51
|
Regula N, Häggman M, Johansson S, Sörensen J. Malignant lipogenesis defined by 11C-acetate PET/CT predicts prostate cancer-specific survival in patients with biochemical relapse after prostatectomy. Eur J Nucl Med Mol Imaging 2016; 43:2131-2138. [PMID: 27392615 DOI: 10.1007/s00259-016-3449-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Malignant de novo lipogenesis is strongly linked to the aggressiveness of prostate cancer (PCa) under experimental conditions. 11C-Acetate PET/CT is a potential noninvasive biomarker of malignant lipogenesis in PCa, but its prognostic value is not known. The objective of this study was to analyse 11C-acetate PET/CT image metrics in relation to survival. METHODS All patients undergoing 11C-acetate PET/CT in one university hospital from 2005 to 2011 due to PSA relapse after previous prostatectomy were retrospectively evaluated. Two groups of patients were compared: those who died from PCa and those who were censored. All previously reported findings of local recurrence, regional or distal lymph node metastases and bone metastases were counted and evaluated regarding 11C-acetate uptake intensity (SUVmax) and tumour volume. Total tumour volume and total lipogenic activity (TLA, summed SUVmax × TV) were calculated. Survival analysis in the entire study population was followed by Cox proportional hazards ratio (HR) analysis. RESULTS A total of 121 patients were included, and 22 PCa-specific deaths were recorded. The mean PSA level at the time of PET was 2.69 ± 4.35 ng/mL. The median follow-up of the study population was 79 ± 28 months. PET identified at least one PCa lesion in 53 % of patients. Five-year PCa-specific survival after PET was 80 % and 100 % in patients with a positive and a negative PET scan, respectively (p < 0.001). Time-to-death was linearly correlated with highest SUVmax (r = -0.55, p = 0.01) and nonlinearly with TLA (r = -0.75, p < 0.001). Multivariate analysis showed statistical significance for number of bone metastases (HR 1.74, p = 0.01), tertile of TLA (HR 5.63, p = 0.029) and postoperative Gleason score (HR 1.84, p = 0.045). CONCLUSION Malignant 11C-acetate accumulation measured with PET/CT is a strong predictor of survival in the setting of PSA relapse after prostatectomy. The study provides further evidence for a quantitative relationship between malignant de novo lipogenesis and early death. 11C-Acetate PET/CT might be useful for identifying a high-risk population of relapsing patients in which therapies targeting malignant lipogenesis might be of particular benefit.
Collapse
Affiliation(s)
- Naresh Regula
- Section of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Häggman
- Section of Urology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Silvia Johansson
- Section of Oncology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Section of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden. .,PET Center Research Department, no. 79, 5th floor, Akademiska Sjukhuset, 751 85, Uppsala, Sweden.
| |
Collapse
|
52
|
Aberrant Lipid Metabolism Promotes Prostate Cancer: Role in Cell Survival under Hypoxia and Extracellular Vesicles Biogenesis. Int J Mol Sci 2016; 17:ijms17071061. [PMID: 27384557 PMCID: PMC4964437 DOI: 10.3390/ijms17071061] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer (PCa) is the leading malignancy among men in United States. Recent studies have focused on the identification of novel metabolic characteristics of PCa, aimed at devising better preventive and therapeutic approaches. PCa cells have revealed unique metabolic features such as higher expression of several enzymes associated with de novo lipogenesis, fatty acid up-take and β-oxidation. This aberrant lipid metabolism has been reported to be important for PCa growth, hormone-refractory progression and treatment resistance. Furthermore, PCa cells effectively use lipid metabolism under adverse environmental conditions for their survival advantage. Specifically, hypoxic cancer cells accumulate higher amount of lipids through a combination of metabolic alterations including high glutamine and fatty acid uptake, as well as decreased fatty acid oxidation. These stored lipids serve to protect cancer cells from oxidative and endoplasmic reticulum stress, and play important roles in fueling cancer cell proliferation following re-oxygenation. Lastly, cellular lipids have also been implicated in extracellular vesicle biogenesis, which play a vital role in intercellular communication. Overall, the new understanding of lipid metabolism in recent years has offered several novel targets to better target and manage clinical PCa.
Collapse
|
53
|
Li L, Che L, Wang C, Blecha JE, Li X, VanBrocklin HF, Calvisi DF, Puchowicz M, Chen X, Seo Y. [(11)C]acetate PET Imaging is not Always Associated with Increased Lipogenesis in Hepatocellular Carcinoma in Mice. Mol Imaging Biol 2016; 18:360-7. [PMID: 26567114 PMCID: PMC4866912 DOI: 10.1007/s11307-015-0915-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Altered metabolism, including increased glycolysis and de novo lipogenesis, is one of the hallmarks of cancer. Radiolabeled nutrients, including glucose and acetate, are extensively used for the detection of various tumors, including hepatocellular carcinomas (HCCs). High signal of [(11)C]acetate positron emission tomography (PET) in tumors is often considered to be associated with increased expression of fatty acid synthase (FASN) and increased de novo lipogenesis in tumor tissues. Defining a subset of tumors with increased [(11)C]acetate PET signal and thus increased lipogenesis was suggested to help select a group of patients, who may benefit from lipogenesis-targeting therapies. PROCEDURES To investigate whether [(11)C]acetate PET imaging is truly associated with increased de novo lipogenesis along with hepatocarcinogenesis, we performed [(11)C]acetate PET imaging in wild-type mice as well as two mouse HCC models, induced by myrAKT/Ras(V12) (AKT/Ras) and PIK3CA(1047R)/c-Met (PI3K/Met) oncogene combinations. In addition, we analyzed FASN expression and de novo lipogenesis rate in these mouse liver tissues. RESULTS We found that while HCCs induced by AKT/Ras co-expression showed high levels of [(11)C]acetate PET signal compared to normal liver, HCCs induced by PI3K/Met overexpression did not. Intriguingly, elevated FASN expression and increased de novo lipogenesis rate were observed in both AKT/Ras and PI3K/Met HCCs. CONCLUSION Altogether, our study suggests that [(11)C]acetate PET imaging can be a useful tool for imaging of a subset of HCCs. However, at molecular level, the increased [(11)C]acetate PET imaging is not always associated with increased FASN expression or de novo lipogenesis.
Collapse
Affiliation(s)
- Lei Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-0912, USA
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-0912, USA
| | - Chunmei Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-0912, USA
| | - Joseph E Blecha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Xiaolei Li
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-0912, USA
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-0912, USA.
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
- Department of Radiation Oncology, University of California, San Francisco, CA, USA.
- UCSF-UC Berkeley Joint Graduate Group in Bioengineering, University of California, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- UCSF Physics Research Laboratory, Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94143-0946, USA.
| |
Collapse
|
54
|
|
55
|
Bouvet V, Wuest M, Jans HS, Janzen N, Genady AR, Valliant JF, Benard F, Wuest F. Automated synthesis of [(18)F]DCFPyL via direct radiofluorination and validation in preclinical prostate cancer models. EJNMMI Res 2016; 6:40. [PMID: 27142881 PMCID: PMC4854855 DOI: 10.1186/s13550-016-0195-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/26/2016] [Indexed: 11/18/2022] Open
Abstract
Background Prostate-specific membrane antigen (PSMA) is frequently overexpressed and upregulated in prostate cancer. To date, various 18F- and 68Ga-labeled urea-based radiotracers for PET imaging of PSMA have been developed and entered clinical trials. Here, we describe an automated synthesis of [18F]DCFPyL via direct radiofluorination and validation in preclinical models of prostate cancer. Methods [18F]DCFPyL was synthesized via direct nucleophilic heteroaromatic substitution reaction in a single reactor TRACERlab FXFN automated synthesis unit. Radiopharmacological evaluation of [18F]DCFPyL involved internalization experiments, dynamic PET imaging in LNCaP (PSMA+) and PC3 (PSMA−) tumor-bearing BALB/c nude mice, biodistribution studies, and metabolic profiling. In addition, reversible two-tissue compartmental model analysis was used to quantify pharmacokinetics of [18F]DCFPyL in LNCaP and PC3 tumor models. Results Automated radiosynthesis afforded radiotracer [18F]DCFPyL in decay-corrected radiochemical yields of 23 ± 5 % (n = 10) within 55 min, including HPLC purification. Dynamic PET analysis revealed rapid and high uptake of radioactivity (SUV5min 0.95) in LNCaP tumors which increased over time (SUV60min 1.1). Radioactivity uptake in LNCaP tumors was blocked in the presence of nonradioactive DCFPyL (SUV60min 0.22). The muscle as reference tissue showed rapid and continuous clearance over time (SUV60min 0.06). Fast blood clearance of radioactivity resulted in tumor-blood ratios of 1.0 after 10 min and 8.3 after 60 min. PC3 tumors also showed continuous clearance of radioactivity over time (SUV60min 0.11). Kinetic analysis of PET data revealed the two-tissue compartmental model as best fit with K1 = 0.12, k2 = 0.18, k3 = 0.08, and k4 = 0.004 min−1, confirming molecular trapping of [18F]DCFPyL in PSMA+ LNCaP cells. Conclusions [18F]DCFPyL can be prepared for clinical applications simply and in good radiochemical yields via a direct radiofluorination synthesis route in a single reactor automated synthesis unit. Radiopharmacological evaluation of [18F]DCFPyL confirmed high PSMA-mediated tumor uptake combined with superior clearance parameters. Compartmental model analysis points to a two-step molecular trapping mechanism based on PSMA binding and subsequent internalization leading to retention of radioactivity in PSMA+ LNCaP tumors. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0195-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vincent Bouvet
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Hans-Soenke Jans
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Nancy Janzen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Afaf R Genady
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - John F Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Francois Benard
- Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada.
