251
|
Bernhardt S, Bayerlová M, Vetter M, Wachter A, Mitra D, Hanf V, Lantzsch T, Uleer C, Peschel S, John J, Buchmann J, Weigert E, Bürrig KF, Thomssen C, Korf U, Beissbarth T, Wiemann S, Kantelhardt EJ. Proteomic profiling of breast cancer metabolism identifies SHMT2 and ASCT2 as prognostic factors. Breast Cancer Res 2017; 19:112. [PMID: 29020998 PMCID: PMC5637318 DOI: 10.1186/s13058-017-0905-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/22/2017] [Indexed: 01/03/2023] Open
Abstract
Background Breast cancer tumors are known to be highly heterogeneous and differences in their metabolic phenotypes, especially at protein level, are less well-understood. Profiling of metabolism-related proteins harbors the potential to establish new patient stratification regimes and biomarkers promoting individualized therapy. In our study, we aimed to examine the relationship between metabolism-associated protein expression profiles and clinicopathological characteristics in a large cohort of breast cancer patients. Methods Breast cancer specimens from 801 consecutive patients, diagnosed between 2009 and 2011, were investigated using reverse phase protein arrays (RPPA). Patients were treated in accordance with national guidelines in five certified German breast centers. To obtain quantitative expression data, 37 antibodies detecting proteins relevant to cancer metabolism, were applied. Hierarchical cluster analysis and individual target characterization were performed. Clustering results and individual protein expression patterns were associated with clinical data. The Kaplan-Meier method was used to estimate survival functions. Univariate and multivariate Cox regression models were applied to assess the impact of protein expression and other clinicopathological features on survival. Results We identified three metabolic clusters of breast cancer, which do not reflect the receptor-defined subtypes, but are significantly correlated with overall survival (OS, p ≤ 0.03) and recurrence-free survival (RFS, p ≤ 0.01). Furthermore, univariate and multivariate analysis of individual protein expression profiles demonstrated the central role of serine hydroxymethyltransferase 2 (SHMT2) and amino acid transporter ASCT2 (SLC1A5) as independent prognostic factors in breast cancer patients. High SHMT2 protein expression was significantly correlated with poor OS (hazard ratio (HR) = 1.53, 95% confidence interval (CI) = 1.10–2.12, p ≤ 0.01) and RFS (HR = 1.54, 95% CI = 1.16–2.04, p ≤ 0.01). High protein expression of ASCT2 was significantly correlated with poor RFS (HR = 1.31, 95% CI = 1.01–1.71, p ≤ 0.05). Conclusions Our data confirm the heterogeneity of breast tumors at a functional proteomic level and dissects the relationship between metabolism-related proteins, pathological features and patient survival. These observations highlight the importance of SHMT2 and ASCT2 as valuable individual prognostic markers and potential targets for personalized breast cancer therapy. Trial registration ClinicalTrials.gov, NCT01592825. Registered on 3 May 2012. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0905-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephan Bernhardt
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Michaela Bayerlová
- Department of Medical Statistics, University Medical Center Goettingen, Humboldtallee 32, 37073, Goettingen, Germany
| | - Martina Vetter
- Department of Gynaecology, Martin-Luther-University, Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Germany
| | - Astrid Wachter
- Department of Medical Statistics, University Medical Center Goettingen, Humboldtallee 32, 37073, Goettingen, Germany
| | - Devina Mitra
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Volker Hanf
- Department of Gynaecology, Hospital Fuerth, Jakob-Henle-Str. 1, 90768, Fuerth, Germany
| | - Tilmann Lantzsch
- Department of Gynaecology, Hospital St. Elisabeth and St. Barbara, Mauerstr. 5, 06110, Halle (Saale), Germany
| | - Christoph Uleer
- Onkologische Praxis Uleer, Bahnhofstr. 5, 31134, Hildesheim, Germany
| | - Susanne Peschel
- Department of Gynaecology, St. Bernward Hospital, Treibestr. 9, 31134, Hildesheim, Germany
| | - Jutta John
- Department of Gynaecology, Helios Hospital Hildesheim, Weinberg 1, 31134, Hildesheim, Germany
| | - Jörg Buchmann
- Institute of Pathology, Hospital Martha-Maria, Roentgenstraße 1, 06120, Halle (Saale), Germany
| | - Edith Weigert
- Institute of Pathology, Hospital Fuerth, Jakob-Henle-Str. 1, 90768, Fuerth, Germany
| | - Karl-Friedrich Bürrig
- Institute of Pathology Hildesheim, Senator-Braun-Allee 35, 31135, Hildesheim, Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University, Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Germany
| | - Ulrike Korf
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Tim Beissbarth
- Department of Medical Statistics, University Medical Center Goettingen, Humboldtallee 32, 37073, Goettingen, Germany.
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany.
| | - Eva Johanna Kantelhardt
- Department of Gynaecology, Martin-Luther-University, Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Germany. .,Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University, Halle-Wittenberg, Magdeburgerstr. 8, 06120, Halle (Saale), Germany.
| |
Collapse
|
252
|
Hashempour Alamdari N, Alaei-Beirami M, Sadat Shandiz SA, Hejazinia H, Rasouli R, Saffari M, Sadat Ebrahimi SE, Assadi A, Shafiee Ardestani M. Gd 3+-Asparagine-Anionic Linear Globular Dendrimer Second-Generation G2 Complexes: Novel Nanobiohybrid Theranostics. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:3625729. [PMID: 29097918 PMCID: PMC5635473 DOI: 10.1155/2017/3625729] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/15/2017] [Indexed: 01/06/2023]
Abstract
Designing a unique theranostic biocompatible, biodegradable, and cost-effective agent which is easy to be synthesized as a biohybrid material was the aim of this study. In this matter, asparagine attached to anionic linear globular dendrimer G2 (as a biocompatible, biodegradable, and cost-effective agent which is negatively charged nanosized and water soluble polymer that outweighs other traditionally used dendrimers) and finally contrast agent (Gd3+) was loaded (which made complexes) in synthesized asparagine-dendrimer. Observations revealed that, in addition to successful colon cancer and brain targeting, Gd3+-dendrimer-asparagine, the proposed theranostic agent, could increase T1 MR relaxation times, decrease T2 MR relaxation times significantly, and improve contrast of image as well as illustrating good cellular uptake based on florescent microscopy/flow cytometry and ICP-mass data. In addition to that, it increased tumor growth inhibition percentage (TGI%) significantly compared to FDA approved contrast agent, Magnevist. Totally, Gd3+-anionic linear globular dendrimer G2-asparagine could be introduced to the cancer imaging/therapy (theranostics) protocols after in vivo MR and fluorescent analysis and passing clinical trials. Hence, this nanotheranostic agent would be a promising candidate for brain drug delivery and imaging in the future.
Collapse
Affiliation(s)
- Nasim Hashempour Alamdari
- Department of Radiopharmacy and Medicinal Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahmood Alaei-Beirami
- Drug Applied Research Center and Students' Research Committee, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Hadi Hejazinia
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Rahimeh Rasouli
- Department of Medical Nanotechnology, School of Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Saffari
- Department of Medical Nanotechnology, School of Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Artin Assadi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
253
|
Zhan B, Wen S, Lu J, Shen G, Lin X, Feng J, Huang H. Identification and causes of metabonomic difference between orthotopic and subcutaneous xenograft of pancreatic cancer. Oncotarget 2017; 8:61264-61281. [PMID: 28977862 PMCID: PMC5617422 DOI: 10.18632/oncotarget.18057] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal tumors. However, the methodological differences between orthotopic and subcutaneous xenograft (OX and SX) models will cause confusion in understanding its pathological mechanism and clinical relevance. In this study, SX and OX models were established by implanting Panc-1 and BxPC-3 cell strains under skin and on the pancreas of mice, respectively. The tumor tissue and serum samples were collected for1H NMR spectroscopy followed by univariate and multivariate statistical analyses. As results, no obvious metabonomic difference was demonstrated in serum between the two models, however, the model- and cell strain-specific metabonomic differences were observed in tumor tissues. According to the KEGG analysis, ABC transporters, glycerophospholipid metabolism, purine metabolism and central carbon metabolism were identified to be the most significant components involved in metabonomic differences. Considering the methodological discrepancy in SX and OX models, such differences should be contributed to tumor microenvironment. In general, SX are not equivalent to OX models at molecular level. Subcutaneous transplantation displayed its inherent limitations though it offered a simple, inexpensive, reproducible and quantifiable advantage. And orthotopic transplantation may be favorable to simulate PDAC in patients due to its similar pathogenesis to human pancreatic cancer.
Collapse
Affiliation(s)
- Bohan Zhan
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jie Lu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Guiping Shen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| |
Collapse
|
254
|
Starkova J, Hermanova I, Hlozkova K, Hararova A, Trka J. Altered Metabolism of Leukemic Cells: New Therapeutic Opportunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 336:93-147. [PMID: 29413894 DOI: 10.1016/bs.ircmb.2017.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The cancer metabolic program alters bioenergetic processes to meet the higher demands of tumor cells for biomass production, nucleotide synthesis, and NADPH-balancing redox homeostasis. It is widely accepted that cancer cells mostly utilize glycolysis, as opposed to normal cells, in which oxidative phosphorylation is the most employed bioenergetic process. Still, studies examining cancer metabolism had been overlooked for many decades, and it was only recently discovered that metabolic alterations affect both the oncogenic potential and therapeutic response. Since most of the published works concern solid tumors, in this comprehensive review, we aim to summarize knowledge about the metabolism of leukemia cells. Leukemia is a malignant disease that ranks first and fifth in cancer-related deaths in children and adults, respectively. Current treatment has reached its limits due to toxicity, and there has been a need for new therapeutic approaches. One of the possible scenarios is improved use of established drugs and another is to introduce new druggable targets. Herein, we aim to describe the complexity of leukemia metabolism and highlight cellular processes that could be targeted therapeutically and enhance the effectiveness of current treatments.
