goshen Center for Cancer Care, Goshen, IN, United States
| |
Collapse
|
2
|
Yang J, Wang T, Zhao L, Rajasekhar VK, Joshi S, Andreou C, Pal S, Hsu HT, Zhang H, Cohen IJ, Huang R, Hendrickson RC, Miele MM, Pei W, Brendel MB, Healey JH, Chiosis G, Kircher MF. Gold/alpha-lactalbumin nanoprobes for the imaging and treatment of breast cancer. Nat Biomed Eng 2020; 4:686-703. [PMID: 32661307 PMCID: PMC8255032 DOI: 10.1038/s41551-020-0584-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/11/2020] [Indexed: 02/03/2023]
Abstract
Theranostic agents should ideally be renally cleared and biodegradable. Here, we report the synthesis, characterization and theranostic applications of fluorescent ultrasmall gold quantum clusters that are stabilized by the milk metalloprotein alpha-lactalbumin. We synthesized three types of these nanoprobes that together display fluorescence across the visible and near-infrared spectra when excited at a single wavelength through optical colour coding. In live tumour-bearing mice, the near-infrared nanoprobe generates contrast for fluorescence, X-ray computed tomography and magnetic resonance imaging, and exhibits long circulation times, low accumulation in the reticuloendothelial system, sustained tumour retention, insignificant toxicity and renal clearance. An intravenously administrated near-infrared nanoprobe with a large Stokes shift facilitated the detection and image-guided resection of breast tumours in vivo using a smartphone with modified optics. Moreover, the partially unfolded structure of alpha-lactalbumin in the nanoprobe helps with the formation of an anti-cancer lipoprotein complex with oleic acid that triggers the inhibition of the MAPK and PI3K-AKT pathways, immunogenic cell death and the recruitment of infiltrating macrophages. The biodegradability and safety profile of the nanoprobes make them suitable for the systemic detection and localized treatment of cancer.
Collapse
Affiliation(s)
- Jiang Yang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tai Wang
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Lina Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | | | - Suhasini Joshi
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Chrysafis Andreou
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suchetan Pal
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsiao-Ting Hsu
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanwen Zhang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan J Cohen
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruimin Huang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbo Pei
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Matthew B Brendel
- Molecular Cytology Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Moritz F Kircher
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Eyvazi S, Hejazi MS, Kahroba H, Abasi M, Zamiri RE, Tarhriz V. CDK9 as an Appealing Target for Therapeutic Interventions. Curr Drug Targets 2020; 20:453-464. [PMID: 30362418 DOI: 10.2174/1389450119666181026152221] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 02/05/2023]
Abstract
Cyclin Dependent Kinase 9 (CDK9) as a serine/threonine kinase belongs to a great number of CDKs. CDK9 is the main core of PTEF-b complex and phosphorylates RNA polymerase (RNAP) II besides other transcription factors which regulate gene transcription elongation in numerous physiological processes. Multi-functional nature of CDK9 in diverse cellular pathways proposes that it is as an appealing target. In this review, we summarized the recent findings on the molecular interaction of CDK9 with critical participant molecules to modulate their activity in various diseases. Furthermore, the presented review provides a rationale supporting the use of CDK9 as a therapeutic target in clinical developments for crucial diseases; particularly cancers will be reviewed.
Collapse
Affiliation(s)
- Shirin Eyvazi
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeid Hejazi
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Homan Kahroba
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozghan Abasi
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Eghdam Zamiri
- Faculty of medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Bowman KR, Kim JH, Lim CS. Narrowing the field: cancer-specific promoters for mitochondrially-targeted p53-BH3 fusion gene therapy in ovarian cancer. J Ovarian Res 2019; 12:38. [PMID: 31039796 PMCID: PMC6492428 DOI: 10.1186/s13048-019-0514-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Despite years of research, the treatment options and mortality rate for ovarian cancer remain relatively stagnant. Resistance to chemotherapy and high heterogeneity in mutations contribute to ovarian cancer's lethality, including many mutations in tumor suppressor p53. Though wild type p53 gene therapy clinical trials failed in ovarian cancer, mitochondrially-targeted p53 fusion constructs, including a fusion with pro-apoptotic protein Bad, have shown much higher apoptotic potential than wild type p53 in vitro. Due to the inherent toxicities of mitochondrial apoptosis, cancer-specificity for the p53 fusion constructs must be developed. Cancer-specific promoters such as hTERT, hTC, Brms1, and Ran have shown promise in ovarian cancer. RESULTS Of five different lengths of hTERT promoter, the - 279/+ 5 length relative to the transcription start site showed the highest activity across a panel of ovarian cancer cells. In addition to - 279/+ 5, promoters hTC (an hTERT/CMV promoter hybrid), Brms1, and Ran were tested as drivers of mitochondrially-targeted p53-Bad and p53-Bad* fusion gene therapy constructs. p53-Bad* displayed cancer-specific killing in all ovarian cancer cell lines when driven by hTC, - 279/+ 5, or Brms1. CONCLUSIONS Cancer-specific promoters hTC, - 279/+ 5, and Brms1 all display promise in driving p53-Bad* gene therapy for treatment of ovarian cancer and should be moved forward into in vivo studies. -279/+ 5 displays lower expression levels in fewer cells, but greater cancer specificity, rendering it most useful for gene therapeutics with high toxicity to normal cells. hTC and Brms1 show higher transfection and expression levels with some cancer specificity, making them ideal for lowering toxicity in order to increase dose without as much of a reduction in the number of cancer cells expressing the gene construct. Having a variety of promoters available means that patient genetic testing can aid in choosing a promoter, thereby increasing cancer-specificity and giving patients with ovarian cancer a greater chance at survival.
