51
|
A high-throughput screen identifies that CDK7 activates glucose consumption in lung cancer cells. Nat Commun 2019; 10:5444. [PMID: 31784510 PMCID: PMC6884612 DOI: 10.1038/s41467-019-13334-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Elevated glucose consumption is fundamental to cancer, but selectively targeting this pathway is challenging. We develop a high-throughput assay for measuring glucose consumption and use it to screen non-small-cell lung cancer cell lines against bioactive small molecules. We identify Milciclib that blocks glucose consumption in H460 and H1975, but not in HCC827 or A549 cells, by decreasing SLC2A1 (GLUT1) mRNA and protein levels and by inhibiting glucose transport. Milciclib blocks glucose consumption by targeting cyclin-dependent kinase 7 (CDK7) similar to other CDK7 inhibitors including THZ1 and LDC4297. Enhanced PIK3CA signaling leads to CDK7 phosphorylation, which promotes RNA Polymerase II phosphorylation and transcription. Milciclib, THZ1, and LDC4297 lead to a reduction in RNA Polymerase II phosphorylation on the SLC2A1 promoter. These data indicate that our high-throughput assay can identify compounds that regulate glucose consumption and that CDK7 is a key regulator of glucose consumption in cells with an activated PI3K pathway. Many cancer cells have increased glucose consumption compared to normal cells, a feature that can be exploited therapeutically. Here, the authors carry out a chemical screen and identify compounds that selectively blocks glucose metabolism in non-small-cell lung cancer cell lines.
Collapse
|
52
|
Ceballos J, Schwalfenberg M, Karageorgis G, Reckzeh ES, Sievers S, Ostermann C, Pahl A, Sellstedt M, Nowacki J, Carnero Corrales MA, Wilke J, Laraia L, Tschapalda K, Metz M, Sehr DA, Brand S, Winklhofer K, Janning P, Ziegler S, Waldmann H. Synthesis of Indomorphan Pseudo-Natural Product Inhibitors of Glucose Transporters GLUT-1 and -3. Angew Chem Int Ed Engl 2019; 58:17016-17025. [PMID: 31469221 PMCID: PMC6900016 DOI: 10.1002/anie.201909518] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/12/2019] [Indexed: 12/22/2022]
Abstract
Bioactive compound design based on natural product (NP) structure may be limited because of partial coverage of NP-like chemical space and biological target space. These limitations can be overcome by combining NP-centered strategies with fragment-based compound design through combination of NP-derived fragments to afford structurally unprecedented "pseudo-natural products" (pseudo-NPs). The design, synthesis, and biological evaluation of a collection of indomorphan pseudo-NPs that combine biosynthetically unrelated indole- and morphan-alkaloid fragments are described. Indomorphane derivative Glupin was identified as a potent inhibitor of glucose uptake by selectively targeting and upregulating glucose transporters GLUT-1 and GLUT-3. Glupin suppresses glycolysis, reduces the levels of glucose-derived metabolites, and attenuates the growth of various cancer cell lines. Our findings underscore the importance of dual GLUT-1 and GLUT-3 inhibition to efficiently suppress tumor cell growth and the cellular rescue mechanism, which counteracts glucose scarcity.
Collapse
Affiliation(s)
- Javier Ceballos
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: Laboratory of Catalysis and Organic SynthesisEPFL SB ISIC LCSO, BCH 42211015LausanneSwitzerland
| | - Melanie Schwalfenberg
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - George Karageorgis
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
| | - Elena S. Reckzeh
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| | - Sonja Sievers
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Claude Ostermann
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Axel Pahl
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Magnus Sellstedt
- Department of ChemistryUmeå University901 87UmeåSweden
- Current address: Clinical Chemistry, Laboratory MedicineUniversity Hospital of Umeå901 85UmeåSweden
| | - Jessica Nowacki
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Marjorie A. Carnero Corrales
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Julian Wilke
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| | - Luca Laraia
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: Department of ChemistryTechnical University of DenmarkKemitorvet, Bygning 2072800Kgs LyngbyDenmark
| | - Kirsten Tschapalda
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Malte Metz
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Dominik A. Sehr
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum44801BochumGermany
| | - Silke Brand
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Konstanze Winklhofer
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum44801BochumGermany
| | - Petra Janning
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Slava Ziegler
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Herbert Waldmann
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| |
Collapse
|
53
|
Ceballos J, Schwalfenberg M, Karageorgis G, Reckzeh ES, Sievers S, Ostermann C, Pahl A, Sellstedt M, Nowacki J, Carnero Corrales MA, Wilke J, Laraia L, Tschapalda K, Metz M, Sehr DA, Brand S, Winklhofer K, Janning P, Ziegler S, Waldmann H. Synthesis of Indomorphan Pseudo‐Natural Product Inhibitors of Glucose Transporters GLUT‐1 and ‐3. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201909518] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Javier Ceballos
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: Laboratory of Catalysis and Organic SynthesisEPFL SB ISIC LCSO, BCH 4221 1015 Lausanne Switzerland
| | - Melanie Schwalfenberg
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - George Karageorgis
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: School of ChemistryUniversity of Leeds Leeds LS2 9JT UK
| | - Elena S. Reckzeh
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Claude Ostermann
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Axel Pahl
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Magnus Sellstedt
- Department of ChemistryUmeå University 901 87 Umeå Sweden
- Current address: Clinical Chemistry, Laboratory MedicineUniversity Hospital of Umeå 901 85 Umeå Sweden
| | - Jessica Nowacki
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Marjorie A. Carnero Corrales
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Julian Wilke
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Luca Laraia
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: Department of ChemistryTechnical University of Denmark Kemitorvet, Bygning 207 2800 Kgs Lyngby Denmark
| | - Kirsten Tschapalda
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Malte Metz
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Dominik A. Sehr
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum 44801 Bochum Germany
| | - Silke Brand
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Konstanze Winklhofer
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum 44801 Bochum Germany
| | - Petra Janning
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Slava Ziegler
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Herbert Waldmann
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| |
Collapse
|
54
|
Reckzeh ES, Karageorgis G, Schwalfenberg M, Ceballos J, Nowacki J, Stroet MC, Binici A, Knauer L, Brand S, Choidas A, Strohmann C, Ziegler S, Waldmann H. Inhibition of Glucose Transporters and Glutaminase Synergistically Impairs Tumor Cell Growth. Cell Chem Biol 2019; 26:1214-1228.e25. [DOI: 10.1016/j.chembiol.2019.06.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/31/2019] [Accepted: 06/17/2019] [Indexed: 12/18/2022]
|
55
|
Sharma A, Boise LH, Shanmugam M. Cancer Metabolism and the Evasion of Apoptotic Cell Death. Cancers (Basel) 2019; 11:E1144. [PMID: 31405035 PMCID: PMC6721599 DOI: 10.3390/cancers11081144] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/29/2019] [Accepted: 08/08/2019] [Indexed: 12/19/2022] Open
Abstract
Cellular growth and proliferation depend upon the acquisition and synthesis of specific metabolites. These metabolites fuel the bioenergy, biosynthesis, and redox potential required for duplication of cellular biomass. Multicellular organisms maintain tissue homeostasis by balancing signals promoting proliferation and removal of cells via apoptosis. While apoptosis is in itself an energy dependent process activated by intrinsic and extrinsic signals, whether specific nutrient acquisition (elevated or suppressed) and their metabolism regulates apoptosis is less well investigated. Normal cellular metabolism is regulated by lineage specific intrinsic features and microenvironment driven extrinsic features. In the context of cancer, genetic abnormalities, unconventional microenvironments and/or therapy engage constitutive pro-survival signaling to re-program and rewire metabolism to maintain survival, growth, and proliferation. It thus becomes particularly relevant to understand whether altered nutrient acquisition and metabolism in cancer can also contribute to the evasion of apoptosis and consequently therapy resistance. Our review attempts to dissect a causal relationship between two cancer hallmarks, i.e., deregulated cellular energetics and the evasion of programmed cell death with primary focus on the intrinsic pathway of apoptosis.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
56
|
Graña-López L, Herranz M, Domínguez-Prado I, Argibay S, Villares Á, Vázquez-Caruncho M. Can dedicated breast PET help to reduce overdiagnosis and overtreatment by differentiating between indolent and potentially aggressive ductal carcinoma in situ? Eur Radiol 2019; 30:514-522. [DOI: 10.1007/s00330-019-06356-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/29/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
|
57
|
Mauceri A, Giansanti L, Bozzuto G, Condello M, Molinari A, Galantini L, Piozzi A, Mancini G. Structurally related glucosylated liposomes: Correlation of physicochemical and biological features. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1468-1475. [DOI: 10.1016/j.bbamem.2019.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 10/26/2022]
|
58
|
Abstract
18-Fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (FDG PET/CT) is currently the criterion standard of lymphoma imaging and recommended through all stages of Hodgkin lymphoma management. Accurate staging is important for risk stratification and initial choice of therapy and also for the planning of postchemoradiotherapy. 18-Fluoro-2-deoxy-D-glucose PET/CT frequently leads to upstaging and potentially a more intensive treatment. Visual-only assessment of staging and interim scans is being accompanied by quantitative and semiquantitative methods to measure metabolic tumor volume, total lesion glycolysis, and so on. It is still unclear if these methods significantly improve the value of FDG PET/CT by visual assessment only. Because of the good prognostic value of FDG PET/CT, a large number of studies have used interim FDG PET to tailor treatment to the individual patients, according to their early metabolic response rather than according to their pretreatment prognostic features. 18-Fluoro-2-deoxy-D-glucose PET/CT is standard of care for posttreatment response assessment but has no place in routine follow-up of Hodgkin lymphoma patients in remission.