Collapse
Affiliation(s)
- Julia Starkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Ivana Hermanova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Hlozkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alzbeta Hararova
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Trka
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
255
|
Carvalho-Cruz P, Alisson-Silva F, Todeschini AR, Dias WB. Cellular glycosylation senses metabolic changes and modulates cell plasticity during epithelial to mesenchymal transition. Dev Dyn 2017; 247:481-491. [PMID: 28722313 DOI: 10.1002/dvdy.24553] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/09/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a developmental program reactivated by tumor cells that leads to the switch from epithelial to mesenchymal phenotype. During EMT, cells are transcriptionally regulated to decrease E-cadherin expression while expressing mesenchymal markers such as vimentin, fibronectin, and N-cadherin. Growing body of evidences suggest that cells engaged in EMT undergo a metabolic reprograming process, redirecting glucose flux toward hexosamine biosynthesis pathway (HBP), which fuels aberrant glycosylation patterns that are extensively observed in cancer cells. HBP depends on nutrient availability to produce its end product UDP-GlcNAc, and for this reason is considered a metabolic sensor pathway. UDP-GlcNAc is the substrate used for the synthesis of major types of glycosylation, including O-GlcNAc and cell surface glycans. In general, the rate limiting enzyme of HBP, GFAT, is overexpressed in many cancer types that present EMT features as well as aberrant glycosylation. Moreover, altered levels of O-GlcNAcylation can modulate cell morphology and favor EMT. In this review, we summarize some of the current knowledge that correlates glucose metabolism, aberrant glycosylation and hyper O-GlcNAcylation supported by HBP that leads to EMT activation. Developmental Dynamics 247:481-491, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Patricia Carvalho-Cruz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Frederico Alisson-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane R Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wagner B Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
256
|
Fluorine-18 labeled amino acids for tumor PET/CT imaging. Oncotarget 2017; 8:60581-60588. [PMID: 28947996 PMCID: PMC5601164 DOI: 10.18632/oncotarget.19943] [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: 05/25/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022] Open
Abstract
Tumor glucose metabolism and amino acid metabolism are usually enhanced, 18F-FDG for tumor glucose metabolism PET imaging has been clinically well known, but tumor amino acid metabolism PET imaging is not clinically familiar. Radiolabeled amino acids (AAs) are an important class of PET/CT tracers that target the upregulated amino acid transporters to show elevated amino acid metabolism in tumor cells. Radiolabeled amino acids were observed to have high uptake in tumor cells but low in normal tissues and inflammatory tissues. The radionuclides used in labeling amino acids include 15O, 13N, 11C, 123I, 18F and 68Ga, among which the most commonly used is 18F [1]. Available data support the use of certain 18F-labeled AAs for PET/CT imaging of gliomas, neuroendocrine tumors, prostate cancer and breast cancer [2, 3]. With the progress of the method of 18F labeling AAs [4-6], 18F-labeled AAs are well established for tumor PET/CT imaging. This review focuses on the current status of key clinical applications of 18F-labeled AAs in tumor PET/CT imaging.
Collapse
|
257
|
Dong J, Xiao D, Zhao Z, Ren P, Li C, Hu Y, Shi J, Su H, Wang L, Liu H, Li B, Gao P, Qing G. Epigenetic silencing of microRNA-137 enhances ASCT2 expression and tumor glutamine metabolism. Oncogenesis 2017; 6:e356. [PMID: 28692032 PMCID: PMC5541711 DOI: 10.1038/oncsis.2017.59] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/16/2017] [Accepted: 05/29/2017] [Indexed: 02/07/2023] Open
Abstract
Tumor cells must activate specific transporters to meet their increased glutamine metabolic demands. Relative to other glutamine transporters, the ASC family transporter 2 (ASCT2, also called SLC1A5) is profoundly elevated in a wide spectrum of human cancers to coordinate metabolic reprogramming and malignant transformation. Understanding the molecular mechanisms whereby tumor cells frequently upregulate this transporter is therefore vital to develop potential strategies for transporter-targeted therapies. Combining in-silico algorithms with systemic experimental screening, we herein identify the tumor suppressor microRNA, miR-137, as an essential regulator that targets ASCT2 and cancer cell glutamine metabolism. Metabolic analysis shows that miR-137 derepression, similar to ASCT2 inactivation, significantly inhibits glutamine consumption and TCA cycle anaplerosis. Mechanistically, methyl-CpG-binding protein 2 (MeCP2) and DNA methyltransferases (DNMTs) cooperate to promote active methylation of the miR-137 promoter and inhibit its transcription, conversely reactivating ASCT2 expression and glutamine metabolism. Moreover, expression between miR-137 and ASCT2 is inversely correlated in tumor specimens from multiple cancer types, and ectopic ASCT2 expression markedly rescued miR-137 suppression of tumorigenesis. These findings thus elucidate a previously unreported mechanism responsible for ASCT2 deregulation in human cancers and identify ASCT2 as a critical downstream effector of miR-137, revealing a molecular link between DNA methylation, microRNA and tumor metabolism.
Collapse
Affiliation(s)
- J Dong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| | - D Xiao
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| | - Z Zhao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, China
| | - P Ren
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - C Li
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Y Hu
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| | - J Shi
- Union Hospital, Tongji Medical College, Huazhong University of Sicence and Technology, Wuhan, China
| | - H Su
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
- Union Hospital, Tongji Medical College, Huazhong University of Sicence and Technology, Wuhan, China
| | - L Wang
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| | - H Liu
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| | - B Li
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - P Gao
- Affiliated Dalian Sixth People’s Hospital, Dalian Medical University, Dalian, China
- Department of Biotechnology, Dalian Institute of Chemical Physics, Dalian, China
| | - G Qing
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cancer Biology, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
258
|
Foulkes MJ, Henry KM, Rougeot J, Hooper-Greenhill E, Loynes CA, Jeffrey P, Fleming A, Savage CO, Meijer AH, Jones S, Renshaw SA. Expression and regulation of drug transporters in vertebrate neutrophils. Sci Rep 2017; 7:4967. [PMID: 28694436 PMCID: PMC5504015 DOI: 10.1038/s41598-017-04785-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/19/2017] [Indexed: 12/16/2022] Open
Abstract
There remains a need to identify novel pro-resolution drugs for treatment of inflammatory disease. To date, there are no neutrophil-specific anti-inflammatory treatments in clinical use, perhaps due to our lack of understanding of how drugs access this complex cell type. Here we present the first comprehensive description and expression of both major classes of drug transporters, SLC and ABC, in resting human blood neutrophils. Moreover, we have studied the expression of these carriers in the tractable model system, the zebrafish (Danio rerio), additionally examining the evolutionary relationship between drug transporters in zebrafish and humans. We anticipate that this will be a valuable resource to the field of inflammation biology and will be an important asset in future anti-inflammatory drug design.
Collapse
Affiliation(s)
- Matthew J Foulkes
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Chemistry, The University of Sheffield, Sheffield, S3 7HF, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Katherine M Henry
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Julien Rougeot
- Institute of Biology, University of Leiden, 2333 CC, Leiden, The Netherlands
| | - Edward Hooper-Greenhill
- Immuno-Inflammation Therapy Area Unit, GlaxoSmithKline Research and Development Ltd., Stevenage, SG1 2NY, UK
| | - Catherine A Loynes
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Phil Jeffrey
- Rare Diseases Research Unit, Pfizer Ltd., Cambridge, CB21 6GP, UK
| | - Angeleen Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, CB2 2XY, UK
| | - Caroline O Savage
- Immuno-Inflammation Therapy Area Unit, GlaxoSmithKline Research and Development Ltd., Stevenage, SG1 2NY, UK
| | - Annemarie H Meijer
- Institute of Biology, University of Leiden, 2333 CC, Leiden, The Netherlands
| | - Simon Jones
- Department of Chemistry, The University of Sheffield, Sheffield, S3 7HF, UK
| | - Stephen A Renshaw
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK.
| |
Collapse
|
259
|
Abstract
Mutations of KRAS are found in a variety of human malignancies, including in pancreatic cancer, colorectal cancer, and non-small cell lung cancer at high frequency. To date, no effective treatments that target mutant variants of KRAS have been introduced into clinical practice. In recent years, a number of studies have shown that the oncogene KRAS plays a critical role in controlling cancer metabolism by orchestrating multiple metabolic changes. One of the metabolic hallmarks of malignant tumor cells is their dependency on aerobic glycolysis, known as the Warburg effect. The role of KRAS signaling in the regulation of aerobic glycolysis has been reported in several types of cancer. KRAS-driven cancers are characterized by altered metabolic pathways involving enhanced nutrients uptake, enhanced glycolysis, enhanced glutaminolysis, and elevated synthesis of fatty acids and nucleotides. However, Just how mutated KRAS can coordinate the metabolic shift to promote tumor growth and whether specific metabolic pathways are essential for the tumorigenesis of KRAS-driven cancers are questions which remain to be answered. In this context, the aim of this review is to summarize current data on KRAS-related metabolic alterations in cancer cells. Given that cancer cells rely on changes in metabolism to support their growth and survival, the targeting of metabolic processes may be a potential strategy for treating KRAS-driven cancers.
Collapse
Affiliation(s)
- Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Kosuke Toda
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
260
|
Kume E, Mutou T, Kansaku N, Takahashi H, Wempe MF, Ikegami M, Kanai Y, Endou H, Wakui S. Ultrastructural immunohistochemical study of L-type amino acid transporter 1-4F2 heavy chain in tumor microvasculatures of N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) induced rat bladder carcinoma. Microscopy (Oxf) 2017; 66:198-203. [PMID: 28339760 DOI: 10.1093/jmicro/dfx008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 02/15/2017] [Indexed: 06/06/2023] Open
Abstract
Angiogenesis is essential for tumor growth, and an enhanced vasculature supplying nutrients and oxygen might reflect malignant potential. L-type amino acid transporter 1 (LAT1/4F2hc) comprises a major nutrient transport system responsible for the Na+-independent transport of large neutral amino acids. Seventy five to seventy eight percent N-butyl-N-(4-hydroxybutyl) nitrosamine-induced rat bladder carcinoma cells showed high LAT1/4F2hc expression. While the intracarcinoma microvasculatures of fenestrated endothelial cells highly expressing LAT1/4F2hc might progressively transport essential amino acids from the microvasculatures to the extracellular matrix, non-fenestrated endothelial cells and pericytes did not. The present study revealed that the tumor angiogenesis is one of target anti-L-type amino acid transporter 1 drug.