| |
Collapse
|
56
|
[ $${}^{11}\hbox {C}$$ 11 C ]acetate and PET/CT assessment of muscle activation in rat studies. Int J Comput Assist Radiol Surg 2016. [DOI: 10.1007/s11548-015-1260-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
57
|
Challapalli A, Aboagye EO. Positron Emission Tomography Imaging of Tumor Cell Metabolism and Application to Therapy Response Monitoring. Front Oncol 2016; 6:44. [PMID: 26973812 PMCID: PMC4770188 DOI: 10.3389/fonc.2016.00044] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/12/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer cells do reprogram their energy metabolism to enable several functions, such as generation of biomass including membrane biosynthesis, and overcoming bioenergetic and redox stress. In this article, we review both established and evolving radioprobes developed in association with positron emission tomography (PET) to detect tumor cell metabolism and effect of treatment. Measurement of enhanced tumor cell glycolysis using 2-deoxy-2-[(18)F]fluoro-D-glucose is well established in the clinic. Analogs of choline, including [(11)C]choline and various fluorinated derivatives are being tested in several cancer types clinically with PET. In addition to these, there is an evolving array of metabolic tracers for measuring intracellular transport of glutamine and other amino acids or for measuring glycogenesis, as well as probes used as surrogates for fatty acid synthesis or precursors for fatty acid oxidation. In addition to providing us with opportunities for examining the complex regulation of reprogramed energy metabolism in living subjects, the PET methods open up opportunities for monitoring pharmacological activity of new therapies that directly or indirectly inhibit tumor cell metabolism.
Collapse
Affiliation(s)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
58
|
Abstract
The field of metabolism research has made a dramatic resurgence in recent years, fueled by a newfound appreciation of the interactions between metabolites and phenotype. Metabolic substrates and their products can be biomarkers of a wide range of pathologies, including cancer, but our understanding of their in vivo interactions and pathways has been hindered by the robustness of noninvasive imaging approaches. The past 3 decades have been flushed with the development of new techniques for the study of metabolism in vivo. These methods include nuclear-based, predominantly positron emission tomography and magnetic resonance imaging, many of which have been translated to the clinic. The purpose of this review was to introduce both long-standing imaging strategies as well as novel approaches to the study of perturbed metabolic pathways in the setting of carcinogenesis. This will involve descriptions of nuclear probes labeled with C and F as well C for study using hyperpolarized magnetic resonance imaging. Highlighting both advantages and disadvantages of each approach, the aim of this summary was to provide the reader with a framework for interrogation of metabolic aberrations in their system of interest.
Collapse
|
59
|
Wang H, Tang G, Hu K, Huang T, Liang X, Wu Z, Li S. Comparison of three ¹⁸F-labeled carboxylic acids with ¹⁸F-FDG of the differentiation tumor from inflammation in model mice. BMC Med Imaging 2016; 16:2. [PMID: 26754531 PMCID: PMC4709996 DOI: 10.1186/s12880-016-0110-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to compare the properties and feasibility of the glucose analog, 2-(18)F-fluoro-2-deoxy-D-glucose ((18)F-FDG), three short (18)F-labeled carboxylic acids, (18)F-fluoroacetate ((18)F-FAC), 2-(18)F-fluoropropionic acid ((18)F-FPA) and 4-((18)F)fluorobenzoic acid ((18)F-FBA), for differentiating tumors from inflammation. METHODS Biodistributions of (18)F-FAC, (18)F-FPA and (18)F-FBA were determined on normal Kunming mice, and positron emission tomography (PET) imaging with these tracers were performed on the separate tumor-bearing mice model and inflammation mice model in comparison with (18)F-FDG. RESULTS Biodistribution results showed that (18)F-FAC and (18)F-FPA had similar biodistribution profiles and the slow radioactivity clearance from most tissues excluding the in vivo defluorination of (18)F-FAC, and (18)F-FBA demonstrated a lower uptake and fast clearance in most tissues. PET imaging with (18)F-FDG, (18)F-FAC and (18)F-FPA revealed the high uptake in both tumor and inflammatory lesions. The ratios of tumor-to-inflammation were 1.63 ± 0.28 for (18)F-FDG, 1.20 ± 0.38 for (18)F-FAC, and 1.41 ± 0.33 for (18)F-FPA at 60 min postinjection, respectively. While clear tumor images with high contrast between tumor and inflammation lesion were observed in (18)F-FBA/PET with the highest ratio of tumor-to-inflammation (1.98 ± 0.15). CONCLUSIONS Our data demonstrated (18)F-FBA is a promising PET probe to distinguish tumor from inflammation. But the further modification of (18)F-FBA structure is required to improve its pharmacokinetics.
Collapse
Affiliation(s)
- Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Ganghua Tang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Kongzhen Hu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tingting Huang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiang Liang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
60
|
Abstract
Imaging of prostate cancer presents many challenges to the imaging community. There has been much progress in this space in large part due to MRI and PET radiopharmaceuticals. Though MRI has been focused on the evaluation of local disease and PET on the detection of metastatic disease, these two areas do converge and will be complementary especially with the growth of new PET/MRI technologies. In this review article, we review novel MRI, MRI/US, and PET radiopharmaceuticals which will offer insight into the future direction of imaging in prostate cancer.
Collapse
Affiliation(s)
- Phillip J Koo
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, University of Colorado School of Medicine, Mail Stop L954, 12401 E. 17th Avenue, Room 1512, Aurora, CO, 80045, USA.
| | - Jennifer J Kwak
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, University of Colorado School of Medicine, Mail Stop L954, 12401 E. 17th Avenue, Room 1512, Aurora, CO, 80045, USA.
| | - Sajal Pokharel
- Division of Abdominal Imaging, Department of Radiology, University of Colorado School of Medicine, Mail Stop L954, 12401 E. 17th Avenue, Room 1512, Aurora, CO, 80045, USA.
| | - Peter L Choyke
- Center for Cancer Research, National Cancer Institute, Building 10, Room B3B69F, Bethesda, MD, 20892-1088, USA.
| |
Collapse
|
61
|
Wibmer AG, Burger IA, Sala E, Hricak H, Weber WA, Vargas HA. Molecular Imaging of Prostate Cancer. Radiographics 2015; 36:142-59. [PMID: 26587888 DOI: 10.1148/rg.2016150059] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most common noncutaneous malignancy among men in the Western world. The natural history and clinical course of prostate cancer are markedly diverse, ranging from small indolent intraprostatic lesions to highly aggressive disseminated disease. An understanding of this biologic heterogeneity is considered a necessary requisite in the quest for the adoption of precise and personalized management strategies. Molecular imaging offers the potential for noninvasive assessment of the biologic interactions underpinning prostate carcinogenesis. Currently, numerous molecular imaging probes are in clinical use or undergoing preclinical or clinical evaluation. These probes can be divided into those that image increased cell metabolism, those that target prostate cancer-specific membrane proteins and receptor molecules, and those that bind to the bone matrix adjacent to metastases to bone. The increased metabolism and vascular changes in prostate cancer cells can be evaluated with radiolabeled analogs of choline, acetate, glucose, amino acids, and nucleotides. The androgen receptor, prostate-specific membrane antigen, and gastrin-releasing peptide receptor (ie, bombesin) are overexpressed in prostate cancer and can be targeted by specific radiolabeled imaging probes. Because metastatic prostate cancer cells induce osteoblastic signaling pathways of adjacent bone tissue, bone-seeking radiotracers are sensitive tools for the detection of metastases to bone. Knowledge about the underlying biologic processes responsible for the phenotypes associated with the different stages of prostate cancer allows an appropriate choice of methods and helps avoid pitfalls.
Collapse
Affiliation(s)
- Andreas G Wibmer
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Irene A Burger
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Evis Sala
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hedvig Hricak
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Wolfgang A Weber
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hebert Alberto Vargas
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| |
Collapse
|
62
|
Björnson E, Mukhopadhyay B, Asplund A, Pristovsek N, Cinar R, Romeo S, Uhlen M, Kunos G, Nielsen J, Mardinoglu A. Stratification of Hepatocellular Carcinoma Patients Based on Acetate Utilization. Cell Rep 2015; 13:2014-26. [PMID: 26655911 DOI: 10.1016/j.celrep.2015.10.045] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/18/2015] [Accepted: 10/14/2015] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly form of liver cancer that is increasingly prevalent. We analyzed global gene expression profiling of 361 HCC tumors and 49 adjacent noncancerous liver samples by means of combinatorial network-based analysis. We investigated the correlation between transcriptome and proteome of HCC and reconstructed a functional genome-scale metabolic model (GEM) for HCC. We identified fundamental metabolic processes required for cell proliferation using the network centric view provided by the GEM. Our analysis revealed tight regulation of fatty acid biosynthesis (FAB) and highly significant deregulation of fatty acid oxidation in HCC. We predicted mitochondrial acetate as an emerging substrate for FAB through upregulation of mitochondrial acetyl-CoA synthetase (ACSS1) in HCC. We analyzed heterogeneous expression of ACSS1 and ACSS2 between HCC patients stratified by high and low ACSS1 and ACSS2 expression and revealed that ACSS1 is associated with tumor growth and malignancy under hypoxic conditions in human HCC.
