1
|
Zhao Y, Bhosale AA, Zhang X. Multimodal surface coils for low field MR imaging. Magn Reson Imaging 2024; 112:107-115. [PMID: 38971265 DOI: 10.1016/j.mri.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Low field MRI is safer and more cost effective than the high field MRI. One of the inherent problems of low field MRI is its low signal-to-noise ratio or sensitivity. In this work, we introduce a multimodal surface coil technique for signal excitation and reception to improve the RF magnetic field (B1) efficiency and potentially improve MR sensitivity. The proposed multimodal surface coil consists of multiple identical resonators that are electromagnetically coupled to form a multimodal resonator. The field distribution of its lowest frequency mode is suitable for MR imaging applications. The prototype multimodal surface coils are built, and the performance is investigated and validated through numerical simulation, standard RF measurements and tests, and comparison with the conventional surface coil at low fields. Our results show that the B1 efficiency of the multimodal surface coil outperforms that of the conventional surface coil which is known to offer the highest B1 efficiency among all coil categories, i.e., volume coil, half-volume coil and surface coil. In addition, in low-field MRI, the required low-frequency coils often use large value capacitance to achieve the low resonant frequency which makes frequency tuning difficult. The proposed multimodal surface coil can be conveniently tuned to the required low frequency for low-field MRI with significantly reduced capacitance value, demonstrating excellent low-frequency operation capability over the conventional surface coil.
Collapse
Affiliation(s)
- Yunkun Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
| | - Aditya A Bhosale
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
| | - Xiaoliang Zhang
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States; Department of Electrical Engineering, State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
2
|
Zhao Y, Bhosale AA, Zhang X. Coupled stack-up volume RF coils for low-field open MR imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.30.24312851. [PMID: 39252906 PMCID: PMC11383509 DOI: 10.1101/2024.08.30.24312851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background Low-field open magnetic resonance imaging (MRI) systems, typically operating at magnetic field strengths below 1 Tesla, has greatly expanded the accessibility of MRI technology to meet a wide range of patient needs. However, the inherent challenges of low-field MRI, such as limited signal-to-noise ratios and limited availability of dedicated radiofrequency (RF) coils, have prompted the need for innovative coil designs that can improve imaging quality and diagnostic capabilities. Purpose In response to these challenges, we introduce the coupled stack-up volume coil, a novel RF coil design that addresses the shortcomings of conventional birdcage in the context of low-field open MRI. Methods The proposed coupled stack-up volume coil design utilizes a unique architecture that optimizes both transmit/receive efficiency and RF field homogeneity and offers the advantage of a simple design and construction, making it a practical and feasible solution for low-field MRI applications. This paper presents a comprehensive exploration of the theoretical framework, design considerations, and experimental validation of this innovative coil design. Results We demonstrate the superior performance of the coupled stack-up volume coil in achieving 47.7% higher transmit/receive efficiency and 68% more uniform magnetic field distribution compared to traditional birdcage coils in electromagnetic simulations. Bench tests results show that the B1 field efficiency of coupled stack-up volume coil is 57.3% higher compared with that of conventional birdcage coil. Conclusions The proposed coupled stack-up volume coil outperforms the conventional birdcage coil in terms of B1 efficiency, imaging coverage, and low-frequency operation capability. This design provides a robust and simple solution to low-field MR RF coil design.
Collapse
Affiliation(s)
- Yunkun Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
| | - Aditya A Bhosale
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
| | - Xiaoliang Zhang
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
- Department of Electrical Engineering, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
3
|
Payne K, Zhao Y, Bhosale AA, Zhang X. Dual-Tuned Coaxial-Transmission-Line RF Coils for Hyperpolarized 13C and Deuterium 2H Metabolic MRS Imaging at Ultrahigh Fields. IEEE Trans Biomed Eng 2024; 71:1521-1530. [PMID: 38090865 PMCID: PMC11095995 DOI: 10.1109/tbme.2023.3341760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
OBJECTIVE Information on the metabolism of tissues in healthy and diseased states plays a significant role in the detection and understanding of tumors, neurodegenerative diseases, diabetes, and other metabolic disorders. Hyperpolarized carbon-13 magnetic resonance imaging (13C-HPMRI) and deuterium metabolic imaging (2H-DMI) are two emerging X-nuclei used as practical imaging tools to investigate tissue metabolism. However due to their low gyromagnetic ratios (ɣ13C = 10.7 MHz/T; ɣ2H = 6.5 MHz/T) and natural abundance, such method required a sophisticated dual-tuned radiofrequency (RF) coil. METHODS Here, we report a dual-tuned coaxial transmission line (CTL) RF coil agile for metabolite information operating at 7T with independent tuning capability. The design analysis has demonstrated how both resonant frequencies can be individually controlled by simply varying the constituent of the design parameters. RESULTS Numerical results have demonstrated a broadband tuning range capability, covering most of the X-nucleus signal, especially the 13C and 2H spectra at 7T. Furthermore, in order to validate the feasibility of the proposed design, both dual-tuned 1H/13C and 1H/2H CTLs RF coils are fabricated using a semi-flexible RG-405 .086" coaxial cable and bench test results (scattering parameters and magnetic field efficiency/distribution) are successfully obtained. CONCLUSION The proposed dual-tuned RF coils reveal highly effective magnetic field obtained from both proton and heteronuclear signal which is crucial for accurate and detailed imaging. SIGNIFICANCE The successful development of this new dual-tuned RF coil technique would provide a tangible and efficient tool for ultrahigh field metabolic MR imaging.
Collapse
|
4
|
Zhao Y, Bhosale AA, Zhang X. Multimodal surface coils for low field MR imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.14.24305802. [PMID: 38699318 PMCID: PMC11065021 DOI: 10.1101/2024.04.14.24305802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Low field MRI is safer and more cost effective than the high field MRI. One of the inherent problems of low field MRI is its low signal-to-noise ratio or sensitivity. In this work, we introduce a multimodal surface coil technique for signal excitation and reception to improve the RF magnetic field (B 1 ) efficiency and potentially improve MR sensitivity. The proposed multimodal surface coil consists of multiple identical resonators that are electromagnetically coupled to form a multimodal resonator. The field distribution of its lowest frequency mode is suitable for MR imaging applications. The prototype multimodal surface coils are built, and the performance is investigated and validated through numerical simulation, standard RF measurements and tests, and comparison with the conventional surface coil at low fields. Our results show that the B 1 efficiency of the multimodal surface coil outperforms that of the conventional surface coil which is known to offer the highest B 1 efficiency among all coil categories, i.e., volume coil, half-volume coil and surface coil. In addition, in low-field MRI, the required low-frequency coils often use large value capacitance to achieve the low resonant frequency which makes frequency tuning difficult. The proposed multimodal surface coil can be conveniently tuned to the required low frequency for low-field MRI with significantly reduced capacitance value, demonstrating excellent low-frequency operation capability over the conventional surface coil.
Collapse
|
5
|
Payne K, Zhao Y, Bhosale AA, Zhang X. Dual-tuned Coaxial-transmission-line RF coils for Hyperpolarized 13C and Deuterium 2H Metabolic MRS Imaging at Ultrahigh Fields. ARXIV 2023:arXiv:2307.11221v3. [PMID: 37502626 PMCID: PMC10370217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Objective Information on the metabolism of tissues in healthy and diseased states plays a significant role in the detection and understanding of tumors, neurodegenerative diseases, diabetes, and other metabolic disorders. Hyperpolarized carbon-13 magnetic resonance imaging (13C-HPMRI) and deuterium metabolic imaging (2H-DMI) are two emerging X-nuclei used as practical imaging tools to investigate tissue metabolism. However due to their low gyromagnetic ratios (ɣ13C = 10.7 MHz/T; ɣ 2H = 6.5 MHz/T) and natural abundance, such method required a sophisticated dual-tuned radiofrequency (RF) coil. Methods Here, we report a dual-tuned coaxial transmission line (CTL) RF coil agile for metabolite information operating at 7T with independent tuning capability. The design analysis has demonstrated how both resonant frequencies can be individually controlled by simply varying the constituent of the design parameters. Results Numerical results have demonstrated a broadband tuning range capability, covering most of the X-nucleus signal, especially the 13C and 2H spectra at 7T. Furthermore, in order to validate the feasibility of the proposed design, both dual-tuned 1H/13C and 1H/2H CTLs RF coils are fabricated using a semi-flexible RG-405 .086" coaxial cable and bench test results (scattering parameters and magnetic field efficiency/distribution) are successfully obtained. Conclusion The proposed dual-tuned RF coils reveal highly effective magnetic field obtained from both proton and heteronuclear signal which is crucial for accurate and detailed imaging. Significance The successful development of this new dual-tuned RF coil technique would provide a tangible and efficient tool for ultrahigh field metabolic MR imaging.
Collapse
Affiliation(s)
- Komlan Payne
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Yunkun Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Aditya Ashok Bhosale
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Xiaoliang Zhang
- Departments of Biomedical Engineering and Electrical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| |
Collapse
|
6
|
Chetta P, Sriram R, Zadra G. Lactate as Key Metabolite in Prostate Cancer Progression: What Are the Clinical Implications? Cancers (Basel) 2023; 15:3473. [PMID: 37444583 PMCID: PMC10340474 DOI: 10.3390/cancers15133473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Advanced prostate cancer represents the fifth leading cause of cancer death in men worldwide. Although androgen-receptor signaling is the major driver of the disease, evidence is accumulating that disease progression is supported by substantial metabolic changes. Alterations in de novo lipogenesis and fatty acid catabolism are consistently reported during prostate cancer development and progression in association with androgen-receptor signaling. Therefore, the term "lipogenic phenotype" is frequently used to describe the complex metabolic rewiring that occurs in prostate cancer. However, a new scenario has emerged in which lactate may play a major role. Alterations in oncogenes/tumor suppressors, androgen signaling, hypoxic conditions, and cells in the tumor microenvironment can promote aerobic glycolysis in prostate cancer cells and the release of lactate in the tumor microenvironment, favoring immune evasion and metastasis. As prostate cancer is composed of metabolically heterogenous cells, glycolytic prostate cancer cells or cancer-associated fibroblasts can also secrete lactate and create "symbiotic" interactions with oxidative prostate cancer cells via lactate shuttling to sustain disease progression. Here, we discuss the multifaceted role of lactate in prostate cancer progression, taking into account the influence of the systemic metabolic and gut microbiota. We call special attention to the clinical opportunities of imaging lactate accumulation for patient stratification and targeting lactate metabolism.