Collapse
Affiliation(s)
- Eisuke Kume
- Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | | | - Norio Kansaku
- Azabu University School of Veterinary Medicine, Kanagawa, Japan
| | - Hitoyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Michael F Wempe
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshikatsu Kanai
- Department of Pharmacology, Osaka University Graduate School of Medicine, Osaka, Japan, and
| | | | - Shin Wakui
- Azabu University School of Veterinary Medicine, Kanagawa, Japan
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
261
|
Li SS, Loor JJ, Liu HY, Liu L, Hosseini A, Zhao WS, Liu JX. Optimal ratios of essential amino acids stimulate β-casein synthesis via activation of the mammalian target of rapamycin signaling pathway in MAC-T cells and bovine mammary tissue explants. J Dairy Sci 2017; 100:6676-6688. [PMID: 28571990 DOI: 10.3168/jds.2017-12681] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/17/2017] [Indexed: 12/26/2022]
Abstract
Amino acids are the building blocks of proteins and serve as key molecular components upstream of the signaling pathways that regulate protein synthesis. The objective of this study was to systematically investigate the effect of essential AA ratios on milk protein synthesis in vitro and to elucidate some of the underlying mechanisms. Triplicate cultures of MAC-T cells and bovine mammary tissue explants (MTE) were incubated with the optimal AA ratio (OPAA; Lys:Met, 2.9:1; Thr:Phe, 1.05:1; Lys:Thr, 1.8:1; Lys:His, 2.38:1; and Lys:Val, 1.23:1) in the presence of rapamycin (control), OPAA, a Lys:Thr ratio of 2.1:1, a Lys:Thr ratio of 1.3:1, a Lys:His ratio of 3.05:1, or a Lys:Val ratio of 1.62:1 for 12 h; the other AA concentrations were equal to OPAA. In some experiments, the cells were cultured with OPAA with or without rapamycin (100 ng/mL) or with mammalian target of rapamycin (mTOR) small interference RNA, and the MTE were exposed to OPAA with rapamycin for β-casein expression. Among the treatments, the expression of β-casein was greatest in the MTE cultured with OPAA. In MAC-T cells, the OPAA upregulated the mRNA expression of SLC1A5 and SLC7A5 but downregulated the expression of IRS1, AKT3, EEF1A1, and EEF2 compared with the control. The OPAA had no effect on the mTOR phosphorylation status but increased the phosphorylation of S6K1 and RPS6. When the MTE were treated with rapamycin in the presence of OPAA, the expression of β-casein was markedly decreased. The phosphorylation of RPS6 and 4EBP1 also was reduced in MAC-T cells. A similar negative effect on the expression of RPS6KB1 and EIF4EBP1 was detected when the cells were cultured with either rapamycin or mTOR small interference RNA. The optimal AA ratio stimulated β-casein expression partly by enhancing the transport of AA into the cells, cross-talk with insulin signaling and a subsequent enhancement of mTOR signaling, or translation elongation in both MAC-T cells and bovine MTE.
Collapse
Affiliation(s)
- S S Li
- Institute of Dairy Science, College of Animal Sciences, MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, P. R. China
| | - J J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| | - H Y Liu
- Institute of Dairy Science, College of Animal Sciences, MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, P. R. China
| | - L Liu
- Institute of Dairy Science, College of Animal Sciences, MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, P. R. China
| | - A Hosseini
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - W S Zhao
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - J X Liu
- Institute of Dairy Science, College of Animal Sciences, MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, P. R. China.
| |
Collapse
|
262
|
Chiu M, Sabino C, Taurino G, Bianchi MG, Andreoli R, Giuliani N, Bussolati O. GPNA inhibits the sodium-independent transport system l for neutral amino acids. Amino Acids 2017; 49:1365-1372. [DOI: 10.1007/s00726-017-2436-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/04/2017] [Indexed: 01/19/2023]
|
263
|
Essential Roles of L-Type Amino Acid Transporter 1 in Syncytiotrophoblast Development by Presenting Fusogenic 4F2hc. Mol Cell Biol 2017; 37:MCB.00427-16. [PMID: 28320871 DOI: 10.1128/mcb.00427-16] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/09/2017] [Indexed: 01/30/2023] Open
Abstract
The layers of the epithelial syncytium, i.e., syncytiotrophoblasts, differentiate from chorionic trophoblasts via cell fusion and separate maternal and fetal circulations in hemochorial placentas. L-type amino acid transporter 1 (LAT1) and its covalently linked ancillary subunit 4F2hc are colocalized on both maternal and fetal surfaces of syncytiotrophoblasts, implying their roles in amino acid transfer through the placental barrier. In this study, LAT1 knockout, in addition, revealed a novel role of LAT1 in syncytiotrophoblast development. LAT1 at midgestation was selectively expressed in trophoblastic lineages in the placenta, exclusively as a LAT1-4F2hc heterodimer. In LAT1 homozygous knockout mice, chorionic trophoblasts remained largely mononucleated, and the layers of syncytiotrophoblasts were almost completely absent. The amount of 4F2hc protein, which possesses a fusogenic function in trophoblastic cells, as well as in virus-infected cells, was drastically reduced by LAT1 knockout, with less affecting the mRNA level. Knockdown of LAT1 in trophoblastic BeWo cells also reduced 4F2hc protein and suppressed forskolin-induced cell fusion. These results demonstrate a novel fundamental role of LAT1 to support the protein expression of 4F2hc via a chaperone-like function in chorionic trophoblasts and to promote syncytiotrophoblast formation by contributing to cell fusion in the developing placenta.
Collapse
|
264
|
Masle-Farquhar E, Bröer A, Yabas M, Enders A, Bröer S. ASCT2 (SLC1A5)-Deficient Mice Have Normal B-Cell Development, Proliferation, and Antibody Production. Front Immunol 2017; 8:549. [PMID: 28553292 PMCID: PMC5427077 DOI: 10.3389/fimmu.2017.00549] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/24/2017] [Indexed: 12/12/2022] Open
Abstract
SLC1A5 (solute carrier family 1, member 5) is a small neutral amino acid exchanger that is upregulated in rapidly proliferating lymphocytes but also in many primary human cancers. Furthermore, cancer cell lines have been shown to require SLC1A5 for their survival in vitro. One of SLC1A5's primary substrates is the immunomodulatory amino acid glutamine, which plays an important role in multiple key processes, such as energy supply, macromolecular synthesis, nucleotide biosynthesis, redox homeostasis, and resistance against oxidative stress. These processes are also essential to immune cells, including neutrophils, macrophages, B and T lymphocytes. We show here that mice with a stop codon in Slc1a5 have reduced glutamine uptake in activated lymphocytes and primary fibroblasts. B and T cell populations and maturation in resting mice were not affected by absence of SLC1A5. Antibody production in resting and immunized mice and the germinal center response to immunization were also found to be normal. SLC1A5 has been recently described as a novel target for the treatment of a variety of cancers, and our results indicate that inhibition of SLC1A5 in cancer therapy may be tolerated well by the immune system of cancer patients.
Collapse
Affiliation(s)
- Etienne Masle-Farquhar
- Research School of Biology, The Australian National University, Canberra, ACT, Australia.,Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Angelika Bröer
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Mehmet Yabas
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Anselm Enders
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
265
|
Schuster DM, Nanni C, Fanti S. Evaluation of Prostate Cancer with Radiolabeled Amino Acid Analogs. J Nucl Med 2017; 57:61S-66S. [PMID: 27694174 DOI: 10.2967/jnumed.115.170209] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
Abstract
Conventional imaging of prostate cancer has limitations related to the frequently indolent biology of the disease. PET is a functional imaging method that can exploit various aspects of tumor biology to enable greater detection of prostate cancer than can be provided by morphologic imaging alone. Radiotracers that are in use or under investigation for targeting salient features of prostate cancer include those directed to glucose, choline, acetate, prostate-specific membrane antigen, bombesin, and amino acids. The tumor imaging features of this last class of radiotracers mirror the upregulation of transmembrane amino acid transport that is necessary in carcinomas because of increased amino acid use for energy requirements and protein synthesis. Natural and synthetic amino acids radiolabeled for PET imaging have been investigated in prostate cancer patients. Early work with naturally occurring amino acid-derived radiotracers, such as l-11C-methionine and l-1-11C-5-hydroxytryptophan, demonstrated promising results, including greater sensitivity than 18F-FDG for intraprostatic and extraprostatic cancer detection. However, limitations with naturally occurring amino acid-derived compounds, including metabolism of the radiotracer itself, led to the development of synthetic amino acid radiotracers, which are not metabolized and therefore more accurately reflect transmembrane amino acid transport. Of the synthetic amino acid-derived PET radiotracers, anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (18F-FACBC or 18F-fluciclovine) has undergone the most promising translation to human use, including the availability of simplified radiosynthesis. Several studies have indicated advantageous biodistribution in the abdomen and pelvis with little renal excretion and bladder activity-characteristics beneficial for prostate cancer imaging. Studies have demonstrated improved lesion detection and diagnostic performance of 18F-fluciclovine in comparison with conventional imaging, especially for recurrent prostate cancer, although issues with nonspecific uptake limit the potential role of 18F-fluciclovine in the diagnosis of primary prostate cancer. Although work is ongoing, recently published intrapatient comparisons of 18F-fluciclovine with 11C-choline reported higher overall diagnostic performance of the former, especially for the detection of disease relapse. This review is aimed at providing a detailed overview of amino acid-derived PET compounds that have been studied for use in prostate cancer imaging.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
266
|
Fasting Enhances the Contrast of Bone Metastatic Lesions in 18F-Fluciclovine-PET: Preclinical Study Using a Rat Model of Mixed Osteolytic/Osteoblastic Bone Metastases. Int J Mol Sci 2017; 18:ijms18050934. [PMID: 28468238 PMCID: PMC5454847 DOI: 10.3390/ijms18050934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 12/21/2022] Open
Abstract
18F-fluciclovine (trans-1-amino-3-18F-fluorocyclobutanecarboxylic acid) is an amino acid positron emission tomography (PET) tracer used for cancer staging (e.g., prostate and breast). Patients scheduled to undergo amino acid-PET are usually required to fast before PET tracer administration. However, there have been no reports addressing whether fasting improves fluciclovine-PET imaging. In this study, the authors investigated the influence of fasting on fluciclovine-PET using triple-tracer autoradiography with 14C-fluciclovine, [5,6-3H]-2-fluoro-2-deoxy-d-glucose (3H-FDG), and 99mTc-hydroxymethylene diphosphonate (99mTc-HMDP) in a rat breast cancer model of mixed osteolytic/osteoblastic bone metastases in which the animals fasted overnight. Lesion accumulation of each tracer was evaluated using the target-to-background (muscle) ratio. The mean ratios of 14C-fluciclovine in osteolytic lesions were 4.6 ± 0.8 and 2.8 ± 0.6, respectively, with and without fasting, while those for 3H-FDG were 6.9 ± 2.5 and 5.1 ± 2.0, respectively. In the peri-tumor bone formation regions (osteoblastic), where 99mTc-HMDP accumulated, the ratios of 14C-fluciclovine were 4.3 ± 1.4 and 2.4 ± 0.7, respectively, and those of 3H-FDG were 6.2 ± 3.8 and 3.3 ± 2.2, respectively, with and without fasting. These results suggest that fasting before 18F-fluciclovine-PET improves the contrast between osteolytic and osteoblastic bone metastatic lesions and background, as well as 18F-FDG-PET.