Collapse
|
59
|
Zambrano A, Molt M, Uribe E, Salas M. Glut 1 in Cancer Cells and the Inhibitory Action of Resveratrol as A Potential Therapeutic Strategy. Int J Mol Sci 2019; 20:ijms20133374. [PMID: 31324056 PMCID: PMC6651361 DOI: 10.3390/ijms20133374] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022] Open
Abstract
An important hallmark in cancer cells is the increase in glucose uptake. GLUT1 is an important target in cancer treatment because cancer cells upregulate GLUT1, a membrane protein that facilitates the basal uptake of glucose in most cell types, to ensure the flux of sugar into metabolic pathways. The dysregulation of GLUT1 is associated with numerous disorders, including cancer and metabolic diseases. There are natural products emerging as a source for inhibitors of glucose uptake, and resveratrol is a molecule of natural origin with many properties that acts as antioxidant and antiproliferative in malignant cells. In the present review, we discuss how GLUT1 is involved in the general scheme of cancer cell metabolism, the mechanism of glucose transport, and the importance of GLUT1 structure to understand the inhibition process. Then, we review the current state-of-the-art of resveratrol and other natural products as GLUT1 inhibitors, focusing on those directed at treating different types of cancer. Targeting GLUT1 activity is a promising strategy for the development of drugs aimed at treating neoplastic growth.
Collapse
Affiliation(s)
- Angara Zambrano
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Concepción, Concepción 4070386, Chile
| | - Matías Molt
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile
| | - Elena Uribe
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Concepción, Concepción 4070386, Chile
| | - Mónica Salas
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile.
| |
Collapse
|
60
|
Koutsioumpa M, Hatziapostolou M, Polytarchou C, Tolosa EJ, Almada LL, Mahurkar-Joshi S, Williams J, Tirado-Rodriguez B, Huerta-Yepez S, Karavias D, Kourea H, Poultsides GA, Struhl K, Dawson DW, Donahue TR, Fernandez-Zapico ME, lliopoulos D. Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming. Gut 2019; 68:1271-1286. [PMID: 30337373 PMCID: PMC6697184 DOI: 10.1136/gutjnl-2017-315690] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 09/06/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Despite advances in the identification of epigenetic alterations in pancreatic cancer, their biological roles in the pathobiology of this dismal neoplasm remain elusive. Here, we aimed to characterise the functional significance of histone lysine methyltransferases (KMTs) and demethylases (KDMs) in pancreatic tumourigenesis. DESIGN DNA methylation sequencing and gene expression microarrays were employed to investigate CpG methylation and expression patterns of KMTs and KDMs in pancreatic cancer tissues versus normal tissues. Gene expression was assessed in five cohorts of patients by reverse transcription quantitative-PCR. Molecular analysis and functional assays were conducted in genetically modified cell lines. Cellular metabolic rates were measured using an XF24-3 Analyzer, while quantitative evaluation of lipids was performed by liquid chromatography-mass spectrometry (LC-MS) analysis. Subcutaneous xenograft mouse models were used to evaluate pancreatic tumour growth in vivo. RESULTS We define a new antitumorous function of the histone lysine (K)-specific methyltransferase 2D (KMT2D) in pancreatic cancer. KMT2D is transcriptionally repressed in human pancreatic tumours through DNA methylation. Clinically, lower levels of this methyltransferase associate with poor prognosis and significant weight alterations. RNAi-based genetic inactivation of KMT2D promotes tumour growth and results in loss of H3K4me3 mark. In addition, KMT2D inhibition increases aerobic glycolysis and alters the lipidomic profiles of pancreatic cancer cells. Further analysis of this phenomenon identified the glucose transporter SLC2A3 as a mediator of KMT2D-induced changes in cellular, metabolic and proliferative rates. CONCLUSION Together our findings define a new tumour suppressor function of KMT2D through the regulation of glucose/fatty acid metabolism in pancreatic cancer.
Collapse
Affiliation(s)
- Marina Koutsioumpa
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| | - Maria Hatziapostolou
- Biological Sciences, University of Southampton,
Southampton, United Kingdom;,Biosciences, School of Science and Technology, Nottingham
Trent University, Nottingham, United Kingdom
| | - Christos Polytarchou
- Interdisciplinary Biomedical Research Centre, School of
Science and Technology, Nottingham Trent University, Nottingham, United
Kingdom
| | - Ezequiel J. Tolosa
- Schulze Center for Novel Therapeutics, Division of Oncology
Research, Mayo Clinic, Rochester, MN
| | - Luciana L. Almada
- Schulze Center for Novel Therapeutics, Division of Oncology
Research, Mayo Clinic, Rochester, MN
| | - Swapna Mahurkar-Joshi
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| | - Jennifer Williams
- Department of Surgery, Division of General Surgery, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | - Belen Tirado-Rodriguez
- Unidad de Investigacion en Enfermedades Oncologicas,
Hospital Infantil de Mexico, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigacion en Enfermedades Oncologicas,
Hospital Infantil de Mexico, Mexico City, Mexico
| | - Dimitrios Karavias
- Department of Pathology, School of Medicine, University of
Patras, Patras, Greece
| | - Helen Kourea
- Department of Pathology, School of Medicine, University of
Patras, Patras, Greece
| | | | - Kevin Struhl
- Department of Biological Chemistry and Molecular
Pharmacology, Harvard Medical School, Boston, MA
| | - David W. Dawson
- Department of Pathology and Laboratory Medicine, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | - Timothy R. Donahue
- Department of Surgery, Division of General Surgery, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | | | - Dimitrios lliopoulos
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| |
Collapse
|
61
|
Kearns FL, Robart C, Kemp MT, Vankayala SL, Chapin BM, Anslyn EV, Woodcock HL, Larkin JD. Modeling Boronic Acid Based Fluorescent Saccharide Sensors: Computational Investigation of d-Fructose Binding to Dimethylaminomethylphenylboronic Acid. J Chem Inf Model 2019; 59:2150-2158. [PMID: 30908030 PMCID: PMC8577280 DOI: 10.1021/acs.jcim.8b00987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Designing organic saccharide sensors for use in aqueous solution is a nontrivial endeavor. Incorporation of hydrogen bonding groups on a sensor's receptor unit to target saccharides is an obvious strategy but not one that is likely to ensure analyte-receptor interactions over analyte-solvent or receptor-solvent interactions. Phenylboronic acids are known to reversibly and covalently bind saccharides (diols in general) with highly selective affinity in aqueous solution. Therefore, recent work has sought to design such sensors and understand their mechanism for allowing fluorescence with bound saccharides. In past work, binding orientations of several saccharides were determined to dimethylaminomethylphenylboronic acid (DMPBA) receptors with an anthracene fluorophore; however, the binding orientation of d-fructose to such a sensor could not be determined. In this work, we investigate the potential binding modes by generating 20 possible bidentate and six possible tridentate modes between fructose and DMPBA, a simplified receptor model. Gas phase and implicit solvent geometry optimizations, with a myriad functional/basis set pairs, were carried out to identify the lowest energy bidentate and tridentate binding modes of d-fructose to DMPBA. An interesting hydrogen transfer was observed during selected bidentate gas phase optimizations; this transfer suggests a strong sharing of the hydrogen atom between the boronate hydroxyl and amine nitrogen.
Collapse
Affiliation(s)
- Fiona L. Kearns
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | - Carrie Robart
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | - M. Trent Kemp
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | - Sai Lakshmana Vankayala
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | - Brette M. Chapin
- Department of Chemistry, Durham University, South Road Durham, Durham DH1 3LE, United Kingdom
| | - Eric V. Anslyn
- Department of Chemistry, The University of Texas at Austin, 100 East 24th Street, Norman Hackerman Building, Austin, Texas 78712, United States
| | - H. Lee Woodcock
- Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE205, Tampa, Florida 33620-5250, United States
| | - Joseph D. Larkin
- Department of Chemistry, Eckerd College, 4200 54th Avenue South, St. Petersburg, Florida 33711, United States
| |
Collapse
|
62
|
Gründker C, Wokoun U, Hellriegel M, Emons G. Inhibition of aerobic glycolysis enhances the anti-tumor efficacy of Zoptarelin Doxorubicin in triple-negative breast cancer cells. J Obstet Gynaecol Res 2019; 45:1334-1342. [PMID: 31016845 DOI: 10.1111/jog.13980] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/06/2019] [Indexed: 11/28/2022]
Abstract
AIM A characteristic of cancer cells including triple-negative breast cancers (TNBC) is an increased aerobic glycolysis for ATP production representing a selective therapeutic target. More than 70% of TNBC express gonadotropin-releasing hormone receptors (GnRH-R). These receptors can be used for targeted chemotherapy with cytotoxic GnRH agonists such as Zoptarelin Doxorubicin, in which doxorubicin is covalently linked to [D-Lys6 ]GnRH. In this study, we have analyzed whether inhibition of aerobic glycolysis can enhance the antitumor efficacy of GnRH-R-targeted chemotherapy using Zoptarelin Doxorubicin. METHODS Triple-negative breast cancers cell lines MDA-MB-231 and HCC1806 were treated with Zoptarelin Doxorubicin, glycolysis inhibitor 2-deoxy-D-glucose (2DG) or the combination of both agents. Cell viability was measured using Alamar blue. Induction of apoptosis was quantified by measurement of loss of mitochondrial membrane potential. In vivo experiments were performed using nude mice bearing xenografted MDA-MB-231 tumors. RESULTS Treatment of TNBC cells with Zoptarelin Doxorubicin or with 2DG resulted in a significant decrease of cell viability and a significant increase of apoptosis. Treatment with Zoptarelin Doxorubicin in combination with 2DG resulted in significantly reduced viability and enhanced apoptosis compared with single-agent treatments. Combinational index (CI) analysis revealed the co-treatment effect as a synergistic. The antitumor effects of Zoptarelin Doxorubicin or 2DG were confirmed in nude mice. The tumor reducing effects of Zoptarelin Doxorubicin were enhanced by combination with 2DG. CONCLUSION The glycolytic phenotype of TNBC can be used to improve antitumor therapies. Co-treatment of Zoptarelin Doxorubicin with glycolysis inhibitor 2DG might be a suitable therapeutic option for GnRH receptor-positive TNBC.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | - Ulrike Wokoun
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | - Martin Hellriegel
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| |
Collapse
|
63
|
Chen LY, Phelix CF. Extracellular gating of glucose transport through GLUT 1. Biochem Biophys Res Commun 2019; 511:573-578. [PMID: 30824189 PMCID: PMC6452493 DOI: 10.1016/j.bbrc.2019.02.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 11/18/2022]
Abstract
The ubiquitous glucose transporter 1 (GLUT1) is physiologically and pathologically relevant in energy metabolism of the CNS, skeletal muscles, cancer cells etc. Extensive experiments on GLUT1 produced thorough understandings of its expressions, functions, and structures which were recently resolved to atomic accuracy. However, theoretical understandings are still controversial about how GLUT1 facilitates glucose diffusion across the cell membrane. Molecular dynamics (MD) simulations of the current literature have GLUT1 embedded in a symmetric bilayer of a single lipid type. They provide atomistic illustrations of the alternating access theory (AAT), but the simulation results are inconsistent with the undisputed experimental data of kinetics showing rapid transport of glucose at near-physiological temperatures, high Arrhenius activation barrier in zero-trans uptake, and large trans-acceleration at sub-physiological temperatures. In this research, we embedded GLUT1 in an asymmetric bilayer of multiple lipids to better mimic the erythrocyte membrane. We ran unbiased MD simulations at 37 °C and at 5 °C and found a new mechanism of glucose transport via GLUT1: The extracellular (EC) gate opened wide for EC glucopyranose at 37 °C and, only in the presence of intracellular (IC) glucose, at 5 °C. In the absence of IC glucose at 5 °C, the EC gate opened narrowly for acyclic glucose, gating out glucopyranose. This EC-gating mechanism is simpler than AAT and yet it well explains for the rapid glucose transport at near-physiological temperatures and large trans-acceleration at sub-physiological temperatures. It also explains why zero-trans uptake (involving the pyranose-to-aldehyde transformation) has an Arrhenius barrier ∼20 kcal/mol higher than the equilibrium exchange transport.
