1
|
Pan F, Liu X, Wan J, Guo Y, Sun P, Zhang X, Wang J, Bao Q, Yang L. Advances and prospects in deuterium metabolic imaging (DMI): a systematic review of in vivo studies. Eur Radiol Exp 2024; 8:65. [PMID: 38825658 PMCID: PMC11144684 DOI: 10.1186/s41747-024-00464-y] [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] [Received: 12/10/2023] [Accepted: 04/02/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Deuterium metabolic imaging (DMI) has emerged as a promising non-invasive technique for studying metabolism in vivo. This review aims to summarize the current developments and discuss the futures in DMI technique in vivo. METHODS A systematic literature review was conducted based on the PRISMA 2020 statement by two authors. Specific technical details and potential applications of DMI in vivo were summarized, including strategies of deuterated metabolites detection, deuterium-labeled tracers and corresponding metabolic pathways in vivo, potential clinical applications, routes of tracer administration, quantitative evaluations of metabolisms, and spatial resolution. RESULTS Of the 2,248 articles initially retrieved, 34 were finally included, highlighting 2 strategies for detecting deuterated metabolites: direct and indirect DMI. Various deuterated tracers (e.g., [6,6'-2H2]glucose, [2,2,2'-2H3]acetate) were utilized in DMI to detect and quantify different metabolic pathways such as glycolysis, tricarboxylic acid cycle, and fatty acid oxidation. The quantifications (e.g., lactate level, lactate/glutamine and glutamate ratio) hold promise for diagnosing malignancies and assessing early anti-tumor treatment responses. Tracers can be administered orally, intravenously, or intraperitoneally, either through bolus administration or continuous infusion. For metabolic quantification, both serial time point methods (including kinetic analysis and calculation of area under the curves) and single time point quantifications are viable. However, insufficient spatial resolution remains a major challenge in DMI (e.g., 3.3-mL spatial resolution with 10-min acquisition at 3 T). CONCLUSIONS Enhancing spatial resolution can facilitate the clinical translation of DMI. Furthermore, optimizing tracer synthesis, administration protocols, and quantification methodologies will further enhance their clinical applicability. RELEVANCE STATEMENT Deuterium metabolic imaging, a promising non-invasive technique, is systematically discussed in this review for its current progression, limitations, and future directions in studying in vivo energetic metabolism, displaying a relevant clinical potential. KEY POINTS • Deuterium metabolic imaging (DMI) shows promise for studying in vivo energetic metabolism. • This review explores DMI's current state, limits, and future research directions comprehensively. • The clinical translation of DMI is mainly impeded by limitations in spatial resolution.
Collapse
Affiliation(s)
- Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinjie Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jiayu Wan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Sun
- MSC Clinical & Technical Solutions, Philips Healthcare, Beijing, 100600, China
| | - Xiaoxiao Zhang
- MSC Clinical & Technical Solutions, Philips Healthcare, Beijing, 100600, China
| | - Jiazheng Wang
- MSC Clinical & Technical Solutions, Philips Healthcare, Beijing, 100600, China
| | - Qingjia Bao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
2
|
Patel RJS, Harlan CJ, Fuentes DT, Bankson JA. A Simulation of the Effects of Diffusion on Hyperpolarized [1- 13C]-Pyruvate Signal Evolution. IEEE Trans Biomed Eng 2023; 70:2905-2913. [PMID: 37097803 PMCID: PMC10538435 DOI: 10.1109/tbme.2023.3269665] [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: 04/26/2023]
Abstract
OBJECTIVE Hyperpolarized [1-13C]-pyruvate magnetic resonance imaging is an emerging metabolic imaging method that offers unprecedented spatiotemporal resolution for monitoring tumor metabolism in vivo. To establish robust imaging biomarkers of metabolism, we must characterize phenomena that may modulate the apparent pyruvate-to-lactate conversion rate (kPL). Here, we investigate the potential effect of diffusion on pyruvate-to-lactate conversion, as failure to account for diffusion in pharmacokinetic analysis may obscure true intracellular chemical conversion rates. METHODS Changes in hyperpolarized pyruvate and lactate signal were calculated using a finite-difference time domain simulation of a two-dimensional tissue model. Signal evolution curves with intracellular kPL values from 0.02 to 1.00 s-1 were analyzed using spatially invariant one-compartment and two-compartment pharmacokinetic models. A second spatially variant simulation incorporating compartmental instantaneous mixing was fit with the same one-compartment model. RESULTS When fitting with the one-compartment model, apparent kPL underestimated intracellular kPL by approximately 50% at an intracellular kPL of 0.02 s-1. This underestimation increased for larger kPL values. However, fitting the instantaneous mixing curves showed that diffusion accounted for only a small part of this underestimation. Fitting with the two-compartment model yielded more accurate intracellular kPL values. SIGNIFICANCE This work suggests diffusion is not a significant rate-limiting factor in pyruvate-to-lactate conversion given that our model assumptions hold true. In higher order models, diffusion effects may be accounted for by a term characterizing metabolite transport. Pharmacokinetic models used to analyze hyperpolarized pyruvate signal evolution should focus on carefully selecting the analytical model for fitting rather than accounting for diffusion effects.