Collapse
|
267
|
Novel insights into the transport mechanism of the human amino acid transporter LAT1 (SLC7A5). Probing critical residues for substrate translocation. Biochim Biophys Acta Gen Subj 2017; 1861:727-736. [DOI: 10.1016/j.bbagen.2017.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/21/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
|
268
|
Abstract
Liver cancer is fundamentally physiologically different from the surrounding liver tissue. Despite multiple efforts to target the altered signaling pathways created by oncogenic mutations, not many have focused on targeting the altered metabolism that allows liver cancer to develop and grow. Still to be resolved is the question of whether the altered metabolic pathways in this cancer differ enough from the surrounding noncancerous cells to allow for the development of potent and specific compounds. Clinical studies of metabolic modulators would provide some more information with regard to the feasibility of this approach. Furthermore, as it appears that oncogenic signaling is essential to this cancer's altered metabolism, it stands to reason that targeting this altered signaling may allow the exploitation of specific metabolic vulnerabilities in combination with other drugs for enhanced efficacy. The identification of biomarkers of metabolic sensitivity will also be essential to determine whether these drugs will have the desired effect.
Collapse
Affiliation(s)
- Ali Zarrinpar
- 1 Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
269
|
Nodwell MB, Yang H, Čolović M, Yuan Z, Merkens H, Martin RE, Bénard F, Schaffer P, Britton R. 18F-Fluorination of Unactivated C-H Bonds in Branched Aliphatic Amino Acids: Direct Synthesis of Oncological Positron Emission Tomography Imaging Agents. J Am Chem Soc 2017; 139:3595-3598. [PMID: 28248493 DOI: 10.1021/jacs.6b11533] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A mild and selective photocatalytic C-H 18F-fluorination reaction has been developed that provides direct access to 18F-fluorinated amino acids. The biodistribution and uptake of three 18F-labeled leucine analogues via LAT1 mediated transport in several cancer cell lines is reported. Positron emission tomography imaging of mice bearing PC3 (prostate) or U87 (glioma) xenografts using 5-[18F]-fluorohomoleucine showed high tumor uptake and excellent tumor visualization, highlighting the utility of this strategy for rapid tracer discovery for oncology.
Collapse
Affiliation(s)
- Matthew B Nodwell
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Milena Čolović
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Zheliang Yuan
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Rainer E Martin
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd , Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Paul Schaffer
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| |
Collapse
|
270
|
Lévy PL, Duponchel S, Eischeid H, Molle J, Michelet M, Diserens G, Vermathen M, Vermathen P, Dufour JF, Dienes HP, Steffen HM, Odenthal M, Zoulim F, Bartosch B. Hepatitis C virus infection triggers a tumor-like glutamine metabolism. Hepatology 2017; 65:789-803. [PMID: 27863447 DOI: 10.1002/hep.28949] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/26/2016] [Accepted: 11/17/2016] [Indexed: 01/02/2023]
Abstract
UNLABELLED Chronic infection with hepatitis C virus (HCV) is one of the main causes of hepatocellular carcinoma. However, the molecular mechanisms linking the infection to cancer development remain poorly understood. Here we used HCV-infected cells and liver biopsies to study how HCV modulates the glutaminolysis pathway, which is known to play an important role in cellular energetics, stress defense, and neoplastic transformation. Transcript levels of glutaminolytic factors were quantified in Huh7.5 cells or primary human hepatocytes infected with the Japanese fulminant hepatitis 1 HCV strain as well as in biopsies of chronic HCV patients. Nutrient deprivation, biochemical analysis, and metabolite quantification were performed with HCV-infected Huh7.5 cells. Furthermore, short hairpin RNA vectors and small molecule inhibitors were used to investigate the dependence of HCV replication on metabolic changes. We show that HCV modulates the transcript levels of key enzymes of glutamine metabolism in vitro and in liver biopsies of chronic HCV patients. Consistently, HCV infection increases glutamine use and dependence. We finally show that inhibiting glutamine metabolism attenuates HCV infection and the oxidative stress associated with HCV infection. CONCLUSION Our data suggest that HCV establishes glutamine dependence, which is required for viral replication, and, importantly, that glutamine addiction is a hallmark of tumor cells. While HCV induces glutaminolysis to create an environment favorable for viral replication, it predisposes the cell to transformation. Glutaminolytic enzymes may be interesting therapeutic targets for prevention of hepatocarcinogenesis in chronic hepatitis C. (Hepatology 2017;65:789-803).
Collapse
Affiliation(s)
- Pierre L Lévy
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France
| | - Sarah Duponchel
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France.,Hospices Civils de Lyon, Lyon, France
| | - Hannah Eischeid
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Jennifer Molle
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France
| | - Maud Michelet
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France
| | - Gaëlle Diserens
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Martina Vermathen
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Peter Vermathen
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | - Hans-Peter Dienes
- Institute of Pathology, University of Cologne, Cologne, Germany.,Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Fabien Zoulim
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France.,Hospices Civils de Lyon, Lyon, France
| | - Birke Bartosch
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, Lyon, France
| |
Collapse
|
271
|
Liu W, Chen H, Wong N, Haynes W, Baker CM, Wang X. Pseudohypoxia induced by miR-126 deactivation promotes migration and therapeutic resistance in renal cell carcinoma. Cancer Lett 2017; 394:65-75. [PMID: 28257806 DOI: 10.1016/j.canlet.2017.02.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 12/19/2022]
Abstract
Pseudohypoxia plays a central role in the progression and therapeutic resistance of clear cell renal cell carcinoma (ccRCC); however, the underlying mechanisms are poorly understood. MicroRNA miR-126 has decreased expression in metastatic or relapsed ccRCC as compared to primary tumors, but the mechanisms by which miR-126 is implicated in RCC remain unknown. Through RNA-seq profiling to evaluate the impact of overexpression or CRISPR knockout of miR-126, we have identified SERPINE1 as a miR-126-5p target regulating cell motility, and SLC7A5 as a miR-126-3p target regulating the mTOR/HIF pathway. Specifically, miR-126 inhibits HIFα protein expression independent of von Hippel-Lindau tumor suppressor (VHL). On the other hand, deactivation of miR-126 induces a pseudohypoxia state due to increased HIFα expression, which further enhances therapeutic resistance and cell motility mediated by SLC7A5 and SERPINE1, respectively. Finally, the clinical relevance of miR-126 modulated gene regulation in ccRCC has been confirmed with profiling data from The Cancer Genome Atlas.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- CRISPR-Cas Systems
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/therapy
- Cell Movement/drug effects
- Cell Movement/radiation effects
- Cisplatin/pharmacology
- Computational Biology
- Databases, Genetic
- Dose-Response Relationship, Drug
- Dose-Response Relationship, Radiation
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- HEK293 Cells
- HeLa Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/therapy
- Large Neutral Amino Acid-Transporter 1/genetics
- Large Neutral Amino Acid-Transporter 1/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplasm Invasiveness
- Plasminogen Activator Inhibitor 1/genetics
- Plasminogen Activator Inhibitor 1/metabolism
- RNA Interference
- Radiation Tolerance
- Signal Transduction/drug effects
- Signal Transduction/radiation effects
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Transfection
- Tumor Hypoxia
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Von Hippel-Lindau Tumor Suppressor Protein/metabolism
Collapse
Affiliation(s)
- Weijun Liu
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hanxiang Chen
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathan Wong
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wesley Haynes
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Callie M Baker
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaowei Wang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
272
|
Kagawa S, Nishii R, Higashi T, Yamauchi H, Ogawa E, Okudaira H, Kobayashi M, Yoshimoto M, Shikano N, Kawai K. Relationship between [ 14C]MeAIB uptake and amino acid transporter family gene expression levels or proliferative activity in a pilot study in human carcinoma cells: Comparison with [ 3H]methionine uptake. Nucl Med Biol 2017; 49:8-15. [PMID: 28284101 DOI: 10.1016/j.nucmedbio.2017.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION To clarify the difference between system A and L amino acid transport imaging in PET clinical imaging, we focused on the use of α-[N-methyl-11C]-methylaminoisobutyric acid ([11C]MeAIB), and compared it with [S-methyl-11C]-L-methionine ([11C]MET). The aim of this study was to assess the correlation of accumulation of these two radioactive amino acid analogs with expression of amino acid transporters and cell proliferative activity in carcinoma cells. METHODS Amino acid uptake inhibitor studies were performed in four human carcinoma cells (epidermal carcinoma A431, colorectal carcinoma LS180, and lung carcinomas PC14/GL and H441/GL) using the radioisotope analogs [3H]MET and [14C]MeAIB. MeAIB was used to inhibit the A system and 2-amino-2-norbornane-carboxylic acid (BCH) was used to inhibit the L system. The carcinoma gene expression levels of a number of amino acid transporters were measured by microarray and quantitative polymerase chain reaction. Carcinoma proliferative activity was assessed using accumulation of [methyl-3H]-3'-deoxy-3'-fluorothymidine ([3H]FLT). RESULTS AND CONCLUSION [14C]MeAIB uptake occurred principally via a Na+-dependent A type mechanism whereas [3H]MET uptake occurred predominantly via a Na+-independent L type mechanism although other transporters were also utilized depending on cell type. There was no correlation between [3H]MET uptake and total system L amino acid transporter (LAT) expression. In contrast, [14C]MeAIB uptake strongly correlated with total system A amino acid transporter (SNAT) expression and proliferative activity in this preliminary study using four human carcinoma cell lines. Carcinoma proliferative activity also correlated with total SNAT expression. Advances in Knowledge and Implications for Patient Care: Because there is a significant correlation between the accumulation of [14C]MeAIB and the gene expression level of total SNAT as well as the accumulation of [3H]FLT, it is suggested that use of the analog [11C]MeAIB in PET may provide an indication of tumor cell proliferative activity. [11C]MeAIB is therefore expected to be very useful in PET imaging.
Collapse
Affiliation(s)
- Shinya Kagawa
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Ryuichi Nishii
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Tatsuya Higashi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroshi Yamauchi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan
| | - Emi Ogawa
- Department of Radiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | | | - Masato Kobayashi
- Wellness Promotion Science Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Naoto Shikano
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, Ibaraki, Japan
| | - Keiichi Kawai
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
273
|
Abstract
Prostate cancer is the most common cancer and the second leading cause of cancer death in men in the United States. Despite high disease prevalence, diagnosis and surveillance of the disease with conventional imaging are limited typically because of indolent biology. Functional imaging with advanced molecular techniques improves the ability to detect disease. Amino acids are building blocks of proteins, and intracellular transport of amino acids is upregulated in prostate cancer. This review provides a detailed overview of the use of F-18 fluciclovine PET in prostate cancer imaging.
Collapse
|
274
|
Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches. Oncogene 2017; 36:3359-3374. [PMID: 28092669 PMCID: PMC5485177 DOI: 10.1038/onc.2016.485] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023]
Abstract
Epigenetic and metabolic alterations in cancer cells are highly intertwined. Oncogene-driven metabolic rewiring modifies the epigenetic landscape via modulating the activities of DNA and histone modification enzymes at the metabolite level. Conversely, epigenetic mechanisms regulate the expression of metabolic genes, thereby altering the metabolome. Epigenetic-metabolomic interplay has a critical role in tumourigenesis by coordinately sustaining cell proliferation, metastasis and pluripotency. Understanding the link between epigenetics and metabolism could unravel novel molecular targets, whose intervention may lead to improvements in cancer treatment. In this review, we summarized the recent discoveries linking epigenetics and metabolism and their underlying roles in tumorigenesis; and highlighted the promising molecular targets, with an update on the development of small molecule or biologic inhibitors against these abnormalities in cancer.