Collapse
Affiliation(s)
- Liao Y Chen
- Department of Physics, University of Texas at San Antonio, San Antonio, TX, 78249, USA.
| | - Clyde F Phelix
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| |
Collapse
|
64
|
Aberrant Metabolism in Hepatocellular Carcinoma Provides Diagnostic and Therapeutic Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7512159. [PMID: 30524660 PMCID: PMC6247426 DOI: 10.1155/2018/7512159] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/03/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) accounts for over 80% of liver cancer cases and is highly malignant, recurrent, drug-resistant, and often diagnosed in the advanced stage. It is clear that early diagnosis and a better understanding of molecular mechanisms contributing to HCC progression is clinically urgent. Metabolic alterations clearly characterize HCC tumors. Numerous clinical parameters currently used to assess liver functions reflect changes in both enzyme activity and metabolites. Indeed, differences in glucose and acetate utilization are used as a valid clinical tool for stratifying patients with HCC. Moreover, increased serum lactate can distinguish HCC from normal subjects, and serum lactate dehydrogenase is used as a prognostic indicator for HCC patients under therapy. Currently, the emerging field of metabolomics that allows metabolite analysis in biological fluids is a powerful method for discovering new biomarkers. Several metabolic targets have been identified by metabolomics approaches, and these could be used as biomarkers in HCC. Moreover, the integration of different omics approaches could provide useful information on the metabolic pathways at the systems level. In this review, we provided an overview of the metabolic characteristics of HCC considering also the reciprocal influences between the metabolism of cancer cells and their microenvironment. Moreover, we also highlighted the interaction between hepatic metabolite production and their serum revelations through metabolomics researches.
Collapse
|
65
|
Zaal EA, Berkers CR. The Influence of Metabolism on Drug Response in Cancer. Front Oncol 2018; 8:500. [PMID: 30456204 PMCID: PMC6230982 DOI: 10.3389/fonc.2018.00500] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
Resistance to therapeutic agents, either intrinsic or acquired, is currently a major problem in the treatment of cancers and occurs in virtually every type of anti-cancer therapy. Therefore, understanding how resistance can be prevented, targeted and predicted becomes increasingly important to improve cancer therapy. In the last decade, it has become apparent that alterations in cellular metabolism are a hallmark of cancer cells and that a rewired metabolism is essential for rapid tumor growth and proliferation. Recently, metabolic alterations have been shown to play a role in the sensitivity of cancer cells to widely-used first-line chemotherapeutics. This suggests that metabolic pathways are important mediators of resistance toward anticancer agents. In this review, we highlight the metabolic alterations associated with resistance toward different anticancer agents and discuss how metabolism may be exploited to overcome drug resistance to classical chemotherapy.
Collapse
Affiliation(s)
- Esther A. Zaal
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Celia R. Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
66
|
Itoh T, Tamura K, Ueda H, Tanaka T, Sato K, Kuroda R, Aoki S. Design and synthesis of boron containing monosaccharides by the hydroboration of d-glucal for use in boron neutron capture therapy (BNCT). Bioorg Med Chem 2018; 26:5922-5933. [PMID: 30420329 DOI: 10.1016/j.bmc.2018.10.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 01/18/2023]
Abstract
Boron neutron capture therapy (BNCT) is one of the radiotherapies that involves the use of boron-containing compounds for the treatment of cancer. Boron-10 (10B) containing compounds that can accumulate in tumor tissue are expected to be suitable agents for BNCT. We report herein on the design and synthesis of some new BNCT agents based on a d-glucose scaffold, since glycoconjugation has been recognized as a useful strategy for the specific targeting of tumors. To introduce a boryl group into a d-glucose scaffold, we focused on the hydroboration of d-glucal derivatives, which have a double bond between the C1 and C2 positions. It was hypothesized that a C-B bond could be introduced at the C2 position of d-glucose by the hydroboration of d-glucal derivatives and that the products could be stabilized by conversion to the corresponding boronic acid ester. To test this hypothesis, we prepared some 2-boryl-1,2-dideoxy-d-glucose derivatives as boron carriers and evaluated their cytotoxicity and cellular uptake activity to cancer cells, especially under hypoxic conditions.
Collapse
Affiliation(s)
- Taiki Itoh
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kei Tamura
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hiroki Ueda
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Tomohiro Tanaka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kyouhei Sato
- Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Reiko Kuroda
- Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Division of Medical-Science-Engineering Cooperation, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Imaging Frontier Center, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
67
|
Eskian M, Alavi A, Khorasanizadeh M, Viglianti BL, Jacobsson H, Barwick TD, Meysamie A, Yi SK, Iwano S, Bybel B, Caobelli F, Lococo F, Gea J, Sancho-Muñoz A, Schildt J, Tatcı E, Lapa C, Keramida G, Peters M, Boktor RR, John J, Pitman AG, Mazurek T, Rezaei N. Effect of blood glucose level on standardized uptake value (SUV) in 18F- FDG PET-scan: a systematic review and meta-analysis of 20,807 individual SUV measurements. Eur J Nucl Med Mol Imaging 2018; 46:224-237. [DOI: 10.1007/s00259-018-4194-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/10/2018] [Indexed: 01/24/2023]
|
68
|
Suh S, Kim YH, Goh TS, Jeong DC, Lee CS, Jang JY, Cha W, Han ME, Kim SJ, Kim IJ, Pak K. mRNA Expression of SLC5A5 and SLC2A Family Genes in Papillary Thyroid Cancer: An Analysis of The Cancer Genome Atlas. Yonsei Med J 2018; 59:746-753. [PMID: 29978611 PMCID: PMC6037592 DOI: 10.3349/ymj.2018.59.6.746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/29/2018] [Accepted: 06/15/2018] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The present study investigated the dynamics and prognostic role of messenger RNA (mRNA) expression responsible for ¹⁸F-fluorodeoxyglucose (FDG) uptake in FDG positron emission tomography (PET) and radioactive iodine (¹³¹I) uptake in whole-body radioactive iodine scans (WBS) in papillary thyroid cancer (PTC) patients. MATERIALS AND METHODS The primary and processed data were downloaded from the Genomic Data Commons Data Portal. Expression data for sodium/iodide symporter (solute carrier family 5 member 5, SLC5A5), hexokinase (HK1-3), glucose-6-phosphate dehydrogenase (G6PD), and glucose transporter (solute carrier family 2, SLC2A1-4) mRNA were collected. RESULTS Expression of SLC5A5 mRNA were negatively correlated with SLC2A1 mRNA and positively correlated with SLC2A4 mRNA. In PTC with BRAF mutations, expressions of SLC2A1, SLC2A3, HK2, and HK3 mRNA were higher than those in PTC without BRAF mutations. Expression of SLC5A5, SLC2A4, HK1, and G6PD mRNA was lower in PTC without BRAF mutation. PTCs with higher expression of SLC5A5 mRNA had more favorable disease-free survival, but no association with overall survival. CONCLUSION Expression of SLC5A5 mRNA was negatively correlated with SLC2A1 mRNA. This finding provides a molecular basis for the management of PTC with negative WBS using ¹⁸F-FDG PET scans. In addition, higher expression of SLC5A5 mRNA was associated with less PTC recurrence, but not with deaths.