Collapse
Affiliation(s)
- Elias Björnson
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Bani Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna Asplund
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Nusa Pristovsek
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, the Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden; Cardiology Department, Sahlgrenska University Hospital, 416 50 Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Mathias Uhlen
- Department of Proteomics, KTH-Royal Institute of Technology, 106 91 Stockholm, Sweden; Science for Life Laboratory, KTH-Royal Institute of Technology, 171 21 Stockholm, Sweden
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Science for Life Laboratory, KTH-Royal Institute of Technology, 171 21 Stockholm, Sweden
| | - Adil Mardinoglu
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Science for Life Laboratory, KTH-Royal Institute of Technology, 171 21 Stockholm, Sweden.
| |
Collapse
|
63
|
Leisser A, Pruscha K, Ubl P, Wadsak W, Mayerhöfer M, Mitterhauser M, Hacker M, Kramer G, Shariat S, Karanikas G, Hartenbach M, Haug AR. Evaluation of fatty acid synthase in prostate cancer recurrence: SUV of [(11) C]acetate PET as a prognostic marker. Prostate 2015; 75:1760-7. [PMID: 26282590 DOI: 10.1002/pros.23061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/28/2015] [Indexed: 11/12/2022]
Abstract
AIM High levels of fatty acid synthase have shown to correlate with the aggressiveness of prostate cancer. As [(11) C]acetate exhibits a close correlation with the level of fatty acid synthase, we aimed to assess whether the SUV in [(11) C]acetate PET serves as a suitable prognostic marker in patients with recurrent prostate cancer. MATERIALS AND METHODS In 123 consecutive patients, examined between 2010 and 2014, the maximum standardized uptake value (SUVmax) of local recurrences as well as lymph node and bone metastases was measured. Choosing the spleen as a standard for relatively high physiological uptake, a ratio of tumor to spleen uptake (SUVts) was calculated for standardizing the uptake, too. The corresponding initial Gleason scores (GS) and serum-PSA levels around the time of the performed PET/CT for each patient were retrospectively collected and PSA doubling together with PSA velocity were determined. For further analysis patients were divided with regard to their initial Gleason score (≤3 + 4 and ≥ 4 + 3). The median of PSA velocity was calculated to separate patients with a high and low PSA velocity and Mann-Whitney U or Student's t-test were used, testing for significant differences. For correlation Spearmen-Rho test was used. RESULTS PET was positive for recurrence in 82/123 patients. PSA was significantly higher in PET-positive than in negative patients (5.9 vs. 3.2 ng/ml; P = 0.006). Initial Gleason score did not differ in PET negative and positive patients (P = 0.3), whereas PSA velocity was markedly higher in PET positive patients (0.4 vs. 0.1 ng/ml/month; P = 0.01). Median SUVmax of PET positive patients was 5.23 (mean 5.78; range 0.9-16.8) and meadian SUVts was 0.78 (mean 0.84, range 0.14-2.50). SUVts was significantly higher in patients with high PSA velocity (SUVts 0.76 vs. 0.92; P = 0.009), whereas SUVmax failed statistical significance (5.4 vs. 6.3 ng/ml/month; P = 0.08). Patients with a high SUVmax proved to have a significantly higher median Gleason score compared to low uptake 8.0 vs. 7.0; P = 0.004). Vice versa both SUVmax (GS 6: 5.0; GS 7: 5.6; GS 8: 5.7; GS 9: 6.5; r = 0.30, P = 0.008) and SUVts (GS 6: 0.63; GS 7: 0.68; GS 8: 0.85; GS 9: 0.89; r = 0.30, P = 0.006) significantly correlated with Gleason score. Patients with a Gleason score ≤ 3 + 4 had a significantly lower SUVmax (4.8 vs. 5.7; P = 0.02) and SUVts (0.67 vs. 0.85; P = 0.02) as compared to a Gleason score ≥ 4 + 3. CONCLUSION [(11) C]acetate uptake demonstrated to correlate with initial Gleason score. Furthermore, patients with a high PSA velocity proved to have higher [(11) C]acetate uptake in tumor lesions.
Collapse
Affiliation(s)
- Asha Leisser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Konstatin Pruscha
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Philipp Ubl
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marius Mayerhöfer
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Paediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Shahrokh Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Georgios Karanikas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Hartenbach
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexander R Haug
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
64
|
Rae C, Haberkorn U, Babich JW, Mairs RJ. Inhibition of Fatty Acid Synthase Sensitizes Prostate Cancer Cells to Radiotherapy. Radiat Res 2015; 184:482-93. [DOI: 10.1667/rr14173.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
65
|
Jeon JY, Lee H, Park J, Lee M, Park SW, Kim JS, Lee M, Cho B, Kim K, Choi AMK, Kim CK, Yun M. The regulation of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase by autophagy in low-glycolytic hepatocellular carcinoma cells. Biochem Biophys Res Commun 2015; 463:440-6. [PMID: 26036577 DOI: 10.1016/j.bbrc.2015.05.103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 05/29/2015] [Indexed: 11/20/2022]
Abstract
The glycolytic phenotype is a dominant metabolic phenomenon in cancer and is reflected in becoming aggressive. Certain hepatocellular carcinoma lack increased glycolysis and prefer to uptake acetate than glucose for metabolism. Autophagy plays a role in preserving energies and nutrients when there is limited external nutrient supply and maintains glucose level of blood though supporting gluconeogenesis in the liver. As the role of autophagy and gluconeogenesis in HCC following the glycolic activity was not clear, we cultured HCC cells with different glycolytic levels in Hank's balanced salt solution (HBSS) to induce autophagy and conducted the activity of gluconeogenesis. Both autophagy and gluconeogenesis were induced in low glycolytic HCC cells (HepG2). In glycolytic Hep3B cells, only autophagy without gluconeogenesis was induced upon starvation. When autophagy was blocked, the level of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) was reduced in HepG2 cells and not in Hep3B. Altogether, we investigated contribution of hepatic gluconeogenesis to the metabolic phenotype of HCC cells and the role of autophagy as a potential mechanism regulating gluconeogenesis in low glycolytic HCC.
Collapse
Affiliation(s)
- Jeong Yong Jeon
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Hyangkyu Lee
- Department of Clinical Nursing Science, Yonsei University College of Nursing, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Jeayeo Park
- Department of Clinical Nursing Science, Yonsei University College of Nursing, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Misu Lee
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Sae Whan Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Ji Sook Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Milim Lee
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Byoungchul Cho
- Division of Medical Oncology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Kyungsup Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project for Medical Science, Institute of Genetic Science, Integrated Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA
| | - Chun K Kim
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02026, USA.
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-752, South Korea.
| |
Collapse
|
66
|
Abstract
Positron emission tomography (PET) is an extraordinarily sensitive clinical imaging modality for interrogating tumor metabolism. Radiolabeled PET substrates can be traced at subphysiological concentrations, allowing noninvasive imaging of metabolism and intratumoral heterogeneity in systems ranging from advanced cancer models to patients in the clinic. There are a wide range of novel and more established PET radiotracers, which can be used to investigate various aspects of the tumor, including carbohydrate, amino acid, and fatty acid metabolism. In this review, we briefly discuss the more established metabolic tracers and describe recent work on the development of new tracers. Some of the unanswered questions in tumor metabolism are considered alongside new technical developments, such as combined PET/magnetic resonance imaging scanners, which could provide new imaging solutions to some of the outstanding diagnostic challenges facing modern cancer medicine.
Collapse
Affiliation(s)
- David Y. Lewis
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Dmitry Soloviev
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|
67
|
Ghosh A, Sengupta A, Chattopadhyay A, Das D. A single probe for sensing both acetate and aluminum(iii): visible region detection, red fluorescence and human breast cancer cell imaging. RSC Adv 2015. [DOI: 10.1039/c4ra16768f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A single probe (L) can recognise both AcO− and Al3+ as prepared by coupling 2-hydroxy-1-naphthaldehyde with hydrazine. The probe allows both colorimetric and fluorescence detection of both the ions.
Collapse
Affiliation(s)
- Abhijit Ghosh
- Department of Chemistry
- The University of Burdwan
- Burdwan
- India
| | - Archya Sengupta
- Department of Zoology
- Visva Bharati University
- Santiniketan
- India
| | | | - Debasis Das
- Department of Chemistry
- The University of Burdwan
- Burdwan
- India
| |
Collapse
|
68
|
Strandberg S, Karlsson CT, Sundström T, Ögren M, Ögren M, Axelsson J, Riklund K. (11)C-acetate PET/CT in pre-therapeutic lymph node staging in high-risk prostate cancer patients and its influence on disease management - a retrospective study. EJNMMI Res 2014; 4:55. [PMID: 26116118 PMCID: PMC4452630 DOI: 10.1186/s13550-014-0055-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/28/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Radiation treatment with simultaneous integrated boost against suspected lymph node metastases may be a curative therapeutic option in patients with high-risk prostate cancer (>15% estimated risk of pelvic lymph node metastases according to the Cagiannos nomogram). (11)C-acetate positron emission tomography/computed tomography (PET/CT) can be used for primary staging as well as for detection of suspected relapse of prostate cancer. The aims of this study were to evaluate the association between positive (11)C-acetate PET/CT findings and the estimated risk of pelvic lymph node metastases and to assess the impact of (11)C-acetate PET/CT on patient management in high-risk prostate cancer patients. METHODS Fifty consecutive prostate cancer patients referred for primary staging with (11)C-acetate PET/CT prior to radiotherapy with curative intention were enrolled in this retrospective study. RESULTS All patients showed increased (11)C-acetate uptake in the prostate. Pelvic lymph node uptake was seen in 42% (21/50) of the patients, with positive external iliac lymph nodes in 71% (15/21) of these. The overall observed proportion of PET/CT-positive pelvic lymph nodes at patient level was higher than the average estimated risk, especially in low-risk groups (<15%). There was a significant association between observed proportion and estimated risk of pelvic lymph node metastases in groups with ≤45 and >45% estimated risk. Treatment strategy was altered due to (11)C-acetate PET/CT findings in 43% (20/47) of the patients. CONCLUSIONS The observed proportion of (11)C-acetate PET/CT findings suggestive of locoregional metastases was higher than the estimated risk, suggesting that the Cagiannos nomogram underestimates the risk for metastases. The imaging results with (11)C-acetate PET/CT have a considerable impact on patient management.