Collapse
|
275
|
Watanabe Y, Kurihara H, Itami J, Sasaki R, Arai Y, Sugimura K. Relationship between the uptake of 18F-borono-L-phenylalanine and L-[methyl- 11C] methionine in head and neck tumors and normal organs. Radiat Oncol 2017; 12:17. [PMID: 28088230 PMCID: PMC5237505 DOI: 10.1186/s13014-017-0763-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE The purpose of this study was to determine the distribution of 4-borono-2-18F-fluoro-phenylalanine (18F-BPA) and L-[methyl-11C] methionine (11C-Met) in normal organs and tumors and to evaluate the usefulness of 11C-Met/PET in screening potential candidates for boron neutron capture therapy (BNCT). MATERIAL METHODS Seven patients who had at least one histologically confirmed head and neck tumor were included in this study. They underwent both whole-body 18F-BPA-PET/CT and 11C-Met-PET/CT within a span of 6 months. Uptake was evaluated using the maximum standardized uptake value (SUVmax). Regions of interest (ROIs) were placed within the tumors and target organs of brain, thyroid, submandibular gland, lung, liver, esophagus, stomach pancreas, spleen, muscle, and bone marrow. RESULTS The tumor SUVmax of FBPA and 11C-Met showed strong correlation (r 2 = 0.72, P = 0.015). Although 18F-BPA and 11C-Met showed markedly different uptake in some organs (submandibular gland, liver, heart, stomach pancreas, spleen, and bone marrow), the uptake of 11C-Met was consistently higher than that of 18F-BPA in these cases. CONCLUSION 11C-Met PET/CT might be used instead of 18F-BPA PET/CT to predict the accumulation of 10B in tumors and to select candidates for BNCT. However, it would not be suitable for evaluating accumulation in some normal organs. Therefore, the 18F-BPA-PET study remains a prerequisite for BNCT. This is the first report of the correlation between 18F-BPA and 11C-Met accumulation.
Collapse
Affiliation(s)
- Yoshiaki Watanabe
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan.,Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Jun Itami
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Yasuaki Arai
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuro Sugimura
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
276
|
Hellsten SV, Lekholm E, Ahmad T, Fredriksson R. The gene expression of numerous SLC transporters is altered in the immortalized hypothalamic cell line N25/2 following amino acid starvation. FEBS Open Bio 2017; 7:249-264. [PMID: 28174690 PMCID: PMC5292668 DOI: 10.1002/2211-5463.12181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 12/20/2022] Open
Abstract
Amino acids are known to play a key role in gene expression regulation, and in mammalian cells, amino acid signaling is mainly mediated via two pathways, the mammalian target of rapamycin complex 1 (mTORC1) pathway and the amino acid responsive (AAR) pathway. It is vital for cells to have a system to sense amino acid levels, in order to control protein and amino acid synthesis and catabolism. Amino acid transporters are crucial in these pathways, due to both their sensing and transport functions. In this large-scale study, an immortalized mouse hypothalamic cell line (N25/2) was used to study the gene expression changes following 1, 2, 3, 5 or 16 h of amino acid starvation. We focused on genes encoding solute carriers (SLCs) and putative SLCs, more specifically on amino acid transporters. The microarray contained 28 270 genes and 86.2% of the genes were expressed in the cell line. At 5 h of starvation, 1001 genes were upregulated and 848 genes were downregulated, and among these, 47 genes from the SLC superfamily or atypical SLCs were found. Of these, 15 were genes encoding amino acid transporters and 32 were genes encoding other SLCs or atypical SLCs. Increased expression was detected for genes encoding amino acid transporters from system A, ASC, L, N, T, xc-, and y+. Using GO annotations, genes involved in amino acid transport and amino acid transmembrane transporter activity were found to be most upregulated at 3 h and 5 h of starvation.
Collapse
Affiliation(s)
- Sofie V Hellsten
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden; Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Emilia Lekholm
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| | - Tauseef Ahmad
- Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| |
Collapse
|
277
|
Rodríguez A, Ortega A. Glutamine/Glutamate Transporters in Glial Cells: Much More Than Participants of a Metabolic Shuttle. ADVANCES IN NEUROBIOLOGY 2017; 16:169-183. [PMID: 28828610 DOI: 10.1007/978-3-319-55769-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Glial glutamine and glutamate transporters play an important role in glial/neuronal interactions. An excellent model to establish the role of these membrane proteins is the cerebellum. The most abundant glutamatergic synapse in the central nervous system is present in the molecular layer of the cerebellar cortex, and it is entirely wrapped by Bergmann glial cells. The recycling of glutamate involves glutamate and glutamine transporters enriched in these radial glial processes. The functional properties of amino acid glial transporters allow, in an activity-dependent manner, the conformation of protein complexes important for the adequate support of glutamatergic neurotransmission. A detailed description of the most important features of glial glutamate and glutamine transporters follows, and a working model of the molecular mechanisms by which these glutamate and glutamine binding proteins interact, and by these means might modulate cerebellar glutamatergic transactions, is presented.
Collapse
Affiliation(s)
- Angelina Rodríguez
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro, México
| | - Arturo Ortega
- Departamento de Toxicología, Cinvestav-IPN, Apartado Postal 14-740, México, DF, 07360, México.
| |
Collapse
|
278
|
Determination of amino acids in colon cancer cells by using UHPLC-MS/MS and [U-13C5]-glutamine as the isotope tracer. Talanta 2017; 162:285-292. [DOI: 10.1016/j.talanta.2016.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/24/2016] [Accepted: 10/02/2016] [Indexed: 12/26/2022]
|
279
|
Structure activity relationships of benzylproline-derived inhibitors of the glutamine transporter ASCT2. Bioorg Med Chem Lett 2016; 27:398-402. [PMID: 28057420 DOI: 10.1016/j.bmcl.2016.12.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 11/20/2022]
Abstract
The glutamine transporter ASCT2 has been identified as a promising target to inhibit rapid growth of cancer cells. However, ASCT2 pharmacology is not well established. In this report, we performed a systematic structure activity analysis of a series of substituted benzylproline derivatives. Substitutions on the phenyl ring resulted in compounds with characteristics of ASCT2 inhibitors. Apparent binding affinity increased with increasing hydrophobicity of the side chain. In contrast, interaction of the ASCT2 binding site with specific positions on the phenyl ring was not observed. The most potent compound inhibits the ASCT2 anion conductance with a Ki of 3μM, which is in the same range as that of more bulky and higher molecular weight inhibitors recently reported by others. The experimental results are consistent with computational analysis based on docking of the inhibitors against an ASCT2 homology model. The benzylproline scaffold provides a valuable tool for further improving binding potency of future ASCT2 inhibitors.
Collapse
|
280
|
Hirasawa K, Moriya S, Miyahara K, Kazama H, Hirota A, Takemura J, Abe A, Inazu M, Hiramoto M, Tsukahara K, Miyazawa K. Macrolide Antibiotics Exhibit Cytotoxic Effect under Amino Acid-Depleted Culture Condition by Blocking Autophagy Flux in Head and Neck Squamous Cell Carcinoma Cell Lines. PLoS One 2016; 11:e0164529. [PMID: 27977675 PMCID: PMC5158196 DOI: 10.1371/journal.pone.0164529] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/27/2016] [Indexed: 01/07/2023] Open
Abstract
Autophagy, a self-digestive system for cytoplasmic components, is required to maintain the amino acid pool for cellular homeostasis. We previously reported that the macrolide antibiotics azithromycin (AZM) and clarithromycin (CAM) have an inhibitory effect on autophagy flux, and they potently enhance the cytocidal effect of various anticancer reagents in vitro. This suggests that macrolide antibiotics can be used as an adjuvant for cancer chemotherapy. Since cancer cells require a larger metabolic demand than normal cells because of their exuberant growth, upregulated autophagy in tumor cells has now become the target for cancer therapy. In the present study, we examined whether macrolides exhibit cytotoxic effect under an amino acid-starving condition in head and neck squamous cancer cell lines such as CAL 27 and Detroit 562 as models of solid tumors with an upregulated autophagy in the central region owing to hypovascularity. AZM and CAM induced cell death under the amino acid-depleted (AAD) culture condition in these cell lines along with CHOP upregulation, although they showed no cytotoxicity under the complete culture medium. CHOP knockdown by siRNA in the CAL 27 cells significantly suppressed macrolide-induced cell death under the AAD culture condition. CHOP-/- murine embryonic fibroblast (MEF) cell lines also attenuated AZM-induced cell death compared with CHOP+/+ MEF cell lines. Using a tet-off atg5 MEF cell line, knockout of atg5, an essential gene for autophagy, also induced cell death and CHOP in the AAD culture medium but not in the complete culture medium. This suggest that macrolide-induced cell death via CHOP induction is dependent on autophagy inhibition. The cytotoxicity of macrolide with CHOP induction was completely cancelled by the addition of amino acids in the culture medium, indicating that the cytotoxicity is due to the insufficient amino acid pool. These data suggest the possibility of using macrolides for "tumor-starving therapy".