Collapse
Affiliation(s)
- Sunghwan Suh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - Tae Sik Goh
- Department of Orthopaedic Surgery and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.
| | | | - Chi Seung Lee
- Biomedical Research Institute, Pusan National University Hospital and School of Medicine, Pusan National University, Busan, Korea
| | - Jeon Yeob Jang
- Department of Otorhinolaryngology-Head and Neck Surgery, Ajou University Hospital, Suwon, Korea
| | - Wonjae Cha
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University Hospital, Busan, Korea
| | - Myoung Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - Seong Jang Kim
- Department of Nuclear Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - In Joo Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.
| |
Collapse
|
69
|
Mechanistic insights into GLUT1 activation and clustering revealed by super-resolution imaging. Proc Natl Acad Sci U S A 2018; 115:7033-7038. [PMID: 29915035 DOI: 10.1073/pnas.1803859115] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The glucose transporter GLUT1, a plasma membrane protein that mediates glucose homeostasis in mammalian cells, is responsible for constitutive uptake of glucose into many tissues and organs. Many studies have focused on its vital physiological functions and close relationship with diseases. However, the molecular mechanisms of its activation and transport are not clear, and its detailed distribution pattern on cell membranes also remains unknown. To address these, we first investigated the distribution and assembly of GLUT1 at a nanometer resolution by super-resolution imaging. On HeLa cell membranes, the transporter formed clusters with an average diameter of ∼250 nm, the majority of which were regulated by lipid rafts, as well as being restricted in size by both the cytoskeleton and glycosylation. More importantly, we found that the activation of GLUT1 by azide or MβCD did not increase its membrane expression but induced the decrease of the large clusters. The results suggested that sporadic distribution of GLUT1 may facilitate the transport of glucose, implying a potential association between the distribution and activation. Collectively, our work characterized the clustering distribution of GLUT1 and linked its spatial structural organization to the functions, which would provide insights into the activation mechanism of the transporter.
Collapse
|
70
|
Metabolic Regulation in Mitochondria and Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1038:149-171. [PMID: 29178075 DOI: 10.1007/978-981-10-6674-0_11] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondria are generally considered as a powerhouse in a cell where the majority of the cellular ATP and metabolite productions occur. Metabolic rewiring and reprogramming may be initiated and regulated by mitochondrial enzymes. The hypothesis that cellular metabolic rewiring and reprogramming processes may occur as cellular microenvironment is disturbed, resulting in alteration of cell phenotype, such as cancer cells resistant to therapeutics seems to be now acceptable. Cancer metabolic reprogramming regulated by mitochondrial enzymes is now one of the hallmarks of cancer. This chapter provides an overview of cancer metabolism and summarizes progress made in mitochondria-mediated metabolic regulation in cancer drug resistance.
Collapse
|
71
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
72
|
Kannan S, Begoyan VV, Fedie JR, Xia S, Weseliński ŁJ, Tanasova M, Rao S. Metabolism-Driven High-Throughput Cancer Identification with GLUT5-Specific Molecular Probes. BIOSENSORS-BASEL 2018; 8:bios8020039. [PMID: 29642606 PMCID: PMC6022918 DOI: 10.3390/bios8020039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/19/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022]
Abstract
Point-of-care applications rely on biomedical sensors to enable rapid detection with high sensitivity and selectivity. Despite advances in sensor development, there are challenges in cancer diagnostics. Detection of biomarkers, cell receptors, circulating tumor cells, gene identification, and fluorescent tagging are time-consuming due to the sample preparation and response time involved. Here, we present a novel approach to target the enhanced metabolism in breast cancers for rapid detection using fluorescent imaging. Fluorescent analogs of fructose target the fructose-specific transporter GLUT5 in breast cancers and have limited to no response from normal cells. These analogs demonstrate a marked difference in adenocarcinoma and premalignant cells leading to a novel detection approach. The vastly different uptake kinetics of the analogs yields two unique signatures for each cell type. We used normal breast cells MCF10A, adenocarcinoma cells MCF7, and premalignant cells MCF10AneoT, with hepatocellular carcinoma cells HepG2 as the negative control. Our data indicated that MCF10AneoT and MCF7 cells had an observable difference in response to only one of the analogs. The response, observed as fluorescence intensity, leads to a two-point assessment of the cells in any sample. Since the treatment time is 10 min, there is potential for use in rapid on-site high-throughput diagnostics.
Collapse
Affiliation(s)
- Srinivas Kannan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA;
| | - Vagarshak V. Begoyan
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA; (V.V.B.); (J.R.F.); (S.X.); (Ł.J.W.)
| | - Joseph R. Fedie
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA; (V.V.B.); (J.R.F.); (S.X.); (Ł.J.W.)
| | - Shuai Xia
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA; (V.V.B.); (J.R.F.); (S.X.); (Ł.J.W.)
| | - Łukasz J. Weseliński
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA; (V.V.B.); (J.R.F.); (S.X.); (Ł.J.W.)
| | - Marina Tanasova
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA; (V.V.B.); (J.R.F.); (S.X.); (Ł.J.W.)
- Correspondence: (M.T.); (S.R.); Tel.: +1-906-487-1163 (M.T.); +1-906-487-3230 (S.R.)
| | - Smitha Rao
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA;
- Correspondence: (M.T.); (S.R.); Tel.: +1-906-487-1163 (M.T.); +1-906-487-3230 (S.R.)
| |
Collapse
|
73
|
MUC13 contributes to rewiring of glucose metabolism in pancreatic cancer. Oncogenesis 2018; 7:19. [PMID: 29467405 PMCID: PMC5833644 DOI: 10.1038/s41389-018-0031-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/04/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Pancreatic tumors are rewired for high-glucose metabolism and typically present with exceptionally poor prognosis. Recently, we have shown that MUC13, which is highly expressed in pancreatic tumors, promotes tumor progression via modulation of HER2 receptor tyrosine kinase activity. Herein, we investigate a novel, MUC13-mediated molecular mechanism responsible for higher glucose metabolism in pancreatic tumors. Our results demonstrate that MUC13 expression leads to the activation/nuclear translocation of NF-κB p65 and phosphorylation of IκB, which in turn upregulates the expression of important proteins (Glut-1, c-Myc, and Bcl-2) that are involved in glucose metabolism. MUC13 functionally interacts and stabilizes Glut-1 to instigate downstream events responsible for higher glucose uptake in pancreatic cancer cells. Altered MUC13 expression by overexpression and knockdown techniques effectively modulated glucose uptake, lactate secretion, and metastatic phenotypes in pancreatic cancer cells. NF-κB inhibitor, Sulfasalazine, abrogates the MUC13 and Glut-1 interaction, and attenuates events associated with MUC13-induced glucose metabolism. Pancreatic ductal adenocarcinoma (PDAC) patient tissue samples also show a positive correlation between the expression of these two proteins. These results delineate how MUC13 rewire aberrant glucose metabolism to enhance aggressiveness of pancreatic cancer and revealed a novel mechanism to develop newer therapeutic strategies for this exceptionally difficult cancer.
Collapse
|
74
|
Poliaková M, Aebersold DM, Zimmer Y, Medová M. The relevance of tyrosine kinase inhibitors for global metabolic pathways in cancer. Mol Cancer 2018; 17:27. [PMID: 29455660 PMCID: PMC5817809 DOI: 10.1186/s12943-018-0798-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor metabolism is a thrilling discipline that focuses on mechanisms used by cancer cells to earn crucial building blocks and energy to preserve growth and overcome resistance to various treatment modalities. At the same time, therapies directed specifically against aberrant signalling pathways driven by protein tyrosine kinases (TKs) involved in proliferation, metastasis and growth count for several years to promising anti-cancer approaches. In this respect, small molecule inhibitors are the most widely used clinically relevant means for targeted therapy, with a rising number of approvals for TKs inhibitors. In this review, we discuss recent observations related to TKs-associated metabolism and to metabolic feedback that is initialized as cellular response to particular TK-targeted therapies. These observations provide collective evidence that therapeutic responses are primarily linked to such pathways as regulation of lipid and amino acid metabolism, TCA cycle and glycolysis, advocating therefore the development of further effective targeted therapies against a broader spectrum of TKs to treat patients whose tumors display deregulated signalling driven by these proteins.
Collapse
Affiliation(s)
- Michaela Poliaková
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland. .,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.
| |
Collapse
|
75
|
Wuest M, Hamann I, Bouvet V, Glubrecht D, Marshall A, Trayner B, Soueidan OM, Krys D, Wagner M, Cheeseman C, West F, Wuest F. Molecular Imaging of GLUT1 and GLUT5 in Breast Cancer: A Multitracer Positron Emission Tomography Imaging Study in Mice. Mol Pharmacol 2018; 93:79-89. [PMID: 29142019 DOI: 10.1124/mol.117.110007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/13/2017] [Indexed: 02/14/2025] Open
Abstract
Use of [18F]FDG-positron emission tomography (PET) in clinical breast cancer (BC) imaging is limited mainly by insufficient expression levels of facilitative glucose transporter (GLUT)1 in up to 50% of all patients. Fructose-specific facilitative hexose transporter GLUT5 represents an alternative biomarker for PET imaging of hexose metabolism in BC. The goal of the present study was to compare the uptake characteristics of selected hexose-based PET radiotracers in murine BC model EMT6. Uptake of 1-deoxy-1-[18F]fluoro-d-fructose (1-[18F]FDF), 6-deoxy-6-[18F]fluoro-d-fructose (6-[18F]FDF), 1-deoxy-1-[18F]fluoro-2,5-anhydro-mannitol (1-[18F]FDAM), 2-deoxy-2-[18F]fluoro-d-glucose (2-[18F]FDG), and 6-deoxy-6-[18F]fluoro-d-glucose (6-[18F]FDG) was studied in EMT6 cells, tumors, and muscle and correlated to GLUT1 and GLUT5 expression levels. Fructose-derivative 6-[18F]FDF revealed greater tumor uptake than did structural analog 1-[18F]FDF, whereas 1-[18F]FDAM with locked anomeric configuration showed similar low tumor uptake to that of 1-[18F]FDF. Glucose-derivative 6-[18F]FDG reached maximum tumor uptake at 20 minutes, with no further accumulation over time. Uptake of 2-[18F]FDG was greatest and continuously increasing owing to metabolic trapping through phosphorylation by hexokinase II. In EMT6 tumors, GLUT5 mRNA expression was 20,000-fold lower compared with GLUT1. Whereas the latter was much greater in tumor than in muscle tissue (GLUT1 50:1), the opposite was found for GLUT5 mRNA expression (GLUT5 1:6). GLUT5 protein levels were higher in tumor versus muscle tissue as determined by Western blot and immunohistochemistry. Our data suggest that tumor uptake of fructose metabolism-targeting radiotracers 1-[18F]FDF, 6-[18F]FDF, and 1-[18F]FDAM does not correlate with GLUT5 mRNA levels but is linked to GLUT5 protein levels. In conclusion, our results highlight the importance of detailed biochemical studies on GLUT protein expression levels in combination with PET imaging studies for functional characterization of GLUTs in BC.