Collapse
|
3
|
Deen SS, Rooney C, Shinozaki A, McGing J, Grist JT, Tyler DJ, Serrão E, Gallagher FA. Hyperpolarized Carbon 13 MRI: Clinical Applications and Future Directions in Oncology. Radiol Imaging Cancer 2023; 5:e230005. [PMID: 37682052 PMCID: PMC10546364 DOI: 10.1148/rycan.230005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/16/2023] [Accepted: 08/02/2023] [Indexed: 09/09/2023]
Abstract
Hyperpolarized carbon 13 MRI (13C MRI) is a novel imaging approach that can noninvasively probe tissue metabolism in both normal and pathologic tissues. The process of hyperpolarization increases the signal acquired by several orders of magnitude, allowing injected 13C-labeled molecules and their downstream metabolites to be imaged in vivo, thus providing real-time information on kinetics. To date, the most important reaction studied with hyperpolarized 13C MRI is exchange of the hyperpolarized 13C signal from injected [1-13C]pyruvate with the resident tissue lactate pool. Recent preclinical and human studies have shown the role of several biologic factors such as the lactate dehydrogenase enzyme, pyruvate transporter expression, and tissue hypoxia in generating the MRI signal from this reaction. Potential clinical applications of hyperpolarized 13C MRI in oncology include using metabolism to stratify tumors by grade, selecting therapeutic pathways based on tumor metabolic profiles, and detecting early treatment response through the imaging of shifts in metabolism that precede tumor structural changes. This review summarizes the foundations of hyperpolarized 13C MRI, presents key findings from human cancer studies, and explores the future clinical directions of the technique in oncology. Keywords: Hyperpolarized Carbon 13 MRI, Molecular Imaging, Cancer, Tissue Metabolism © RSNA, 2023.
Collapse
Affiliation(s)
- Surrin S Deen
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Catriona Rooney
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Ayaka Shinozaki
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Jordan McGing
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - James T Grist
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Damian J Tyler
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Eva Serrão
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| | - Ferdia A Gallagher
- From the Department of Radiology, Cambridge University Hospitals, Biomedical Campus, Cambridge, CB2 0QQ, England (S.S.D., E.S., F.A.G.); Department of Physiology, Anatomy, and Genetics (C.R., A.S., J.T.G., D.J.T.) and the Oxford Centre for Clinical Magnetic Resonance Research (A.S., J.T.G., D.J.T.), University of Oxford, Oxford, England; Department of Radiology, Oxford University Hospitals, Oxford, England (J.M., J.T.G.); Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, England (J.T.G.); Department of Radiology, University of Cambridge, Cambridge, England (E.S., F.A.G.); Cancer Research UK Cambridge Centre, Cambridge, England (F.A.G.); and Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada (E.S.)
| |
Collapse
|
4
|
Han Z, MacCuaig WM, Gurcan MN, Claros-Sorto J, Garwe T, Henson C, Holter-Chakrabarty J, Hannafon B, Chandra V, Wellberg E, McNally LR. Dynamic 2-deoxy-D-glucose-enhanced multispectral optoacoustic tomography for assessing metabolism and vascular hemodynamics of breast cancer. PHOTOACOUSTICS 2023; 32:100531. [PMID: 37485041 PMCID: PMC10362308 DOI: 10.1016/j.pacs.2023.100531] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/22/2023] [Accepted: 07/08/2023] [Indexed: 07/25/2023]
Abstract
Clinical tools for measuring tumor vascular hemodynamics, such as dynamic contrast-enhanced MRI, are clinically important to assess tumor properties. Here we explored the use of multispectral optoacoustic tomography (MSOT), which has a high spatial and temporal resolution, to measure the intratumoral pharmacokinetics of a near-infrared-dye-labeled 2-Deoxyglucose, 2-DG-800, in orthotropic 2-LMP breast tumors in mice. As uptake of 2-DG-800 is dependent on both vascular properties, and glucose transporter activity - a widely-used surrogate for metabolism, we evaluate hemodynamics of 2-DG-MP by fitting the dynamic MSOT signal of 2-DG-800 into two-compartment models including the extended Tofts model (ETM) and reference region model (RRM). We showed that dynamic 2-DG-enhanced MSOT (DGE-MSOT) is powerful in acquiring hemodynamic rate constants, including Ktrans and Kep, via systemically injecting a low dose of 2-DG-800 (0.5 µmol/kg b.w.). In our study, both ETM and RRM are efficient in deriving hemodynamic parameters in the tumor. Area-under-curve (AUC) values (which correlate to metabolism), and Ktrans and Kep values, can effectively distinguish tumor from muscle. Hemodynamic parameters also demonstrated correlations to hemoglobin, oxyhemoglobin, and blood oxygen level (SO2) measurements by spectral unmixing of the MSOT data. Together, our study for the first time demonstrated the capability of DGE-MSOT in assessing vascular hemodynamics of tumors.
Collapse
Affiliation(s)
- Zheng Han
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Center for Health Systems Innovation, Oklahoma State University, Stillwater, OK 74078, USA
| | - William M. MacCuaig
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Department of Bioengineering, University of Oklahoma, Norman, OK 73019, USA
| | - Metin N. Gurcan
- Center for Biomedical Informatics, Wake Forest Baptist Health, Winston-Salem, NC 27101, USA
| | - Juan Claros-Sorto
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Tabitha Garwe
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Christina Henson
- Department of Internal Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | | | - Bethany Hannafon
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Vishal Chandra
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Elizabeth Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Lacey R. McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
5
|
Woitek R, Brindle KM. Hyperpolarized Carbon-13 MRI in Breast Cancer. Diagnostics (Basel) 2023; 13:2311. [PMID: 37443703 DOI: 10.3390/diagnostics13132311] [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: 03/31/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
One of the hallmarks of cancer is metabolic reprogramming, including high levels of aerobic glycolysis (the Warburg effect). Pyruvate is a product of glucose metabolism, and 13C-MR imaging of the metabolism of hyperpolarized (HP) [1-13C]pyruvate (HP 13C-MRI) has been shown to be a potentially versatile tool for the clinical evaluation of tumor metabolism. Hyperpolarization of the 13C nuclear spin can increase the sensitivity of detection by 4-5 orders of magnitude. Therefore, following intravenous injection, the location of hyperpolarized 13C-labeled pyruvate in the body and its subsequent metabolism can be tracked using 13C-MRI. Hyperpolarized [13C]urea and [1,4-13C2]fumarate are also likely to translate to the clinic in the near future as tools for imaging tissue perfusion and post-treatment tumor cell death, respectively. For clinical breast imaging, HP 13C-MRI can be combined with 1H-MRI to address the need for detailed anatomical imaging combined with improved functional tumor phenotyping and very early identification of patients not responding to standard and novel neoadjuvant treatments. If the technical complexity of the hyperpolarization process and the relatively high associated costs can be reduced, then hyperpolarized 13C-MRI has the potential to become more widely available for large-scale clinical trials.