Collapse
Affiliation(s)
- Paolo Chetta
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Giorgia Zadra
- Institute of Molecular Genetics, National Research Council (IGM-CNR), 27100 Pavia, Italy
| |
Collapse
|
7
|
Minami N, Hong D, Stevers N, Barger CJ, Radoul M, Hong C, Chen L, Kim Y, Batsios G, Gillespie AM, Pieper RO, Costello JF, Viswanath P, Ronen SM. Imaging biomarkers of TERT or GABPB1 silencing in TERT-positive glioblastoma. Neuro Oncol 2022; 24:1898-1910. [PMID: 35460557 PMCID: PMC9629440 DOI: 10.1093/neuonc/noac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND TERT promoter mutations are observed in 80% of wild-type IDH glioblastoma (GBM). Moreover, the upstream TERT transcription factor GABPB1 was recently identified as a cancer-specific therapeutic target for tumors harboring a TERT promoter mutation. In that context, noninvasive imaging biomarkers are needed for the detection of TERT modulation. METHODS Multiple GBM models were investigated as cells and in vivo tumors and the impact of TERT silencing, either directly or by targeting GABPB1, was determined using 1H and hyperpolarized 13C magnetic resonance spectroscopy (MRS). Changes in associated metabolic enzymes were also investigated. RESULTS 1H-MRS revealed that lactate and glutathione (GSH) were the most significantly altered metabolites when either TERT or GABPB1 was silenced, and lactate and GSH levels were correlated with cellular TERT expression. Consistent with the drop in lactate, 13C-MRS showed that hyperpolarized [1-13C]lactate production from [1-13C]pyruvate was also reduced when TERT was silenced. Mechanistically, the reduction in GSH was associated with a reduction in pentose phosphate pathway flux, reduced activity of glucose-6-phosphate dehydrogenase, and reduced NADPH. The drop in lactate and hyperpolarized lactate were associated with reductions in glycolytic flux, NADH, and expression/activity of GLUT1, monocarboxylate transporters, and lactate dehydrogenase A. CONCLUSIONS Our study indicates that MRS-detectable GSH, lactate, and lactate production could serve as metabolic biomarkers of response to emerging TERT-targeted therapies for GBM with activating TERT promoter mutations. Importantly these biomarkers are readily translatable to the clinic, and thus could ultimately improve GBM patient management.
Collapse
Affiliation(s)
- Noriaki Minami
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Donghyun Hong
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Nicholas Stevers
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Carter J Barger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Marina Radoul
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Lee Chen
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Yaewon Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Russel O Pieper
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| |
Collapse
|
8
|
Multinuclear MRI in Drug Discovery. Molecules 2022; 27:molecules27196493. [PMID: 36235031 PMCID: PMC9572840 DOI: 10.3390/molecules27196493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/17/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
The continuous development of magnetic resonance imaging broadens the range of applications to newer areas. Using MRI, we can not only visualize, but also track pharmaceutical substances and labeled cells in both in vivo and in vitro tests. 1H is widely used in the MRI method, which is determined by its high content in the human body. The potential of the MRI method makes it an excellent tool for imaging the morphology of the examined objects, and also enables registration of changes at the level of metabolism. There are several reports in the scientific publications on the use of clinical MRI for in vitro tracking. The use of multinuclear MRI has great potential for scientific research and clinical studies. Tuning MRI scanners to the Larmor frequency of a given nucleus, allows imaging without tissue background. Heavy nuclei are components of both drugs and contrast agents and molecular complexes. The implementation of hyperpolarization techniques allows for better MRI sensitivity. The aim of this review is to present the use of multinuclear MRI for investigations in drug delivery.
Collapse
|
9
|
Wei Y, Yang C, Jiang H, Li Q, Che F, Wan S, Yao S, Gao F, Zhang T, Wang J, Song B. Multi-nuclear magnetic resonance spectroscopy: state of the art and future directions. Insights Imaging 2022; 13:135. [PMID: 35976510 PMCID: PMC9382599 DOI: 10.1186/s13244-022-01262-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/04/2022] [Indexed: 12/16/2022] Open
Abstract
With the development of heteronuclear fluorine, sodium, phosphorus, and other probes and imaging technologies as well as the optimization of magnetic resonance imaging (MRI) equipment and sequences, multi-nuclear magnetic resonance (multi-NMR) has enabled localize molecular activities in vivo that are central to a variety of diseases, including cardiovascular disease, neurodegenerative pathologies, metabolic diseases, kidney, and tumor, to shift from the traditional morphological imaging to the molecular imaging, precision diagnosis, and treatment mode. However, due to the low natural abundance and low gyromagnetic ratios, the clinical application of multi-NMR has been hampered. Several techniques have been developed to amplify the NMR sensitivity such as the dynamic nuclear polarization, spin-exchange optical pumping, and brute-force polarization. Meanwhile, a wide range of nuclei can be hyperpolarized, such as 2H, 3He, 13C, 15 N, 31P, and 129Xe. The signal can be increased and allows real-time observation of biological perfusion, metabolite transport, and metabolic reactions in vivo, overcoming the disadvantages of conventional magnetic resonance of low sensitivity. HP-NMR imaging of different nuclear substrates provides a unique opportunity and invention to map the metabolic changes in various organs without invasive procedures. This review aims to focus on the recent applications of multi-NMR technology not only in a range of preliminary animal experiments but also in various disease spectrum in human. Furthermore, we will discuss the future challenges and opportunities of this multi-NMR from a clinical perspective, in the hope of truly bridging the gap between cutting-edge molecular biology and clinical applications.
Collapse
Affiliation(s)
- Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Caiwei Yang
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Hanyu Jiang
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Feng Che
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Shang Wan
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Shan Yao
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Feifei Gao
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Tong Zhang
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Jiazheng Wang
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, People's Republic of China. .,Department of Radiology, Sanya People's Hospital, Sanya, China.
| |
Collapse
|
10
|
Woitek R, McLean MA, Ursprung S, Rueda OM, Manzano Garcia R, Locke MJ, Beer L, Baxter G, Rundo L, Provenzano E, Kaggie J, Patterson A, Frary A, Field-Rayner J, Papalouka V, Kane J, Benjamin AJV, Gill AB, Priest AN, Lewis DY, Russell R, Grimmer A, White B, Latimer-Bowman B, Patterson I, Schiller A, Carmo B, Slough R, Lanz T, Wason J, Schulte RF, Chin SF, Graves MJ, Gilbert FJ, Abraham JE, Caldas C, Brindle KM, Sala E, Gallagher FA. Hyperpolarized Carbon-13 MRI for Early Response Assessment of Neoadjuvant Chemotherapy in Breast Cancer Patients. Cancer Res 2021; 81:6004-6017. [PMID: 34625424 PMCID: PMC7612070 DOI: 10.1158/0008-5472.can-21-1499] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/14/2021] [Accepted: 10/06/2021] [Indexed: 01/09/2023]
Abstract
Hyperpolarized 13C-MRI is an emerging tool for probing tissue metabolism by measuring 13C-label exchange between intravenously injected hyperpolarized [1-13C]pyruvate and endogenous tissue lactate. Here, we demonstrate that hyperpolarized 13C-MRI can be used to detect early response to neoadjuvant therapy in breast cancer. Seven patients underwent multiparametric 1H-MRI and hyperpolarized 13C-MRI before and 7-11 days after commencing treatment. An increase in the lactate-to-pyruvate ratio of approximately 20% identified three patients who, following 5-6 cycles of treatment, showed pathological complete response. This ratio correlated with gene expression of the pyruvate transporter MCT1 and lactate dehydrogenase A (LDHA), the enzyme catalyzing label exchange between pyruvate and lactate. Analysis of approximately 2,000 breast tumors showed that overexpression of LDHA and the hypoxia marker CAIX was associated with reduced relapse-free and overall survival. Hyperpolarized 13C-MRI represents a promising method for monitoring very early treatment response in breast cancer and has demonstrated prognostic potential. SIGNIFICANCE: Hyperpolarized carbon-13 MRI allows response assessment in patients with breast cancer after 7-11 days of neoadjuvant chemotherapy and outperformed state-of-the-art and research quantitative proton MRI techniques.
Collapse
Affiliation(s)
- Ramona Woitek
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Mary A McLean
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
| | - Stephan Ursprung
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Oscar M Rueda
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Raquel Manzano Garcia
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
| | - Matthew J Locke
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Lucian Beer
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gabrielle Baxter
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Leonardo Rundo
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Elena Provenzano
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Oncology, Cambridge Breast Cancer Research Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Joshua Kaggie
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Andrew Patterson
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Amy Frary
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Johanna Field-Rayner
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Vasiliki Papalouka
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Justine Kane
- Department of Oncology, Cambridge Breast Cancer Research Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Arnold J V Benjamin
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Andrew B Gill
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Andrew N Priest
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - David Y Lewis
- Molecular Imaging Laboratory Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Roslin Russell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
| | - Ashley Grimmer
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Brian White
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Beth Latimer-Bowman
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Ilse Patterson
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Amy Schiller
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Bruno Carmo
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Rhys Slough
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - James Wason
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | | | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Fiona J Gilbert
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jean E Abraham
- Department of Oncology, Cambridge Breast Cancer Research Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
- Department of Oncology, Cambridge Breast Cancer Research Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Evis Sala
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom.
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
11
|
Choi Y, Lee J, Lee H, Song JE, Kim D, Song H. Offset of apparent hyperpolarized 13 C lactate flux by the use of adjuvant metformin in ionizing radiation therapy in vivo. NMR IN BIOMEDICINE 2021; 34:e4561. [PMID: 34080736 PMCID: PMC8365667 DOI: 10.1002/nbm.4561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 05/13/2023]
Abstract
An increase in hyperpolarized (HP) [1-13 C]lactate production has been suggested as a biomarker for cancer occurrence as well as for response monitoring of cancer treatment. Recently, the use of metformin has been suggested as an anticancer or adjuvant treatment. By regulating the cytosolic NAD+ /NADH redox state, metformin stimulates lactate production and increases the HP [1-13 C]lactate conversion rate in the kidney, liver, and heart. In general, increased HP [1-13 C]lactate is regarded as a sign of cancer occurrence or tumor growth. Thus, the relationship between the tumor suppression effect of metformin and the change in metabolism monitored by HP [1-13 C]pyruvate MRS in cancer treatment needs to be investigated. The present study was performed using a brain metastasis animal model with MDA-MB-231(BR)-Luc breast cancer cells. HP [1-13 C]pyruvate MRS, T2 -weighted MRI, and bioluminescence imaging were performed in groups treated with metformin or adjuvant metformin and radiation therapy. Metformin treatment alone did not display a tumor suppression effect, and the HP [1-13 C]lactate conversion rate increased. In radiation therapy, the HP [1-13 C]lactate conversion rate decreased with tumor suppression, with a p-value of 0.028. In the adjuvant metformin and radiation treatment, the tumor suppression effect increased, with a p-value of 0.001. However, the apparent HP [1-13 C]lactate conversion rate (Kpl ) was observed to be offset by two opposite effects: a decrease on radiation therapy and an increase caused by metformin treatment. Although HP [1-13 C]pyruvate MRS could not evaluate the tumor suppression effect of adjuvant metformin and radiation therapy due to the offset phenomenon, metabolic changes following only metformin pre-treatment could be monitored. Therefore, our results indicate that the interpretation of HP [1-13 C]pyruvate MRS for response monitoring of cancer treatment should be carried out with caution when metformin is used as an adjuvant cancer therapy.