Collapse
Affiliation(s)
| | - Ji Hoon Kim
- New York University, 31 Washington Pl, New York, NY 10003 USA
| | - Carol S. Lim
- University of Utah, 30 S 2000 E Room #301, Salt Lake City, UT 84112 USA
| |
Collapse
|
5
|
Hsin IL, Hsu JC, Wu WJ, Lu HJ, Wu MF, Ko JL. GMI, a fungal immunomodulatory protein from Ganoderma microsporum, induce apoptosis via β-catenin suppression in lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:955-961. [PMID: 29974605 DOI: 10.1002/tox.22582] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
β-catenin is important in development of lung cancer. In our previous study, GMI, a fungal immunomodulatory protein, inhibits lung cancer cell survival. The aim of this study is to evaluate the effect of GMI on β-catenin inhibition and apoptosis induction. GMI induced apoptosis in lung cancer cells bearing wild-type and mutated EGFR. GMI did not reduce the β-catenin mRNA expression but suppressed the protein expressions of β-catenin that resulted in the transcriptional downregulation of its target genes: survivin and cyclin-D1. The transcriptional activation activity of β-catenin was demonstrated by TOPFLASH/FOPFLASH luciferase reporter assay. Inhibition of GSK-3β and proteasome blocked the inhibiting effect of GMI on β-catenin and its target genes. β-catenin silencing increased activation of apoptosis in GMI-treated H1355 cells. This is the first study to reveal the novel function of GMI in inducing apoptosis via β-catenin inhibition. These results provide a new potential of GMI in against lung cancer.
Collapse
Affiliation(s)
- I-Lun Hsin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Inflammation Research & Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Jen-Chieh Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Jun Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsueh-Ju Lu
- Division of Medical Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Fang Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Medical Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Medical Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
6
|
DNA-dependent protein kinase modulates the anti-cancer properties of silver nanoparticles in human cancer cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017; 824:32-41. [PMID: 29150048 DOI: 10.1016/j.mrgentox.2017.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
Abstract
Silver nanoparticles (Ag-np) were reported to be toxic to eukaryotic cells. These potentially detrimental effects of Ag-np can be advantageous in experimental therapeutics. They are currently being employed to enhance the therapeutic efficacy of cancer drugs. In this study, we demonstrate that Ag-np treatment trigger the activation of DNA-PKcs and JNK pathway at selected doses, presumably as a physiologic response to DNA damage and repair in normal and malignant cells. Ag-np altered the telomere dynamics by disrupting the shelterin complex located at the telomeres and telomere lengths. The genotoxic effect of Ag-np was not restricted to telomeres but the entire genome as Ag-np induced γ-H2AX foci formation, an indicator of global DNA damage. Inhibition of DNA-PKcs activity sensitised the cancer cells towards the cytotoxicity of Ag-np and substantiated the damaging effect of Ag-np at telomeres in human cancer cells. Abrogation of JNK mediated DNA repair and extensive damage of telomeres led to greater cell death following Ag-np treatment in DNA-PKcs inhibited cancer cells. Collectively, this study suggests that improved anti-proliferative and cytotoxic effects of Ag-np treatment in cancer cells can be achieved by the inhibition of DNA-PKcs.