Collapse
Affiliation(s)
- Ramona Woitek
- Research Centre for Medical Image Analysis and AI, Danube Private University, 3500 Krems, Austria
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
6
|
Sharma G, Enriquez JS, Armijo R, Wang M, Bhattacharya P, Pudakalakatti S. Enhancing Cancer Diagnosis with Real-Time Feedback: Tumor Metabolism through Hyperpolarized 1- 13C Pyruvate MRSI. Metabolites 2023; 13:606. [PMID: 37233647 PMCID: PMC10224418 DOI: 10.3390/metabo13050606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/27/2023] Open
Abstract
This review article discusses the potential of hyperpolarized (HP) 13C magnetic resonance spectroscopic imaging (MRSI) as a noninvasive technique for identifying altered metabolism in various cancer types. Hyperpolarization significantly improves the signal-to-noise ratio for the identification of 13C-labeled metabolites, enabling dynamic and real-time imaging of the conversion of [1-13C] pyruvate to [1-13C] lactate and/or [1-13C] alanine. The technique has shown promise in identifying upregulated glycolysis in most cancers, as compared to normal cells, and detecting successful treatment responses at an earlier stage than multiparametric MRI in breast and prostate cancer patients. The review provides a concise overview of the applications of HP [1-13C] pyruvate MRSI in various cancer systems, highlighting its potential for use in preclinical and clinical investigations, precision medicine, and long-term studies of therapeutic response. The article also discusses emerging frontiers in the field, such as combining multiple metabolic imaging techniques with HP MRSI for a more comprehensive view of cancer metabolism, and leveraging artificial intelligence to develop real-time, actionable biomarkers for early detection, assessing aggressiveness, and interrogating the early efficacy of therapies.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Cardiovascular & Thoracic Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA;
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - José S. Enriquez
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA; (J.S.E.); (R.A.); (M.W.); (P.B.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA
| | - Ryan Armijo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA; (J.S.E.); (R.A.); (M.W.); (P.B.)
| | - Muxin Wang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA; (J.S.E.); (R.A.); (M.W.); (P.B.)
| | - Pratip Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA; (J.S.E.); (R.A.); (M.W.); (P.B.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA
| | - Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 75390, USA; (J.S.E.); (R.A.); (M.W.); (P.B.)
| |
Collapse
|
7
|
Skorda A, Bay ML, Hautaniemi S, Lahtinen A, Kallunki T. Kinase Inhibitors in the Treatment of Ovarian Cancer: Current State and Future Promises. Cancers (Basel) 2022; 14:6257. [PMID: 36551745 PMCID: PMC9777107 DOI: 10.3390/cancers14246257] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Ovarian cancer is the deadliest gynecological cancer, the high-grade serous ovarian carcinoma (HGSC) being its most common and most aggressive form. Despite the latest therapeutical advancements following the introduction of vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors and poly-ADP-ribose-polymerase (PARP) inhibitors to supplement the standard platinum- and taxane-based chemotherapy, the expected overall survival of HGSC patients has not improved significantly from the five-year rate of 42%. This calls for the development and testing of more efficient treatment options. Many oncogenic kinase-signaling pathways are dysregulated in HGSC. Since small-molecule kinase inhibitors have revolutionized the treatment of many solid cancers due to the generality of the increased activation of protein kinases in carcinomas, it is reasonable to evaluate their potential against HGSC. Here, we present the latest concluded and on-going clinical trials on kinase inhibitors in HGSC, as well as the recent work concerning ovarian cancer patient organoids and xenograft models. We discuss the potential of kinase inhibitors as personalized treatments, which would require comprehensive assessment of the biological mechanisms underlying tumor spread and chemoresistance in individual patients, and their connection to tumor genome and transcriptome to establish identifiable subgroups of patients who are most likely to benefit from a given therapy.
Collapse
Affiliation(s)
- Aikaterini Skorda
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Marie Lund Bay
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Sampsa Hautaniemi
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Lahtinen
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Tuula Kallunki
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Esfahani SA, Callahan C, Rotile NJ, Heidari P, Mahmood U, Caravan PD, Grant AK, Yen YF. Hyperpolarized [1- 13C]Pyruvate Magnetic Resonance Spectroscopic Imaging for Evaluation of Early Response to Tyrosine Kinase Inhibition Therapy in Gastric Cancer. Mol Imaging Biol 2022; 24:769-779. [PMID: 35467249 PMCID: PMC9588528 DOI: 10.1007/s11307-022-01727-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/13/2023]
Abstract
PURPOSE To evaluate the use of hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopic imaging (HP-13C MRSI) for quantitative measurement of early changes in glycolytic metabolism and its ability to predict response to pan-tyrosine kinase inhibitor (Pan-TKI) therapy in gastric cancer (GCa). PROCEDURES Pan-TKI afatinib-sensitive NCI-N87 and resistant SNU16 human GCa cells were assessed for GLUT1, hexokinase-II (HKII), lactate dehydrogenase (LDHA), phosphorylated AKT (pAKT), and phosphorylated MAPK (pMAPK) at 0-72 h of treatment with 0.1 μM afatinib. Subcutaneous NCI-N87 tumor-bearing nude mice underwent [18F]FDG PET/MRI and HP-13C MRSI at baseline and 4 days after treatment with afatinib 10 mg/kg/day or vehicle (n = 10/group). Changes in PET and HP-13C MRSI metabolic parameters were compared between the two groups. Imaging findings were correlated with tumor growth and histopathology over 3 weeks of treatment. RESULTS In vitro analysis showed a continuous decrease in LDHA, pAKT, and pMAPK in NCI-N87 compared to SNU16 cells within 72 h of treatment with afatinib, without a significant change in GLUT1 and HKII in either cell type. [18F]FDG PET of NCI-N87 tumors showed no significant change in PET measures at baseline and day 4 of treatment in either treatment group (SUVmean day 4/day 0: 2.7 ± 0.42/2.34 ± 0.38, p = 0.57 in the treated group vs. 1.73 ± 0.66/2.24 ± 0.43, p = 0.4 in the control group). HP-13C MRSI demonstrated significantly decreased lactate-to-pyruvate ratio (L/P) in treated tumors (L/P day 4/day 0: 0.83 ± 0.30/1.10 ± 0.20, p = 0.012 vs. 0.94 ± 0.20/0.98 ± 0.30, p = 0.75, in the treated vs. control group, respectively). Response to afatinib was confirmed with decreased tumor size over 3 weeks (11.10 ± 16.50 vs. 293.00 ± 79.30 mm3, p < 0.001, treated group vs. control group, respectively) and histopathologic evaluation. CONCLUSIONS HP-13C MRSI is a more representative biomarker of early metabolic changes in response to pan-TKI in GCa than [18F]FDG PET and could be used for early prediction of response to targeted therapies.