Collapse
Affiliation(s)
- Young‐Suk Choi
- Department of Radiology and Research Institute of Radiological ScienceYonsei University College of MedicineSeoulSouth Korea
| | - Joonsung Lee
- Biomedical Science InstituteYonsei University College of MedicineSeoulSouth Korea
- GE HealthcareSeoulSouth Korea
| | - Han‐Sol Lee
- Department of Electrical and Electronic EngineeringYonsei UniversitySeoulSouth Korea
| | - Jae Eun Song
- Department of Electrical and Electronic EngineeringYonsei UniversitySeoulSouth Korea
| | - Dong‐Hyun Kim
- Department of Electrical and Electronic EngineeringYonsei UniversitySeoulSouth Korea
| | - Ho‐Taek Song
- Department of Radiology and Research Institute of Radiological ScienceYonsei University College of MedicineSeoulSouth Korea
| |
Collapse
|
12
|
Nguyen NT, Bae EH, Do LN, Nguyen TA, Park I, Shin SS. In Vivo Assessment of Metabolic Abnormality in Alport Syndrome Using Hyperpolarized [1- 13C] Pyruvate MR Spectroscopic Imaging. Metabolites 2021; 11:metabo11040222. [PMID: 33917329 PMCID: PMC8067337 DOI: 10.3390/metabo11040222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 01/23/2023] Open
Abstract
Alport Syndrome (AS) is a genetic disorder characterized by impaired kidney function. The development of a noninvasive tool for early diagnosis and monitoring of renal function during disease progression is of clinical importance. Hyperpolarized 13C MRI is an emerging technique that enables non-invasive, real-time measurement of in vivo metabolism. This study aimed to investigate the feasibility of using this technique for assessing changes in renal metabolism in the mouse model of AS. Mice with AS demonstrated a significant reduction in the level of lactate from 4- to 7-week-old, while the levels of lactate were unchanged in the control mice over time. This reduction in lactate production in the AS group accompanied a significant increase of PEPCK expression levels, indicating that the disease progression in AS triggered the gluconeogenic pathway and might have resulted in a decreased lactate pool size and a subsequent reduction in pyruvate-to-lactate conversion. Additional metabolic imaging parameters, including the level of lactate and pyruvate, were found to be different between the AS and control groups. These preliminary results suggest that hyperpolarized 13C MRI might provide a potential noninvasive tool for the characterization of disease progression in AS.
Collapse
Affiliation(s)
- Nguyen-Trong Nguyen
- Department of Biomedical Science, Chonnam National University, Gwangju 61469, Korea;
| | - Eun-Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea;
| | - Luu-Ngoc Do
- Department of Radiology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (L.-N.D.); (T.-A.N.)
| | - Tien-Anh Nguyen
- Department of Radiology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (L.-N.D.); (T.-A.N.)
| | - Ilwoo Park
- Department of Radiology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (L.-N.D.); (T.-A.N.)
- Department of Artificial Intelligence Convergence, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (I.P.); (S.-S.S.); Tel.: +82-62-220-5744 (I.P.); +82-62-220-5882 (S.-S.S.)
| | - Sang-Soo Shin
- Department of Radiology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (L.-N.D.); (T.-A.N.)
- Correspondence: (I.P.); (S.-S.S.); Tel.: +82-62-220-5744 (I.P.); +82-62-220-5882 (S.-S.S.)
| |
Collapse
|
13
|
Lim H, Martínez-Santiesteban F, Jensen MD, Chen A, Wong E, Scholl TJ. Monitoring Early Changes in Tumor Metabolism in Response to Therapy Using Hyperpolarized 13C MRSI in a Preclinical Model of Glioma. ACTA ACUST UNITED AC 2020; 6:290-300. [PMID: 32879899 PMCID: PMC7442089 DOI: 10.18383/j.tom.2020.00024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study shows the use of hyperpolarized 13C magnetic resonance spectroscopic imaging (MRSI) to assess therapeutic efficacy in a preclinical tumor model. 13C-labeled pyruvate was used to monitor early changes in tumor metabolism based on the Warburg effect. High-grade malignant tumors exhibit increased glycolytic activity and lactate production to promote proliferation. A rodent glioma model was used to explore altered lactate production after therapy as an early imaging biomarker for therapeutic response. Rodents were surgically implanted with C6 glioma cells and separated into 4 groups, namely, no therapy, radiotherapy, chemotherapy and combined therapy. Animals were imaged serially at 6 different time points with magnetic resonance imaging at 3 T using hyperpolarized [1-13C]pyruvate MRSI and conventional 1H imaging. Using hyperpolarized [1-13C]pyruvate MRSI, alterations in tumor metabolism were detected as changes in the conversion of lactate to pyruvate (measured as Lac/Pyr ratio) and compared with the conventional method of detecting therapeutic response using the Response Evaluation Criteria in Solid Tumors. Moreover, each therapy group expressed different characteristic changes in tumor metabolism. The group that received no therapy showed a gradual increase of Lac/Pyr ratio within the tumor. The radiotherapy group showed large variations in tumor Lac/Pyr ratio. The chemo- and combined-therapy groups showed a statistically significant reduction in tumor Lac/Pyr ratio; however, only combined therapy was capable of suppressing tumor growth, which resulted in low endpoint mortality rate. Hyperpolarized 13C MRSI detected a prompt reduction in Lac/Pyr ratio as early as 2 days post combined chemo- and radiotherapies.
Collapse
Affiliation(s)
- Heeseung Lim
- Department of Medical Biophysics, Western University, London, ON, Canada
| | | | - Michael D Jensen
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Albert Chen
- General Electric Healthcare, Toronto, ON, Canada
| | - Eugene Wong
- Department of Medical Biophysics, Western University, London, ON, Canada.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and
| | - Timothy J Scholl
- Department of Medical Biophysics, Western University, London, ON, Canada.,Departments of Physics and Astronomy; Oncology; and Robarts Research Institute, Western University, London, ON, Canada, and.,Ontario Institute for Cancer Research, Toronto, ON, Canada
| |
Collapse
|
14
|
Topping GJ, Hundshammer C, Nagel L, Grashei M, Aigner M, Skinner JG, Schulte RF, Schilling F. Acquisition strategies for spatially resolved magnetic resonance detection of hyperpolarized nuclei. MAGMA (NEW YORK, N.Y.) 2020; 33:221-256. [PMID: 31811491 PMCID: PMC7109201 DOI: 10.1007/s10334-019-00807-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/08/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Hyperpolarization is an emerging method in magnetic resonance imaging that allows nuclear spin polarization of gases or liquids to be temporarily enhanced by up to five or six orders of magnitude at clinically relevant field strengths and administered at high concentration to a subject at the time of measurement. This transient gain in signal has enabled the non-invasive detection and imaging of gas ventilation and diffusion in the lungs, perfusion in blood vessels and tissues, and metabolic conversion in cells, animals, and patients. The rapid development of this method is based on advances in polarizer technology, the availability of suitable probe isotopes and molecules, improved MRI hardware and pulse sequence development. Acquisition strategies for hyperpolarized nuclei are not yet standardized and are set up individually at most sites depending on the specific requirements of the probe, the object of interest, and the MRI hardware. This review provides a detailed introduction to spatially resolved detection of hyperpolarized nuclei and summarizes novel and previously established acquisition strategies for different key areas of application.
Collapse
Affiliation(s)
- Geoffrey J Topping
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian Hundshammer
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Luca Nagel
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martin Grashei
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maximilian Aigner
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jason G Skinner
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Franz Schilling
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
| |
Collapse
|
15
|
Ntziachristos V, Pleitez MA, Aime S, Brindle KM. Emerging Technologies to Image Tissue Metabolism. Cell Metab 2019; 29:518-538. [PMID: 30269982 DOI: 10.1016/j.cmet.2018.09.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/24/2018] [Accepted: 09/02/2018] [Indexed: 12/19/2022]
Abstract
Due to the implication of altered metabolism in a large spectrum of tissue function and disease, assessment of metabolic processes becomes essential in managing health. In this regard, imaging can play a critical role in allowing observation of biochemical and physiological processes. Nuclear imaging methods, in particular positron emission tomography, have been widely employed for imaging metabolism but are mainly limited by the use of ionizing radiation and the sensing of only one parameter at each scanning session. Observations in healthy individuals or longitudinal studies of disease could markedly benefit from non-ionizing, multi-parameter imaging methods. We therefore focus this review on progress with the non-ionizing radiation methods of MRI, hyperpolarized magnetic resonance and magnetic resonance spectroscopy, chemical exchange saturation transfer, and emerging optoacoustic (photoacoustic) imaging. We also briefly discuss the role of nuclear and optical imaging methods for research and clinical protocols.
Collapse
Affiliation(s)
- Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg 85764, Germany; Chair of Biological Imaging, TranslaTUM, Technical University of Munich, Ismaningerstr. 22, Munich 81675, Germany.
| | - Miguel A Pleitez
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg 85764, Germany; Chair of Biological Imaging, TranslaTUM, Technical University of Munich, Ismaningerstr. 22, Munich 81675, Germany
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin 10126, Italy
| | - Kevin M Brindle
- Department of Biochemistry, University of Cambridge, Old Addenbrooke's Site, Cambridge CB2 1GA, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
16
|
Lin C, Salzillo TC, Bader DA, Wilkenfeld SR, Awad D, Pulliam TL, Dutta P, Pudakalakatti S, Titus M, McGuire SE, Bhattacharya PK, Frigo DE. Prostate Cancer Energetics and Biosynthesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:185-237. [PMID: 31900911 PMCID: PMC8096614 DOI: 10.1007/978-3-030-32656-2_10] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers must alter their metabolism to satisfy the increased demand for energy and to produce building blocks that are required to create a rapidly growing tumor. Further, for cancer cells to thrive, they must also adapt to an often changing tumor microenvironment, which can present new metabolic challenges (ex. hypoxia) that are unfavorable for most other cells. As such, altered metabolism is now considered an emerging hallmark of cancer. Like many other malignancies, the metabolism of prostate cancer is considerably different compared to matched benign tissue. However, prostate cancers exhibit distinct metabolic characteristics that set them apart from many other tumor types. In this chapter, we will describe the known alterations in prostate cancer metabolism that occur during initial tumorigenesis and throughout disease progression. In addition, we will highlight upstream regulators that control these metabolic changes. Finally, we will discuss how this new knowledge is being leveraged to improve patient care through the development of novel biomarkers and metabolically targeted therapies.
Collapse
Affiliation(s)
- Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis C Salzillo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - David A Bader
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Molecular Medicine Program, The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
17
|
Razza EM, Pedersen HS, Stroebech L, Fontes PK, Kadarmideen HN, Callesen H, Pihl M, Nogueira MFG, Hyttel P. Simulated physiological oocyte maturation has side effects on bovine oocytes and embryos. J Assist Reprod Genet 2018; 36:413-424. [PMID: 30443692 DOI: 10.1007/s10815-018-1365-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/02/2018] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Oocyte maturation is a complex process involving nuclear and cytoplasmic modulations, during which oocytes acquire their ability to become fertilized and support embryonic development. The oocyte is apparently "primed" for maturation during its development in the dominant follicle. As bovine oocytes immediately resume meiosis when cultured, it was hypothesized that delaying resumption of meiosis with cyclic nucleotide modulators before in vitro maturation (IVM) would allow the oocytes to acquire improved developmental competence. METHODS We tested the Simulated Physiological Oocyte Maturation (SPOM) system that uses forskolin and 3-isobutyl-1-methylxanthine for 2 h prior to IVM against two different systems of conventional IVM (Con-IVM). We evaluated the ultrastructure of matured oocytes and blastocysts and also assessed the expression of 96 genes related to embryo quality in the blastocysts. RESULTS In summary, the SPOM system resulted in lower blastocyst rates than both Con-IVM systems (30 ± 9.1 vs. 35 ± 8.7; 29 ± 2.6 vs. 38 ± 2.8). Mature SPOM oocytes had significantly increased volume and number of vesicles, reduced volume and surface density of large smooth endoplasmic reticulum clusters, and lower number of mitochondria than Con-IVM oocytes. SPOM blastocysts showed only subtle differences with parallel undulations of adjacent trophectoderm plasma membranes and peripherally localized ribosomes in cells of the inner cell mass compared with Con-IVM blastocysts. SPOM blastocysts, however, displayed significant downregulation of genes related to embryonic developmental potential when compared to Con-IVM blastocysts. CONCLUSIONS Our results show that the use of the current version of the SPOM system may have adverse effects on oocytes and blastocysts calling for optimized protocols for improving oocyte competence.