Collapse
Affiliation(s)
- Kazuhiro Hirasawa
- Department of Otolaryngology (Head and Neck Surgery), Tokyo Medical University, Tokyo, Japan
| | - Shota Moriya
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Kana Miyahara
- Department of Breast Surgery, Tokyo Medical University, Tokyo, Japan
| | - Hiromi Kazama
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Ayako Hirota
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Jun Takemura
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Akihisa Abe
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Masato Inazu
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Kiyoaki Tsukahara
- Department of Otolaryngology (Head and Neck Surgery), Tokyo Medical University, Tokyo, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
281
|
Khosravian P, Shafiee Ardestani M, Khoobi M, Ostad SN, Dorkoosh FA, Akbari Javar H, Amanlou M. Mesoporous silica nanoparticles functionalized with folic acid/methionine for active targeted delivery of docetaxel. Onco Targets Ther 2016; 9:7315-7330. [PMID: 27980423 PMCID: PMC5144897 DOI: 10.2147/ott.s113815] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mesoporous silica nanoparticles (MSNs) are known as carriers with high loading capacity and large functionalizable surface area for target-directed delivery. In this study, a series of docetaxel-loaded folic acid- or methionine-functionalized mesoporous silica nanoparticles (DTX/MSN-FA or DTX/MSN-Met) with large pores and amine groups at inner pore surface properties were prepared. The results showed that the MSNs were successfully synthesized, having good pay load and pH-sensitive drug release kinetics. The cellular investigation on MCF-7 cells showed better performance of cytotoxicity and cell apoptosis and an increase in cellular uptake of targeted nanoparticles. In vivo fluorescent imaging on healthy BALB/c mice proved that bare MSN-NH2 are mostly accumulated in the liver but MSN-FA or MSN-Met are more concentrated in the kidney. Importantly, ex vivo fluorescent images of tumor-induced BALB/c mice organs revealed the ability of MSN-FA to reach the tumor tissues. In conclusion, DTX/MSNs exhibited a good anticancer activity and enhanced the possibility of targeted drug delivery for breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences Research Center
- Drug Design and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
282
|
May glutamine addiction drive the delivery of antitumor cisplatin-based Pt(IV) prodrugs? J Inorg Biochem 2016; 167:27-35. [PMID: 27898344 DOI: 10.1016/j.jinorgbio.2016.11.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/09/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022]
Abstract
A small series of Pt(IV) prodrugs containing Gln-like (Gln=glutamine) axial ligands has been designed with the aim to take advantage of the increased demand of Gln showed by some cancer cells (glutamine addiction). In complex 4 the Gln, linked through the α-carboxylic group is recognized by the Gln transporters, in particular by the solute carrier transporter SLC1A5. All compounds showed cellular accumulation, as well as antiproliferative activity, related to their lipophilicity, as already demonstrated for the majority of Pt(IV) prodrugs, that enter cells mainly by passive diffusion. On the contrary, when the Gln concentration in cell medium is near or lower to the physiological value, complex 4 acts as a Trojan horse: it enters SLC1A5-overexpressing cells, where, upon reduction, it releases the active metabolite cisplatin and the Gln-containing ligand, thus preventing any possible extrusion by the L-type amino acid transporter LAT1. This selective mechanism could decrease off-target accumulation of 4 and, consequently, Pt-associated side-effects.
Collapse
|
283
|
Deshmukh A, Deshpande K, Arfuso F, Newsholme P, Dharmarajan A. Cancer stem cell metabolism: a potential target for cancer therapy. Mol Cancer 2016; 15:69. [PMID: 27825361 PMCID: PMC5101698 DOI: 10.1186/s12943-016-0555-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/01/2016] [Indexed: 12/19/2022] Open
Abstract
Cancer Stem cells (CSCs) are a unipotent cell population present within the tumour cell mass. CSCs are known to be highly chemo-resistant, and in recent years, they have gained intense interest as key tumour initiating cells that may also play an integral role in tumour recurrence following chemotherapy. Cancer cells have the ability to alter their metabolism in order to fulfil bio-energetic and biosynthetic requirements. They are largely dependent on aerobic glycolysis for their energy production and also are associated with increased fatty acid synthesis and increased rates of glutamine utilisation. Emerging evidence has shown that therapeutic resistance to cancer treatment may arise due to dysregulation in glucose metabolism, fatty acid synthesis, and glutaminolysis. To propagate their lethal effects and maintain survival, tumour cells alter their metabolic requirements to ensure optimal nutrient use for their survival, evasion from host immune attack, and proliferation. It is now evident that cancer cells metabolise glutamine to grow rapidly because it provides the metabolic stimulus for required energy and precursors for synthesis of proteins, lipids, and nucleic acids. It can also regulate the activities of some of the signalling pathways that control the proliferation of cancer cells. This review describes the key metabolic pathways required by CSCs to maintain a survival advantage and highlights how a combined approach of targeting cellular metabolism in conjunction with the use of chemotherapeutic drugs may provide a promising strategy to overcome therapeutic resistance and therefore aid in cancer therapy.
Collapse
Affiliation(s)
- Abhijeet Deshmukh
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Kedar Deshpande
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia.
| |
Collapse
|
284
|
Timosenko E, Ghadbane H, Silk JD, Shepherd D, Gileadi U, Howson LJ, Laynes R, Zhao Q, Strausberg RL, Olsen LR, Taylor S, Buffa FM, Boyd R, Cerundolo V. Nutritional Stress Induced by Tryptophan-Degrading Enzymes Results in ATF4-Dependent Reprogramming of the Amino Acid Transporter Profile in Tumor Cells. Cancer Res 2016; 76:6193-6204. [PMID: 27651314 PMCID: PMC5096689 DOI: 10.1158/0008-5472.can-15-3502] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023]
Abstract
Tryptophan degradation is an immune escape strategy shared by many tumors. However, cancer cells' compensatory mechanisms remain unclear. We demonstrate here that a shortage of tryptophan caused by expression of indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) resulted in ATF4-dependent upregulation of several amino acid transporters, including SLC1A5 and its truncated isoforms, which in turn enhanced tryptophan and glutamine uptake. Importantly, SLC1A5 failed to be upregulated in resting human T cells kept under low tryptophan conditions but was enhanced upon cognate antigen T-cell receptor engagement. Our results highlight key differences in the ability of tumor and T cells to adapt to tryptophan starvation and provide important insights into the poor prognosis of tumors coexpressing IDO and SLC1A5. Cancer Res; 76(21); 6193-204. ©2016 AACR.
Collapse
Affiliation(s)
- Elina Timosenko
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hemza Ghadbane
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jonathan D Silk
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Dawn Shepherd
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lauren J Howson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robert Laynes
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Qi Zhao
- Ludwig Cancer Research, New York, New York
| | | | - Lars R Olsen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Stephen Taylor
- The Computational Biology Research Group (CBRG), Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Francesca M Buffa
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom
| | - Richard Boyd
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
285
|
Abstract
Conventional anatomical imaging with CT and MRI has limitations in the evaluation of prostate cancer. PET is a powerful imaging technique, which can be directed toward molecular targets as diverse as glucose metabolism, density of prostate-specific membrane antigen receptors, and skeletal osteoblastic activity. Although 2-deoxy-2-18F-FDG-PET is the mainstay of molecular imaging, FDG has limitations in typically indolent prostate cancer. Yet, there are many useful and emerging PET tracers beyond FDG, which provide added value. These include radiotracers interrogating prostate cancer via molecular mechanisms related to the biology of choline, acetate, amino acids, bombesin, and dihydrotestosterone, among others. Choline is used for cell membrane synthesis and its metabolism is upregulated in prostate cancer. 11C-choline and 18F-choline are in wide clinical use outside the United States, and they have proven most beneficial for detection of recurrent prostate cancer. 11C-acetate is an indirect biomarker of fatty acid synthesis, which is also upregulated in prostate cancer. Imaging of prostate cancer with 11C-acetate is overall similar to the choline radiotracers yet is not as widely used. Upregulation of amino acid transport in prostate cancer provides the biologic basis for amino acid-based radiotracers. Most recent progress has been made with the nonnatural alicyclic amino acid analogue radiotracer anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (FACBC or fluciclovine) also proven most useful for the detection of recurrent prostate cancer. Other emerging PET radiotracers for prostate cancer include the bombesin group directed to the gastrin-releasing peptide receptor, 16β-18F-fluoro-5α-dihydrotestosterone (FDHT) that binds to the androgen receptor, and those targeting the vasoactive intestinal polypeptide receptor 1 (VPAC-1) and urokinase plasminogen activator receptor (uPAR), which are also overexpressed in prostate cancer.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA.
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
286
|
Assessment of Amino Acid/Drug Transporters for Renal Transport of [ 18F]Fluciclovine (anti-[ 18F]FACBC) in Vitro. Int J Mol Sci 2016; 17:ijms17101730. [PMID: 27754421 PMCID: PMC5085761 DOI: 10.3390/ijms17101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/14/2016] [Accepted: 10/08/2016] [Indexed: 01/19/2023] Open
Abstract
[18F]Fluciclovine (trans-1-amino-3-[18F]fluorocyclobutanecarboxylic acid; anti-[18F]FACBC), a positron emission tomography tracer used for the diagnosis of recurrent prostate cancer, is transported via amino acid transporters (AATs) with high affinity (Km: 97-230 μM). However, the mechanism underlying urinary excretion is unknown. In this study, we investigated the involvement of AATs and drug transporters in renal [18F]fluciclovine reuptake. [14C]Fluciclovine (trans-1-amino-3-fluoro[1-14C]cyclobutanecarboxylic acid) was used because of its long half-life. The involvement of AATs in [14C]fluciclovine transport was measured by apical-to-basal transport using an LLC-PK1 monolayer as model for renal proximal tubules. The contribution of drug transporters herein was assessed using vesicles/cells expressing the drug transporters P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), multidrug resistance-associated protein 4 (MRP4), organic anion transporter 1 (OAT1), organic anion transporter 3 (OAT3) , organic cation transporter 2 (OCT2), organic anion transporting polypeptide 1B1 (OATP1B1), and organic anion transporting polypeptide 1B3 (OATP1B3). The apical-to-basal transport of [14C]fluciclovine was attenuated by l-threonine, the substrate for system alanine-serine-cysteine (ASC) AATs. [14C]Fluciclovine uptake by drug transporter-expressing vesicles/cells was not significantly different from that of control vesicles/cells. Fluciclovine inhibited P-gp, MRP4, OAT1, OCT2, and OATP1B1 (IC50 > 2.95 mM). Therefore, system ASC AATs may be partly involved in the renal reuptake of [18F]fluciclovine. Further, given that [18F]fluciclovine is recognized as an inhibitor with millimolar affinity for the tested drug transporters, slow urinary excretion of [18F]fluciclovine may be mediated by system ASC AATs, but not by drug transporters.
Collapse
|
287
|
Filp U, Pekošak A, Poot AJ, Windhorst AD. Enantioselective synthesis of carbon-11 labeled l-alanine using phase transfer catalysis of Schiff bases. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.08.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
288
|
Recent Advances in Understanding Amino Acid Sensing Mechanisms that Regulate mTORC1. Int J Mol Sci 2016; 17:ijms17101636. [PMID: 27690010 PMCID: PMC5085669 DOI: 10.3390/ijms17101636] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 11/25/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is the central regulator of mammalian cell growth, and is essential for the formation of two structurally and functionally distinct complexes: mTORC1 and mTORC2. mTORC1 can sense multiple cues such as nutrients, energy status, growth factors and hormones to control cell growth and proliferation, angiogenesis, autophagy, and metabolism. As one of the key environmental stimuli, amino acids (AAs), especially leucine, glutamine and arginine, play a crucial role in mTORC1 activation, but where and how AAs are sensed and signal to mTORC1 are not fully understood. Classically, AAs activate mTORC1 by Rag GTPases which recruit mTORC1 to lysosomes, where AA signaling initiates. Plasma membrane transceptor L amino acid transporter 1 (LAT1)-4F2hc has dual transporter-receptor function that can sense extracellular AA availability upstream of mTORC1. The lysosomal AA sensors (PAT1 and SLC38A9) and cytoplasmic AA sensors (LRS, Sestrin2 and CASTOR1) also participate in regulating mTORC1 activation. Importantly, AAs can be sensed by plasma membrane receptors, like G protein-coupled receptor (GPCR) T1R1/T1R3, and regulate mTORC1 without being transported into the cells. Furthermore, AA-dependent mTORC1 activation also initiates within Golgi, which is regulated by Golgi-localized AA transporter PAT4. This review provides an overview of the research progress of the AA sensing mechanisms that regulate mTORC1 activity.