Collapse
Affiliation(s)
- Shadi A Esfahani
- Divisionof Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Cody Callahan
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Pedram Heidari
- Divisionof Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Umar Mahmood
- Divisionof Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Boston, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Peter D Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aaron K Grant
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
9
|
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: 6.5] [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
|
10
|
Fiedorowicz M, Wieteska M, Rylewicz K, Kossowski B, Piątkowska-Janko E, Czarnecka AM, Toczylowska B, Bogorodzki P. Hyperpolarized 13C tracers: Technical advancements and perspectives for clinical applications. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
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: 1.3] [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
|
Goodwin JS, Tsai LL, Mwin D, Coutinho de Souza P, Dialani S, Moon JT, Zhang Z, Grant AK, Ahmed M. In vivo detection of distal tumor glycolytic flux stimulated by hepatic ablation in a breast cancer model using hyperpolarized 13C MRI. Magn Reson Imaging 2021; 80:90-97. [PMID: 33901585 DOI: 10.1016/j.mri.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Hepatic thermal ablation therapy can result in c-Met-mediated off-target stimulation of distal tumor growth. The purpose of this study was to determine if a similar effect on tumor metabolism could be detected in vivo with hyperpolarized 13C MRI. MATERIALS AND METHODS In this prospective study, female Fisher rats (n = 28, 120-150 g) were implanted with R3230 rat breast adenocarcinoma cells and assigned to either: sham surgery, hepatic radiofrequency ablation (RFA), or hepatic RFA + adjuvant c-Met inhibition with PHA-665752 (RFA + PHA). PHA-665752 was administered at 0.83 mg/kg at 24 h post-RFA. Tumor growth was measured daily. MRI was performed 24 h before and 72 h after treatment on 14 rats, and the conversion of 13C-pyruvate into 13C-lactate within each tumor was quantified as lactate:pyruvate ratio (LPR). Comparisons of tumor growth and LPR were performed using paired and unpaired t-tests. RESULTS Hepatic RFA alone resulted in increased growth of the distant tumor compared to sham treatment (0.50 ± 0.13 mm/day versus 0.11 ± 0.07 mm/day; p < 0.001), whereas RFA + PHA (0.06 ± 0.13 mm/day) resulted in no significant change from sham treatment (p = 0.28). A significant increase in LPR was seen following hepatic RFA (+0.016 ± 0.010, p = 0.02), while LPR was unchanged for sham treatment (-0.048 ± 0.051, p = 0.10) or RFA + PHA (0.003 ± 0.041, p = 0.90). CONCLUSION In vivo hyperpolarized 13C MRI can detect hepatic RFA-induced increase in lactate flux within a distant R3230 tumor, which correlates with increased tumor growth. Adjuvant inhibition of c-Met suppresses these off-target effects, supporting a role for the HGF/c-Met signaling axis in these tumorigenic responses.
Collapse
Affiliation(s)
- J Scott Goodwin
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; UT Austin Dell Medical School Transitional Program, 1400 IH-35, CEC 2.404, Austin, TX 78701, USA
| | - Leo L Tsai
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA.
| | - David Mwin
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Patricia Coutinho de Souza
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Genmab, 777 Scudders Mill Rd, Plainsboro, NJ 08536, USA
| | - Svayam Dialani
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Northwestern University, 2145 Ridge Ave, Evanston, IL 60201, USA
| | - John T Moon
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA; Division of Interventional Radiology, Emory University School of Medicine, Emory University Hospital, 1364 Clifton Road NE, Atlanta, GA 30322, USA
| | - Zheng Zhang
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Aaron K Grant
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Muneeb Ahmed
- Beth Israel Deaconess Medical Center, Department of Radiology, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
13
|
Woitek R, Gallagher FA. The use of hyperpolarised 13C-MRI in clinical body imaging to probe cancer metabolism. Br J Cancer 2021; 124:1187-1198. [PMID: 33504974 PMCID: PMC8007617 DOI: 10.1038/s41416-020-01224-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/19/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of cancer and includes the Warburg effect, which is exhibited by many tumours. This can be exploited by positron emission tomography (PET) as part of routine clinical cancer imaging. However, an emerging and alternative method to detect altered metabolism is carbon-13 magnetic resonance imaging (MRI) following injection of hyperpolarised [1-13C]pyruvate. The technique increases the signal-to-noise ratio for the detection of hyperpolarised 13C-labelled metabolites by several orders of magnitude and facilitates the dynamic, noninvasive imaging of the exchange of 13C-pyruvate to 13C-lactate over time. The method has produced promising preclinical results in the area of oncology and is currently being explored in human imaging studies. The first translational studies have demonstrated the safety and feasibility of the technique in patients with prostate, renal, breast and pancreatic cancer, as well as revealing a successful response to treatment in breast and prostate cancer patients at an earlier stage than multiparametric MRI. This review will focus on the strengths of the technique and its applications in the area of oncological body MRI including noninvasive characterisation of disease aggressiveness, mapping of tumour heterogeneity, and early response assessment. A comparison of hyperpolarised 13C-MRI with state-of-the-art multiparametric MRI is likely to reveal the unique additional information and applications offered by the technique.