Collapse
Affiliation(s)
- Eduardo M Razza
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), Distrito de Rubião Junior s/n, Botucatu, São Paulo, 18618970, Brazil.
| | - Hanne S Pedersen
- Department of Animal Science, Aarhus University, DK-8830, Tjele, Denmark
| | - Lotte Stroebech
- EmbryoTrans Biotech, Frederiksberg C, DK-1851, Copenhagen, Denmark
| | - Patricia K Fontes
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), Distrito de Rubião Junior s/n, Botucatu, São Paulo, 18618970, Brazil
| | - Haja N Kadarmideen
- Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet, 2800, Kgs. Lyngby, Denmark
| | - Henrik Callesen
- Department of Animal Science, Aarhus University, DK-8830, Tjele, Denmark
| | - Maria Pihl
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Marcelo F G Nogueira
- Department of Pharmacology, Institute of Bioscience, São Paulo State University (UNESP), Distrito de Rubião Junior s/n, Botucatu, São Paulo, 18618970, Brazil.,Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Avenida Dom Antonio, 2100, Assis, São Paulo, 19806900, Brazil
| | - Poul Hyttel
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
18
|
Taglang C, Korenchan DE, von Morze C, Yu J, Najac C, Wang S, Blecha JE, Subramaniam S, Bok R, VanBrocklin HF, Vigneron DB, Ronen SM, Sriram R, Kurhanewicz J, Wilson DM, Flavell RR. Late-stage deuteration of 13C-enriched substrates for T 1 prolongation in hyperpolarized 13C MRI. Chem Commun (Camb) 2018; 54:5233-5236. [PMID: 29726563 PMCID: PMC6054790 DOI: 10.1039/c8cc02246a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A robust and selective late-stage deuteration methodology was applied to 13C-enriched amino and alpha hydroxy acids to increase spin-lattice relaxation constant T1 for hyperpolarized 13C magnetic resonance imaging. For the five substrates with 13C-labeling on the C1-position ([1-13C]alanine, [1-13C]serine, [1-13C]lactate, [1-13C]glycine, and [1-13C]valine), significant increase of their T1 was observed at 3 T with deuterium labeling (+26%, 22%, +16%, +25% and +29%, respectively). Remarkably, in the case of [2-13C]alanine, [2-13C]serine and [2-13C]lactate, deuterium labeling led to a greater than four fold increase in T1. [1-13C,2-2H]alanine, produced using this method, was applied to in vitro enzyme assays with alanine aminotransferase, demonstrating a kinetic isotope effect.
Collapse
Affiliation(s)
- Céline Taglang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - David E. Korenchan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Cornelius von Morze
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Justin Yu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Chloé Najac
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Joseph E. Blecha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Sukumar Subramaniam
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Daniel B. Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Sabrina M. Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA.
| |
Collapse
|
19
|
MR Molecular Imaging of Brain Cancer Metabolism Using Hyperpolarized 13C Magnetic Resonance Spectroscopy. Top Magn Reson Imaging 2017; 25:187-196. [PMID: 27748711 DOI: 10.1097/rmr.0000000000000104] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolic reprogramming is an important hallmark of cancer. Alterations in many metabolic pathways support the requirement for cellular building blocks that are essential for cancer cell proliferation. This metabolic reprogramming can be imaged using magnetic resonance spectroscopy (MRS). H MRS can inform on alterations in the steady-state levels of cellular metabolites, but the emergence of hyperpolarized C MRS has now also enabled imaging of metabolic fluxes in real-time, providing a new method for tumor detection and monitoring of therapeutic response. In the case of glioma, preclinical cell and animal studies have shown that the hyperpolarized C MRS metabolic imaging signature is specific to tumor type and can distinguish between mutant IDH1 glioma and primary glioblastoma. Here, we review these findings, first describing the main metabolic pathways that are altered in the different glioma subtypes, and then reporting on the use of hyperpolarized C MRS and MR spectroscopic imaging (MRSI) to probe these pathways. We show that the future translation of this hyperpolarized C MRS molecular metabolic imaging method to the clinic promises to improve the noninvasive detection, characterization, and response-monitoring of brain tumors resulting in improved patient diagnosis and clinical management.
Collapse
|
20
|
Serrao EM, Brindle KM. Dynamic nuclear polarisation: The future of imaging in oncology? Porto Biomed J 2017; 2:71-75. [PMID: 32258590 PMCID: PMC6806983 DOI: 10.1016/j.pbj.2017.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/02/2017] [Indexed: 12/19/2022] Open
Abstract
As clinical oncology evolves with new treatment options becoming available, there is an increasing demand on anatomic imaging for the assessment of patients at different stages. Imaging with hyperpolarized 13C-labelled cell substrates has the potential to become a powerful tool in many steps of clinical evaluation, offering a new metabolic metric and therefore a more personalised approach to treatment response. This articles explores the metabolic basis and potential for translation of hyperpolarised pyruvate as a dynamic nuclear polarisation probe in clinical oncology.
Collapse
Affiliation(s)
- Eva M Serrao
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Miyahira AK, Roychowdhury S, Goswami S, Ippolito JE, Priceman SJ, Pritchard CC, Sfanos KS, Subudhi SK, Simons JW, Pienta KJ, Soule HR. Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer; Report From the 2016 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2017; 77:123-144. [PMID: 27679977 DOI: 10.1002/pros.23260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The 2016 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer," was held from June 23 to June 26, 2016, in Coronado, California. METHODS For the 4th year in a row, the Prostate Cancer Foundation (PCF) hosted the CHPCA Meeting, a think tank-structured scientific conference, which focuses on a specific topic of critical unmet need on the biology and treatment of advanced prostate cancer. The 2016 CHPCA Meeting was attended by 71 investigators from prostate cancer and other fields, who discussed the biology, study methodologies, treatment strategies, and critical unmet needs concerning metastatic prostate cancer, with the ultimate goal of advancing strategies to treat and eliminate this disease. RESULTS The major topics of discussion included: the molecular landscape and molecular heterogeneity of metastatic prostate cancer, the role of the metastatic microenvironment, optimizing immunotherapy in metastatic prostate cancer, learning from exceptional responders and non-responders, targeting DNA repair deficiency in advanced prostate cancer, developing and applying novel biomarkers and imaging techniques, and potential roles for the microbiome in prostate cancer. DISCUSSION This article reviews the topics presented and discussions held at the CHPCA Meeting, with a focus on the unknowns and next steps needed to advance our understanding of the biology and most effective treatment strategies for metastatic prostate cancer. Prostate 77:123-144, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Sameek Roychowdhury
- Comprehensive Cancer Center, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Division of Medical Oncology, Ohio State University, Cincinnati, Ohio
| | - Sangeeta Goswami
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Joseph E Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Saul J Priceman
- Departments of Hematology and Hematopoietic Cell Transplantation, and Immuno-Oncology, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Kenneth J Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
22
|
Kubala E, Muñoz-Álvarez KA, Topping G, Hundshammer C, Feuerecker B, Gómez PA, Pariani G, Schilling F, Glaser SJ, Schulte RF, Menzel MI, Schwaiger M. Hyperpolarized 13C Metabolic Magnetic Resonance Spectroscopy and Imaging. J Vis Exp 2016:54751. [PMID: 28060330 PMCID: PMC5226623 DOI: 10.3791/54751] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
In the past decades, new methods for tumor staging, restaging, treatment response monitoring, and recurrence detection of a variety of cancers have emerged in conjunction with the state-of-the-art positron emission tomography with 18F-fluorodeoxyglucose ([18F]-FDG PET). 13C magnetic resonance spectroscopic imaging (13CMRSI) is a minimally invasive imaging method that enables the monitoring of metabolism in vivo and in real time. As with any other method based on 13C nuclear magnetic resonance (NMR), it faces the challenge of low thermal polarization and a subsequent low signal-to-noise ratio due to the relatively low gyromagnetic ratio of 13C and its low natural abundance in biological samples. By overcoming these limitations, dynamic nuclear polarization (DNP) with subsequent sample dissolution has recently enabled commonly used NMR and magnetic resonance imaging (MRI) systems to measure, study, and image key metabolic pathways in various biological systems. A particularly interesting and promising molecule used in 13CMRSI is [1-13C]pyruvate, which, in the last ten years, has been widely used for in vitro, preclinical, and, more recently, clinical studies to investigate the cellular energy metabolism in cancer and other diseases. In this article, we outline the technique of dissolution DNP using a 3.35 T preclinical DNP hyperpolarizer and demonstrate its usage in in vitro studies. A similar protocol for hyperpolarization may be applied for the most part in in vivo studies as well. To do so, we used lactate dehydrogenase (LDH) and catalyzed the metabolic reaction of [1-13C]pyruvate to [1-13C]lactate in a prostate carcinoma cell line, PC3, in vitro using 13CMRSI.
Collapse
Affiliation(s)
- Eugen Kubala
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München; Department of Chemistry, Technische Universität München; GE Global Research;
| | - Kim A Muñoz-Álvarez
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München
| | - Geoffrey Topping
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München
| | - Christian Hundshammer
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München; Department of Chemistry, Technische Universität München
| | - Benedikt Feuerecker
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München
| | - Pedro A Gómez
- GE Global Research; Zentralinstitut für Medizintechnik der Technischen Universität München (IMETUM), Technische Universität München
| | - Giorgio Pariani
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München; Institute for Biological and Medical Imaging (IBMI), Helmholtz Zentrum München; IDG Institute of Developmental Genetics, Helmholtz Zentrum München
| | - Franz Schilling
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München
| | | | | | | | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München
| |
Collapse
|
23
|
Reed GD, von Morze C, Verkman AS, Koelsch BL, Chaumeil MM, Lustig M, Ronen SM, Bok RA, Sands JM, Larson PEZ, Wang ZJ, Larsen JHA, Kurhanewicz J, Vigneron DB. Imaging Renal Urea Handling in Rats at Millimeter Resolution using Hyperpolarized Magnetic Resonance Relaxometry. Tomography 2016; 2:125-135. [PMID: 27570835 PMCID: PMC4996281 DOI: 10.18383/j.tom.2016.00127] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In vivo spin spin relaxation time (T2) heterogeneity of hyperpolarized [13C,15N2]urea in the rat kidney was investigated. Selective quenching of the vascular hyperpolarized 13C signal with a macromolecular relaxation agent revealed that a long-T2 component of the [13C,15N2]urea signal originated from the renal extravascular space, thus allowing the vascular and renal filtrate contrast agent pools of the [13C,15N2]urea to be distinguished via multi-exponential analysis. The T2 response to induced diuresis and antidiuresis was performed with two imaging agents: hyperpolarized [13C,15N2]urea and a control agent hyperpolarized bis-1,1-(hydroxymethyl)-1-13C-cyclopropane-2H8. Large T2 increases in the inner-medullar and papilla were observed with the former agent and not the latter during antidiuresis. Therefore, [13C,15N2]urea relaxometry is sensitive to two steps of the renal urea handling process: glomerular filtration and the inner-medullary urea transporter (UT)-A1 and UT-A3 mediated urea concentrating process. Simple motion correction and subspace denoising algorithms are presented to aid in the multi exponential data analysis. Furthermore, a T2-edited, ultra long echo time sequence was developed for sub-2 mm3 resolution 3D encoding of urea by exploiting relaxation differences in the vascular and filtrate pools.