Collapse
|
7
|
Lee J, Kim MS, Kim MA, Jang YK. Calmidazolium chloride inhibits growth of murine embryonal carcinoma cells, a model of cancer stem-like cells. Toxicol In Vitro 2016; 35:86-92. [PMID: 27247146 DOI: 10.1016/j.tiv.2016.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/07/2016] [Accepted: 05/27/2016] [Indexed: 01/01/2023]
Abstract
Calmidazolium chloride (CMZ) is widely used as a calmodulin (CaM) antagonist, but is also known to induce apoptosis in certain cancer cell lines. However, in spite of the importance of cancer stem cells (CSCs) in cancer therapy, the effects of CMZ on CSCs are not yet well understood. We investigated the effects of CMZ on the F9 embryonal carcinoma cell (ECC) line as a surrogate model of CSCs. To avoid bias due to culture conditions, F9 ECCs and E14 embryonic stem cells (ESCs) were grown in the same culture medium. Results obtained using a cell-counting kit showed that CMZ significantly inhibited growth in F9 ECCs compared with growth in E14 ESCs. CMZ also induced apoptosis of F9 ECCs, but not of E14 ESCs, which was associated with caspase-3 activation and an increased fraction of the sub-G1 cell population. In addition, our data revealed that the expression of stemness-related genes including c-Myc was selectively down regulated in CMZ-treated F9 ECCs. Our results suggest that CMZ can inhibit the growth of ECCs by inducing apoptosis and down regulating stemness-related genes, without causing any harm to normal stem cells. These findings indicate a potential application of CMZ in the development of anti-CSC therapeutics.
Collapse
Affiliation(s)
- Jina Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| | - Min Seong Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| | - Min Aeh Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea
| | - Yeun Kyu Jang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Initiative for Biological Function & Systems, Yonsei University, Seoul 120-749, Republic of Korea.
| |
Collapse
|
8
|
Rosso M, Lapyckyj L, Amiano N, Besso MJ, Sánchez M, Chuluyan E, Vazquez-Levin MH. Secretory Leukocyte Protease Inhibitor (SLPI) expression downregulates E-cadherin, induces β-catenin re-localisation and triggers apoptosis-related events in breast cancer cells. Biol Cell 2014; 106:308-22. [PMID: 25039920 DOI: 10.1111/boc.201300075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 07/01/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND INFORMATION Epithelial cadherin (E-cadherin) is involved in cell-cell adhesion through its extracellular domain, whereas the intracellular domain interacts with adaptor proteins, i.e. β-catenin, links E-cadherin to the actin cytoskeleton and participates in signal transduction events. E-cadherin protects mammary epithelial cells from apoptosis and its loss during tumour progression has been documented. Secretory Leukocyte Protease Inhibitor (SLPI) has anti- and pro-tumourigenic activities but its role in breast cancer has not been fully elucidated. Notwithstanding its relevance, how SLPI affects E-cadherin in breast cancer is still unknown. This study evaluated the effect of SLPI upon E-cadherin/β-catenin expression and apoptosis-related markers in murine (F3II) and human (MCF-7) breast tumour cells either treated with exogenous recombinant human SLPI (rhSLPI) or stably transfected with a plasmid encoding its sequence. RESULTS Addition of rhSLPI to F3II cells caused a decrease (P < 0.05) in E-cadherin transcript and protein levels. Similar results were observed in SLPI-stable F3II transfectants (2C1), and treatment of 2C1 cells with a siRNA toward SLPI restored E-cadherin to control levels. SLPI-expressing cells showed disruption of E-cadherin/β-catenin complex and increased (P < 0.05) percentage of cells depicting nuclear β-catenin localisation. Associated to these changes, 2C1 cells showed increased Bax/Bcl-2 ratio and p21 protein levels, decreased c-Myc protein levels and decreased Cyclin D1 and Claudin-1 transcript levels. No differences in N- and P-cadherin were observed between SLPI-transfected cells and controls. Addition of rhSLPI to MCF-7 cells or stable transfection with SLPI caused a decrease (P < 0.05) in E-cadherin expression (transcript/protein) and its redistribution to the cytoplasm, as well as β-catenin re-localisation to the cell nucleus. CONCLUSIONS Expression of SLPI was associated to a decrease in E-cadherin expression and re-localisation of E-cadherin to the cell cytoplasm and β-catenin to the cell cytoplasm and nucleus, and had pro-apoptotic and cell cycle-arrest effects.