Collapse
Affiliation(s)
- Ramona Woitek
- Department of Radiology, University of Cambridge, Cambridge, UK.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
- Cancer Research UK Cambridge Centre, Cambridge, UK.
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge, UK
| |
Collapse
|
14
|
van Heijster FH, Heskamp S, Breukels V, Veltien A, Franssen GM, Jansen K(C, Boerman OC, Schalken JA, Scheenen TW, Heerschap A. Pyruvate-lactate exchange and glucose uptake in human prostate cancer cell models. A study in xenografts and suspensions by hyperpolarized [1- 13 C]pyruvate MRS and [ 18 F]FDG-PET. NMR IN BIOMEDICINE 2020; 33:e4362. [PMID: 32662543 PMCID: PMC7507209 DOI: 10.1002/nbm.4362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 06/06/2020] [Accepted: 06/07/2020] [Indexed: 05/04/2023]
Abstract
Reprogramming of energy metabolism in the development of prostate cancer can be exploited for a better diagnosis and treatment of the disease. The goal of this study was to determine whether differences in glucose and pyruvate metabolism of human prostate cancer cells with dissimilar aggressivenesses can be detected using hyperpolarized [1-13 C]pyruvate MRS and [18 F]FDG-PET imaging, and to evaluate whether these measures correlate. For this purpose, we compared murine xenografts of human prostate cancer LNCaP cells with those of more aggressive PC3 cells. [1-13 C]pyruvate was hyperpolarized by dissolution dynamic nuclear polarization (dDNP) and [1-13 C]pyruvate to lactate conversion was followed by 13 C MRS. Subsequently [18 F]FDG uptake was investigated by static and dynamic PET measurements. Standard uptake values (SUVs) for [18 F]FDG were significantly higher for xenografts of PC3 compared with those of LNCaP. However, we did not observe a difference in the average apparent rate constant kpl of 13 C label exchange from pyruvate to lactate between the tumor variants. A significant negative correlation was found between SUVs from [18 F]FDG PET measurements and kpl values for the xenografts of both tumor types. The kpl rate constant may be influenced by various factors, and studies with a range of prostate cancer cells in suspension suggest that LDH inhibition by pyruvate may be one of these. Our results indicate that glucose and pyruvate metabolism in the prostate cancer cell models differs from that in other tumor models and that [18 F]FDG-PET can serve as a valuable complementary tool in dDNP studies of aggressive prostate cancer with [1-13 C]pyruvate.
Collapse
Affiliation(s)
- Frits H.A. van Heijster
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Vincent Breukels
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Andor Veltien
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Gerben M. Franssen
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Otto C. Boerman
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Jack A. Schalken
- Department of UrologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Tom W.J. Scheenen
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
15
|
In vivo magnetic resonance imaging of orthotopic prostate cancer. Biotechniques 2020; 69:395-403. [PMID: 32363906 DOI: 10.2144/btn-2020-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Methods for imaging orthotopic prostate tumors within the prostate or small tumors with extension outside the prostate are needed to more closely model human prostate tumors, which are most commonly located within the gland or may extend just through the gland. By comparing MR sequences, we found that the T2-based Dixon 'water only' sequence best visualized tumors within the prostate of mouse models in both young and old mice and that tumor weight derived from this sequence correlated highly with ex vivo tumor weight (r2 = 0.98, p < 0.001, n = 12). This should aid tumor detection, margin delineation and evaluation of tumor burden to enable studies including, but not limited to, evaluating the natural history of the disease, the mechanisms of action and the efficacy of therapeutic interventions.
Collapse
|
16
|
Biegger P, Ladd ME, Komljenovic D. Multifunctional Magnetic Resonance Imaging Probes. Recent Results Cancer Res 2020; 216:189-226. [PMID: 32594388 DOI: 10.1007/978-3-030-42618-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Magnetic resonance imaging is characterized by high spatial resolution and unsurpassed soft tissue discrimination. Development and characterization of both intrinsic and extrinsic magnetic resonance (MR) imaging probes in the last decade has further strengthened the pivotal role MR imaging holds in the assessment of cancer in preclinical and translational settings. Sophisticated chemical modifications of a variety of nanoparticulate probes hold the potential to deliver valuable multifunctional tools applicable in diagnostics and/or treatment in human oncology. MR imaging suffers from a lack of sensitivity achievable by, e.g., nuclear medicine imaging methods. Advantages of including additional functionality/functionalities in a probe suitable for MR imaging are thus numerous, comprising the addition of fundamentally different imaging information (diagnostics), drug delivery (therapy), or the combination of both (theranostics). In recent years, we have witnessed a plethora of preclinical multimodal or multifunctional imaging probes being published mainly as proof-of-principle studies, yet only a handful are readily applicable in clinical settings. This chapter summarizes recent innovations in the development of multifunctional MR imaging probes and discusses the suitability of these probes for clinical transfer.