Collapse
Affiliation(s)
- Galen D Reed
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Cornelius von Morze
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, California, USA
| | - Bertram L Koelsch
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Myriam M Chaumeil
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Michael Lustig
- Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA; Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, California, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Robert A Bok
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jan Henrik Ardenkjær Larsen
- GE Healthcare, Brøndby, Denmark; Department of Electrical Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
24
|
Reed GD, von Morze C, Verkman AS, Koelsch BL, Chaumeil MM, Lustig M, Ronen SM, Bok RA, Sands JM, Larson PEZ, Wang ZJ, Larsen JHA, Kurhanewicz J, Vigneron DB. Imaging Renal Urea Handling in Rats at Millimeter Resolution using Hyperpolarized Magnetic Resonance Relaxometry. Tomography 2016. [PMID: 27570835 DOI: 10.18383/j.tom2016.00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2023] Open
Abstract
In vivo spin spin relaxation time (T2) heterogeneity of hyperpolarized [13C,15N2]urea in the rat kidney was investigated. Selective quenching of the vascular hyperpolarized 13C signal with a macromolecular relaxation agent revealed that a long-T2 component of the [13C,15N2]urea signal originated from the renal extravascular space, thus allowing the vascular and renal filtrate contrast agent pools of the [13C,15N2]urea to be distinguished via multi-exponential analysis. The T2 response to induced diuresis and antidiuresis was performed with two imaging agents: hyperpolarized [13C,15N2]urea and a control agent hyperpolarized bis-1,1-(hydroxymethyl)-1-13C-cyclopropane-2H8. Large T2 increases in the inner-medullar and papilla were observed with the former agent and not the latter during antidiuresis. Therefore, [13C,15N2]urea relaxometry is sensitive to two steps of the renal urea handling process: glomerular filtration and the inner-medullary urea transporter (UT)-A1 and UT-A3 mediated urea concentrating process. Simple motion correction and subspace denoising algorithms are presented to aid in the multi exponential data analysis. Furthermore, a T2-edited, ultra long echo time sequence was developed for sub-2 mm3 resolution 3D encoding of urea by exploiting relaxation differences in the vascular and filtrate pools.
Collapse
Affiliation(s)
- Galen D Reed
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Cornelius von Morze
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, California, USA
| | - Bertram L Koelsch
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Myriam M Chaumeil
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Michael Lustig
- Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA; Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, California, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Robert A Bok
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jan Henrik Ardenkjær Larsen
- GE Healthcare, Brøndby, Denmark; Department of Electrical Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA; Graduate Group in Bioengineering University of California San Francisco, San Francisco, California, USA, and University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
25
|
Radoul M, Chaumeil MM, Eriksson P, Wang AS, Phillips JJ, Ronen SM. MR Studies of Glioblastoma Models Treated with Dual PI3K/mTOR Inhibitor and Temozolomide:Metabolic Changes Are Associated with Enhanced Survival. Mol Cancer Ther 2016; 15:1113-22. [PMID: 26883274 PMCID: PMC4873419 DOI: 10.1158/1535-7163.mct-15-0769] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/02/2016] [Indexed: 12/29/2022]
Abstract
The current standard of care for glioblastoma (GBM) is surgical resection, radiotherapy, and treatment with temozolomide (TMZ). However, resistance to current therapies and recurrence are common. To improve survival, agents that target the PI3K signaling pathway, which is activated in approximately 88% of GBM, are currently in clinical trials. A challenge with such therapies is that tumor shrinkage is not always observed. New imaging methods are therefore needed to monitor response to therapy and predict survival. The goal of this study was to determine whether hyperpolarized (13)C magnetic resonance spectroscopic imaging (MRSI) and (1)H magnetic resonance spectroscopy (MRS) can be used to monitor response to the second-generation dual PI3K/mTOR inhibitor voxtalisib (XL765, SAR245409), alone or in combination with TMZ. We investigated GS-2 and U87-MG GBM orthotopic tumors in mice, and used MRI, hyperpolarized (13)C MRSI, and (1)H MRS to monitor the effects of treatment. In our study, (1)H MRS could not predict tumor response to therapy. However, in both our models, we observed a significantly lower hyperpolarized lactate-to-pyruvate ratio in animals treated with voxtalisib, TMZ, or combination therapy, when compared with controls. This metabolic alteration was observed prior to MRI-detectable changes in tumor size, was consistent with drug action, and was associated with enhanced animal survival. Our findings confirm the potential translational value of the hyperpolarized lactate-to-pyruvate ratio as a biomarker for noninvasively assessing the effects of emerging therapies for patients with GBM. Mol Cancer Ther; 15(5); 1113-22. ©2016 AACR.
Collapse
Affiliation(s)
- Marina Radoul
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Myriam M Chaumeil
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Pia Eriksson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Alan S Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Joanna J Phillips
- Brain Tumor Research Center, University of California San Francisco, San Francisco, California. Neuropathology Division, Department of Pathology, UCSF School of Medicine, UCSF Medical Center, San Francisco, California
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California. Brain Tumor Research Center, University of California San Francisco, San Francisco, California.
| |
Collapse
|
26
|
Serrao EM, Brindle KM. Potential Clinical Roles for Metabolic Imaging with Hyperpolarized [1-(13)C]Pyruvate. Front Oncol 2016; 6:59. [PMID: 27014634 PMCID: PMC4786548 DOI: 10.3389/fonc.2016.00059] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/28/2016] [Indexed: 01/06/2023] Open
Affiliation(s)
- Eva M. Serrao
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Kevin M. Brindle
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
Hosseini SM, Dufort I, Caballero J, Moulavi F, Ghanaei HR, Sirard MA. Transcriptome profiling of bovine inner cell mass and trophectoderm derived from in vivo generated blastocysts. BMC DEVELOPMENTAL BIOLOGY 2015; 15:49. [PMID: 26681441 PMCID: PMC4683974 DOI: 10.1186/s12861-015-0096-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/22/2015] [Indexed: 12/24/2022]
Abstract
Background This study describes the generation and analysis of the transcriptional profile of bovine inner cell mass (ICM) and trophectoderm (TE), obtained from in vivo developed embryos by using a bovine-embryo specific array (EmbryoGENE) containing 37,238 probes. Results A total of 4,689 probes were differentially expressed between ICM and TE, among these, 2,380 and 2,309 probes were upregulated in ICM and TE tissues, respectively (P ≤ 0.01, FC ≥ 2.0, FDR: 2.0). Ontological classification of the genes predominantly expressed in ICM emerged a range of functional categories with a preponderance of genes involved in basal and developmental signaling pathways including P53, TGFβ, IL8, mTOR, integrin, ILK, and ELF2 signalings. Cross-referencing of microarray data with two available in vitro studies indicated a marked reduction in ICM vs. TE transcriptional difference following in vitro culture of bovine embryos. Moreover, a great majority of genes that were found to be misregulated following in vitro culture of bovine embryos were known genes involved in epigenetic regulation of pluripotency and cell differentiation including DNMT1, GADD45, CARM1, ELF5 HDAC8, CCNB1, KDM6A, PRDM9, CDX2, ARID3A, IL6, GADD45A, FGFR2, PPP2R2B, and SMARCA2. Cross-species referencing of microarray data revealed substantial divergence between bovine and mouse and human in signaling pathways involved in early lineage specification. Conclusions The transcriptional changes occur during ICM and TE lineages specification in bovine is greater than previously understood. Therefore, this array data establishes a standard to evaluate the in vitro imprint on the transcriptome and to hypothesize the cross-species differences that allow in vitro acquisition of pluripotent ICM in human and mice but hinder that process in bovine. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0096-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- S M Hosseini
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran. .,Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, QC, G1V 0A6, Canada.
| | - I Dufort
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, QC, G1V 0A6, Canada.
| | - J Caballero
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, QC, G1V 0A6, Canada.
| | - F Moulavi
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - H R Ghanaei
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - M A Sirard
- Centre de Recherche en Biologie de la Reproduction, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
28
|
Gutte H, Hansen AE, Johannesen HH, Clemmensen AE, Ardenkjær-Larsen JH, Nielsen CH, Kjær A. The use of dynamic nuclear polarization (13)C-pyruvate MRS in cancer. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2015; 5:548-60. [PMID: 26550544 PMCID: PMC4620180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/31/2015] [Indexed: 06/05/2023]
Abstract
In recent years there has been an immense development of new targeted anti-cancer drugs. For practicing precision medicine, a sensitive method imaging for non-invasive, assessment of early treatment response and for assisting in developing new drugs is warranted. Magnetic Resonance Spectroscopy (MRS) is a potent technique for non-invasive in vivo investigation of tissue chemistry and cellular metabolism. Hyperpolarization by Dynamic Nuclear Polarization (DNP) is capable of creating solutions of molecules with polarized nuclear spins in a range of biological molecules and has enabled the real-time investigation of in vivo metabolism. The development of this new method has been demonstrated to enhance the nuclear polarization more than 10,000-fold, thereby significantly increasing the sensitivity of the MRS with a spatial resolution to the millimeters and a temporal resolution at the subsecond range. Furthermore, the method enables measuring kinetics of conversion of substrates into cell metabolites and can be integrated with anatomical proton magnetic resonance imaging (MRI). Many nuclei and substrates have been hyperpolarized using the DNP method. Currently, the most widely used compound is (13)C-pyruvate due to favoring technicalities. Intravenous injection of the hyperpolarized (13)C-pyruvate results in appearance of (13)C-lactate, (13)C-alanine and (13)C-bicarbonate resonance peaks depending on the tissue, disease and the metabolic state probed. In cancer, the lactate level is increased due to increased glycolysis. The use of DNP enhanced (13)C-pyruvate has in preclinical studies shown to be a sensitive method for detecting cancer and for assessment of early treatment response in a variety of cancers. Recently, a first-in-man 31-patient study was conducted with the primary objective to assess the safety of hyperpolarized (13)C-pyruvate in healthy subjects and prostate cancer patients. The study showed an elevated (13)C-lactate/(13)C-pyruvate ratio in regions of biopsy-proven prostate cancer compared to noncancerous tissue. However, more studies are needed in order to establish use of hyperpolarized (13)C MRS imaging of cancer.
Collapse
Affiliation(s)
- Henrik Gutte
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| | - Adam Espe Hansen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| | - Helle Hjorth Johannesen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| | - Andreas Ettrup Clemmensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| | - Jan Henrik Ardenkjær-Larsen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre Denmark ; Department of Electrical Engineering, Technical University of Denmark Kgs Lyngby, Denmark ; GE Healthcare, Brøndby Denmark
| | - Carsten Haagen Nielsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen Denmark
| |
Collapse
|
29
|
Integration of imaging into clinical practice to assess the delivery and performance of macromolecular and nanotechnology-based oncology therapies. J Control Release 2015; 219:295-312. [PMID: 26403800 DOI: 10.1016/j.jconrel.2015.09.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/19/2015] [Accepted: 09/19/2015] [Indexed: 01/02/2023]
Abstract
Functional and molecular imaging has become increasingly used to evaluate interpatient and intrapatient tumor heterogeneity. Imaging allows for assessment of microenvironment parameters including tumor hypoxia, perfusion and proliferation, as well as tumor metabolism and the intratumoral distribution of specific molecular markers. Imaging information may be used to stratify patients for targeted therapies, and to define patient populations that may benefit from alternative therapeutic approaches. It also provides a method for non-invasive monitoring of treatment response at earlier time-points than traditional cues, such as tumor shrinkage. Further, companion diagnostic imaging techniques are becoming progressively more important for development and clinical implementation of targeted therapies. Imaging-based companion diagnostics are likely to be essential for the validation and FDA approval of targeted nanotherapies and macromolecular medicines. This review describes recent clinical advances in the use of functional and molecular imaging to evaluate the tumor microenvironment. Additionally, this article focuses on image-based assessment of distribution and anti-tumor effect of nano- and macromolecular systems.