Collapse
Affiliation(s)
- Melinda Wuest
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Ingrit Hamann
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Vincent Bouvet
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Darryl Glubrecht
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Alison Marshall
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Brendan Trayner
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Olivier-Mohamad Soueidan
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Krys
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Michael Wagner
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Chris Cheeseman
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Frederick West
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| | - Frank Wuest
- Departments of Oncology and Cross Cancer Institute (M.Wu., I.H., V.B., D.G., A.M., D.K., M.Wa., F.Wu.), Chemistry (O.-M.S., F.We.), and Physiology (B.T., O.-M.S., C.C.), University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
76
|
Bannasch P, Ribback S, Su Q, Mayer D. Clear cell hepatocellular carcinoma: origin, metabolic traits and fate of glycogenotic clear and ground glass cells. Hepatobiliary Pancreat Dis Int 2017; 16:570-594. [PMID: 29291777 DOI: 10.1016/s1499-3872(17)60071-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/14/2017] [Indexed: 02/05/2023]
Abstract
Clear cell hepatocellular carcinoma (CCHCC) has hitherto been considered an uncommon, highly differentiated variant of hepatocellular carcinoma (HCC) with a relatively favorable prognosis. CCHCC is composed of mixtures of clear and/or acidophilic ground glass hepatocytes with excessive glycogen and/or fat and shares histology, clinical features and etiology with common HCCs. Studies in animal models of chemical, hormonal and viral hepatocarcinogenesis and observations in patients with chronic liver diseases prone to develop HCC have shown that the majority of HCCs are preceded by, or associated with, focal or diffuse excessive storage of glycogen (glycogenosis) which later may be replaced by fat (lipidosis/steatosis). In ground glass cells, the glycogenosis is accompanied by proliferation of the smooth endoplasmic reticulum, which is closely related to glycogen particles and frequently harbors the hepatitis B surface antigen (HBsAg). From the findings in animal models a sequence of changes has been established, commencing with preneoplastic glycogenotic liver lesions, often containing ground glass cells, and progressing to glycogen-poor neoplasms via various intermediate stages, including glycogenotic/lipidotic clear cell foci, clear cell hepatocellular adenomas (CCHCA) rich in glycogen and/or fat, and CCHCC. A similar process seems to take place in humans, with clear cells frequently persisting in CCHCC and steatohepatitic HCC, which presumably represent intermediate stages in the development rather than particular variants of HCC. During the progression of the preneoplastic lesions, the clear and ground glass cells transform into cells characteristic of common HCC. The sequential cellular changes are associated with metabolic aberrations, which start with an activation of the insulin signaling cascade resulting in pre-neoplastic hepatic glycogenosis. The molecular and metabolic changes underlying the glycogenosis/lipidosis are apparently responsible for the dramatic metabolic shift from gluconeogenesis to the pentose phosphate pathway and Warburg-type glycolysis, which provide precursors and energy for an ever increasing cell proliferation during progression.
Collapse
Affiliation(s)
| | - Silvia Ribback
- Institut für Pathologie, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Qin Su
- Cell Marque, Millipore-Sigma Rocklin, USA
| | - Doris Mayer
- German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
77
|
Důra M, Němejcová K, Jakša R, Bártů M, Kodet O, Tichá I, Michálková R, Dundr P. Expression of Glut-1 in Malignant Melanoma and Melanocytic Nevi: an Immunohistochemical Study of 400 Cases. Pathol Oncol Res 2017; 25:361-368. [PMID: 29128957 DOI: 10.1007/s12253-017-0363-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/08/2017] [Indexed: 11/26/2022]
Abstract
The glucose transporter-1 (Glut-1) is a cell membrane glycoprotein involved in glucose uptake. An increased expression of Glut-1 is an important cell adaptation mechanism against hypoxia. An upregulation of Glut-1 can be found in several types of malignant tumors, which are able to reprogram their metabolism from oxidative phosphorylation to aerobic glycolysis (Warburg effect). However, the data regarding melanocytic lesions is equivocal. We performed comprehensive immunohistochemical analysis of the Glut-1 expression in 225 malignant melanomas (MM) and 175 benign nevi. Only the membranous expression of Glut-1 was regarded as positive. The expression of Glut-1 (the cut-off for positivity was determined as H-score 15) was found in 69/225 malignant melanomas. The number of positive cases and the H-score of Glut-1 increased where there was a higher Breslow thickness (p < 0.00001) when comparing pT1- pT4 MM groups. All benign nevi were classified as negative. In conclusion, the membranous expression of Glut-1 is a common feature of a malignant melanoma but this type of expression is very rare in benign melanocytic nevi. Our results suggest that the membranous expression of Glut-1 can be used as a surrogate marker in the assessing of the biological nature of benign and malignant melanocytic lesions. However, despite its high specificity, the sensitivity of this marker is relatively low. Moreover, due to the fact that the increased expression of Glut-1 correlates with a shorter survival period (10-year disease free survival, recurrence free survival and metastasis free survival and MFS), it can be used as a prognostically adverse factor.
Collapse
Affiliation(s)
- Miroslav Důra
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
- Department of Dermatology and Venereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kristýna Němejcová
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Radek Jakša
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Michaela Bártů
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Ondřej Kodet
- Department of Dermatology and Venereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ivana Tichá
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Romana Michálková
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic.
| |
Collapse
|
78
|
Chandra D, Selvanesan BC, Yuan Z, Libutti SK, Koba W, Beck A, Zhu K, Casadevall A, Dadachova E, Gravekamp C. 32-Phosphorus selectively delivered by listeria to pancreatic cancer demonstrates a strong therapeutic effect. Oncotarget 2017; 8:20729-20740. [PMID: 28186976 PMCID: PMC5400540 DOI: 10.18632/oncotarget.15117] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/24/2017] [Indexed: 12/21/2022] Open
Abstract
Our laboratory has developed a novel delivery platform using an attenuated non-toxic and non-pathogenic bacterium Listeria monocytogenes that infects tumor cells and selectively survives and multiplies in metastases and primary tumors with help of myeloid-derived suppressor cells (MDSC) and immune suppression in the tumor microenvironment (TME). 32P was efficiently incorporated into the Listeria bacteria by starvation of the bacteria in saline, and then cultured in phosphorus-free medium complemented with 32P as a nutrient. Listeria-32P kills tumor cells through both 32P-induced ionizing radiation and Listeria-induced reactive oxygen species (ROS). The levels of 32P and Listeria were studied in various normal and tumor tissues, at sequential time points after injection of mice with pancreatic cancer (syngeneic model Panc-02). We found that 32P and Listeria predominantly accumulated in tumors and metastases, with their highest accumulation 4 hrs (32P) and 3 days (Listeria) after injection. Listeria also penetrated the transgenic KPC (conditionally express endogenous Kras-G12D and p53-R172H mutant alleles) pancreatic tumors and metastases. This is remarkable since KPC tumors, like human tumors, exhibit a stromal barrier, which prevents most drugs from penetrating the pancreatic tumors. Therapeutic treatment with Listeria -32P resulted in a strong reduction of the growth of pancreatic cancer at early and late stages in Panc-02 and KPC mice. These results highlight the power of Listeria as new delivery platform of anticancer agents to the TME. Not only were therapeutic levels of radioactive Listeria reached in tumors and metastases but the selective delivery also led to minimal side effects.
Collapse
Affiliation(s)
- Dinesh Chandra
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, NY 10461, USA
| | | | - Ziqiang Yuan
- Montefiore Medical Center, Medical Arts Pavilion, MMC-MAP, Bronx, NY 10467, USA
| | - Steven K Libutti
- Montefiore Medical Center, Medical Arts Pavilion, MMC-MAP, Bronx, NY 10467, USA
| | - Wade Koba
- Albert Einstein College of Medicine, Department of Radiology, MRRC, Bronx, NY 10461, USA
| | - Amanda Beck
- Albert Einstein College of Medicine, Department of Pathology, Michael F. Price Center, Bronx, NY 10461, USA
| | - Kun Zhu
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, NY 10461, USA
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, MD 21205, USA
| | - Ekaterina Dadachova
- Albert Einstein College of Medicine, Department of Radiology/Department of Microbiology and Immunology, Bronx, NY 10461, USA
| | - Claudia Gravekamp
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, NY 10461, USA
| |
Collapse
|
79
|
Bonomi M, De Filippis C, Lopci E, Gianoncelli L, Rizzardi G, Cerchiaro E, Bortolotti L, Zanello A, Ceresoli GL. Clinical staging of malignant pleural mesothelioma: current perspectives. LUNG CANCER-TARGETS AND THERAPY 2017; 8:127-139. [PMID: 28860886 PMCID: PMC5571821 DOI: 10.2147/lctt.s102113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a disease with limited therapeutic options, the management of which is still controversial. Diagnosis is usually made by thoracoscopy, which allows multiple biopsies with histological subtyping and is indicated for staging purposes in surgical candidates. The recommended and recently updated classification for clinical use is the TNM staging system established by the International Mesothelioma Interest Group and the International Association for the Study of Lung Cancer, which is based mainly on surgical and pathological variables, as well as on cross-sectional imaging. Contrast-enhanced computed tomography is the primary imaging procedure. Currently, the most used measurement system for MPM is the modified Response Evaluation Criteria in Solid Tumors (RECIST) method, which is based on unidimensional measurements of tumor thickness perpendicular to the chest wall or mediastinum. Magnetic resonance imaging and functional imaging with 18F-fluoro-2-deoxy-D-glucose positron-emission tomography can provide additional staging information in selected cases, although the usefulness of this method is limited in patients undergoing pleurodesis. Molecular reclassification of MPM and gene expression or miRNA prognostic models have the potential to improve prognostication and patient selection for a proper treatment algorithm; however, they await prospective validation to be introduced in clinical practice.