Collapse
Affiliation(s)
- Sara Strandberg
- Department of Radiation Sciences, Umeå University, 90185, Umeå, Sweden
| | | | | | - Mattias Ögren
- Department of Radiation Sciences, Umeå University, 90185, Umeå, Sweden
| | - Margareta Ögren
- Department of Radiation Sciences, Umeå University, 90185, Umeå, Sweden
| | - Jan Axelsson
- Department of Radiation Sciences, Umeå University, 90185, Umeå, Sweden
| | - Katrine Riklund
- Department of Radiation Sciences, Umeå University, 90185, Umeå, Sweden
| |
Collapse
|
69
|
Schlaepfer IR, Rider L, Rodrigues LU, Gijón MA, Pac CT, Romero L, Cimic A, Sirintrapun SJ, Glodé LM, Eckel RH, Cramer SD. Lipid catabolism via CPT1 as a therapeutic target for prostate cancer. Mol Cancer Ther 2014; 13:2361-71. [PMID: 25122071 DOI: 10.1158/1535-7163.mct-14-0183] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy among Western men and accounts for the second leading cause of cancer-related deaths. Prostate cancer tends to grow slowly and recent studies suggest that it relies on lipid fuel more than on aerobic glycolysis. However, the biochemical mechanisms governing the relationships between lipid synthesis, lipid utilization, and cancer growth remain unknown. To address the role of lipid metabolism in prostate cancer, we have used etomoxir and orlistat, clinically safe drugs that block lipid oxidation and lipid synthesis/lipolysis, respectively. Etomoxir is an irreversible inhibitor of the carnitine palmitoyltransferase (CPT1) enzyme that decreases β oxidation in the mitochondria. Combinatorial treatments using etomoxir and orlistat resulted in synergistic decreased viability in LNCaP, VCaP, and patient-derived benign and prostate cancer cells. These effects were associated with decreased androgen receptor expression, decreased mTOR signaling, and increased caspase-3 activation. Knockdown of CPT1A enzyme in LNCaP cells resulted in decreased palmitate oxidation but increased sensitivity to etomoxir, with inactivation of AKT kinase and activation of caspase-3. Systemic treatment with etomoxir in nude mice resulted in decreased xenograft growth over 21 days, underscoring the therapeutic potential of blocking lipid catabolism to decrease prostate cancer tumor growth.
Collapse
Affiliation(s)
- Isabel R Schlaepfer
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado.
| | - Leah Rider
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | | | - Miguel A Gijón
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Colton T Pac
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Lina Romero
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Adela Cimic
- Department of Pathology, Wake Forest University School of Medicine Winston-Salem, North Carolina
| | - S Joseph Sirintrapun
- Department of Pathology, Wake Forest University School of Medicine Winston-Salem, North Carolina
| | - L Michael Glodé
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Robert H Eckel
- Division of Endocrinology Metabolism and Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Scott D Cramer
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
70
|
Lewis DY, Boren J, Shaw GL, Bielik R, Ramos-Montoya A, Larkin TJ, Martins CP, Neal DE, Soloviev D, Brindle KM. Late Imaging with [1-(11)C]Acetate Improves Detection of Tumor Fatty Acid Synthesis with PET. J Nucl Med 2014; 55:1144-9. [PMID: 24777291 DOI: 10.2967/jnumed.113.134437] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 03/19/2014] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED Tumors are often characterized by high levels of de novo fatty acid synthesis. The kinetics of acetate incorporation into tricarboxylic acid cycle intermediates and into lipids suggest that detection of tumors with [1-(11)C]acetate PET could be improved by imaging at later time points. METHODS The uptake and metabolism of [1-(11)C], [1-(13)C], and [1-(14)C]acetate were measured in mouse prostate and lung cancer models to investigate the time course of (11)C label incorporation into tumor metabolites. RESULTS Radioactivity in the lipid fraction, as compared with the aqueous fraction, in extracts of C4-2B human prostate xenografts peaked at 90 min after [1-(14)C]acetate injection, which coincided with peak (13)C label incorporation into the fatty acids palmitate and stearate. Contrast between the tumor and tissues, such as blood and muscle, increased in PET images acquired over a period of 120 min after [1-(11)C]acetate injection, and Patlak plots were linear from 17.5 min after injection. Similar results were obtained in a genetically engineered K-ras(G12D); p53(null) lung cancer model, in which the mean tumor-to-lung ratio at 90 min after [1-(14)C]acetate injection was 4.4-fold higher than at 15 min. CONCLUSION These findings suggest that when imaging de novo fatty acid synthesis with [1-(11)C]acetate it is preferable to measure uptake at later time points, when the effects of perfusion and (11)C incorporation into tricarboxylic acid cycle intermediates and bicarbonate are declining. The data presented here suggest that future clinical PET scans of tumors should be acquired later than 30 min, when tracer accumulation due to de novo fatty acid synthesis prevails.
Collapse
Affiliation(s)
- David Y Lewis
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Joan Boren
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Greg L Shaw
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Robert Bielik
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Antonio Ramos-Montoya
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Timothy J Larkin
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom; and
| | - Carla P Martins
- MRC Cancer Unit, University of Cambridge, Cambridge, United Kingdom
| | - David E Neal
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Dmitry Soloviev
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Kevin M Brindle
- Cancer Research United Kingdom-Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom; and
| |
Collapse
|
71
|
Rapid diagnosis and staging of colorectal cancer via high-resolution magic angle spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy of intact tissue biopsies. Ann Surg 2014; 259:1138-49. [PMID: 23860197 DOI: 10.1097/sla.0b013e31829d5c45] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To develop novel metabolite-based models for diagnosis and staging in colorectal cancer (CRC) using high-resolution magic angle spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy. BACKGROUND Previous studies have demonstrated that cancer cells harbor unique metabolic characteristics relative to healthy counterparts. This study sought to characterize metabolic properties in CRC using HR-MAS NMR spectroscopy. METHODS Between November 2010 and January 2012, 44 consecutive patients with confirmed CRC were recruited to a prospective observational study. Fresh tissue samples were obtained from center of tumor and 5 cm from tumor margin from surgical resection specimens. Samples were run in duplicate where tissue volume permitted to compensate for anticipated sample heterogeneity. Samples were subjected to HR-MAS NMR spectroscopic profiling and acquired spectral data were imported into SIMCA and MATLAB statistical software packages for unsupervised and supervised multivariate analysis. RESULTS A total of 171 spectra were acquired (center of tumor, n = 88; 5 cm from tumor margin, n = 83). Cancer tissue contained significantly increased levels of lactate (P < 0.005), taurine (P < 0.005), and isoglutamine (P < 0.005) and decreased levels of lipids/triglycerides (P < 0.005) relative to healthy mucosa (R2Y = 0.94; Q2Y = 0.72; area under the curve, 0.98). Colon cancer samples (n = 49) contained higher levels of acetate (P < 0.005) and arginine (P < 0.005) and lower levels of lactate (P < 0.005) relative to rectal cancer samples (n = 39). In addition unique metabolic profiles were observed for tumors of differing T-stage. CONCLUSIONS HR-MAS NMR profiling demonstrates cancer-specific metabolic signatures in CRC and reveals metabolic differences between colonic and rectal cancers. In addition, this approach reveals that tumor metabolism undergoes modification during local tumor advancement, offering potential in future staging and therapeutic approaches.
Collapse
|
72
|
Yoshii Y, Furukawa T, Saga T, Fujibayashi Y. Acetate/acetyl-CoA metabolism associated with cancer fatty acid synthesis: overview and application. Cancer Lett 2014; 356:211-6. [PMID: 24569091 DOI: 10.1016/j.canlet.2014.02.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 02/17/2014] [Accepted: 02/17/2014] [Indexed: 12/29/2022]
Abstract
Understanding cancer-specific metabolism is important for identifying novel targets for cancer diagnosis and therapy. Induced acetate/acetyl CoA metabolism is a notable feature that is related to fatty acid synthesis supporting tumor growth. In this review, we focused on the recent findings related to cancer acetate/acetyl CoA metabolism. We also introduce [1-¹¹C]acetate positron emission tomography (PET), which is a useful tool to visualize up-regulation of acetate/acetyl CoA metabolism in cancer, and discuss the utility of [1-¹¹C]acetate PET in cancer diagnosis and its application to personalized medicine.
Collapse
Affiliation(s)
- Yukie Yoshii
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan.
| | - Takako Furukawa
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Yasuhisa Fujibayashi
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
73
|
Merchant S, Witney TH, Aboagye EO. Imaging as a pharmacodynamic and response biomarker in cancer. Clin Transl Imaging 2014. [DOI: 10.1007/s40336-014-0049-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
74
|
Zadra G, Photopoulos C, Tyekucheva S, Heidari P, Weng QP, Fedele G, Liu H, Scaglia N, Priolo C, Sicinska E, Mahmood U, Signoretti S, Birnberg N, Loda M. A novel direct activator of AMPK inhibits prostate cancer growth by blocking lipogenesis. EMBO Mol Med 2014; 6:519-38. [PMID: 24497570 PMCID: PMC3992078 DOI: 10.1002/emmm.201302734] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
5′AMP-activated kinase (AMPK) constitutes a hub for cellular metabolic and growth control, thus representing an ideal therapeutic target for prostate cancers (PCas) characterized by increased lipogenesis and activation of mTORC1 pathway. However, whether AMPK activation itself is sufficient to block cancer cell growth remains to be determined. A small molecule screening was performed and identified MT 63–78, a specific and potent direct AMPK activator. Here, we show that direct activation of AMPK inhibits PCa cell growth in androgen sensitive and castration resistant PCa (CRPC) models, induces mitotic arrest, and apoptosis. In vivo, AMPK activation is sufficient to reduce PCa growth, whereas the allelic loss of its catalytic subunits fosters PCa development. Importantly, despite mTORC1 blockade, the suppression of de novo lipogenesis is the underpinning mechanism responsible for AMPK-mediated PCa growth inhibition, suggesting AMPK as a therapeutic target especially for lipogenesis-driven PCas. Finally, we demonstrate that MT 63–78 enhances the growth inhibitory effect of AR signaling inhibitors MDV3100 and abiraterone. This study thus provides a rationale for their combined use in CRPC treatment.