Collapse
|
30
|
Chaumeil MM, Najac C, Ronen SM. Studies of Metabolism Using (13)C MRS of Hyperpolarized Probes. Methods Enzymol 2015; 561:1-71. [PMID: 26358901 DOI: 10.1016/bs.mie.2015.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
First described in 2003, the dissolution dynamic nuclear polarization (DNP) technique, combined with (13)C magnetic resonance spectroscopy (MRS), has since been used in numerous metabolic studies and has become a valuable metabolic imaging method. DNP dramatically increases the level of polarization of (13)C-labeled compounds resulting in an increase in the signal-to-noise ratio (SNR) of over 50,000 fold for the MRS spectrum of hyperpolarized compounds. The high SNR enables rapid real-time detection of metabolism in cells, tissues, and in vivo. This chapter will present a comprehensive review of the DNP approaches that have been used to monitor metabolism in living systems. First, the list of (13)C DNP probes developed to date will be presented, with a particular focus on the most commonly used probe, namely [1-(13)C] pyruvate. In the next four sections, we will then describe the different factors that need to be considered when designing (13)C DNP probes for metabolic studies, conducting in vitro or in vivo hyperpolarized experiments, as well as acquiring, analyzing, and modeling hyperpolarized (13)C data.
Collapse
Affiliation(s)
- Myriam M Chaumeil
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Chloé Najac
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA.
| |
Collapse
|
31
|
Saito K, Matsumoto S, Takakusagi Y, Matsuo M, Morris HD, Lizak MJ, Munasinghe JP, Devasahayam N, Subramanian S, Mitchell JB, Krishna MC. 13C-MR Spectroscopic Imaging with Hyperpolarized [1-13C]pyruvate Detects Early Response to Radiotherapy in SCC Tumors and HT-29 Tumors. Clin Cancer Res 2015; 21:5073-81. [PMID: 25673698 DOI: 10.1158/1078-0432.ccr-14-1717] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 01/24/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE X-ray irradiation of tumors causes diverse effects on the tumor microenvironment, including metabolism. Recent developments of hyperpolarized (13)C-MRI enabled detecting metabolic changes in tumors using a tracer [1-(13)C]pyruvate, which participates in important bioenergetic processes that are altered in cancers. Here, we investigated the effects of X-ray irradiation on pyruvate metabolism in squamous cell carcinoma (SCCVII) and colon cancer (HT-29) using hyperpolarized (13)C-MRI. EXPERIMENTAL DESIGN SCCVII and HT-29 tumors were grown by injecting tumor cells into the hind legs of mice. [1-(13)C]pyruvate was hyperpolarized and injected intravenously into tumor-bearing mice, and (13)C-MR signals were acquired using a 4.7 T scanner. RESULTS [1-(13)C]pyruvate and [1-(13)C]lactate were detected in the tumor-bearing legs immediately after hyperpolarized [1-(13)C]pyruvate administration. The [1-(13)C]lactate to [1-(13)C]pyruvate ratio (Lac/Pyr) increased as the tumors grew in nonirradiated SCCVII tumors. The increase in Lac/Pyr was suppressed modestly with a single 10 Gy of irradiation, but it significantly decreased by further irradiation (10 Gy × 3). Similar results were obtained in HT-29; Lac/Pyr significantly dropped with fractionated 30 Gy irradiation. Independent ex vivo measurements revealed that the lactate dehydrogenase (LDH) activity and protein level were significantly smaller in the irradiated SCCVII tumors compared with the nonirradiated tumors, indicating that a decrease in LDH activity was one of the main factors responsible for the decrease of Lac/Pyr observed on (13)C-MRI. CONCLUSIONS Robust changes of Lac/Pyr observed in the HT-29 after the radiation suggested that lactate conversion from pyruvate monitored with hyperpolarized (13)C-MRI could be useful for the evaluation of early response to radiotherapy. See related commentary by Lai et al., p. 4996.
Collapse
Affiliation(s)
- Keita Saito
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Shingo Matsumoto
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Yoichi Takakusagi
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Masayuki Matsuo
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - H Douglas Morris
- National Institute of Neurological Disorder and Stroke, NIH, Bethesda, Maryland
| | - Martin J Lizak
- National Institute of Neurological Disorder and Stroke, NIH, Bethesda, Maryland
| | - Jeeva P Munasinghe
- National Institute of Neurological Disorder and Stroke, NIH, Bethesda, Maryland
| | | | - Sankaran Subramanian
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
32
|
Park I, Mukherjee J, Ito M, Chaumeil MM, Jalbert LE, Gaensler K, Ronen SM, Nelson SJ, Pieper RO. Changes in pyruvate metabolism detected by magnetic resonance imaging are linked to DNA damage and serve as a sensor of temozolomide response in glioblastoma cells. Cancer Res 2014; 74:7115-24. [PMID: 25320009 PMCID: PMC4253720 DOI: 10.1158/0008-5472.can-14-0849] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent findings show that exposure to temozolomide (TMZ), a DNA-damaging drug used to treat glioblastoma (GBM), can suppress the conversion of pyruvate to lactate. To understand the mechanistic basis for this effect and its potential utility as a TMZ response biomarker, we compared the response of isogenic GBM cell populations differing only in expression of the DNA repair protein methyltransferase (MGMT), a TMZ-sensitivity determinant, after exposure to TMZ in vitro and in vivo. Hyperpolarized [1-((13))C]-pyruvate-based MRI was used to monitor temporal effects on pyruvate metabolism in parallel with DNA-damage responses and tumor cell growth. TMZ exposure decreased conversion of pyruvate to lactate only in MGMT-deficient cells. This effect coincided temporally with TMZ-induced increases in levels of the DNA-damage response protein pChk1. Changes in pyruvate to lactate conversion triggered by TMZ preceded tumor growth suppression and were not associated with changes in levels of NADH or lactate dehydrogenase activity in tumors. Instead, they were associated with a TMZ-induced decrease in the expression and activity of pyruvate kinase PKM2, a glycolytic enzyme that indirectly controls pyruvate metabolism. PKM2 silencing decreased PK activity, intracellular lactate levels, and conversion of pyruvate to lactate in the same manner as TMZ, and Chk1 silencing blocked the TMZ-induced decrease in PKM2 expression. Overall, our findings showed how TMZ-induced DNA damage is linked through PKM2 to changes in pyruvate metabolism, and how these changes can be exploited by MRI methods as an early sensor of TMZ therapeutic response.
Collapse
Affiliation(s)
- Ilwoo Park
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Joydeep Mukherjee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Brain Tumor Research Centre, University of California, San Francisco, San Francisco, California
| | - Motokazu Ito
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Brain Tumor Research Centre, University of California, San Francisco, San Francisco, California
| | - Myriam M Chaumeil
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Llewellyn E Jalbert
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Karin Gaensler
- Department of Hematology/Oncology, University of California, San Francisco, San Francisco, California
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.
| | - Russell O Pieper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Brain Tumor Research Centre, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
33
|
Comment A, Merritt ME. Hyperpolarized magnetic resonance as a sensitive detector of metabolic function. Biochemistry 2014; 53:7333-57. [PMID: 25369537 PMCID: PMC4255644 DOI: 10.1021/bi501225t] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
Hyperpolarized magnetic resonance
allows for noninvasive measurements
of biochemical reactions in vivo. Although this technique
provides a unique tool for assaying enzymatic activities in intact
organs, the scope of its application is still elusive for the wider
scientific community. The purpose of this review is to provide key
principles and parameters to guide the researcher interested in adopting
this technology to address a biochemical, biomedical, or medical issue.
It is presented in the form of a compendium containing the underlying
essential physical concepts as well as suggestions to help assess
the potential of the technique within the framework of specific research
environments. Explicit examples are used to illustrate the power as
well as the limitations of hyperpolarized magnetic resonance.
Collapse
Affiliation(s)
- Arnaud Comment
- Institute of Physics of Biological Systems, Ecole Polytechnique Fédérale de Lausanne , CH-1015 Lausanne, Switzerland
| | | |
Collapse
|
34
|
Lerche MH, Jensen PR, Karlsson M, Meier S. NMR insights into the inner workings of living cells. Anal Chem 2014; 87:119-32. [PMID: 25084065 DOI: 10.1021/ac501467x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Mathilde H Lerche
- Albeda Research , Gamle Carlsberg Vej 10, 1799 Copenhagen V, Denmark
| | | | | | | |
Collapse
|
35
|
Chowdhury R, Ganeshan B, Irshad S, Lawler K, Eisenblätter M, Milewicz H, Rodriguez-Justo M, Miles K, Ellis P, Groves A, Punwani S, Ng T. The use of molecular imaging combined with genomic techniques to understand the heterogeneity in cancer metastasis. BJR Case Rep 2014. [DOI: 10.1259/bjrcr.20140065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
36
|
Canapè C, Catanzaro G, Terreno E, Karlsson M, Lerche MH, Jensen PR. Probing treatment response of glutaminolytic prostate cancer cells to natural drugs with hyperpolarized [5-13C]glutamine. Magn Reson Med 2014; 73:2296-305. [DOI: 10.1002/mrm.25360] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/19/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Carolina Canapè
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center; University of Torino; Torino Italy
| | | | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center; University of Torino; Torino Italy
| | - Magnus Karlsson
- Albeda Research Aps; Gamle Carlsberg Vej 10 Copenhagen Denmark
| | | | | |
Collapse
|
37
|
Suzuki Y, Iida M, Miura I, Inubushi T, Morikawa S. A polymer-based magnetic resonance tracer for visualization of solid tumors by 13C spectroscopic imaging. PLoS One 2014; 9:e102132. [PMID: 25007334 PMCID: PMC4090184 DOI: 10.1371/journal.pone.0102132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/16/2014] [Indexed: 12/22/2022] Open
Abstract
Morphological imaging precedes lesion-specific visualization in magnetic resonance imaging (MRI) because of the superior ability of this technique to depict tissue morphology with excellent spatial and temporal resolutions. To achieve lesion-specific visualization of tumors by MRI, we investigated the availability of a novel polymer-based tracer. Although the 13C nucleus is a candidate for a detection nucleus because of its low background signal in the body, the low magnetic resonance sensitivity of the nucleus needs to be resolved before developing a 13C-based tracer. In order to overcome this problem, we enriched polyethylene glycol (PEG), a biocompatible polymer, with 13C atoms. 13C-PEG40,000 (13C-PEG with an average molecular weight of 40 kDa) emitted a single 13C signal with a high signal-to-noise ratio due to its ability to maintain signal sharpness, as was confirmed by in vivo investigation, and displayed a chemical shift sufficiently distinct from that of endogenous fat. 13C-PEG40,000 intravenously injected into mice showed long retention in circulation, leading to its effective accumulation in tumors reflecting the well-known phenomenon that macromolecules accumulate in tumors because of leaky tumor capillaries. These properties of 13C-PEG40,000 allowed visualization of tumors in mice by 13C spectroscopic imaging. These findings suggest that a technique based on 13C-PEG is a promising strategy for tumor detection.