Collapse
Affiliation(s)
- Marina Rosso
- Instituto de Biología & Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Vuelta de Obligado 2490, Buenos Aires, C1428ADN, Argentina
| | | | | | | | | | | | | |
Collapse
|
9
|
Yang L, Moss T, Mangala LS, Marini J, Zhao H, Wahlig S, Armaiz-Pena G, Jiang D, Achreja A, Win J, Roopaimoole R, Rodriguez-Aguayo C, Mercado-Uribe I, Lopez-Berestein G, Liu J, Tsukamoto T, Sood AK, Ram PT, Nagrath D. Metabolic shifts toward glutamine regulate tumor growth, invasion and bioenergetics in ovarian cancer. Mol Syst Biol 2014; 10:728. [PMID: 24799285 PMCID: PMC4188042 DOI: 10.1002/msb.20134892] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Glutamine can play a critical role in cellular growth in multiple cancers. Glutamine‐addicted cancer cells are dependent on glutamine for viability, and their metabolism is reprogrammed for glutamine utilization through the tricarboxylic acid (TCA) cycle. Here, we have uncovered a missing link between cancer invasiveness and glutamine dependence. Using isotope tracer and bioenergetic analysis, we found that low‐invasive ovarian cancer (OVCA) cells are glutamine independent, whereas high‐invasive OVCA cells are markedly glutamine dependent. Consistent with our findings, OVCA patients’ microarray data suggest that glutaminolysis correlates with poor survival. Notably, the ratio of gene expression associated with glutamine anabolism versus catabolism has emerged as a novel biomarker for patient prognosis. Significantly, we found that glutamine regulates the activation of STAT3, a mediator of signaling pathways which regulates cancer hallmarks in invasive OVCA cells. Our findings suggest that a combined approach of targeting high‐invasive OVCA cells by blocking glutamine's entry into the TCA cycle, along with targeting low‐invasive OVCA cells by inhibiting glutamine synthesis and STAT3 may lead to potential therapeutic approaches for treating OVCAs.
Collapse
Affiliation(s)
- Lifeng Yang
- Laboratory for Systems Biology of Human Diseases, Rice University, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cui J, Waltman P, Le VH, Lewis EA. The effect of molecular crowding on the stability of human c-MYC promoter sequence I-motif at neutral pH. Molecules 2013; 18:12751-67. [PMID: 24132198 PMCID: PMC6270392 DOI: 10.3390/molecules181012751] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/26/2013] [Accepted: 10/10/2013] [Indexed: 12/20/2022] Open
Abstract
We have previously shown that c-MYC promoter sequences can form stable i-motifs in acidic solution (pH 4.5-5.5). In terms of drug targeting, the question is whether c-MYC promoter sequence i-motifs will exist in the nucleus at neutral pH. In this work, we have investigated the stability of a mutant c-MYC i-motif in solutions containing a molecular crowding agent. The crowded nuclear environment was modeled by the addition of up to 40% w/w polyethylene glycols having molecular weights up to 12,000 g/mol. CD and DSC were used to establish the presence and stability of c-MYC i-motifs in buffer solutions over the pH range 4 to 7. We have shown that the c-MYC i-motif can exist as a stable structure at pH values as high as 6.7 in crowded solutions. Generic dielectric constant effects, e.g., a shift in the pKa of cytosine by more than 2 units (e.g., 4.8 to 7.0), or the formation of non-specific PEG/DNA complexes appear to contribute insignificantly to i-motif stabilization. Molecular crowding, largely an excluded volume effect of added PEG, having a molecular weight in excess of 1,000 g/mol, appears to be responsible for stabilizing the more compact i-motif over the random coil at higher pH values.
Collapse
Affiliation(s)
- Jingjing Cui
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, USA.
| | | | | | | |
Collapse
|
11
|
Zhang L, Sang H, Liu Y, Li J. Manganese activates caspase-9-dependent apoptosis in human bronchial epithelial cells. Hum Exp Toxicol 2012; 32:1155-63. [PMID: 23263852 DOI: 10.1177/0960327112470272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Acute inhalation exposure to high levels of manganese (Mn) is associated with pulmonary edema and impaired function. The immune-mediated lung epithelium injury of Mn in vivo and in vitro experiments has been well characterized, whereas its apoptotic effect is not well defined. Our results show that human bronchial epithelial (16HBE) cells undergo caspase-9-mediated cell death in response to Mn. Loss of mitochondrial membrane potential (ΔΨm), the formation of reactive oxygen species and release of cytochrome c were regulated during this process. In addition, decreasing c-Myc level and increasing of phosphorylated p53 (Ser 15) and WAF1/p21 were also taken part in Mn-mediated lung toxicity. Proteasome inhibitor MG132 could increase c-Myc protein in abundance. Taking together, our results demonstrate that caspase-9-dependent intrinsic pathway, the downregulation of c-Myc and the upregulation of p53 and phosphorylated p53 might be responsible for Mn-mediated apoptosis in 16HBE cells. Moreover, c-Myc decrease might be due to increased degradation through the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- L Zhang
- 1Department of Prevention, Tongji University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
12
|
Thayanithy V, Sarver AL, Kartha RV, Lihua L, Angstadt AY, Breen M, Steer CJ, Modiano JF, Subramanian S. Perturbation of 14q32 miRNAs-cMYC gene network in osteosarcoma. Bone 2012; 50:171-81. [PMID: 22037351 PMCID: PMC3755949 DOI: 10.1016/j.bone.2011.10.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 10/04/2011] [Accepted: 10/10/2011] [Indexed: 01/07/2023]
Abstract
Osteosarcoma (OS) is the common histological form of primary bone cancer and one of the leading aggressive cancers in children under age fifteen. Although several genetic predisposing conditions have been associated with OS the understanding of its molecular etiology is limited. Here, we show that microRNAs (miRNAs) at the chr.14q32 locus are significantly downregulated in osteosarcoma compared to normal bone tissues. Bioinformatic predictions identified that a subset of 14q32 miRNAs (miR-382, miR-369-3p, miR-544 and miR-134) could potentially target cMYC transcript. The physical interaction between these 14q32 miRNAs and cMYC was validated using reporter assays. Further, restoring expression of these four 14q32 miRNAs decreased cMYC levels and induced apoptosis in Saos2 cells. We also show that exogenous expression of 14q32 miRNAs in Saos2 cells significantly downregulated miR-17-92, a transcriptional target of cMYC. The pro-apoptotic effect of 14q32 miRNAs in Saos2 cells was rescued either by overexpression of cMYC cDNA without the 3'UTR or with miR-17-92 cluster. Further, array comparative genomic hybridization studies showed no DNA copy number changes at 14q32 locus in OS patient samples suggesting that downregulation of 14q32 miRNAs are not due to deletion at this locus. Together, our data support a model where the deregulation of a network involving 14q32 miRNAs, cMYC and miR-17-92 miRNAs could contribute to osteosarcoma pathogenesis.