Collapse
|
289
|
Hansson J, Lindgren D, Nilsson H, Johansson E, Johansson M, Gustavsson L, Axelson H. Overexpression of Functional SLC6A3 in Clear Cell Renal Cell Carcinoma. Clin Cancer Res 2016; 23:2105-2115. [PMID: 27663598 DOI: 10.1158/1078-0432.ccr-16-0496] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/29/2016] [Accepted: 08/29/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Renal cell carcinoma (RCC) is derived from a tissue with a remarkable capacity for vectorial transport. We therefore performed an unbiased exploration of transporter proteins in normal kidney and kidney cancer to discover novel clinical targets.Experimental Design: Using The Cancer Genome Atlas (TCGA) database, we investigated differences in membrane transporter expression in clear cell RCC (ccRCC) and normal kidney. We identified the dopamine transporter SLC6A3 as a specific biomarker for ccRCC. To investigate the functionality of SLC6A3, we used a [3H]-dopamine uptake assay on ccRCC cells. We further explored the effect of hypoxia-inducible factor (HIF) proteins on SLC6A3 expression by introducing siRNA in ccRCC cells and by hypoxic treatment of nonmalignant cells.Results: We show that ccRCC expresses very high transcript levels of SLC6A3 in contrast to normal kidney tissue and other tumor types, which do not express appreciable levels of this transporter. Importantly, we demonstrate that the elevated expression of SLC6A3 in ccRCC cells is associated with specific uptake of dopamine. By targeting the expression of HIF-1α and HIF-2α, we could show that SLC6A3 expression is primarily influenced by HIF-2α and that hypoxia can induce SLC6A3 expression in normal renal cells.Conclusions: We conclude that the dopamine transporter SLC6A3 constitutes a novel biomarker that is highly specific for ccRCC. We further postulate that the protein can be exploited for diagnostic or therapeutic purposes for detection or treatment of ccRCC. Clin Cancer Res; 23(8); 2105-15. ©2016 AACR.
Collapse
Affiliation(s)
- Jennifer Hansson
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - David Lindgren
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Helén Nilsson
- Center for Molecular Pathology, Department of Translational Medicine, Lund University, Department of Pathology, Skåne University Hospital, Malmö, Sweden
| | - Elinn Johansson
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Martin Johansson
- Center for Molecular Pathology, Department of Translational Medicine, Lund University, Department of Pathology, Skåne University Hospital, Malmö, Sweden
| | - Lena Gustavsson
- Department of Drug Metabolism, H. Lundbeck A/S, Valby, Denmark
| | - Håkan Axelson
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden.
| |
Collapse
|
290
|
Enhanced Cellular Uptake and Pharmacokinetic Characteristics of Doxorubicin-Valine Amide Prodrug. Molecules 2016; 21:molecules21101272. [PMID: 27669201 PMCID: PMC6274118 DOI: 10.3390/molecules21101272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/17/2016] [Indexed: 11/17/2022] Open
Abstract
In this study, we synthesized the valine (Val)-conjugated amide prodrug of doxorubicin (DOX) by the formation of amide bonds between DOX and Val. The synthesis of the DOX-Val prodrug was identified by a proton nuclear magnetic resonance (1H-NMR) assay. In the MCF-7 cells (human breast adenocarcinoma cell; amino acid transporter–positive cell), the cellular accumulation efficiency of DOX-Val was higher than that of DOX according to the flow cytometry analysis data. Using confocal laser scanning microscopy (CLSM) imaging, it was confirmed that DOX-Val as well as DOX was mainly distributed in the nucleus of cancer cells. DOX-Val was intravenously administered to rats at a dose of 4 mg/kg, and the plasma concentrations of DOX-Val (prodrug) and DOX (formed metabolite) were quantitatively determined. Based on the systemic exposure (represented as area under the curve (AUC) values) of DOX-Val (prodrug) and DOX (formed metabolite), approximately half of DOX-Val seemed to be metabolized into DOX. However, it is expected that the remaining DOX-Val may exert improved cellular uptake efficiency in cancer cells after its delivery to the cancer region.
Collapse
|
291
|
Detour J, Pierre A, Boisson F, Kreutter G, Lavaux T, Namer IJ, Kessler L, Brasse D, Marchand P, Imperiale A. Effect of Carbidopa on 18F-FDOPA Uptake in Insulinoma: From Cell Culture to Small-Animal PET Imaging. J Nucl Med 2016; 58:36-41. [PMID: 27609787 DOI: 10.2967/jnumed.116.180588] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/10/2016] [Indexed: 01/13/2023] Open
Abstract
Patient premedication with carbidopa seems to improve the accuracy of 6-18F-fluoro-3,4-dihydroxy-l-phenylalanine (18F-FDOPA) PET for insulinoma diagnosis. However, the risk of PET false-negative results in the presence of carbidopa is a concern. Consequently, we aimed to evaluate the effect of carbidopa on 18F-FDOPA uptake in insulinoma β-cells and an insulinoma xenograft model in mice. METHODS 18F-FDOPA in vitro accumulation was assessed in the murine β-cell line RIN-m5F. In vivo small-animal PET experiments were performed on tumor-bearing nude mice after subcutaneous injection of RIN-m5F cells. Experiments were conducted with and without carbidopa pretreatment. RESULTS Incubation of RIN-m5F cells with 80 μM carbidopa did not significantly affect the cellular accumulation of 18F-FDOPA. Tumor xenografts were clearly detectable by small-animal PET in all cases. Insulinoma xenografts in carbidopa-treated mice showed significantly higher 18F-FDOPA uptake than those in nontreated mice. Regardless of carbidopa premedication, the xenografts were characterized by an early increase in 18F-FDOPA uptake and then a progressive reduction over time. CONCLUSION Carbidopa did not influence in vitro 18F-FDOPA accumulation in RIN-m5F cells but improved insulinoma imaging in vivo. Our findings increase current knowledge about the 18F-FDOPA uptake profile of RIN-m5F cells and a related xenograft model. To our knowledge, the present work represents the first preclinical research specifically focused on insulinomas, with potential translational implications.
Collapse
Affiliation(s)
- Julien Detour
- Department of Radiopharmacy, Strasbourg University Hospitals, Strasbourg, France.,Department of Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France
| | - Alice Pierre
- Department of Radiopharmacy, Strasbourg University Hospitals, Strasbourg, France.,Department of Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France
| | - Fréderic Boisson
- Institut Pluridisciplinaire Hubert Curien (IPHC), CNRS/UMR7178, Strasbourg University, Strasbourg, France
| | - Guillaume Kreutter
- EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France.,Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France
| | - Thomas Lavaux
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospitals, Strasbourg, France
| | - Izzie Jacques Namer
- Department of Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France.,Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France.,ICube, CNRS/UMR7357, Strasbourg University, Strasbourg, France; and
| | - Laurence Kessler
- EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France.,Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France.,Department of Diabetology, Strasbourg University Hospitals, Strasbourg, France
| | - David Brasse
- Institut Pluridisciplinaire Hubert Curien (IPHC), CNRS/UMR7178, Strasbourg University, Strasbourg, France
| | - Patrice Marchand
- Institut Pluridisciplinaire Hubert Curien (IPHC), CNRS/UMR7178, Strasbourg University, Strasbourg, France
| | - Alessio Imperiale
- Department of Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France .,Institut Pluridisciplinaire Hubert Curien (IPHC), CNRS/UMR7178, Strasbourg University, Strasbourg, France.,Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France.,ICube, CNRS/UMR7357, Strasbourg University, Strasbourg, France; and
| |
Collapse
|
292
|
Kaste SC, Snyder SE, Metzger ML, Sandlund JT, Howard SC, Krasin M, Shulkin BL. Comparison of 11C-Methionine and 18F-FDG PET/CT for Staging and Follow-up of Pediatric Lymphoma. J Nucl Med 2016; 58:419-424. [PMID: 27609791 DOI: 10.2967/jnumed.116.178640] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/22/2016] [Indexed: 02/03/2023] Open
Abstract
Methionine transport across plasma membranes occurs via the large amino acid transporter, which is overexpressed in malignant cells, leading to tracer accumulation within tumors. We investigated the uptake of 11C-methionine (11C-MET) in children and young adults with Hodgkin lymphoma (HL) or non-Hodgkin lymphoma (NHL) and compared the biodistribution of 11C-MET PET/CT with that of 18F-FDG PET/CT. Methods: Conducted under an investigational new drug authorization, we prospectively enrolled patients with newly diagnosed HL (n = 19) and NHL (n = 2) onto the Institutional Review Board-approved investigation of 11C-MET PET/CT. After a minimum 4-h fast, patients received 740 MBq/1.7 m2 (maximum, 740 MBq [20 mCi/1.7 m2; maximum, 20 mCi]) of 11C-methionine intravenously. PET/CT was performed 5 min after injection from the vertex to thighs at 3 min per bed position. In a separate session, patients received 5.5 MBq/kg (maximum, 485 MBq [0.15 mCi/kg; maximum, 12 mCi]) of 18F-FDG with imaging initiated approximately 1 h after radiopharmaceutical administration. All studies were reviewed by consensus of 2 senior imaging specialists. The presence of metabolic activity on baseline studies was compared among 17 nodal groups. Results: Eighteen patients (11 male; median age, 15.2 y; age range, 9.5-22.6 y) comprised the study cohort. All had paired 11C-MET PET/CT and 18F-FDG PET/CT studies at diagnosis. At baseline, 3 nodal groups demonstrating discordant metabolic activity by both 18F-FDG PET/CT and 11C-MET PET/CT were Waldeyer's ring, paraaortic region, and the liver. All others were found to have concordant metabolic activity. Normal intense 11C-MET uptake in the pancreas and liver reduced sensitivity for disease detection in these regions. At follow-up, 14 of 15 study pairs had concordant results. Conclusion:11C-MET uptake is elevated in most regions involved with lymphoma at diagnosis and follow-up. Its utility in the abdomen is limited by uptake in normal structures.