Collapse
Affiliation(s)
- Yoshikazu Suzuki
- Diagnostic Division, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
- * E-mail:
| | - Mitsuru Iida
- Diagnostic Division, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Iwao Miura
- Pharmaceutical Division, Otsuka Pharmaceutical Co., Ltd., Tokushima, Tokushima, Japan
| | - Toshiro Inubushi
- Biomedical MR Science Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shigehiro Morikawa
- Biomedical MR Science Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
38
|
Chowdhury R, Ganeshan B, Irshad S, Lawler K, Eisenblätter M, Milewicz H, Rodriguez-Justo M, Miles K, Ellis P, Groves A, Punwani S, Ng T. The use of molecular imaging combined with genomic techniques to understand the heterogeneity in cancer metastasis. Br J Radiol 2014; 87:20140065. [PMID: 24597512 PMCID: PMC4075563 DOI: 10.1259/bjr.20140065] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/03/2014] [Indexed: 01/10/2023] Open
Abstract
Tumour heterogeneity has, in recent times, come to play a vital role in how we understand and treat cancers; however, the clinical translation of this has lagged behind advances in research. Although significant advancements in oncological management have been made, personalized care remains an elusive goal. Inter- and intratumour heterogeneity, particularly in the clinical setting, has been difficult to quantify and therefore to treat. The histological quantification of heterogeneity of tumours can be a logistical and clinical challenge. The ability to examine not just the whole tumour but also all the molecular variations of metastatic disease in a patient is obviously difficult with current histological techniques. Advances in imaging techniques and novel applications, alongside our understanding of tumour heterogeneity, have opened up a plethora of non-invasive biomarker potential to examine tumours, their heterogeneity and the clinical translation. This review will focus on how various imaging methods that allow for quantification of metastatic tumour heterogeneity, along with the potential of developing imaging, integrated with other in vitro diagnostic approaches such as genomics and exosome analyses, have the potential role as a non-invasive biomarker for guiding the treatment algorithm.
Collapse
Affiliation(s)
- R Chowdhury
- Richard Dimbleby Department of Cancer Research, Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lodi A, Woods SM, Ronen SM. MR-detectable metabolic consequences of mitogen-activated protein kinase kinase (MEK) inhibition. NMR IN BIOMEDICINE 2014; 27:700-708. [PMID: 24706368 PMCID: PMC4154568 DOI: 10.1002/nbm.3109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/08/2014] [Accepted: 03/09/2014] [Indexed: 06/03/2023]
Abstract
Metabolic reprogramming is increasingly being viewed as a hallmark of cancer. Accordingly, metabolic readouts can serve as biomarkers of response to therapy. The goal of this study was to investigate some of the MRS-detectable metabolic consequences of mitogen-activated protein kinase kinase (MEK) inhibition. We investigated PC3 prostate cancer, MCF-7 breast cancer and A375 melanoma cells, and determined that, consistent with previous studies, MRS-detectable levels of phosphocholine decreased significantly in all cell lines (to 63%, 50% and 18% of the control, respectively) following MEK inhibition with U0126. This effect was mediated by a decrease in the expression of choline kinase α, the enzyme that catalyzes the phosphorylation of choline. In contrast, the impact of MEK inhibition on glycolysis was cell line dependent. A375 cells, which express mutant BRAF, demonstrated significant decreases in glucose uptake (to 36% of control) and lactate production (to 42% of control) in line with positron emission tomography data. In contrast, in PC3 and MCF-7 cells, increases in glucose uptake (to 198% and 192% of control, respectively) and lactate production (to 177% and 212% of control, respectively) were observed, in line with a previous hyperpolarized (13) C MRS study. This effect is probably mediated by the activation of the phosphoinositide 3-kinase pathway and AMP-activated protein kinase. Our findings demonstrate the value of translatable non-invasive MRS methods for the provision of information on cellular metabolism as an indication of the activation of potential feedback loops following MEK inhibition.
Collapse
Affiliation(s)
- Alessia Lodi
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
40
|
Chaumeil MM, Larson PEZ, Woods SM, Cai L, Eriksson P, Robinson AE, Lupo JM, Vigneron DB, Nelson SJ, Pieper RO, Phillips JJ, Ronen SM. Hyperpolarized [1-13C] glutamate: a metabolic imaging biomarker of IDH1 mutational status in glioma. Cancer Res 2014; 74:4247-57. [PMID: 24876103 DOI: 10.1158/0008-5472.can-14-0680] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mutations of the isocitrate dehydrogenase 1 (IDH1) gene are among the most prevalent in low-grade glioma and secondary glioblastoma, represent an early pathogenic event, and are associated with epigenetically driven modulations of metabolism. Of particular interest is the recently uncovered relationship between the IDH1 mutation and decreased activity of the branched-chain amino acid transaminase 1 (BCAT1) enzyme. Noninvasive imaging methods that can assess BCAT1 activity could therefore improve detection of mutant IDH1 tumors and aid in developing and monitoring new targeted therapies. BCAT1 catalyzes the transamination of branched-chain amino acids while converting α-ketoglutarate (α-KG) to glutamate. Our goal was to use (13)C magnetic resonance spectroscopy to probe the conversion of hyperpolarized [1-(13)C] α-KG to hyperpolarized [1-(13)C] glutamate as a readout of BCAT1 activity. We investigated two isogenic glioblastoma lines that differed only in their IDH1 status and performed experiments in live cells and in vivo in rat orthotopic tumors. Following injection of hyperpolarized [1-(13)C] α-KG, hyperpolarized [1-(13)C] glutamate production was detected both in cells and in vivo, and the level of hyperpolarized [1-(13)C] glutamate was significantly lower in mutant IDH1 cells and tumors compared with their IDH1-wild-type counterparts. Importantly however, in our cells the observed drop in hyperpolarized [1-(13)C] glutamate was likely mediated not only by a drop in BCAT1 activity, but also by reductions in aspartate transaminase and glutamate dehydrogenase activities, suggesting additional metabolic reprogramming at least in our model. Hyperpolarized [1-(13)C] glutamate could thus inform on multiple mutant IDH1-associated metabolic events that mediate reduced glutamate production.
Collapse
Affiliation(s)
| | | | | | - Larry Cai
- Departments of Radiology and Biomedical Imaging
| | | | - Aaron E Robinson
- Pathology, and Neurological Surgery, Helen Diller Research Center; and Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| | | | | | | | - Russell O Pieper
- Neurological Surgery, Helen Diller Research Center; and Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| | - Joanna J Phillips
- Pathology, and Neurological Surgery, Helen Diller Research Center; and Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| | - Sabrina M Ronen
- Departments of Radiology and Biomedical Imaging, Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
41
|
Nelson SJ, Kurhanewicz J, Vigneron DB, Larson PEZ, Harzstark AL, Ferrone M, van Criekinge M, Chang JW, Bok R, Park I, Reed G, Carvajal L, Small EJ, Munster P, Weinberg VK, Ardenkjaer-Larsen JH, Chen AP, Hurd RE, Odegardstuen LI, Robb FJ, Tropp J, Murray JA. Metabolic imaging of patients with prostate cancer using hyperpolarized [1-¹³C]pyruvate. Sci Transl Med 2014; 5:198ra108. [PMID: 23946197 DOI: 10.1126/scitranslmed.3006070] [Citation(s) in RCA: 963] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This first-in-man imaging study evaluated the safety and feasibility of hyperpolarized [1-¹³C]pyruvate as an agent for noninvasively characterizing alterations in tumor metabolism for patients with prostate cancer. Imaging living systems with hyperpolarized agents can result in more than 10,000-fold enhancement in signal relative to conventional magnetic resonance (MR) imaging. When combined with the rapid acquisition of in vivo ¹³C MR data, it is possible to evaluate the distribution of agents such as [1-¹³C]pyruvate and its metabolic products lactate, alanine, and bicarbonate in a matter of seconds. Preclinical studies in cancer models have detected elevated levels of hyperpolarized [1-¹³C]lactate in tumor, with the ratio of [1-¹³C]lactate/[1-¹³C]pyruvate being increased in high-grade tumors and decreased after successful treatment. Translation of this technology into humans was achieved by modifying the instrument that generates the hyperpolarized agent, constructing specialized radio frequency coils to detect ¹³C nuclei, and developing new pulse sequences to efficiently capture the signal. The study population comprised patients with biopsy-proven prostate cancer, with 31 subjects being injected with hyperpolarized [1-¹³C]pyruvate. The median time to deliver the agent was 66 s, and uptake was observed about 20 s after injection. No dose-limiting toxicities were observed, and the highest dose (0.43 ml/kg of 230 mM agent) gave the best signal-to-noise ratio for hyperpolarized [1-¹³C]pyruvate. The results were extremely promising in not only confirming the safety of the agent but also showing elevated [1-¹³C]lactate/[1-¹³C]pyruvate in regions of biopsy-proven cancer. These findings will be valuable for noninvasive cancer diagnosis and treatment monitoring in future clinical trials.
Collapse
Affiliation(s)
- Sarah J Nelson
- Surbeck Laboratory of Advanced Imaging, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang H. The potential of hyperpolarized (13)C MRI in assessing signaling pathways in cancer. Acad Radiol 2014; 21:215-22. [PMID: 24439335 DOI: 10.1016/j.acra.2013.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/16/2013] [Accepted: 11/18/2013] [Indexed: 12/28/2022]
Abstract
Advances in genomics are enabling integration of various -omics to reveal the complexities underneath carcinogenesis. Multivariate signaling pathways are deregulated and evolve spatially and temporally depending on the tumor microenvironment. This finding shifts the focus of cancer research from "one disease-one target and drug" to "one disease-multiple pathway targets and combinational therapy" and imposes new challenges on the imaging community in terms of imaging targets, scales and information levels. In current clinical settings, most imaging modalities assess cancer risk through alternations in anatomy, function, metabolism, cellularity, or limited molecular events. Few clinical-translatable imaging modalities are capable of detecting aberrations in signaling pathways at the level of tissue biology. An exception to this is hyperpolarized (13)C magnetic resonance spectroscopic imaging (HP (13)C MRI), which is capable of imaging the molecular signatures of special metabolic enzymes using HP (13)C-labeled substrates. HP (13)C MRI can identify multiple metabolites including intermediates and products simultaneously to allow extraction of critical parameters such as flux alterations for multiple metabolic pathways. Meanwhile, recent progress in cancer metabolism research affirms that metabolic alterations are directly controlled by signaling pathways. Thus, in vivo assessment of aberrations occurring in signaling pathways becomes feasible through HP (13)C imaging. This report briefly reviews the connections between signaling pathways and cancer metabolic phenotypes, the current status of HP (13)C MRI in assessing signal pathways, and recent advances in HP (13)C MRI techniques. Integrated with cancer genomics and animal models, HP (13)C MRI may hold high promise in exploring important issues in cancer that are linked to functionality of signaling pathways. Examples include genomic-driven therapy, intratumoral heterogeneity, and drug resistances.
Collapse
|
43
|
Asghar Butt S, Søgaard LV, Ardenkjaer-Larsen JH, Lauritzen MH, Engelholm LH, Paulson OB, Mirza O, Holck S, Magnusson P, Åkeson P. Monitoring mammary tumor progression and effect of tamoxifen treatment in MMTV-PymT using MRI and magnetic resonance spectroscopy with hyperpolarized [1-13C]pyruvate. Magn Reson Med 2014; 73:51-8. [PMID: 24435823 DOI: 10.1002/mrm.25095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 11/20/2013] [Accepted: 12/04/2013] [Indexed: 01/19/2023]
Abstract
PURPOSE To use dynamic magnetic resonance spectroscopy (MRS) of hyperpolarized (13)C-pyruvate to follow the progress over time in vivo of breast cancer metabolism in the MMTV-PymT model, and to follow the response to the anti-estrogen drug tamoxifen. METHODS Tumor growth was monitored by anatomical MRI by measuring tumor volumes. Dynamic MRS of hyperpolarized (13)C was used to measure an "apparent" pyruvate-to-lactate rate constant (kp) of lactate dehydrogenase (LDH) in vivo. Further, ex vivo pathology and in vitro LDH initial reaction velocity were evaluated. RESULTS Tamoxifen significantly halted the tumor growth measured as tumor volume by MRI. In the untreated animals, kp correlated with tumor growth. The kP was somewhat but not significantly lower in the treated group. Studies in vitro confirmed the effects of tamoxifen on tumor growth, and here the LDH reaction velocity was reduced significantly in the treated group. CONCLUSION These hyperpolarized (13)C MRS findings indicate that tumor metabolic changes affects kP. The measured kp did not relate to treatment response to the same extent as did tumor growth, histological evaluation, and in vitro determination of LDH activity.