Collapse
Affiliation(s)
- Maria Bonomi
- Department of Oncology, Thoracic and GU Oncology Unit
| | | | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, Milan
| | | | - Giovanna Rizzardi
- Department of Thoracic Surgery, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | | | - Luigi Bortolotti
- Department of Thoracic Surgery, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | | | | |
Collapse
|
80
|
Nagarajan A, Dogra SK, Sun L, Gandotra N, Ho T, Cai G, Cline G, Kumar P, Cowles RA, Wajapeyee N. Paraoxonase 2 Facilitates Pancreatic Cancer Growth and Metastasis by Stimulating GLUT1-Mediated Glucose Transport. Mol Cell 2017; 67:685-701.e6. [PMID: 28803777 DOI: 10.1016/j.molcel.2017.07.014] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/07/2017] [Accepted: 07/11/2017] [Indexed: 12/28/2022]
Abstract
Metabolic deregulation is a hallmark of human cancers, and the glycolytic and glutamine metabolism pathways were shown to be deregulated in pancreatic ductal adenocarcinoma (PDAC). To identify new metabolic regulators of PDAC tumor growth and metastasis, we systematically knocked down metabolic genes that were overexpressed in human PDAC tumor samples using short hairpin RNAs. We found that p53 transcriptionally represses paraoxonase 2 (PON2), which regulates GLUT1-mediated glucose transport via stomatin. The loss of PON2 initiates the cellular starvation response and activates AMP-activated protein kinase (AMPK). In turn, AMPK activates FOXO3A and its transcriptional target, PUMA, which induces anoikis to suppress PDAC tumor growth and metastasis. Pharmacological or genetic activation of AMPK, similar to PON2 inhibition, blocks PDAC tumor growth. Collectively, our results identify PON2 as a new modulator of glucose transport that regulates a pharmacologically tractable pathway necessary for PDAC tumor growth and metastasis.
Collapse
Affiliation(s)
- Arvindhan Nagarajan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shaillay Kumar Dogra
- Singapore Institute of Clinical Sciences, Agency for Science Technology and Research (A(∗)STAR), Brenner Center for Molecular Medicine, Singapore 117609, Singapore
| | - Lisha Sun
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Neeru Gandotra
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thuy Ho
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gary Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Priti Kumar
- Department of Internal Medicine and Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Robert A Cowles
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Narendra Wajapeyee
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
81
|
Abstract
OBJECTIVE In this article, we provide a general overview of how cancer cells subvert critical metabolic pathways to support their growth and unchecked division. Furthermore, we outline how molecular imaging can diagnostically exploit the resulting differences between cancer and normal cells. CONCLUSION Molecular PET can provide valuable information about the metabolic dysregulation in cancer.
Collapse
|
82
|
Implications of Resveratrol on Glucose Uptake and Metabolism. Molecules 2017; 22:molecules22030398. [PMID: 28272357 PMCID: PMC6155386 DOI: 10.3390/molecules22030398] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
Resveratrol—a polyphenol of natural origin—has been the object of massive research in the past decade because of its potential use in cancer therapy. However, resveratrol has shown an extensive range of cellular targets and effects, which hinders the use of the molecule for medical applications including cancer and type 2 diabetes. Here, we review the latest advances in understanding how resveratrol modulates glucose uptake, regulates cellular metabolism, and how this may be useful to improve current therapies. We discuss challenges and findings regarding the inhibition of glucose uptake by resveratrol and other polyphenols of similar chemical structure. We review alternatives that can be exploited to improve cancer therapies, including the use of other polyphenols, or the combination of resveratrol with other molecules and their impact on glucose homeostasis in cancer and diabetes.
Collapse
|
83
|
Wokoun U, Hellriegel M, Emons G, Gründker C. Co-treatment of breast cancer cells with pharmacologic doses of 2-deoxy-D-glucose and metformin: Starving tumors. Oncol Rep 2017; 37:2418-2424. [PMID: 28350075 DOI: 10.3892/or.2017.5491] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/26/2017] [Indexed: 11/05/2022] Open
Abstract
A characteristic of tumor cells is the increased aerobic glycolysis for energy production. Thus, inhibition of glycolysis represents a selective therapeutic option. It has been shown that glycolysis inhibitor 2-deoxy-D-glucose (2DG) induces apoptotic cell death in different tumor entities. In addition, the antitumor activity of the anti-diabetic drug metformin has been demonstrated. In the present study, we aimed to ascertain whether the combination of pharmacologic doses of 2DG with metformin increases the antitumor efficacy. Cell viability of MDA-MB-231 and HCC1806 triple-negative breast cancer (TNBC) cells treated without or with 2DG or with metformin alone or with the combination of both agents was measured using Alamar Blue assay. Induction of apoptosis was quantified by measurement of the loss of mitochondrial membrane potential and cleavage of PARP. Treatment of breast cancer cells with glycolysis inhibitor 2DG or with the anti-diabetic drug metformin resulted in a significant decrease in cell viability and an increase in apoptosis. Treatment with 2DG in combination with metformin resulted in significantly reduced viability compared with the single agent treatments. The observed reduction in viability was due to induction of apoptosis. In addition, in regards to apoptosis induction a stronger effect in the case of co-treatment compared with single agent treatments was observed. The glycolytic phenotype of human breast cancer cells can be targeted for therapeutic intervention. Co-treatment with doses of the glycolysis inhibitor 2DG and anti-diabetic drug metformin is tolerable in humans and may be a suitable therapy for human breast cancers.
Collapse
Affiliation(s)
- Ulrike Wokoun
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Martin Hellriegel
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| |
Collapse
|
84
|
Upregulation of SLC2 (GLUT) family genes is related to poor survival outcomes in papillary thyroid carcinoma: Analysis of data from The Cancer Genome Atlas. Surgery 2017; 161:188-194. [DOI: 10.1016/j.surg.2016.04.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/06/2016] [Accepted: 04/10/2016] [Indexed: 01/08/2023]
|
85
|
Němejcová K, Rosmusová J, Bártů M, Důra M, Tichá I, Dundr P. Expression of Glut-1 in Normal Endometrium and Endometrial Lesions: Analysis of 336 Cases. Int J Surg Pathol 2016; 25:389-396. [DOI: 10.1177/1066896916683510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Glucose transporter-1 (Glut-1) is a membrane glycoprotein that is, together with other glucose transporters, responsible for the regulation of glucose uptake. An increased expression of this protein seems to be a general feature of several malignant tumors that are able to reprogram their metabolism and switch from oxidative phosphorylation to aerobic glycolysis. Methods: We performed comprehensive immunohistochemical analysis of Glut-1 expression in 336 endometrial samples, including tumors, nontumor lesions, and normal tissues. Results: Expression of Glut-1 was found in 87% of endometrioid carcinomas (160/184 cases), 100% of serous carcinomas (29/29 cases), 100% of clear cell carcinomas (17/17 cases), 50% of polyps with atypical hyperplasia (8/16 cases), 12.5% of polyps with non-atypical hyperplasia (3/24 cases), 77% of hyperplasias with atypias (10/13 cases), 9% of hyperplasias without atypias (1/11 cases), 87% of secretory endometrium samples (13/15 cases), and in none of the nonsecretory endometrium samples (0/27 cases). In endometrioid carcinomas, Glut-1 was expressed in a marked geographical pattern. In nontumor lesions, its expression was more common in atypical hyperplasia and polyps with atypical hyperplasia compared with polyps with non-atypical hyperplasia and hyperplasias without atypia ( P = .00032). Conclusion: Our study confirms the high expression of Glut-1 not only in endometrioid carcinomas but also in other carcinomas of endometrium including clear cell and serous types. Glut-1 expression can be used as a surrogate marker in differential diagnosis between hyperplasia with and without atypia. Because of common Glut-1 expression in malignant tumors, therapeutic strategies influencing this protein or its signaling pathways can be beneficial.
Collapse
Affiliation(s)
- Kristýna Němejcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jana Rosmusová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Michaela Bártů
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Miroslav Důra
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Dermatovenerology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ivana Tichá
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Dundr
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
86
|
Koh YW, Lee SJ, Park SY. Differential expression and prognostic significance of GLUT1 according to histologic type of non-small-cell lung cancer and its association with volume-dependent parameters. Lung Cancer 2016; 104:31-37. [PMID: 28212997 DOI: 10.1016/j.lungcan.2016.12.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/09/2016] [Accepted: 12/10/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND We evaluated glucose transporter type 1 (GLUT1) and carbonic anhydrase IX (CAIX) expression, together with volume-based18F-fluorodeoxyglucose positron emission tomography (FDG-PET) parameters, in non-small cell lung cancer (NSCLC) patients, and examined the prognostic significance of those parameters according to its histologic subtype. METHOD A total of 269 patients, who underwent surgical resection for NSCLC, were reviewed retrospectively. Metabolic tumor volume (MTV) and total lesion glycolysis (TLG) values were measured by preoperative 18F-fluorodeoxyglucose positron emission tomography computed tomography. GLUT1 and CAIX expression was evaluated using immunohistochemical method. RESULTS The mean MTV and TLG values were 30.0±57.1 and 165.4±361.3, respectively, and were significantly higher in patients with squamous cell carcinoma than with adenocarcinoma (p=0.047 for MTV; p=0.042 for TLG). GLUT1 expression was identified in 99% of squamous cell carcinoma and 50% of adenocarcinoma patients. MTV and TLG values were significantly higher in GLUT1-positive than GLUT-negative adenocarcinomas; however, CAIX expression did not show this pattern. GLUT1-positive adenocarcinoma patients had a lower OS than GLUT1-negative patients (p<0.001), whereas CAIX-positive and CAIX-negative patients showed similar OS rates (p=0.226). Patients with high MTV and TLG values showed lower OS rates than those with low MTV and TLG values. Multivariate analysis showed that GLUT1 positivity was an independent risk factor for a lower OS rate in lung adenocarcinoma patients (hazard ratio=2.574, p=0.016). GLUT1 expression was associated with micropapillary/solid histology, lymphovascular invasion, and advanced pTNM stage. CONCLUSIONS MTV and TLG values, and GLUT1 expression, significantly differed between patients with squamous cell carcinoma and adenocarcinoma. High GLUT1 expression levels were significantly associated with MTV and TLG values and adverse clinical outcomes in patients with adenocarcinoma.