Collapse
Affiliation(s)
- Giorgia Zadra
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Andersen KF, Divilov V, Sevak K, Koziorowski J, Lewis JS, Pillarsetty N. Influence of free fatty acids on glucose uptake in prostate cancer cells. Nucl Med Biol 2013; 41:254-8. [PMID: 24440212 DOI: 10.1016/j.nucmedbio.2013.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 11/18/2013] [Accepted: 12/10/2013] [Indexed: 10/25/2022]
Abstract
INTRODUCTION The study focuses on the interaction between glucose and free fatty acids (FFA) in malignant human prostate cancer cell lines by an in vitro observation of uptake of fluoro-2-deoxy-D-glucose (FDG) and acetate. METHODS Human prostate cancer cell lines (PC3, CWR22Rv1, LNCaP, and DU145) were incubated for 2 h and 24 h in glucose-containing (5.5 mM) Dulbecco's Modified Eagle's Medium (DMEM) with varying concentrations of the free fatty acid palmitate (0-1.0 mM). Then the cells were incubated with [(18)F]-FDG (1 μCi/mL; 0.037 MBq/mL) in DMEM either in presence or absence of glucose and in presence of varying concentrations of palmitate for 1 h. Standardized procedures regarding cell counting and measuring for (18)F radioactivity were applied. Cell uptake studies with (14)C-1-acetate under the same conditions were performed on PC3 cells. RESULTS In glucose containing media there was significantly increased FDG uptake after 24 h incubation in all cell lines, except DU145, when upper physiological levels of palmitate were added. A 4-fold increase of FDG uptake in PC3 cells (15.11% vs. 3.94%/10(6) cells) was observed in media with 1.0 mM palmitate compared to media with no palmitate. The same tendency was observed in PC3 and CWR22Rv1 cells after 2 h incubation. In glucose-free media no significant differences in FDG uptake after 24 h incubation were observed. The significant differences after 2 h incubation all pointed in the direction of increased FDG uptake when palmitate was added. Acetate uptake in PC3 cells was significantly lower when palmitate was added in glucose-free DMEM. No clear tendency when comparing FDG or acetate uptake in the same media at different time points of incubation was observed. CONCLUSIONS Our results indicate a FFA dependent metabolic boost/switch of glucose uptake in PCa, with patterns reflecting the true heterogeneity of the disease.
Collapse
Affiliation(s)
- Kim Francis Andersen
- Department of Clinical Physiology & Nuclear Medicine, Herlev Hospital, University Hospital of Copenhagen, Herlev, Denmark.
| | - Vadim Divilov
- Radiochemistry & Imaging Sciences Services, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Kuntalkumar Sevak
- Radiochemistry & Imaging Sciences Services, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Jacek Koziorowski
- Department of Clinical Physiology & Nuclear Medicine, Herlev Hospital, University Hospital of Copenhagen, Herlev, Denmark
| | - Jason S Lewis
- Radiochemistry & Imaging Sciences Services, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - NagaVarakishore Pillarsetty
- Radiochemistry & Imaging Sciences Services, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
76
|
Metabolic shifts induced by fatty acid synthase inhibitor orlistat in non-small cell lung carcinoma cells provide novel pharmacodynamic biomarkers for positron emission tomography and magnetic resonance spectroscopy. Mol Imaging Biol 2013; 15:136-47. [PMID: 22886728 PMCID: PMC3591534 DOI: 10.1007/s11307-012-0587-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Purpose Abnormal fatty acid (FA) synthesis is one of the common features of cancer. Fatty acid synthase (FASN), a multifunctional enzyme playing a key role in biosynthesis of FA, is up-regulated in prostate, breast, and lung carcinomas. Orlistat is a FDA-approved anti-obesity drug that inhibits the thioesterase domain of FASN, interferes with cellular FA synthesis, can arrest tumor cell proliferation, and induces tumor cell apoptosis. The current study was aimed to investigate the metabolic changes associated with FASN inhibition by orlistat and to understand the molecular mechanisms behind the observed metabolic changes in non-small cell lung carcinoma (NSCLC) cell lines. Procedures Changes in metabolite pools in four NSCLC cell lines (H441, H1975, H3255, and PC14) with different mutational profiles were studied using NMR spectroscopy before and after in vitro incubation with sub-toxic concentration of orlistat and [1-13C]d-glucose or [1,2-13C2]choline. In vitro radiotracer accumulation assays in cells were performed with [3H]acetate, [14C]fluoroacetate, and 2-deoxy-2-[18F]fluoro-d-glucose. In parallel, microarray profiling of genes involved in the regulation of carbohydrate and lipid metabolism was performed. Results In orlistat-treated NSCLC cells, FASN inhibition results in characteristic changes in intermediary metabolites (FAs, choline, phospholipids, and TCA cycle metabolites) as observed by magnetic resonance spectroscopy. Further, FASN inhibition by orlistat induces multiple adaptive changes in FA synthetic pathway and associated metabolic pathways, including induction of ketone metabolism and glutaminolysis, as well as the up-regulation of 5' adenosine monophosphate-activated protein kinase. Conclusions These observed changes in metabolic pools in orlistat-treated cells demonstrate the critical role of fatty acid de novo synthesis and metabolism for cellular energy production, especially in tumor cells with low glycolytic activity, which goes beyond the widely accepted concept that FA synthesis is important for cell membrane biosynthesis in rapidly proliferating tumor cells. Electronic supplementary material The online version of this article (doi:10.1007/s11307-012-0587-6) contains supplementary material, which is available to authorized users.
Collapse
|
77
|
Goswami S, Maity S, Das AK, Maity AC. Single chemosensor for highly selective colorimetric and fluorometric dual sensing of Cu(II) as well as ‘NIRF’ to acetate ion. Tetrahedron Lett 2013. [DOI: 10.1016/j.tetlet.2013.09.126] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
78
|
Deford-Watts LM, Mintz A, Kridel SJ. The potential of ¹¹C-acetate PET for monitoring the Fatty acid synthesis pathway in Tumors. Curr Pharm Biotechnol 2013; 14:300-12. [PMID: 23597406 DOI: 10.2174/1389201011314030006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 09/17/2010] [Indexed: 11/22/2022]
Abstract
Positron emission tomography (PET) is a molecular imaging modality that provides the opportunity to rapidly and non-invasively visualize tumors derived from multiple organs. In order to do so, PET utilizes radiotracers, such as ¹⁸F-FDG and ¹¹C-acetate, whose uptake coincides with altered metabolic pathways within tumors. Increased expression and activity of enzymes in the fatty acid synthesis pathway is a frequent hallmark of cancer cells. As a result, this pathway has become a prime target for therapeutic intervention. Although multiple drugs have been developed that both directly and indirectly interfere with fatty acid synthesis, an optimal means to assess their efficacy is lacking. Given that ¹¹Cacetate is directly linked to the fatty acid synthesis pathway, this probe provides a unique opportunity to monitor lipogenic tumors by PET. Herein, we review the relevance of the fatty acid synthesis pathway in cancer. Furthermore, we address the potential utility of ¹¹C-acetate PET in imaging tumors, especially those that are not FDG-avid. Last, we discuss several therapeutic interventions that could benefit from ¹¹C-acetate PET to monitor therapeutic response in patients with certain types of cancers.
Collapse
Affiliation(s)
- Laura M Deford-Watts
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | |
Collapse
|
79
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 PMCID: PMC3799232 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
80
|
Abstract
Prostate cancer is a heterogeneous disease, and its management is now evolving to become more personalized and to incorporate new targeted therapies. With these new changes comes a demand for molecular imaging techniques that can not only detect disease but also assess biology and treatment response. This review article summarizes current molecular imaging approaches in prostate cancer (e.g. 99mTc bone scintigraphy and 18F-fluorodeoxyglucose positron emission tomography) and highlights emerging clinical and preclinical imaging agents, with an emphasis on mechanism and clinical application. Emerging agents at various stages of clinical translation include radiolabeled analogs of lipid, amino acid, and nucleoside metabolism, as well as agents more specifically targeting prostate cancer biomarkers including androgen receptor, prostate-specific membrane antigen and others. We also highlight new techniques and targeted contrast agents for magnetic resonance imaging and spectroscopy. For all these imaging techniques, a growing and important unmet need is for well-designed prospective clinical trials to establish clear indications with clinical benefit in prostate cancer.
Collapse
Affiliation(s)
- Ana P. Kiess
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Steve Y. Cho
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
81
|
Fortuin A, de Rooij M, Zamecnik P, Haberkorn U, Barentsz J. Molecular and functional imaging for detection of lymph node metastases in prostate cancer. Int J Mol Sci 2013; 14:13842-75. [PMID: 23823804 PMCID: PMC3742221 DOI: 10.3390/ijms140713842] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 12/11/2022] Open
Abstract
Knowledge on lymph node metastases is crucial for the prognosis and treatment of prostate cancer patients. Conventional anatomic imaging often fails to differentiate benign from metastatic lymph nodes. Pelvic lymph node dissection is an invasive technique and underestimates the extent of lymph node metastases. Therefore, there is a need for more accurate non-invasive diagnostic techniques. Molecular and functional imaging has been subject of research for the last decades, in this respect. Therefore, in this article the value of imaging techniques to detect lymph node metastases is reviewed. These techniques include scintigraphy, sentinel node imaging, positron emission tomography/computed tomography (PET/CT), diffusion weighted magnetic resonance imaging (DWI MRI) and magnetic resonance lymphography (MRL). Knowledge on pathway and size of lymph node metastases has increased with molecular and functional imaging. Furthermore, improved detection and localization of lymph node metastases will enable (focal) treatment of the positive nodes only.