Collapse
Affiliation(s)
- Sadia Asghar Butt
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise V Søgaard
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Jan H Ardenkjaer-Larsen
- GE Healthcare, Brøndby, Denmark.,Department of Electrical Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mette H Lauritzen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Lars H Engelholm
- The Finsen Laboratory/BRIC, Rigshospitalet/Copenhagen University, Copenhagen, Denmark
| | - Olaf B Paulson
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Osman Mirza
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Holck
- Department of Pathology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Peter Magnusson
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Per Åkeson
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| |
Collapse
|
44
|
Bokacheva L, Ackerstaff E, LeKaye HC, Zakian K, Koutcher JA. High-field small animal magnetic resonance oncology studies. Phys Med Biol 2013; 59:R65-R127. [PMID: 24374985 DOI: 10.1088/0031-9155/59/2/r65] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review focuses on the applications of high magnetic field magnetic resonance imaging (MRI) and spectroscopy (MRS) to cancer studies in small animals. High-field MRI can provide information about tumor physiology, the microenvironment, metabolism, vascularity and cellularity. Such studies are invaluable for understanding tumor growth and proliferation, response to treatment and drug development. The MR techniques reviewed here include (1)H, (31)P, chemical exchange saturation transfer imaging and hyperpolarized (13)C MRS as well as diffusion-weighted, blood oxygen level dependent contrast imaging and dynamic contrast-enhanced MRI. These methods have been proven effective in animal studies and are highly relevant to human clinical studies.
Collapse
Affiliation(s)
- Louisa Bokacheva
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, 415 East 68 Street, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
45
|
Mignion L, Dutta P, Martinez GV, Foroutan P, Gillies RJ, Jordan BF. Monitoring chemotherapeutic response by hyperpolarized 13C-fumarate MRS and diffusion MRI. Cancer Res 2013; 74:686-94. [PMID: 24285723 DOI: 10.1158/0008-5472.can-13-1914] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Targeted chemotherapeutic agents often do not result in tumor shrinkage, so new biomarkers that correlate with clinical efficacy are needed. In this study, we investigated noninvasive imaging protocols to monitor responses to sorafenib, a multikinase inhibitor approved for treatment of renal cell and hepatocellular carcinoma. Healthy cells are impermeable to fumarate, so conversion of this metabolite to malate as detected by (13)C-magnetic resonance spectroscopy (MRS) has been suggested as one marker for cell death and treatment response in tumors. Diffusion MRI also has been suggested as a measure of therapy-induced cytotoxic edema because viable cells act as a diffusion barrier in tissue. For these reasons, we assessed sorafenib responses using hyperpolarized (13)C-fumarate, diffusion-weighted MRI (DW-MRI) in a xenograft model of human breast cancer in which daily administration of sorafenib was sufficient to stabilize tumor growth. We detected signals from fumarate and malate following intravenous administration of hyperpolarized fumarate with a progressive increase in the malate-to-fumarate (MA/FA) ratio at days 2 to 5 after sorafenib infusion. The apparent diffusion coefficient (ADC) measured by DW-MRI increased in the treated group consistent with cytotoxic edema. However, the MA/FA ratio was a more sensitive marker of therapeutic response than ADC, with 2.8-fold versus 1.3-fold changes, respectively, by day 5 of drug treatment. Histologic analyses confirmed cell death in the sorafenib-treated cohort. Notably, (13)C-pyruvate-to-lactate conversion was not affected by sorafenib in the breast cancer model examined. Our results illustrate how combining hyperpolarized substrates with DW-MRI can allow noninvasive monitoring of targeted therapeutic responses at relatively early times after drug administration.
Collapse
Affiliation(s)
- Lionel Mignion
- Authors' Affiliations: Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium; and Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | | | | | | | | |
Collapse
|
46
|
Spin hyperpolarization in NMR to address enzymatic processes in vivo. MENDELEEV COMMUNICATIONS 2013. [DOI: 10.1016/j.mencom.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
47
|
Nelson SJ, Kurhanewicz J, Vigneron DB, Larson PEZ, Harzstark AL, Ferrone M, van Criekinge M, Chang JW, Bok R, Park I, Reed G, Carvajal L, Small EJ, Munster P, Weinberg VK, Ardenkjaer-Larsen JH, Chen AP, Hurd RE, Odegardstuen LI, Robb FJ, Tropp J, Murray JA. Metabolic imaging of patients with prostate cancer using hyperpolarized [1-¹³C]pyruvate. Sci Transl Med 2013. [PMID: 23946197 DOI: 10.1126/scitranslmed3006070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
This first-in-man imaging study evaluated the safety and feasibility of hyperpolarized [1-¹³C]pyruvate as an agent for noninvasively characterizing alterations in tumor metabolism for patients with prostate cancer. Imaging living systems with hyperpolarized agents can result in more than 10,000-fold enhancement in signal relative to conventional magnetic resonance (MR) imaging. When combined with the rapid acquisition of in vivo ¹³C MR data, it is possible to evaluate the distribution of agents such as [1-¹³C]pyruvate and its metabolic products lactate, alanine, and bicarbonate in a matter of seconds. Preclinical studies in cancer models have detected elevated levels of hyperpolarized [1-¹³C]lactate in tumor, with the ratio of [1-¹³C]lactate/[1-¹³C]pyruvate being increased in high-grade tumors and decreased after successful treatment. Translation of this technology into humans was achieved by modifying the instrument that generates the hyperpolarized agent, constructing specialized radio frequency coils to detect ¹³C nuclei, and developing new pulse sequences to efficiently capture the signal. The study population comprised patients with biopsy-proven prostate cancer, with 31 subjects being injected with hyperpolarized [1-¹³C]pyruvate. The median time to deliver the agent was 66 s, and uptake was observed about 20 s after injection. No dose-limiting toxicities were observed, and the highest dose (0.43 ml/kg of 230 mM agent) gave the best signal-to-noise ratio for hyperpolarized [1-¹³C]pyruvate. The results were extremely promising in not only confirming the safety of the agent but also showing elevated [1-¹³C]lactate/[1-¹³C]pyruvate in regions of biopsy-proven cancer. These findings will be valuable for noninvasive cancer diagnosis and treatment monitoring in future clinical trials.
Collapse
Affiliation(s)
- Sarah J Nelson
- Surbeck Laboratory of Advanced Imaging, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Falck Miniotis M, Arunan V, Eykyn TR, Marais R, Workman P, Leach MO, Beloueche-Babari M. MEK1/2 inhibition decreases lactate in BRAF-driven human cancer cells. Cancer Res 2013; 73:4039-49. [PMID: 23639941 DOI: 10.1158/0008-5472.can-12-1969] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The RAS/BRAF/MEK/ERK signaling pathway is a central driver in cancer with many BRAF and MEK inhibitors being evaluated in clinical trials. Identifying noninvasive biomarkers of early pharmacodynamic responses is important for development of these targeted drugs. As increased aerobic glycolysis is often observed in cancer, we hypothesized that MEK1/2 (MAP2K1/MAP2K2) inhibitors may reduce lactate levels as detected by magnetic resonance spectroscopy (MRS), as a metabolic biomarker for the pharmacodynamic response. MRS was used to monitor intracellular and extracellular levels of lactate in human cancer cells in vitro and in melanoma tumors ex vivo. In addition, we used (1)H MRS and a fluorescent glucose analog to evaluate the effect of MEK inhibition on glucose uptake. MEK1/2 signaling inhibition reduced extracellular lactate levels in BRAF-dependent cells but not BRAF-independent cells. The reduction in extracellular lactate in BRAF-driven melanoma cells was time-dependent and associated with reduced expression of hexokinase-II driven by c-Myc depletion. Taken together, these results reveal how MEK1/2 inhibition affects cancer cell metabolism in the context of BRAF oncogene addiction. Furthermore, they offer a preclinical proof-of-concept for the use of MRS to measure lactate as a noninvasive metabolic biomarker for pharmacodynamic response to MEK1/2 inhibition in BRAF-driven cancers.
Collapse
Affiliation(s)
- Maria Falck Miniotis
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
49
|
Witte C, Schröder L. NMR of hyperpolarised probes. NMR IN BIOMEDICINE 2013; 26:788-802. [PMID: 23033215 DOI: 10.1002/nbm.2873] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/23/2012] [Accepted: 08/29/2012] [Indexed: 06/01/2023]
Abstract
Increasing the sensitivity of NMR experiments is an ongoing field of research to help realise the exquisite molecular specificity of this technique. Hyperpolarisation of various nuclei is a powerful approach that enables the use of NMR for molecular and cellular imaging. Substantial progress has been achieved over recent years in terms of both tracer preparation and detection schemes. This review summarises recent developments in probe design and optimised signal encoding, and promising results in sensitive disease detection and efficient therapeutic monitoring. The different methods have great potential to provide molecular specificity not available by other diagnostic modalities.
Collapse
Affiliation(s)
- Christopher Witte
- ERC Project BiosensorImaging, Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | | |
Collapse
|
50
|
Cabella C, Karlsson M, Canapè C, Catanzaro G, Colombo Serra S, Miragoli L, Poggi L, Uggeri F, Venturi L, Jensen PR, Lerche MH, Tedoldi F. In vivo and in vitro liver cancer metabolism observed with hyperpolarized [5-(13)C]glutamine. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2013; 232:45-52. [PMID: 23689113 DOI: 10.1016/j.jmr.2013.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/15/2013] [Accepted: 04/17/2013] [Indexed: 05/10/2023]
Abstract
Glutamine metabolism is, with its many links to oncogene expression, considered a crucial step in cancer metabolism and it is thereby a key target for alteration in cancer development. In particular, strong correlations have been reported between oncogene expression and expression and activity of the enzyme glutaminase. This mitochondrial enzyme, which is responsible for the deamidation of glutamine to form glutamate, is overexpressed in many tumour tissues. In animal models, glutaminase expression is correlated with tumour growth rate and it is readily possible to limit tumour growth by suppression of glutaminase activity. In principle, hyperpolarized (13)C MR spectroscopy can provide insight to glutamine metabolism and should hence be a valuable tool to study changes in glutaminase activity as tumours progress. However, no such successful in vivo studies have been reported, even though several good biological models have been tested. This may, at least partly, be due to problems in preparing glutamine for hyperpolarization. This paper reports a new and improved preparation of hyperpolarized [5-(13)C]glutamine, which provides a highly sensitive (13)C MR marker. With this preparation of hyperpolarized [5-(13)C]glutamine, glutaminase activity in vivo in a rat liver tumour was investigated. Moreover, this marker was also used to measure response to drug treatment in vitro in cancer cells. These examples of [5-(13)C]glutamine used in tumour models warrant the new preparation to allow metabolic studies with this conditionally essential amino acid.
Collapse
Affiliation(s)
- C Cabella
- Centro Ricerche Bracco, Bracco Imaging Spa, Via Ribes 5, 10010 Colleretto Giacosa (TO), Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|