Collapse
Affiliation(s)
- Philipp Biegger
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark E Ladd
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, University of Heidelberg, Heidelberg, Germany.,Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
| | - Dorde Komljenovic
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Julià-Sapé M, Candiota AP, Arús C. Cancer metabolism in a snapshot: MRS(I). NMR IN BIOMEDICINE 2019; 32:e4054. [PMID: 30633389 DOI: 10.1002/nbm.4054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
The contribution of MRS(I) to the in vivo evaluation of cancer-metabolism-derived metrics, mostly since 2016, is reviewed here. Increased carbon consumption by tumour cells, which are highly glycolytic, is now being sampled by 13 C magnetic resonance spectroscopic imaging (MRSI) following the injection of hyperpolarized [1-13 C] pyruvate (Pyr). Hot-spots of, mostly, increased lactate dehydrogenase activity or flow between Pyr and lactate (Lac) have been seen with cancer progression in prostate (preclinical and in humans), brain and pancreas (both preclinical) tumours. Therapy response is usually signalled by decreased Lac/Pyr 13 C-labelled ratio with respect to untreated or non-responding tumour. For therapeutic agents inducing tumour hypoxia, the 13 C-labelled Lac/bicarbonate ratio may be a better metric than the Lac/Pyr ratio. 31 P MRSI may sample intracellular pH changes from brain tumours (acidification upon antiangiogenic treatment, basification at fast proliferation and relapse). The steady state tumour metabolome pattern is still in use for cancer evaluation. Metrics used for this range from quantification of single oncometabolites (such as 2-hydroxyglutarate in mutant IDH1 glial brain tumours) to selected metabolite ratios (such as total choline to N-acetylaspartate (plain ratio or CNI index)) or the whole 1 H MRSI(I) pattern through pattern recognition analysis. These approaches have been applied to address different questions such as tumour subtype definition, following/predicting the response to therapy or defining better resection or radiosurgery limits.
Collapse
Affiliation(s)
- Margarida Julià-Sapé
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Carles Arús
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| |
Collapse
|
18
|
Katopodis P, Chudasama D, Wander G, Sales L, Kumar J, Pandhal M, Anikin V, Chatterjee J, Hall M, Karteris E. Kinase Inhibitors and Ovarian Cancer. Cancers (Basel) 2019; 11:E1357. [PMID: 31547471 PMCID: PMC6770231 DOI: 10.3390/cancers11091357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is fifth in the rankings of cancer deaths among women, and accounts for more deaths than any other gynecological malignancy. Despite some improvement in overall-(OS) and progression-free survival (PFS) following surgery and first-line chemotherapy, there is a need for development of novel and more effective therapeutic strategies. In this mini review, we provide a summary of the current landscape of the clinical use of tyrosine kinase inhibitors (TKIs) and mechanistic target of rapamycin (mTOR) inhibitors in ovarian cancer. Emerging data from phase I and II trials reveals that a combinatorial treatment that includes TKIs and chemotherapy agents seems promising in terms of PFS despite some adverse effects recorded; whereas the use of mTOR inhibitors seems less effective. There is a need for further research into the inhibition of multiple signaling pathways in ovarian cancer and progression to phase III trials for drugs that seem most promising.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
| | - Dimple Chudasama
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Gurleen Wander
- Chelsea and Westminster Hospital NHS Trust, London UB9 6JH, UK.
| | - Louise Sales
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Juhi Kumar
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Manreen Pandhal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Jayanta Chatterjee
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| | - Marcia Hall
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK.
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
19
|
Hesketh RL, Wang J, Wright AJ, Lewis DY, Denton AE, Grenfell R, Miller JL, Bielik R, Gehrung M, Fala M, Ros S, Xie B, Hu DE, Brindle KM. Magnetic Resonance Imaging Is More Sensitive Than PET for Detecting Treatment-Induced Cell Death-Dependent Changes in Glycolysis. Cancer Res 2019; 79:3557-3569. [PMID: 31088837 PMCID: PMC6640042 DOI: 10.1158/0008-5472.can-19-0182] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/30/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022]
Abstract
Metabolic imaging has been widely used to measure the early responses of tumors to treatment. Here, we assess the abilities of PET measurement of [18F]FDG uptake and MRI measurement of hyperpolarized [1-13C]pyruvate metabolism to detect early changes in glycolysis following treatment-induced cell death in human colorectal (Colo205) and breast adenocarcinoma (MDA-MB-231) xenografts in mice. A TRAIL agonist that binds to human but not mouse cells induced tumor-selective cell death. Tumor glycolysis was assessed by injecting [1,6-13C2]glucose and measuring 13C-labeled metabolites in tumor extracts. Injection of hyperpolarized [1-13C]pyruvate induced rapid reduction in lactate labeling. This decrease, which correlated with an increase in histologic markers of cell death and preceded decrease in tumor volume, reflected reduced flux from glucose to lactate and decreased lactate concentration. However, [18F]FDG uptake and phosphorylation were maintained following treatment, which has been attributed previously to increased [18F]FDG uptake by infiltrating immune cells. Quantification of [18F]FDG uptake in flow-sorted tumor and immune cells from disaggregated tumors identified CD11b+/CD45+ macrophages as the most [18F]FDG-avid cell type present, yet they represented <5% of the cells present in the tumors and could not explain the failure of [18F]FDG-PET to detect treatment response. MRI measurement of hyperpolarized [1-13C]pyruvate metabolism is therefore a more sensitive marker of the early decreases in glycolytic flux that occur following cell death than PET measurements of [18F]FDG uptake. SIGNIFICANCE: These findings demonstrate superior sensitivity of MRI measurement of hyperpolarized [1-13C]pyruvate metabolism versus PET measurement of 18F-FDG uptake for detecting early changes in glycolysis following treatment-induced tumor cell death.