Collapse
Affiliation(s)
- Ansje Fortuin
- Department of Radiology, Nijmegen Medical Center, Radboud University, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; E-Mails: (M.R.); (P.Z.); (J.B.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +31-24-3611-111; Fax: +31-24-3540-866
| | - Maarten de Rooij
- Department of Radiology, Nijmegen Medical Center, Radboud University, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; E-Mails: (M.R.); (P.Z.); (J.B.)
- Department of Operation Rooms, Nijmegen Medical Center, Radboud University, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Patrik Zamecnik
- Department of Radiology, Nijmegen Medical Center, Radboud University, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; E-Mails: (M.R.); (P.Z.); (J.B.)
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University of Heidelberg, Im Neuenheimer Feld 400, Heidelberg 69120, Germany; E-Mail:
| | - Jelle Barentsz
- Department of Radiology, Nijmegen Medical Center, Radboud University, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; E-Mails: (M.R.); (P.Z.); (J.B.)
| |
Collapse
|
82
|
Yoshii Y, Furukawa T, Oyama N, Hasegawa Y, Kiyono Y, Nishii R, Waki A, Tsuji AB, Sogawa C, Wakizaka H, Fukumura T, Yoshii H, Fujibayashi Y, Lewis JS, Saga T. Fatty acid synthase is a key target in multiple essential tumor functions of prostate cancer: uptake of radiolabeled acetate as a predictor of the targeted therapy outcome. PLoS One 2013; 8:e64570. [PMID: 23741342 PMCID: PMC3669310 DOI: 10.1371/journal.pone.0064570] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 04/15/2013] [Indexed: 11/18/2022] Open
Abstract
Fatty acid synthase (FASN) expression is elevated in several cancers, and this over-expression is associated with poor prognosis. Inhibitors of FASN, such as orlistat, reportedly show antitumor effects against cancers that over-express FASN, making FASN a promising therapeutic target. However, large variations in FASN expression levels in individual tumors have been observed, and methods to predict FASN-targeted therapy outcome before treatment are required to avoid unnecessary treatment. In addition, how FASN inhibition affects tumor progression remains unclear. Here, we showed the method to predict FASN-targeted therapy outcome using radiolabeled acetate uptake and presented mechanisms of FASN inhibition with human prostate cancer cell lines, to provide the treatment strategy of FASN-targeted therapy. We revealed that tumor uptake of radiolabeled acetate reflected the FASN expression levels and sensitivity to FASN-targeted therapy with orlistat in vitro and in vivo. FASN-targeted therapy was noticeably effective against tumors with high FASN expression, which was indicated by high acetate uptake. To examine mechanisms, we established FASN knockdown prostate cancer cells by transduction of short-hairpin RNA against FASN and investigated the characteristics by analyses on morphology and cell behavior and microarray-based gene expression profiling. FASN inhibition not only suppressed cell proliferation but prevented pseudopodia formation and suppressed cell adhesion, migration, and invasion. FASN inhibition also suppressed genes involved in production of intracellular second messenger arachidonic acid and androgen hormones, both of which promote tumor progression. Collectively, our data demonstrated that uptake of radiolabeled acetate is a useful predictor of FASN-targeted therapy outcome. This suggests that [1-(11)C]acetate positron emission tomography (PET) could be a powerful tool to accomplish personalized FASN-targeted therapy by non-invasive visualization of tumor acetate uptake and selection of responsive tumors. FASN-targeted therapy could be an effective treatment to suppress multiple steps related to tumor progression in prostate cancers selected by [1-(11)C]acetate PET.
Collapse
Affiliation(s)
- Yukie Yoshii
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Tsai IL, Kuo TC, Ho TJ, Harn YC, Wang SY, Fu WM, Kuo CH, Tseng YJ. Metabolomic Dynamic Analysis of Hypoxia in MDA-MB-231 and the Comparison with Inferred Metabolites from Transcriptomics Data. Cancers (Basel) 2013; 5:491-510. [PMID: 24216987 PMCID: PMC3730319 DOI: 10.3390/cancers5020491] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 04/24/2013] [Accepted: 04/24/2013] [Indexed: 01/04/2023] Open
Abstract
Hypoxia affects the tumor microenvironment and is considered important to metastasis progression and therapy resistance. Thus far, the majority of global analyses of tumor hypoxia responses have been limited to just a single omics level. Combining multiple omics data can broaden our understanding of tumor hypoxia. Here, we investigate the temporal change of the metabolite composition with gene expression data from literature to provide a more comprehensive insight into the system level in response to hypoxia. Nuclear magnetic resonance spectroscopy was used to perform metabolomic profiling on the MDA-MB-231 breast cancer cell line under hypoxic conditions. Multivariate statistical analysis revealed that the metabolic difference between hypoxia and normoxia was similar over 24 h, but became distinct over 48 h. Time dependent microarray data from the same cell line in the literature displayed different gene expressions under hypoxic and normoxic conditions mostly at 12 h or earlier. The direct metabolomic profiles show a large overlap with theoretical metabolic profiles deduced from previous transcriptomic studies. Consistent pathways are glycolysis/gluconeogenesis, pyruvate, purine and arginine and proline metabolism. Ten metabolic pathways revealed by metabolomics were not covered by the downstream of the known transcriptomic profiles, suggesting new metabolic phenotypes. These results confirm previous transcriptomics understanding and expand the knowledge from existing models on correlation and co-regulation between transcriptomic and metabolomics profiles, which demonstrates the power of integrated omics analysis.
Collapse
Affiliation(s)
- I-Lin Tsai
- Department of Pharmacy, National Taiwan University, No. 1, Jen-Ai Road, Section 1 Taipei 10051, Taiwan; E-Mail:
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Center for Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Tien-Chueh Kuo
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Graduate Institute of Biomedical Electronic and Bioinformatics, National Taiwan University, Room 410 BL Building, No. 1, Roosevelt Road, Sec. 4, Taipei 106, Taiwan
| | - Tsung-Jung Ho
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Department of Computer Science and Information Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan
| | - Yeu-Chern Harn
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Graduate Institute of Networking and Multimedia, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan
| | - San-Yuan Wang
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Department of Computer Science and Information Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan
| | - Wen-Mei Fu
- Department of Pharmacology, National Taiwan University, 11 F No. 1 Sec. 1, Ren-ai Rd., Taipei 10051, Taiwan; E-Mail:
| | - Ching-Hua Kuo
- Department of Pharmacy, National Taiwan University, No. 1, Jen-Ai Road, Section 1 Taipei 10051, Taiwan; E-Mail:
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Center for Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan
- Authors to whom correspondence should be addressed; E-Mails: (C.-H.K.); (Y.J.T.); Tel: +886-2-3366-4888 (Y.J.T.); Fax: +886-2-2362-8167 (Y.J.T.)
| | - Yufeng Jane Tseng
- Department of Pharmacy, National Taiwan University, No. 1, Jen-Ai Road, Section 1 Taipei 10051, Taiwan; E-Mail:
- The Metabolomics Group, National Taiwan University, Taipei 106, Taiwan; E-Mails: (T.-C.K.); (T.-J.H.); (Y.-C.H.); (S.-Y.W.)
- Center for Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Biomedical Electronic and Bioinformatics, National Taiwan University, Room 410 BL Building, No. 1, Roosevelt Road, Sec. 4, Taipei 106, Taiwan
- Department of Computer Science and Information Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan
- Authors to whom correspondence should be addressed; E-Mails: (C.-H.K.); (Y.J.T.); Tel: +886-2-3366-4888 (Y.J.T.); Fax: +886-2-2362-8167 (Y.J.T.)
| |
Collapse
|
84
|
Brogsitter C, Zöphel K, Kotzerke J. 18F-Choline, 11C-choline and 11C-acetate PET/CT: comparative analysis for imaging prostate cancer patients. Eur J Nucl Med Mol Imaging 2013; 40 Suppl 1:S18-27. [PMID: 23579863 DOI: 10.1007/s00259-013-2358-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 01/24/2013] [Indexed: 11/26/2022]
Affiliation(s)
- Claudia Brogsitter
- Department of Nuclear Medicine, Carl Gustav Carus Medical School, University of Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | | | | |
Collapse
|
85
|
Zadra G, Photopoulos C, Loda M. The fat side of prostate cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1518-32. [PMID: 23562839 DOI: 10.1016/j.bbalip.2013.03.010] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 03/23/2013] [Accepted: 03/24/2013] [Indexed: 12/28/2022]
Abstract
Prostate cancer (PCa) metabolism appears to be unique in comparison with other types of solid cancers. Normal prostate cells mainly rely on glucose oxidation to provide precursors for the synthesis and secretion of citrate, resulting in an incomplete Krebs cycle and minimal oxidative phosphorylation for energy production. In contrast, during transformation, PCa cells no longer secrete citrate and they reactivate the Krebs cycle as energy source. Moreover, primary PCas do not show increased aerobic glycolysis and therefore they are not efficiently detectable with (18)F-FDG-PET. However, increased de novo lipid synthesis, strictly intertwined with deregulation in classical oncogenes and oncosuppressors, is an early event of the disease. Up-regulation and increased activity of lipogenic enzymes (including fatty acid synthase and choline kinase) occurs throughout PCa carcinogenesis and correlates with worse prognosis and poor survival. Thus, lipid precursors such as acetate and choline have been successfully used as alternative tracers for PET imaging. Lipid synthesis intermediates and FA catabolism also emerged as important players in PCa maintenance. Finally, epidemiologic studies suggested that systemic metabolic disorders including obesity, metabolic syndrome, and diabetes as well as hypercaloric and fat-rich diets might increase the risk of PCa. However, how metabolic disorders contribute to PCa development and whether dietary lipids and de novo lipids synthesized intra-tumor are differentially metabolized still remains unclear. In this review, we examine the switch in lipid metabolism supporting the development and progression of PCa and we discuss how we can exploit its lipogenic nature for therapeutic and diagnostic purposes. This article is part of a Special Issue entitled Lipid Metabolism in Cancer.