Collapse
Affiliation(s)
- Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Su Jin Lee
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, 206 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea.
| | - Seong Yong Park
- Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, 206 Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea.
| |
Collapse
|
87
|
The HK2 Dependent "Warburg Effect" and Mitochondrial Oxidative Phosphorylation in Cancer: Targets for Effective Therapy with 3-Bromopyruvate. Molecules 2016; 21:molecules21121730. [PMID: 27983708 PMCID: PMC6273842 DOI: 10.3390/molecules21121730] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/09/2016] [Accepted: 12/11/2016] [Indexed: 12/30/2022] Open
Abstract
This review summarizes the current state of knowledge about the metabolism of cancer cells, especially with respect to the "Warburg" and "Crabtree" effects. This work also summarizes two key discoveries, one of which relates to hexokinase-2 (HK2), a major player in both the "Warburg effect" and cancer cell immortalization. The second discovery relates to the finding that cancer cells, unlike normal cells, derive as much as 60% of their ATP from glycolysis via the "Warburg effect", and the remaining 40% is derived from mitochondrial oxidative phosphorylation. Also described are selected anticancer agents which generally act as strong energy blockers inside cancer cells. Among them, much attention has focused on 3-bromopyruvate (3BP). This small alkylating compound targets both the "Warburg effect", i.e., elevated glycolysis even in the presence oxygen, as well as mitochondrial oxidative phosphorylation in cancer cells. Normal cells remain unharmed. 3BP rapidly kills cancer cells growing in tissue culture, eradicates tumors in animals, and prevents metastasis. In addition, properly formulated 3BP shows promise also as an effective anti-liver cancer agent in humans and is effective also toward cancers known as "multiple myeloma". Finally, 3BP has been shown to significantly extend the life of a human patient for which no other options were available. Thus, it can be stated that 3BP is a very promising new anti-cancer agent in the process of undergoing clinical development.
Collapse
|
88
|
Yang HJ, Xu WJ, Guan YH, Zhang HW, Ding WQ, Rong L, Qiu ZB, Zhong L. Expression of Glut-1 and HK-II in Pancreatic Cancer and Their Impact on Prognosis and FDG Accumulation. Transl Oncol 2016; 9:583-591. [PMID: 27916293 PMCID: PMC5143352 DOI: 10.1016/j.tranon.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/09/2016] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE: The purpose of this article is to analyze the expression of Glut-1 and HK-II, the association between their expression and 18F-FDG accumulation in pancreatic cancer. METHODS: Fifty patients with histologically proven pancreatic cancer were included in this preliminary study, all of whom received 18F-FDG PET/CT performance before surgery. Immunohistochemical staining of tumor tissue and adjacent normal tissue was performed for Glut-1 and HK-II. By combining proportions and intensity of immunochemical staining, we obtained the modified immunohistological scores for Glut-1 and HK-II respectively. The relationship between expression of Glut-1, HK-II and series of parameters was analyzed, i.e. clinicopathological characteristics, prognosis of patients and SUVmax of PET-CT. RESULTS: Compared with normal tissue, the Glut-1 and HK-II expression in pancreatic cancer tissue was significantly increased (P < .001). There was no correlation between expression of Glut-1, HK-II and age, gender, tumor size, tumor location, tumor histological type, tumor differentiation, the nerve infiltration, vascular invasion, local infiltration, lymph node metastasis or tumor staging in pancreatic cancer (P > .05). During the follow-up period, the survival curves of low Glut-1 group and high Glut-1 group were statistically different (P = .049). Multivariate analysis (Cox regression) revealed that Glut-1 expression was not associated with mortality (P > .05). No statistical difference was found in the survival curves of negative HK-II group and positive HK-II group (P = .545). There was no correlation between 18F-FDG uptake and expression of Glut-1 and HK-II(P > .05). CONCLUSION: The Glut-1 and HK-II expression in pancreatic cancer tissue was significantly increased. There was no correlation between expression of Glut-1, HK-II and clinicopathological characteristics, prognosis and 18F-FDG uptake.
Collapse
Affiliation(s)
- Hai-Jing Yang
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| | - Wei-Jia Xu
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| | - Yi-Hui Guan
- PET Center of Huashan Hospital, Fudan University, 518 Wuzhong East Road, Shanghai 200235, China
| | - Hui-Wei Zhang
- PET Center of Huashan Hospital, Fudan University, 518 Wuzhong East Road, Shanghai 200235, China
| | - Wei-Qun Ding
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Lan Rong
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Zhi-Bing Qiu
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Liang Zhong
- Department of Gastroenterology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| |
Collapse
|
89
|
Toward a cancer-specific diet. Clin Nutr 2016; 35:1188-95. [DOI: 10.1016/j.clnu.2015.01.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/13/2015] [Accepted: 01/18/2015] [Indexed: 01/01/2023]
|
90
|
Sarisozen C, Dhokai S, Tsikudo EG, Luther E, Rachman IM, Torchilin VP. Nanomedicine based curcumin and doxorubicin combination treatment of glioblastoma with scFv-targeted micelles: In vitro evaluation on 2D and 3D tumor models. Eur J Pharm Biopharm 2016; 108:54-67. [PMID: 27569031 DOI: 10.1016/j.ejpb.2016.08.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/09/2016] [Accepted: 08/21/2016] [Indexed: 11/29/2022]
Abstract
NF-κB is strongly associated with poor prognosis of different cancer types and an important factor responsible for the malignant phenotype of glioblastoma. Overcoming chemotherapy-induced resistance caused by activation of PI3K/Akt and NF-κB pathways is crucial for successful glioblastoma therapy. We developed an all-in-one nanomedicine formulation for co-delivery of a chemotherapeutic agent (topoisomerase II inhibitor, doxorubicin) and a multidrug resistance modulator (NF-κB inhibitor, curcumin) for treatment of glioblastoma due to their synergism. Both agents were incorporated into PEG-PE-based polymeric micelles. The glucose transporter-1 (GLUT1) is overexpressed in many tumors including glioblastoma. The micellar system was decorated with GLUT1 antibody single chain fragment variable (scFv) as the ligand to promote blood brain barrier transport and glioblastoma targeting. The combination treatment was synergistic (combination index, CI of 0.73) against U87MG glioblastoma cells. This synergism was improved by micellar encapsulation (CI: 0.63) and further so with GLUT1 targeting (CI: 0.46). Compared to non-targeted micelles, GLUT1 scFv surface modification increased the association of micelles (>20%, P<0.01) and the nuclear localization of doxorubicin (∼3-fold) in U87MGcells, which also translated into enhanced cytotoxicity. The increased caspase 3/7 activation by targeted micelles indicates successful apoptosis enhancement by combinatory treatment. Moreover, GLUT1 targeted micelles resulted in deeper penetration into the 3D spheroid model. The increased efficacy of combination nanoformulations on the spheroids compared to a single agent loaded, or to non-targeted formulations, reinforces the rationale for selection of this combination and successful utilization of GLUT1 scFv as a targeting agent for glioblastoma treatment.
Collapse
Affiliation(s)
- Can Sarisozen
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Shekhar Dhokai
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Edcar G Tsikudo
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Ed Luther
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
91
|
Guo H, Nan Y, Zhen Y, Zhang Y, Guo L, Yu K, Huang Q, Zhong Y. miRNA-451 inhibits glioma cell proliferation and invasion by downregulating glucose transporter 1. Tumour Biol 2016; 37:13751-13761. [DOI: 10.1007/s13277-016-5219-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/14/2016] [Indexed: 01/19/2023] Open
|
92
|
A bioluminescent assay for measuring glucose uptake. Anal Biochem 2016; 505:43-50. [DOI: 10.1016/j.ab.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 11/18/2022]
|
93
|
Abstract
Oncogenes are key drivers of tumor growth. Although several cancer-driving mechanisms have been identified, the role of oncogenes in shaping metabolic patterns in cancer cells is only beginning to be appreciated. Recent studies show that oncogenes directly regulate critical metabolic enzymes and metabolic signaling pathways. Here, we present evidence for oncogene-directed cancer metabolic regulation and discuss the importance of identifying underlying mechanisms that can be targeted for developing precision cancer therapies.
Collapse
|
94
|
Kue CS, Kamkaew A, Burgess K, Kiew LV, Chung LY, Lee HB. Small Molecules for Active Targeting in Cancer. Med Res Rev 2016; 36:494-575. [PMID: 26992114 DOI: 10.1002/med.21387] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/29/2022]
Abstract
For the purpose of this review, active targeting in cancer research encompasses strategies wherein a ligand for a cell surface receptor expressed on tumor cells is used to deliver a cytotoxic or imaging cargo. This area of research is more than two decades old, but in those 20 and more years, how many receptors have been studied extensively? What kinds of the ligands are used for active targeting? Are they mostly naturally occurring molecules such as folic acid, or synthetic substances developed in campaigns for medicinal chemistry efforts? This review outlines the most important receptor or ligand combinations that have been used in active targeting to answer these questions, and therefore to address the most important one of all: is research in active targeting affording diminishing returns, or is this an area for which the potential far exceeds progress made so far?