Collapse
Affiliation(s)
- Richard L Hesketh
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Jiazheng Wang
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Alan J Wright
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - David Y Lewis
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Alice E Denton
- Lymphocyte Signalling and Development, Babraham Hall House, Babraham, Cambridge, United Kingdom
| | - Richard Grenfell
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Jodi L Miller
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Robert Bielik
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Marcel Gehrung
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Maria Fala
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Susana Ros
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Bangwen Xie
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - De-En Hu
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Kevin M Brindle
- CRUK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom.
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
Feuerecker B, Michalik M, Hundshammer C, Schwaiger M, Bruchertseifer F, Morgenstern A, Seidl C. Assessment of 213Bi-anti-EGFR MAb treatment efficacy in malignant cancer cells with [1- 13C]pyruvate and [ 18F]FDG. Sci Rep 2019; 9:8294. [PMID: 31165773 PMCID: PMC6549183 DOI: 10.1038/s41598-019-44484-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 05/13/2019] [Indexed: 12/02/2022] Open
Abstract
Evaluation of response to therapy is among the key objectives of oncology. A new method to evaluate this response includes magnetic resonance spectroscopy (MRS) with hyperpolarized 13C-labelled metabolites, which holds promise to provide new insights in terms of both therapeutic efficacy and tumor cell metabolism. Human EJ28Luc urothelial carcinoma and LN18 glioma cells were treated with lethal activity concentrations of a 213Bi-anti-EGFR immunoconjugate. Treatment efficacy was controlled via analysis of DNA double-strand breaks (immunofluorescence γH2AX staining) and clonogenic survival of cells. To investigate changes in metabolism of treated cells vs controls we analyzed conversion of hyperpolarized [1-13C]pyruvate to [1-13C]lactate via MRS as well as viability of cells, lactate formation and lactate dehydrogenase activity in the cellular supernatants and [18F]FDG uptake in treated cells vs controls, respectively. Treatment of malignant cancer cells with 213Bi-anti-EGFR-MAb induced intense DNA double-strand breaks, resulting in cell death as monitored via clonogenic survival. Moreover, treatment of EJ28Luc bladder cancer cells resulted in decreased cell viability, [18F]FDG-uptake and an increased lactate export. In both EJ28Luc and LN18 carcinoma cells treatment with 213Bi-anti-EGFR-MAb triggered a significant increase in lactate/pyruvate ratios, as measured with hyperpolarized [1-13C]pyruvate. Treatment with 213Bi-anti-EGFR-MAb resulted in an effective induction of cell death in EJ28Luc and LN18 cells. Lactate/pyruvate ratios of hyperpolarized [1-13C]pyruvate proved to detect early treatment response effects, holding promise for future clinical applications in early therapy monitoring.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany. .,German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Michael Michalik
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Christian Hundshammer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany.,Department of Chemistry, Technical University of Munich, Garching, Germany.,Munich School of Bioengineering, Technical University of Munich, Garching, Germany
| | - Markus Schwaiger
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Christof Seidl
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany.,Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Obstetrics and Gynecology, Munich, Germany
| |
Collapse
|
21
|
Wang ZJ, Ohliger MA, Larson PEZ, Gordon JW, Bok RA, Slater J, Villanueva-Meyer JE, Hess CP, Kurhanewicz J, Vigneron DB. Hyperpolarized 13C MRI: State of the Art and Future Directions. Radiology 2019; 291:273-284. [PMID: 30835184 DOI: 10.1148/radiol.2019182391] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hyperpolarized (HP) carbon 13 (13C) MRI is an emerging molecular imaging method that allows rapid, noninvasive, and pathway-specific investigation of dynamic metabolic and physiologic processes that were previously inaccessible to imaging. This technique has enabled real-time in vivo investigations of metabolism that are central to a variety of diseases, including cancer, cardiovascular disease, and metabolic diseases of the liver and kidney. This review provides an overview of the methods of hyperpolarization and 13C probes investigated to date in preclinical models of disease. The article then discusses the progress that has been made in translating this technology for clinical investigation. In particular, the potential roles and emerging clinical applications of HP [1-13C]pyruvate MRI will be highlighted. The future directions to enable the adoption of this technology to advance the basic understanding of metabolism, to improve disease diagnosis, and to accelerate treatment assessment are also detailed.
Collapse
Affiliation(s)
- Zhen J Wang
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Michael A Ohliger
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Peder E Z Larson
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Jeremy W Gordon
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Robert A Bok
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - James Slater
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Javier E Villanueva-Meyer
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Christopher P Hess
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - John Kurhanewicz
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| | - Daniel B Vigneron
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143
| |
Collapse
|
22
|
Hundshammer C, Braeuer M, Müller CA, Hansen AE, Schillmaier M, Düwel S, Feuerecker B, Glaser SJ, Haase A, Weichert W, Steiger K, Cabello J, Schilling F, Hövener JB, Kjær A, Nekolla SG, Schwaiger M. Simultaneous characterization of tumor cellularity and the Warburg effect with PET, MRI and hyperpolarized 13C-MRSI. Am J Cancer Res 2018; 8:4765-4780. [PMID: 30279736 PMCID: PMC6160766 DOI: 10.7150/thno.25162] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023] Open
Abstract
Modern oncology aims at patient-specific therapy approaches, which triggered the development of biomedical imaging techniques to synergistically address tumor biology at the cellular and molecular level. PET/MR is a new hybrid modality that allows acquisition of high-resolution anatomic images and quantification of functional and metabolic information at the same time. Key steps of the Warburg effect-one of the hallmarks of tumors-can be measured non-invasively with this emerging technique. The aim of this study was to quantify and compare simultaneously imaged augmented glucose uptake and LDH activity in a subcutaneous breast cancer model in rats (MAT-B-III) and to study the effect of varying tumor cellularity on image-derived metabolic information. Methods: For this purpose, we established and validated a multimodal imaging workflow for a clinical PET/MR system including proton magnetic resonance (MR) imaging to acquire accurate morphologic information and diffusion-weighted imaging (DWI) to address tumor cellularity. Metabolic data were measured with dynamic [18F]FDG-PET and hyperpolarized (HP) 13C-pyruvate MR spectroscopic imaging (MRSI). We applied our workflow in a longitudinal study and analyzed the effect of growth dependent variations of cellular density on glycolytic parameters. Results: Tumors of similar cellularity with similar apparent diffusion coefficients (ADC) showed a significant positive correlation of FDG uptake and pyruvate-to-lactate exchange. Longitudinal DWI data indicated a decreasing tumor cellularity with tumor growth, while ADCs exhibited a significant inverse correlation with PET standard uptake values (SUV). Similar but not significant trends were observed with HP-13C-MRSI, but we found that partial volume effects and point spread function artifacts are major confounders for the quantification of 13C-data when the spatial resolution is limited and major blood vessels are close to the tumor. Nevertheless, analysis of longitudinal data with varying tumor cellularity further detected a positive correlation between quantitative PET and 13C-data. Conclusions: Our workflow allows the quantification of simultaneously acquired PET, MRSI and DWI data in rodents on a clinical PET/MR scanner. The correlations and findings suggest that a major portion of consumed glucose is metabolized by aerobic glycolysis in the investigated tumor model. Furthermore, we conclude that variations in cell density affect PET and 13C-data in a similar manner and correlations of longitudinal metabolic data appear to reflect both biochemical processes and tumor cellularity.