Collapse
Affiliation(s)
- Venugopal Thayanithy
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, MN 55455 USA
| | - Aaron L. Sarver
- Masonic Cancer Center, University of Minnesota, MN 55455 USA
| | - Reena V. Kartha
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, MN 55455 USA
| | - Li Lihua
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, MN 55455 USA
| | - Andrea Y. Angstadt
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, NC-27606, USA
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, NC-27606, USA
| | - Clifford J. Steer
- Masonic Cancer Center, University of Minnesota, MN 55455 USA
- Department of Medicine, University of Minnesota, MN 55455 USA
- Department of Genetics Cell Biology & Development, University of Minnesota, MN 55455 USA
| | - Jaime F. Modiano
- Masonic Cancer Center, University of Minnesota, MN 55455 USA
- Department of Veterinary Clinical Sciences, University of Minnesota, MN 55455 USA
| | - Subbaya Subramanian
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, MN 55455 USA
- Masonic Cancer Center, University of Minnesota, MN 55455 USA
- Manuscript correspondence to: Subbaya Subramanian, PhD, 11-212 Moos Tower, 515 Delaware Street S.E, Minneapolis, MN 55455, , Tel: 612-626-4330; Fax: 612-626-7031
| |
Collapse
|
13
|
Clausen DM, Guo J, Parise RA, Beumer JH, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. In vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of 10074-G5, a novel small-molecule inhibitor of c-Myc/Max dimerization. J Pharmacol Exp Ther 2010; 335:715-27. [PMID: 20801893 DOI: 10.1124/jpet.110.170555] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The c-Myc oncoprotein is overexpressed in many tumors and is essential for maintaining the proliferation of transformed cells. To function as a transcription factor, c-Myc must dimerize with Max via the basic helix-loop-helix leucine zipper protein (bHLH-ZIP) domains in each protein. The small molecule 7-nitro-N-(2-phenylphenyl)-2,1,3-benzoxadiazol-4-amine (10074-G5) binds to and distorts the bHLH-ZIP domain of c-Myc, thereby inhibiting c-Myc/Max heterodimer formation and inhibiting its transcriptional activity. We report in vitro cytotoxicity and in vivo efficacy, pharmacodynamics, pharmacokinetics, and metabolism of 10074-G5 in human xenograft-bearing mice. In vitro, 10074-G5 inhibited the growth of Daudi Burkitt's lymphoma cells and disrupted c-Myc/Max dimerization. 10074-G5 had no effect on the growth of Daudi xenografts in C.B-17 SCID mice that were treated with 20 mg/kg 10074-G5 intravenously for 5 consecutive days. Inhibition of c-Myc/Max dimerization in Daudi xenografts was not seen 2 or 24 h after treatment. Concentrations of 10074-G5 in various matrices were determined by high-performance liquid chromatography-UV, and metabolites of 10074-G5 were identified by liquid chromatography/tandem mass spectrometry. The plasma half-life of 10074-G5 in mice treated with 20 mg/kg i.v. was 37 min, and peak plasma concentration was 58 μM, which was 10-fold higher than peak tumor concentration. The lack of antitumor activity probably was caused by the rapid metabolism of 10074-G5 to inactive metabolites, resulting in tumor concentrations of 10074-G5 insufficient to inhibit c-Myc/Max dimerization. Our identification of 10074-G5 metabolites in mice will help design new, more metabolically stable small-molecule inhibitors of c-Myc.