Collapse
Affiliation(s)
- Sue C Kaste
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee .,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Radiology, University of Tennessee, School of Health Sciences, Memphis, Tennessee
| | - Scott E Snyder
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Monika L Metzger
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Pediatrics, University of Tennessee, School of Health Sciences, Memphis, Tennessee
| | - John T Sandlund
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Pediatrics, University of Tennessee, School of Health Sciences, Memphis, Tennessee
| | - Scott C Howard
- School of Health Studies, University of Memphis, Memphis, Tennessee; and
| | - Matthew Krasin
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Barry L Shulkin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Radiology, University of Tennessee, School of Health Sciences, Memphis, Tennessee
| |
Collapse
|
293
|
Ryu JM, Lee SH, Seong JK, Han HJ. Glutamine contributes to maintenance of mouse embryonic stem cell self-renewal through PKC-dependent downregulation of HDAC1 and DNMT1/3a. Cell Cycle 2016; 14:3292-305. [PMID: 26375799 DOI: 10.1080/15384101.2015.1087620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although glutamine (Gln) is not an essential amino acid, it is considered a critical substrate in many key metabolic processes that control a variety of physiological functions and are involved in regulating early embryonic development. Thus, we investigated the effect of Gln on regulation of mouse embryonic stem cell (mESC) self-renewal and related signaling pathways. Gln deprivation decreased Oct4 expression as well as expression of cell cycle regulatory proteins. However, Gln treatment retained the expression of cell cycle regulatory proteins and the Oct4 in mESCs, which were blocked by compound 968 (a glutaminase inhibitor). In addition, Gln stimulated PI3K/Akt pathway, which subsequently elicited PKCϵ translocation to membrane without an influx of intracellular Ca(2+). Inhibition of Akt and PKC blocked Gln-induced Oct4 expression and proliferation. Gln also stimulated mTOR phosphorylation in a time-dependent manner, which abolished by PKC inhibition. Furthermore, Gln increased the cellular population of both Oct4 and bromodeoxyuridine positive cells, suggesting that Gln regulates self-renewal ability of mESCs. Gln induced a decrease in HDAC1, but not in HDAC2, which were blocked by PKC inhibitors. Gln treatment resulted in an increase in global histone acetylation and methylation. In addition, Gln significantly reduced methylation of the Oct4 promoter region through decrease in DNMT1 and DNMT3a expression, which were blocked by PKC and HDAC inhibitors. In conclusion, Gln stimulates mESC proliferation and maintains mESC undifferentiation status through transcription regulation via the Akt, PKCϵ, and mTOR signaling pathways.
Collapse
Affiliation(s)
- Jung Min Ryu
- a Department of Veterinary Physiology ; College of Veterinary Medicine, Seoul National University ; Seoul , Korea
| | - Sang Hun Lee
- b Medical Science Research Institute, Soonchunhyang University Seoul Hospital ; Seoul , Korea
| | - Je Kyung Seong
- c BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University ; Seoul , Korea.,d Department of Anatomy and Cell Biology ; Korea Mouse Phenotyping Center (KMPC), College of Veterinary Medicine, Seoul National University ; Seoul , Korea
| | - Ho Jae Han
- a Department of Veterinary Physiology ; College of Veterinary Medicine, Seoul National University ; Seoul , Korea.,c BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University ; Seoul , Korea
| |
Collapse
|
294
|
Böhme I, Bosserhoff AK. Acidic tumor microenvironment in human melanoma. Pigment Cell Melanoma Res 2016; 29:508-23. [PMID: 27233233 DOI: 10.1111/pcmr.12495] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/25/2016] [Indexed: 12/18/2022]
Abstract
One characteristic of solid tumors such as malignant melanoma is the acidification of the tumor microenvironment. The deregulation of cancer cell metabolism is considered a main cause of extracellular acidosis. Here, cancer cells utilize aerobic glycolysis instead of oxidative phosphorylation even under normoxic conditions, as originally described by Otto Warburg. These metabolic alterations cause enhanced acid production, especially of lactate and carbon dioxide (CO2 ). The extensive production of acidic metabolites and the enhanced acid export to the extracellular space cause a consistent acidification of the tumor microenvironment, thus promoting the formation of an acid-resistant tumor cell population with increased invasive and metastatic potential. As melanoma is one of the deadliest and most metastatic forms of cancer, understanding the effects of this extracellular acidosis on human melanoma cells with distinct metastatic properties is important. The aim of this review was to summarize recent studies of the acidification of the tumor microenvironment, focusing on the specific effects of the acidic milieu on melanoma cells and to give a short overview of therapeutic approaches.
Collapse
Affiliation(s)
- Ines Böhme
- Institute of Biochemistry, Emil-Fischer-Centrum, Friedrich Alexander University Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Centrum, Friedrich Alexander University Erlangen-Nürnberg, Erlangen-Nürnberg, Germany. .,Comprehensive Cancer Center Erlangen-EMN, University of Erlangen, Erlangen, Germany.
| |
Collapse
|
295
|
Physical and functional interactions between the serotonin transporter and the neutral amino acid transporter ASCT2. Biochem J 2016; 473:1953-65. [DOI: 10.1042/bcj20160315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/03/2016] [Indexed: 01/23/2023]
Abstract
The activity of serotonergic systems depends on the reuptake of extracellular serotonin via its plasma membrane serotonin [5-HT (5-hydroxytryptamine)] transporter (SERT), a member of the Na+/Cl−-dependent solute carrier 6 family. SERT is finely regulated by multiple molecular mechanisms including its physical interaction with intracellular proteins. The majority of previously identified SERT partners that control its functional activity are soluble proteins, which bind to its intracellular domains. SERT also interacts with transmembrane proteins, but its association with other plasma membrane transporters remains to be established. Using a proteomics strategy, we show that SERT associates with ASCT2 (alanine–serine–cysteine–threonine 2), a member of the solute carrier 1 family co-expressed with SERT in serotonergic neurons and involved in the transport of small neutral amino acids across the plasma membrane. Co-expression of ASCT2 with SERT in HEK (human embryonic kidney)-293 cells affects glycosylation and cell-surface localization of SERT with a concomitant reduction in its 5-HT uptake activity. Conversely, depletion of cellular ASCT2 by RNAi enhances 5-HT uptake in both HEK-293 cells and primary cultured mesencephalon neurons. Mimicking the effect of ASCT2 down-regulation, treatment of HEK-293 cells and neurons with the ASCT2 inhibitor D-threonine also increases 5-HT uptake. Moreover, D-threonine does not enhance further the maximal velocity of 5-HT uptake in cells depleted of ASCT2. Collectively, these findings provide evidence for a complex assembly involving SERT and a member of another solute carrier family, which strongly influences the subcellular distribution of SERT and the reuptake of 5-HT.
Collapse
|
296
|
Huttunen KM, Gynther M, Huttunen J, Puris E, Spicer JA, Denny WA. A Selective and Slowly Reversible Inhibitor of l-Type Amino Acid Transporter 1 (LAT1) Potentiates Antiproliferative Drug Efficacy in Cancer Cells. J Med Chem 2016; 59:5740-51. [PMID: 27253989 DOI: 10.1021/acs.jmedchem.6b00190] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The l-type amino acid transporter 1 (LAT1) is a transmembrane protein carrying bulky and neutral amino acids into cells. LAT1 is overexpressed in several types of tumors, and its inhibition can result in reduced cancer cell growth. However, known LAT1 inhibitors lack selectivity over other transporters. In the present study, we designed and synthesized a novel selective LAT1 inhibitor (1), which inhibited the uptake of LAT1 substrate, l-leucin as well as cell growth. It also significantly potentiated the efficacy of bestatin and cisplatin even at low concentrations (25 μM). Inhibition was slowly reversible, as the inhibitor was able to be detached from the cell surface and blood-brain barrier. Moreover, the inhibitor was metabolically stable and selective toward LAT1. Since the inhibitor was readily accumulated into the prostate after intraperitoneal injection to the healthy mice, this compound may be a promising agent or adjuvant especially for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Elena Puris
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Julie A Spicer
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
297
|
Colas C, Ung PMU, Schlessinger A. SLC Transporters: Structure, Function, and Drug Discovery. MEDCHEMCOMM 2016; 7:1069-1081. [PMID: 27672436 PMCID: PMC5034948 DOI: 10.1039/c6md00005c] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human Solute Carrier (SLC) transporters are important targets for drug development. Structure-based drug discovery for SLC transporters requires the description of their structure, dynamics, and mechanism of interaction with small molecule ligands and ions. The recent determination of atomic structures of human SLC transporters and their homologs, combined with improved computational power and prediction methods have led to an increased applicability of structure-based drug design methods for human SLC members. In this review, we provide an overview of the SLC transporters' structures and transport mechanisms. We then describe computational techniques, such as homology modeling and virtual screening that are emerging as key tools to discover chemical probes for human SLC members. We illustrate the utility of these methods by presenting case studies in which rational integration of computation and experiment was used to characterize SLC members that transport key nutrients and metabolites, including the amino acid transporters LAT-1 and ASCT2, the SLC13 family of citric acid cycle intermediate transporters, and the glucose transporter GLUT1. We conclude with a brief discussion about future directions in structure-based drug discovery for the human SLC superfamily, one of the most structurally and functionally diverse protein families in human.
Collapse
Affiliation(s)
- Claire Colas
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter Man-Un Ung
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avner Schlessinger
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
298
|
Chiotellis A, Müller Herde A, Rössler SL, Brekalo A, Gedeonova E, Mu L, Keller C, Schibli R, Krämer SD, Ametamey SM. Synthesis, Radiolabeling, and Biological Evaluation of 5-Hydroxy-2-[18F]fluoroalkyl-tryptophan Analogues as Potential PET Radiotracers for Tumor Imaging. J Med Chem 2016; 59:5324-40. [DOI: 10.1021/acs.jmedchem.6b00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Aristeidis Chiotellis
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Adrienne Müller Herde
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Simon L. Rössler
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Ante Brekalo
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Erika Gedeonova
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Linjing Mu
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Department of Nuclear
Medicine, University Hospital Zurich, Zurich 8091, Switzerland
| | - Claudia Keller
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Roger Schibli
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Stefanie D. Krämer
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Simon M. Ametamey
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| |
Collapse
|
299
|
McConathy J. 18F-Fluciclovine (FACBC) and Its Potential Use for Breast Cancer Imaging. J Nucl Med 2016; 57:1329-30. [DOI: 10.2967/jnumed.116.175489] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/25/2016] [Indexed: 12/17/2022] Open
|
300
|
Abstract
This article provides an overview of the key considerations for the development and application of molecular imaging agents for brain tumors and the major classes of PET tracers that have been used for imaging brain tumors in humans. The mechanisms of uptake, biological implications, primary applications, and limitations of PET tracers in neuro-oncology are reviewed. The available data indicate that several of these classes of tracers, including radiolabeled amino acids, have imaging properties superior to those of (18)F-fluorodeoxyglucose, and can complement contrast-enhanced magnetic resonance imaging in the evaluation of brain tumors.
Collapse
|