Collapse
Affiliation(s)
- Giorgia Zadra
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
86
|
Haseebuddin M, Dehdashti F, Siegel BA, Liu J, Roth EB, Nepple KG, Siegel CL, Fischer KC, Kibel AS, Andriole GL, Miller TR. 11C-acetate PET/CT before radical prostatectomy: nodal staging and treatment failure prediction. J Nucl Med 2013; 54:699-706. [PMID: 23471311 DOI: 10.2967/jnumed.112.111153] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Despite early detection programs, many patients with prostate cancer present with intermediate- or high-risk disease. We prospectively investigated whether (11)C-acetate PET/CT predicts lymph node (LN) metastasis and treatment failure in men for whom radical prostatectomy is planned. METHODS 107 men with intermediate- or high-risk localized prostate cancer and negative conventional imaging findings underwent PET/CT with (11)C-acetate. Five underwent LN staging only, and 102 underwent LN staging and prostatectomy. PET/CT findings were correlated with pathologic nodal status. Treatment-failure-free survival was estimated by the Kaplan-Meier method. The ability of PET/CT to predict outcomes was evaluated by multivariate Cox proportional hazards analysis. RESULTS PET/CT was positive for pelvic LN or distant metastasis in 36 of 107 patients (33.6%). LN metastasis was present histopathologically in 25 (23.4%). The sensitivity, specificity, and positive and negative predictive values of PET/CT for detecting LN metastasis were 68.0%, 78.1%, 48.6%, and 88.9%, respectively. Treatment failed in 64 patients: 25 with metastasis, 17 with a persistent postprostatectomy prostate-specific antigen level greater than 0.20 ng/mL, and 22 with biochemical recurrence (prostate-specific antigen level > 0.20 ng/mL after nadir) during follow-up for a median of 44.0 mo. Treatment-failure-free survival was worse in PET-positive than in PET-negative patients (P < 0.0001) and in those with false-positive than in those with true-negative scan results (P < 0.01), suggesting that PET may have demonstrated nodal disease not removed surgically or identified pathologically. PET positivity independently predicted failure in preoperative (hazard ratio, 3.26; P < 0.0001) and postoperative (hazard ratio, 3.07; P = 0.0001) multivariate models. CONCLUSION In patients planned for or completing prostatectomy, (11)C-acetate PET/CT detects LN metastasis not identified by conventional imaging and independently predicts treatment-failure-free survival.
Collapse
Affiliation(s)
- Mohammed Haseebuddin
- Division of Urologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Roivainen A, Naum A, Nuutinen H, Leino R, Nurmi H, Någren K, Parkkola R, Virtanen J, Kallajoki M, Kujari H, Ovaska J, Roberts P, Seppänen M. Characterization of hepatic tumors using [11C]metomidate through positron emission tomography: comparison with [11C]acetate. EJNMMI Res 2013; 3:13. [PMID: 23442595 PMCID: PMC3598556 DOI: 10.1186/2191-219x-3-13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 02/15/2013] [Indexed: 12/14/2022] Open
Abstract
Background Using positron emission tomography (PET), we compared two tracers, [11C]metomidate ([11C]MTO) and [11C]acetate ([11C]ACE), for the characterization of hepatic tumors. Methods Thirty-three patients underwent PET with [11C]MTO and [11C]ACE and magnetic resonance imaging (MRI). Based on the histology of the tumor biopsy, 14 patients had hepatocellular carcinoma (HCC), 9 patients had focal nodular hyperplasia (FNH), and 10 patients had other types of hepatic tumors. Tumor uptake was evaluated by calculating the maximum and mean standardized uptake value and tumor-to-liver ratio. Results Altogether, 120 hepatic lesions (59 HCC, 18 FNH, 30 metastases of different primaries, 9 adenomas, and 4 regenerating nodules of liver cirrhosis) were detected by MRI. The overall tumor detection rate was slightly higher for [11C]MTO (39%) than for [11C]ACE (33%). [11C]ACE was more sensitive for HCC detection (50% versus 43%, respectively), whereas [11C]MTO was more sensitive for FNH detection (78% versus 44%, respectively). In HCC patients, the tumor grade correlated with [11C]ACE, but not with [11C]MTO. All of the patients with liver metastases, from various primary tumors (n = 10), were negative for both tracers. Conclusions Due to low sensitivity, [11C]MTO and [11C]ACE PET have only limited value in diagnosing hepatic tumors.
Collapse
Affiliation(s)
- Anne Roivainen
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, Turku 20521, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Takesh M. The Potential Benefit by Application of Kinetic Analysis of PET in the Clinical Oncology. ISRN ONCOLOGY 2012; 2012:349351. [PMID: 23326682 PMCID: PMC3541563 DOI: 10.5402/2012/349351] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 11/25/2012] [Indexed: 12/22/2022]
Abstract
PET is an appropriate method to display the functional activities in target tissue using many types of traces. The visual assessment of PET images plus the semiquantitative parameter (SUV) are the main diagnostic standards considered in identifying the malignant lesion. However, these standards lack occasionally the proper specificity and/or sensitivity. That emphasizes the importance of considering supplemental diagnostic criteria such as the kinetic parameter. The latter gives the way to image the ongoing metabolic processes within the target tissue as well as to identify the alterations occurring at the microscale level before they become observable in the conventional PET-imaging.
The importance of kinetic analysis of PET imaging has increased with newly developed PET devices that offer images of good quality and high spatial resolution.
In this paper, we highlighted the potential contribution of kinetic analysis in improving the diagnostic accuracy in intracranial tumour, lung tumour, liver tumour, colorectal tumour, bone and soft tissue tumours, and prostate cancer. Moreover, we showed that the appropriate therapy monitoring can be best achieved after considering the kinetic parameters. These promising results indicate that the kinetic analysis of PET imaging may become an essential part in preclinical and clinical molecular imaging as well.
Collapse
Affiliation(s)
- Mustafa Takesh
- Nuclear Medicine Department, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| |
Collapse
|
89
|
Chen HW, Chang YF, Chuang HY, Tai WT, Hwang JJ. Targeted therapy with fatty acid synthase inhibitors in a human prostate carcinoma LNCaP/tk-luc-bearing animal model. Prostate Cancer Prostatic Dis 2012; 15:260-4. [PMID: 22565411 DOI: 10.1038/pcan.2012.15] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Fatty acid synthase (FASN) is highly upregulated in human prostate carcinomas. Inhibition of FASN could arrest cell cycle and trigger apoptosis rapidly, implying the reliance of cancer cell survival on FASN. However, little is known about the effect of C75, a FASN inhibitor, and siFASN (that is, small interfering RNA targeted at FASN) on prostate cancer in living subjects. METHODS We used C75 and siFASN to mediate the endogenous fatty acid metabolism in LNCaP human prostate cancer cells stably expressing herpes simplex virus type 1 thymidine kinase (HSV1-tk) and luciferase (luc) reporter genes, and assessed the effect of FASN blockade with different schedules of administration on tumor growth using noninvasive molecular imaging. RESULTS FASN blockade exhibited the proliferative inhibition and induced G1-phase cell cycle arrest of LNCaP cells. For in vivo studies, the tumor growth inhibition by C75 (total 120 mgkg(-1); 30 mgkg(-1) once a week or 15 mgkg(-1) twice a week for 4 weeks) and siFASN (1.4 mgkg(-1) every alternate day up to 16 days) treatments were 80% and 70%, respectively, compared with that of the control. CONCLUSION The results suggest that C75 may be superior to siFASN in anticancer effect on prostate cancer.
Collapse
Affiliation(s)
- H-W Chen
- Department of Radiation Oncology, Mackay Memorial Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
90
|
Abstract
Functional radionuclide imaging modalities, now commonly combined with anatomical imaging modalities computed tomography (CT) or magnetic resonance imaging (single photon emission computed tomography [SPECT]/CT, positron emission tomography [PET]/CT, and PET/magnetic resonance imaging), are promising tools for the management of prostate cancer, particularly for therapeutic implications. Sensitive detection capability of prostate cancer using these imaging modalities is one issue; however, the treatment of prostate cancer using the information that can be obtained from functional radionuclide imaging techniques is another challenging area. There are not many SPECT or PET radiotracers that can cover the full spectrum of the management of prostate cancer from initial detection to staging, prognosis predictor, and all the way to treatment response assessment. However, when used appropriately, the information from functional radionuclide imaging improves, and sometimes significantly changes, the whole course of the cancer management. The limitations of using SPECT and PET radiotracers with regard to therapeutic implications are not so much different from their limitations solely for the task of detecting prostate cancer; however, the specific imaging target and how this target is reliably imaged by SPECT and PET can potentially make significant impact in the treatment of prostate cancer. Finally, although the localized prostate cancer is considered manageable, there is still significant need for improvement in noninvasive imaging of metastatic prostate cancer, in treatment guidance, and in response assessment from functional imaging, including radionuclide-based techniques. In this review article, we present the rationale of using functional radionuclide imaging and the therapeutic implications for each of radionuclide imaging agent that have been studied in human subjects.