Collapse
Affiliation(s)
- Dana M Clausen
- Molecular Therapeutics/Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
c-MYC is an important regulator of a wide array of cellular processes necessary for normal cell growth and differentiation, and its dysregulation is one of the hallmarks of many cancers. Consequently, understanding c-MYC transcriptional activation is critical for understanding developmental and cancer biology, as well as for the development of new anticancer drugs. The nuclease hypersensitive element (NHE) III(1) region of the c-MYC promoter has been shown to be particularly important in regulating c-MYC expression. Specifically, the formation of a G-quadruplex structure appears to promote repression of c-MYC transcription. This review focuses on what is known about the formation of a G-quadruplex in the NHE III(1) region of the c-MYC promoter, as well as on those factors that are known to modulate its formation. Last, we discuss the development of small molecules that stabilize or induce the formation of G-quadruplex structures and could potentially be used as anticancer agents.
Collapse
|
15
|
Shao GL, Wu AG, Jiao DC, Ji SF. Effect of c-myc antisense oligodeoxynucleotides on proliferation, apoptosis and chemosensitivity of colorectal cancer cell line HT-29. Shijie Huaren Xiaohua Zazhi 2009; 17:459-463. [DOI: 10.11569/wcjd.v17.i5.459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of c-myc antisense oligodeoxynucleotides (ASODN) on the proliferation, apoptosis and chemosensitivity to human colorectal cancer cell line HT-29 in vitro.
METHODS: c-myc ASODN and scrambled oligodeoxynueleotide (SODN) were transfected with lipofectamineTM 2000 into human colorectal cancer cell line HT-29. The expression of c-myc mRNA and protein in human colorectal cancer cell line HT-29 was detected by semi quantitive reverse transcription-polymerase chain reaction (RT-PCR) and Western blot, respectively. The effects of the c-myc ASODN on the proliferation and sensitivity to oxaliplatin of HT-29 cell line were studied by MTT assay and flow cytometry (FCM).
RESULTS: The expression of c-myc mRNA level in HT-29 was down-regulated remarkably between the antisense group and control group (0.464 ± 0.029 vs 0.974 ± 0.027, 0.945 ± 0.012, both P < 0.01). Protein expression of c-myc in HT-29 cells was confirmed and it was clear that specific band on PVDF membrane in ASODN group was significantly weakened than those in control groups; MTT and FCM showed the proliferation of HT-29 transfected with c-myc ASODN was retarded obviously in contrast to control groups after 48 hours' transfection (P < 0.05). Compared with the control groups after 72 hours' transfection, the percentage of the survival cells significantly decreased in the presence of both oxaliplatin and c-myc ASODNs (P < 0.05).
CONCLUSION: The c-myc ASODN can inhibit c-myc expression and inhibit the proliferation of colonic cancer HT-29 cells. Combination of c-myc ASODN and oxaliplatin might improve the treatment outcome of colorectal carcinoma. c-myc may be a potential target of gene therapy for human colorectal cancer.
Collapse
|
16
|
Lee SJ, Lee CS. Combined effect of protein kinase B inhibitor or extracellular signal-regulated kinase inhibitor against farnesyltransferase inhibition-induced apoptosis in SiHa cells. Naunyn Schmiedebergs Arch Pharmacol 2008; 379:291-303. [DOI: 10.1007/s00210-008-0359-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 09/25/2008] [Indexed: 01/08/2023]
|
17
|
Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as novel targets for cancer therapy (part III): transcription factors. ACTA ACUST UNITED AC 2005; 5:327-38. [PMID: 16196502 DOI: 10.2165/00129785-200505050-00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This is the third paper in a four-part serial review on potential therapeutic targeting of oncogenes. The previous parts described the involvement of oncogenes in different aspects of cancer growth and development, and considered the new technologies responsible for the advancement of oncogene identification, target validation, and drug design. Because of such advances, new specific and more efficient therapeutic agents can be developed for cancer. This part of the review continues the exploration of various oncogenes that we have grouped within seven categories: growth factors, tyrosine kinases, intermediate signaling molecules, transcription factors, cell cycle regulators, DNA damage repair genes, and genes involved in apoptosis. Part one discussed growth factors and tyrosine kinases and part two discussed intermediate signaling molecules. This portion of the review covers transcription factors and the various strategies being used to inhibit their expression or decrease their activities.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology and Division of Clinical Pharmacology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | |
Collapse
|
18
|
Russo P, Catassi A, Cesario A, Imperatori A, Rotolo N, Fini M, Granone P, Dominioni L. Molecular mechanisms of hexavalent chromium-induced apoptosis in human bronchoalveolar cells. Am J Respir Cell Mol Biol 2005; 33:589-600. [PMID: 16166740 PMCID: PMC2715333 DOI: 10.1165/rcmb.2005-0213oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hexavalent chromium (Cr[VI]) is classified by the International Agency for Research on Cancer as a group I carcinogen. Although the U.S. Occupational Safety and Health Administration was obliged to reduce the permissible exposure limit (PEL), it was reported that U.S. workers continue to be exposed to dangerously high Cr(VI) levels. In this study, we examined the role of p53 and target genes in a bronchoalveolar carcinoma isogenic cell line system and in primary human bronchial epithelial cells. p53-Negative parental H358 cell line, the same line in which the wild-type p53 expression vector (pC53-SN3) was introduced, and cells obtained from biopsies of human bronchus were exposed to chromate. Induction of DNA strand breaks were evaluated by alkaline elution assay, and apoptosis was analyzed by gel ladder, annexin V-PI staining, and ELISA, whereas p53 and target genes were evaluated by Western blots. Although Cr(VI) induced DNA strand breaks in both H358 cell clones, apoptosis was present only in the p53-transfected cells (H358p53(+/+)). In these cells, Cr(VI)-induced apoptosis is mediated by p53 upregulation of p53-upregulated modulator of apoptosis (PUMA), BAX translocation to mitochondria, cytochrome c release, and caspase-3 activation. In primary human bronchial epithelial cells expressing functional p53, Cr(VI) induced expression of PUMA and Noxa, which promote apoptosis through BAX. This result establishes p53 as the "necessary" player in Cr(VI)-induced apoptosis. To the best of our knowledge, this is the first report indicating strict correlation of Cr(VI) apoptosis to PUMA induction on primary human bronchoalveolar cells in short-term cultures.
Collapse
Affiliation(s)
- Patrizia Russo
- Department of Integrated Medical Oncology (DOMI), Laboratory of Translational Research B (Lung Cancer), National Cancer Institute, Largo Rosanna Benzi 10, I-16132 Genoa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Cesario A, Catassi A, Festi L, Imperatori A, Pericelli A, Galetta D, Margaritora S, Porziella V, Cardaci V, Granone P, Dominioni L, Russo P. Farnesyltransferase inhibitors and human malignant pleural mesothelioma: a first-step comparative translational study. Clin Cancer Res 2005; 11:2026-37. [PMID: 15756029 DOI: 10.1158/1078-0432.ccr-04-1450] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is known that the potential clinical use of farnesyltransferase inhibitors (FTI) could be expanded to include cancers harboring activated receptor tyrosine kinases. Approximately 70% of malignant pleural mesotheliomas (MPM) overexpress epidermal growth factor receptors (EGFR) and a subset express both EGFR and transforming growth factor alpha (TGF-alpha), suggesting an autocrine role for EGFR in MPM. We checked on MPM cells (10 human cell lines, 11 primary cultures obtained by human biopsies, and 7 short-term normal mesothelial cell cultures) concerning the following: (a) the relative overexpression of EGFR (Western blotting, flow cytometry, immunohistochemistry), (b) the relative expression of EGFR ligands (EGF, amphiregulin, TGF-alpha, ELISA), (c) the relative increase of the activated form of Ras (Ras-bound GTP) after EGF stimulation (Ras activation assay), (d) the efficacy of five different FTIs (HDJ2 prenylation, cell cytotoxicity, and apoptosis using ApopTag and gel ladder). EGFR was overexpressed in MPM cells compared with normal pleural mesothelial cells in equivalent levels as in non-small cell lung cancer cells A459. MPM cells constitutively expressed EGFR ligands; however, Ras activation was attenuated at high EGF concentrations (100 ng/mL). Growth of MPM cells was substantially not affected by treatment with different FTIs (SCH66336, BMS-214662, R115777, RPR-115135, and Manumycin). Among these, BMS-214662 was the only one moderately active. BMS-214662 triggered apoptosis in a small fraction of cells (not higher than 30%) that was paralleled by a slight decrease in the levels of TGF-alpha secreted by treated MPM cells. Our data highlighted the concept that the same signaling pathway can be regulated in different ways and these regulations can differ between different cells of different origin.