Collapse
Affiliation(s)
- Chin S Kue
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Anyanee Kamkaew
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Lik V Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lip Y Chung
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hong B Lee
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
95
|
Barron CC, Bilan PJ, Tsakiridis T, Tsiani E. Facilitative glucose transporters: Implications for cancer detection, prognosis and treatment. Metabolism 2016; 65:124-39. [PMID: 26773935 DOI: 10.1016/j.metabol.2015.10.007] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/22/2015] [Accepted: 10/01/2015] [Indexed: 12/11/2022]
Abstract
It is long recognized that cancer cells display increased glucose uptake and metabolism. In a rate-limiting step for glucose metabolism, the glucose transporter (GLUT) proteins facilitate glucose uptake across the plasma membrane. Fourteen members of the GLUT protein family have been identified in humans. This review describes the major characteristics of each member of the GLUT family and highlights evidence of abnormal expression in tumors and cancer cells. The regulation of GLUTs by key proliferation and pro-survival pathways including the phosphatidylinositol 3-kinase (PI3K)-Akt, hypoxia-inducible factor-1 (HIF-1), Ras, c-Myc and p53 pathways is discussed. The clinical utility of GLUT expression in cancer has been recognized and evidence regarding the use of GLUTs as prognostic or predictive biomarkers is presented. GLUTs represent attractive targets for cancer therapy and this review summarizes recent studies in which GLUT1, GLUT3, GLUT5 and others are inhibited to decrease cancer growth.
Collapse
Affiliation(s)
- Carly C Barron
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Philip J Bilan
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Theodoros Tsakiridis
- Department of Oncology, and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
96
|
Yamamoto N, Ueda-Wakagi M, Sato T, Kawasaki K, Sawada K, Kawabata K, Akagawa M, Ashida H. Measurement of Glucose Uptake in Cultured Cells. ACTA ACUST UNITED AC 2015; 71:12.14.1-12.14.26. [PMID: 26646194 DOI: 10.1002/0471141755.ph1214s71] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Facilitative glucose uptake transport systems are ubiquitous in animal cells and are responsible for transporting glucose across cell surface membranes. Evaluation of glucose uptake is crucial in the study of numerous diseases and metabolic disorders such as myocardial ischemia, diabetes mellitus, and cancer. Detailed in this unit are laboratory methods for assessing glucose uptake into mammalian cells. The unit is divided into five sections: (1) a brief overview of glucose uptake assays in cultured cells; (2) a method for measuring glucose uptake using radiolabeled 3-O-methylglucose; (3) a method for measuring glucose uptake using radiolabeled 2-deoxyglucose (2DG); (4) a microplate method for measuring 2DG-uptake using an enzymatic, fluorometric assay; and (5) a microplate-based method using a fluorescent analog of 2DG.
Collapse
Affiliation(s)
- Norio Yamamoto
- Research & Development Institute, House Wellness Foods Corporation, Hyogo, Japan
| | - Manabu Ueda-Wakagi
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Takuya Sato
- Research & Development Institute, House Wellness Foods Corporation, Hyogo, Japan
| | - Kengo Kawasaki
- Research & Development Institute, House Wellness Foods Corporation, Hyogo, Japan
| | - Keisuke Sawada
- Department of Agrobiosciences, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Kyuichi Kawabata
- Department of Bioscience, Fukui Prefectural University, Fukui, Japan
| | - Mitsugu Akagawa
- Department of Biological Chemistry, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Japan
| | - Hitoshi Ashida
- Department of Agrobiosciences, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| |
Collapse
|
97
|
Bell C, Dowson N, Fay M, Thomas P, Puttick S, Gal Y, Rose S. Hypoxia imaging in gliomas with 18F-fluoromisonidazole PET: toward clinical translation. Semin Nucl Med 2015; 45:136-50. [PMID: 25704386 DOI: 10.1053/j.semnuclmed.2014.10.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
There is significant interest in the development of improved image-guided therapy for neuro-oncology applications. Glioblastomas (GBM) in particular present a considerable challenge because of their pervasive nature, propensity for recurrence, and resistance to conventional therapies. MRI is routinely used as a guide for planning treatment strategies. However, this imaging modality is not able to provide images that clearly delineate tumor boundaries and affords only indirect information about key tumor pathophysiology. With the emergence of PET imaging with new oncology radiotracers, mapping of tumor infiltration and other important molecular events such as hypoxia is now feasible within the clinical setting. In particular, the importance of imaging hypoxia levels within the tumoral microenvironment is gathering interest, as hypoxia is known to play a central role in glioma pathogenesis and resistance to treatment. One of the hypoxia radiotracers known for its clinical utility is (18)F-fluoromisodazole ((18)F-FMISO). In this review, we highlight the typical causes of treatment failure in gliomas that may be linked to hypoxia and outline current methods for the detection of hypoxia. We also provide an overview of the growing body of studies focusing on the clinical translation of (18)F-FMISO PET imaging, strengthening the argument for the use of (18)F-FMISO hypoxia imaging to help optimize and guide treatment strategies for patients with glioblastoma.
Collapse
Affiliation(s)
- Christopher Bell
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; School of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Nicholas Dowson
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Mike Fay
- Department of Radiation Oncology, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
| | - Paul Thomas
- Specialised PET Services Queensland, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
| | - Simon Puttick
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Yaniv Gal
- Centre for Medical Diagnostic Technologies in Queensland, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Stephen Rose
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; School of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia.
| |
Collapse
|
98
|
Wu S, Yin X, Fang X, Zheng J, Li L, Liu X, Chu L. c-MYC responds to glucose deprivation in a cell-type-dependent manner. Cell Death Discov 2015; 1:15057. [PMID: 27551483 PMCID: PMC4979460 DOI: 10.1038/cddiscovery.2015.57] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/14/2015] [Accepted: 10/21/2015] [Indexed: 12/17/2022] Open
Abstract
Metabolic reprogramming supports cancer cells’ demands for rapid proliferation and growth. Previous work shows that oncogenes, such as MYC, hypoxia-inducible factor 1 (HIF1), have a central role in driving metabolic reprogramming. A lot of metabolic enzymes, which are deregulated in most cancer cells, are the targets of these oncogenes. However, whether metabolic change affects these oncogenes is still unclear. Here we show that glucose deprivation (GD) affects c-MYC protein levels in a cell-type-dependent manner regardless of P53 mutation status. GD dephosphorylates and then decreases c-MYC protein stability through PI3K signaling pathway in HeLa cells, but not in MDA-MB-231 cells. Role of c-MYC in sensitivity of GD also varies with cell types. c-MYC-mediated glutamine metabolism partially improves the sensitivity of GD in MDA-MB-231 cells. Our results reveal that the heterogeneity of cancer cells in response to metabolic stress should be considered in metabolic therapy for cancer.
Collapse
Affiliation(s)
- S Wu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - X Yin
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - X Fang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - J Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| | - L Li
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| | - X Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - L Chu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
99
|
Differentiation and apoptosis induction by lovastatin and γ-tocotrienol in HL-60 cells via Ras/ERK/NF-κB and Ras/Akt/NF-κB signaling dependent down-regulation of glyoxalase 1 and HMG-CoA reductase. Cell Signal 2015. [DOI: 10.1016/j.cellsig.2015.07.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
100
|
Micro Regional Heterogeneity of 64Cu-ATSM and 18F-FDG Uptake in Canine Soft Tissue Sarcomas: Relation to Cell Proliferation, Hypoxia and Glycolysis. PLoS One 2015; 10:e0141379. [PMID: 26501874 PMCID: PMC4621038 DOI: 10.1371/journal.pone.0141379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Tumour microenvironment heterogeneity is believed to play a key role in cancer progression and therapy resistance. However, little is known about micro regional distribution of hypoxia, glycolysis and proliferation in spontaneous solid tumours. The overall aim was simultaneous investigation of micro regional heterogeneity of 64Cu-ATSM (hypoxia) and 18F-FDG (glycolysis) uptake and correlation to endogenous markers of hypoxia, glycolysis, proliferation and angiogenesis to better therapeutically target aggressive tumour regions and prognosticate outcome. METHODS Exploiting the different half-lives of 64Cu-ATSM (13 h) and 18F-FDG (2 h) enabled simultaneous investigation of micro regional distribution of hypoxia and glycolysis in 145 tumour pieces from four spontaneous canine soft tissue sarcomas. Pairwise measurements of radioactivity and gene expression of endogenous markers of hypoxia (HIF-1α, CAIX), glycolysis (HK2, GLUT1 and GLUT3), proliferation (Ki-67) and angiogenesis (VEGFA and TF) were performed. Dual tracer autoradiography was compared with Ki-67 immunohistochemistry. RESULTS Micro regional heterogeneity in hypoxia and glycolysis within and between tumour sections of each tumour piece was observed. The spatial distribution of 64Cu-ATSM and 18F-FDG was rather similar within each tumour section as reflected in moderate positive significant correlations between the two tracers (ρ = 0.3920-0.7807; p = 0.0180 -<0.0001) based on pixel-to-pixel comparisons of autoradiographies and gamma counting of tumour pieces. 64Cu-ATSM and 18F-FDG correlated positively with gene expression of GLUT1 and GLUT3, but negatively with HIF-1α and CAIX. Significant positive correlations were seen between Ki-67 gene expression and 64Cu-ATSM (ρ = 0.5578, p = 0.0004) and 18F-FDG (ρ = 0.4629-0.7001, p = 0.0001-0.0151). Ki-67 gene expression more consistently correlated with 18F-FDG than with 64Cu-ATSM. CONCLUSIONS Micro regional heterogeneity of hypoxia and glycolysis was documented in spontaneous canine soft tissue sarcomas. 64Cu-ATSM and 18F-FDG uptakes and distributions showed significant moderate correlations at the micro regional level indicating overlapping, yet different information from the tracers.18F-FDG better reflected cell proliferation as measured by Ki-67 gene expression than 64Cu-ATSM.
Collapse
|