Collapse
Affiliation(s)
- Carina Mari Aparici
- Department of Radiology and Biomedical Imaging, University of California, and Nuclear Medicine Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94107, USA
| | | |
Collapse
|
91
|
Abstract
OBJECTIVE Recent advances in the fundamental understanding of the complex biology of prostate cancer have provided an increasing number of potential targets for imaging and treatment. The imaging evaluation of prostate cancer needs to be tailored to the various phases of this remarkably heterogeneous disease. CONCLUSION In this article, I review the current state of affairs on a range of PET radiotracers for potential use in the imaging evaluation of men with prostate cancer.
Collapse
|
92
|
Foss CA, Mease RC, Cho SY, Kim HJ, Pomper MG. GCPII imaging and cancer. Curr Med Chem 2012; 19:1346-59. [PMID: 22304713 DOI: 10.2174/092986712799462612] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 12/26/2011] [Accepted: 12/27/2011] [Indexed: 12/11/2022]
Abstract
Glutamate carboxypeptidase II (GCPII) in the central nervous system is referred to as the prostate-specific membrane antigen (PSMA) in the periphery. PSMA serves as a target for imaging and treatment of prostate cancer and because of its expression in solid tumor neovasculature has the potential to be used in this regard for other malignancies as well. An overview of GCPII/PSMA in cancer, as well as a discussion of imaging and therapy of prostate cancer using a wide variety of PSMA-targeting agents is provided.
Collapse
Affiliation(s)
- C A Foss
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical School, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
93
|
Suburu J, Chen YQ. Lipids and prostate cancer. Prostaglandins Other Lipid Mediat 2012; 98:1-10. [PMID: 22503963 DOI: 10.1016/j.prostaglandins.2012.03.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 03/13/2012] [Accepted: 03/30/2012] [Indexed: 12/25/2022]
Abstract
The role of lipid metabolism has gained particular interest in prostate cancer research. A large body of literature has outlined the unique upregulation of de novo lipid synthesis in prostate cancer. Concordant with this lipogenic phenotype is a metabolic shift, in which cancer cells use alternative enzymes and pathways to facilitate the production of fatty acids. These newly synthesized lipids may support a number of cellular processes to promote cancer cell proliferation and survival. Hence, de novo lipogenesis is under intense investigation as a therapeutic target. Epidemiologic studies suggest dietary fat may also contribute to prostate cancer; however, whether dietary lipids and de novo synthesized lipids are differentially metabolized remains unclear. Here, we highlight the lipogenic nature of prostate cancer, especially the promotion of de novo lipid synthesis, and the significance of various dietary lipids in prostate cancer development and progression.
Collapse
Affiliation(s)
- Janel Suburu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
94
|
Mena E, Turkbey B, Mani H, Adler S, Valera VA, Bernardo M, Shah V, Pohida T, McKinney Y, Kwarteng G, Daar D, Lindenberg ML, Eclarinal P, Wade R, Linehan WM, Merino MJ, Pinto PA, Choyke PL, Kurdziel KA. 11C-Acetate PET/CT in localized prostate cancer: a study with MRI and histopathologic correlation. J Nucl Med 2012; 53:538-45. [PMID: 22343504 DOI: 10.2967/jnumed.111.096032] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED This work characterizes the uptake of (11)C-acetate in prostate cancer (PCa), benign prostate hyperplasia, and normal prostate tissue in comparison with multiparametric MRI, whole-mount histopathology, and clinical markers to evaluate the potential utility of (11)C-acetate for delineating intraprostatic tumors in a population of patients with localized PCa. METHODS Thirty-nine men with presumed localized PCa underwent dynamic-static abdominal-pelvic (11)C-acetate PET/CT for 30 min and 3-T multiparametric MRI before prostatectomy. PET/CT images were registered to MR images using pelvic bones for initial rotation-translation, followed by manual adjustments to account for prostate motion and deformation from the MRI endorectal coil. Whole-mount pathology specimens were sectioned using an MRI-based patient-specific mold resulting in improved registration between the MRI, PET, and pathology. (11)C-acetate PET standardized uptake values were compared with multiparametric MRI and pathology. RESULTS (11)C-acetate uptake was rapid but reversible, peaking at 3-5 min after injection and reaching a relative plateau at approximately 10 min. The average maximum standardized uptake value (10-12 min) of tumors was significantly higher than that of normal prostate tissue (4.4 ± 2.05 [range, 1.8-9.2] vs. 2.1 ± 0.94 [range, 0.7-3.4], respectively; P < 0.001); however, it was not significantly different from that of benign prostatic hyperplasia (4.8 ± 2.01 [range, 1.8-8.8]). A sector-based comparison with histopathology, including all tumors greater than 0.5 cm, revealed a sensitivity and specificity of 61.6% and 80.0%, respectively, for (11)C-acetate PET/CT and 82.3% and 95.1%, respectively, for MRI. The (11)C-acetate accuracy was comparable to that of MRI when only tumors greater than 0.9 cm were considered. In a small cohort (n = 9), (11)C-acetate uptake was independent of fatty acid synthase expression using immunohistochemistry. CONCLUSION (11)C-acetate PET/CT demonstrates higher uptake in tumor foci than in normal prostate tissue; however, (11)C-acetate uptake in tumors is similar to that in benign prostate hyperplasia nodules. Although (11)C-acetate PET/CT is not likely to have utility as an independent modality for evaluation of localized PCa, the high uptake in tumors may make it useful for monitoring focal therapy when tissue damage after therapy may limit anatomic imaging methods.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Sola A, Tárraga A, Molina P. A ferrocenyl-guanidine derivative as a highly selective electrochemical and colorimetric chemosensor molecule for acetate anions. Dalton Trans 2012; 41:8401-9. [DOI: 10.1039/c2dt30632h] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
96
|
Lodi F, Malizia C, Castellucci P, Cicoria G, Fanti S, Boschi S. Synthesis of oncological [11C]radiopharmaceuticals for clinical PET. Nucl Med Biol 2011; 39:447-60. [PMID: 22172394 DOI: 10.1016/j.nucmedbio.2011.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 10/14/2011] [Accepted: 10/22/2011] [Indexed: 01/05/2023]
Abstract
Positron emission tomography (PET) is a nuclear medicine modality which provides quantitative images of biological processes in vivo at the molecular level. Several PET radiopharmaceuticals labeled with short-lived isotopes such as (18)F and (11)C were developed in order to trace specific cellular and molecular pathways with the aim of enhancing clinical applications. Among these [(11)C]radiopharmaceuticals are N-[(11)C]methyl-choline ([(11)C]choline), l-(S-methyl-[(11)C])methionine ([(11)C]methionine) and 1-[(11)C]acetate ([(11)C]acetate), which have gained an important role in oncology where the application of 2-[(18)F]fluoro-2-deoxy-d-glucose ([(18)F]FDG) is suboptimal. Nevertheless, the production of these radiopharmaceuticals did not reach the same level of standardization as for [(18)F]FDG synthesis. This review describes the most recent developments in the synthesis of the above-mentioned [(11)C]radiopharmaceuticals aiming to increase the availability and hence the use of [(11)C]choline, [(11)C]methionine and [(11)C]acetate in clinical practice.
Collapse
Affiliation(s)
- Filippo Lodi
- PET Radiopharmacy, Nuclear Medicine Unit, Azienda Ospedaliero Universitaria di Bologna, Policlinico S. Orsola-Malpighi, Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
97
|
Application of positron emission tomography molecular probes in hepatocellular carcinoma biological imaging. ACTA ACUST UNITED AC 2011; 26:113-8. [PMID: 21703120 DOI: 10.1016/s1001-9294(11)60030-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Biological behavior is a hot issue in hepatocellular carcinoma (HCC) study. Positron emission tomography (PET), a biological imaging technique, has been widely applied in many types of tumors. It is capable of noninvasive detection of biological behavior. Different radiotracers provide different information of HCC, including glucose/lipid metabolism, DNA synthesis, and apoptosis. In addition, radiotracer uptake relates to biological and clinical prognostic markers. In this article we review the application of several existing and novel radiotracers in PET in HCC study.
Collapse
|
98
|
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, Michigan 48201, USA.
| |
Collapse
|
99
|
Chuang HY, Chang YF, Hwang JJ. Antitumor effect of orlistat, a fatty acid synthase inhibitor, is via activation of caspase-3 on human colorectal carcinoma-bearing animal. Biomed Pharmacother 2011; 65:286-92. [PMID: 21723078 DOI: 10.1016/j.biopha.2011.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 02/08/2011] [Indexed: 12/30/2022] Open
Abstract
We established a HT-29/tk-luc human colorectal carcinoma-bearing animal model for the study of the inhibition effect and mechanism of orlistat, a fatty acid synthase (FASN) inhibitor. The results showed that orlistat caused cell cycle arrest at G1 phase, and triggered apoptosis through caspase-3 activation. The tumor inhibition effect of orlistat may also due to the inhibition of fatty acid synthesis without altering FASN activity. The tumor size of orlistat-treated mice in vivo was significantly smaller than that of the controls with 55% inhibition. The therapeutic efficacy was further confirmed with the bioluminescent imaging and nuclear molecular imaging with ¹³¹I-FIAU/gamma scintigraphy and ¹¹C-acetate/microPET. We suggest that FASN is a potential target for the treatment of human colorectal carcinoma.
Collapse
Affiliation(s)
- Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, No. 155, Sec. 2, Li-Nong St., Beitou, Taipei 112, Taiwan
| | | | | |
Collapse
|
100
|
Initial Results with 11C-Acetate Positron Emission Tomography/Computed Tomography (PET/CT) in the Staging of Urinary Bladder Cancer. Mol Imaging Biol 2011; 14:245-51. [DOI: 10.1007/s11307-011-0488-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|