Collapse
Affiliation(s)
- Alfredo Cesario
- Department of Surgical Science, Division of General Thoracic Surgery, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Winquist E, Moore MJ, Chi KN, Ernst DS, Hirte H, North S, Powers J, Walsh W, Boucher T, Patton R, Seymour L. A multinomial Phase II study of lonafarnib (SCH 66336) in patients with refractory urothelial cancer☆. Urol Oncol 2005; 23:143-9. [PMID: 15907712 DOI: 10.1016/j.urolonc.2004.12.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2003] [Revised: 11/12/2004] [Accepted: 11/15/2004] [Indexed: 11/28/2022]
Abstract
PURPOSE Protein farnesylation by farnesyltransferase (FTase) is required for membrane localization and effective signal transduction by G-proteins, including Ras. Lonafarnib inhibits FTase and has shown antitumor activity in both preclinical and clinical settings. As disturbances in Ras signaling pathways have been implicated in the pathogenesis of transitional cell carcinoma (TCC), the antitumor activity of lonafarnib was studied in a National Cancer Institute of Canada Clinical Trials Group Phase II trial in patients with previously treated TCC. PATIENTS AND METHODS Patients had at least 1 prior chemotherapy regimen for advanced unresectable or metastatic TCC, or recurrence less than 1 year after adjuvant or neoadjuvant chemotherapy. Lonafarnib was given at a dose of 200 mg PO twice daily continuously, with cycles repeated every 4 weeks. RESULTS Between December 1999 and December 2000, 19 eligible patients were enrolled at 8 National Cancer Institute of Canada Clinical Trials Group centers. Median time on treatment was 7.1 weeks (range, 0.6-23.9). Drug-related Grade 3 toxicities included fatigue, anorexia, nausea, confusion, dehydration, muscle weakness, depression, headache, and dyspnea. Five patients discontinued the study protocol due to toxicity. No responses were observed in 10 patients who were evaluable. Of 9 patients not evaluable for response, 5 had symptomatic progression, fulfilling multinomial criteria to stop the study after the first stage. CONCLUSION No single-agent activity of lonafarnib was observed in this study of patients with aggressive TCC failing prior chemotherapy.
Collapse
Affiliation(s)
- Eric Winquist
- London Regional Cancer Centre, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Trombino S, Cesario A, Margaritora S, Granone P, Motta G, Falugi C, Russo P. Alpha7-nicotinic acetylcholine receptors affect growth regulation of human mesothelioma cells: role of mitogen-activated protein kinase pathway. Cancer Res 2004; 64:135-45. [PMID: 14729617 DOI: 10.1158/0008-5472.can-03-1672] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study presents data suggesting that both human mesothelioma (cell lines and human mesothelioma biopsies) and human normal mesothelial cells express receptors for acetylcholine and that stimulation of these receptors by nicotine prompted cell growth via activation of nicotinic cholinergic receptors. Thus, these data demonstrate that: (a) human mesothelioma cells and human biopsies of mesothelioma as well as of normal pleural mesothelial cells express functionally alpha-7 nicotinic acethlycholine receptors, evaluated by alpha-bungarotoxin-FITC binding, receptor binding assay, Western blot, and reverse transcription-PCR; (b) choline acetyltransferase immunostaining is present in mesothelioma cells; (c) mesothelioma cell growth is modulated by the cholinergic system in which agonists (i.e., nicotine) has a proliferative effect, and antagonists (i.e., curare) has an inhibitory effect, evaluated by cell cloning, DNA synthesis and cell cycle; (d) nicotine induces Ca(+2) influx, evaluated by [(45)Ca(2+)] uptake, and consequently activation of mitogen-activated protein kinase pathway (extracellular signal-regulated kinase and p90(RSK) phosphorylation), evaluated by Western blot; and (e) apoptosis mechanisms in mesothelioma cells are under the control of the cholinergic system (nicotine antiapoptotic via induction of nuclear factor-kappaB complexes and phosphorylation of Bad at Ser(112); curare proapoptotic via G(0)-G(1) arrest p21(waf-1) dependent but p53 independent). The involvement of the nonneuronal cholinergic system in mesothelioma appears reasonable and open up new therapeutic strategies.
Collapse
Affiliation(s)
- Sonya Trombino
- Department of Biology, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Bellmeyer A, Krase J, Lindgren J, LaBonne C. The protooncogene c-myc is an essential regulator of neural crest formation in xenopus. Dev Cell 2003; 4:827-39. [PMID: 12791268 DOI: 10.1016/s1534-5807(03)00160-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The neural crest, a population of multipotent progenitor cells, is a defining feature of vertebrate embryos. Neural crest precursor cells arise at the neural plate border in response to inductive signals, but much remains to be learned about the molecular mechanisms underlying their induction. Here we show that the protooncogene c-Myc is an essential early regulator of neural crest cell formation in Xenopus. c-myc is localized at the neural plate border prior to the expression of early neural crest markers, such as slug. A morpholino-mediated "knockdown" of c-Myc protein results in the absence of neural crest precursor cells and a resultant loss of neural crest derivatives. These effects are not dependent upon changes in cell proliferation or cell death. Instead, our findings reveal an important and unexpected role for c-Myc in the specification of cell fates in the early ectoderm.
Collapse
Affiliation(s)
- Amy Bellmeyer
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | | | | | | |
Collapse
|