Collapse
|
23
|
Lacroix R, Rozeman EA, Kreutz M, Renner K, Blank CU. Targeting tumor-associated acidity in cancer immunotherapy. Cancer Immunol Immunother 2018; 67:1331-1348. [PMID: 29974196 PMCID: PMC11028141 DOI: 10.1007/s00262-018-2195-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/29/2018] [Indexed: 12/21/2022]
Abstract
Checkpoint inhibitors, such as cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed cell death-1 (PD-1) monoclonal antibodies have changed profoundly the treatment of melanoma, renal cell carcinoma, non-small cell lung cancer, Hodgkin lymphoma, and bladder cancer. Currently, they are tested in various tumor entities as monotherapy or in combination with chemotherapies or targeted therapies. However, only a subgroup of patients benefit from checkpoint blockade (combinations). This raises the question, which all mechanisms inhibit T cell function in the tumor environment, restricting the efficacy of these immunotherapeutic approaches. Serum activity of lactate dehydrogenase, likely reflecting the glycolytic activity of the tumor cells and thus acidity within the tumor microenvironment, turned out to be one of the strongest markers predicting response to checkpoint inhibition. In this review, we discuss the impact of tumor-associated acidity on the efficacy of T cell-mediated cancer immunotherapy and possible approaches to break this barrier.
Collapse
Affiliation(s)
- Ruben Lacroix
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Elisa A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Magnetic resonance imaging of cancer metabolism with hyperpolarized 13C-labeled cell metabolites. Curr Opin Chem Biol 2018; 45:187-194. [DOI: 10.1016/j.cbpa.2018.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 02/06/2023]
|
25
|
Combined hyperpolarized 13C-pyruvate MRS and 18F-FDG PET (hyperPET) estimates of glycolysis in canine cancer patients. Eur J Radiol 2018; 103:6-12. [PMID: 29803387 DOI: 10.1016/j.ejrad.2018.02.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 02/13/2018] [Accepted: 02/24/2018] [Indexed: 01/13/2023]
Abstract
13C Magnetic Resonance Spectroscopy (MRS) using hyperpolarized 13C-labeled pyruvate as a substrate offers a measure of pyruvate-lactate interconversion and is thereby a marker of the elevated aerobic glycolysis (Warburg effect) generally exhibited by cancer cells. Here, we aim to compare hyperpolarized [1-13C]pyruvate MRS with simultaneous 18F-2-fluoro-2-deoxy-d-glucose (FDG) PET in a cross-sectional study of canine cancer patients. METHODS Canine cancer patients underwent integrated PET/MRI using a clinical whole-body system. Hyperpolarized [1-13C]pyruvate was obtained using dissolution-DNP. 18F-FDG PET, dynamic 13C MRS, 13C MRS Imaging (MRSI) and anatomical 1H MRI was acquired from 17 patients. Apparent pyruvate-to-lactate rate constants were estimated from dynamic 13C MRS. 18F-FDG Standard Uptake Values and maximum [1-13C]lactate-to-total-13C ratios were obtained from tumor regions of interest. Following inspection of data, patients were grouped according to main cancer type and linear regression between measures of lactate generation and 18F-FDG uptake were tested within groups. Between groups, the same measures were tested for group differences. RESULTS The main cancer types of the 17 patients were sarcoma (n = 11), carcinoma (n = 5) and mastocytoma (n = 1). Significant correlations between pyruvate-to-lactate rate constants and 18F-FDG uptake were found for sarcoma patients, whereas no significant correlations appeared for carcinoma patients. The sarcoma patients showed a non-significant trend towards lower 18F-FDG uptake and higher lactate generation than carcinoma patients. However, the ratio of lactate generation to 18F-FDG uptake was found to be significantly higher in sarcoma as compared to carcinoma. The results were found both when lactate generation was estimated as an apparent pyruvate-to-lactate rate constant from dynamic 13C MRS and as an [1-13C]lactate to total 13C ratio from 13C MRSI. CONCLUSIONS A comparison of hyperpolarized [1-13C]pyruvate MRS with simultaneous 18F-FDG PET indicate that lactate generation and 18F-FDG uptake in cancers can be related and that their relation depend on cancer type. This finding could be important for the interpretation and eventual clinical implementation of hyperpolarized 13C. In addition, the differences between the two modalities may allow for better metabolic phenotyping performing hybrid imaging in the form of hyperPET.
Collapse
|