1
|
Sun PZ. Physics-guided multi-dimensional scan optimization and quasi-steady-state reconstruction to enhance CEST MRI sensitivity efficiency and quantification accuracy. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2025; 370:107821. [PMID: 39689390 PMCID: PMC11725439 DOI: 10.1016/j.jmr.2024.107821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Chemical exchange saturation transfer (CEST) MRI has become increasingly utilized for detecting dilute labile protons and characterizing microenvironment properties. However, the CEST MRI effect is only a few percent, and there is a need for a systematic approach to optimize scan parameters for sensitive and accurate CEST quantification. We propose multi-dimensional adjustments of key parameters such as the repetition time (TR) and RF duty cycle to optimize CEST MRI sensitivity per unit of time and utilization of quasi-steady-state (QUASS) reconstruction to recover the full CEST effect during postprocessing. Our work herein derived the CEST effect based on the generalized spin-lock CEST model and determined the interdependency of the optimal RF duty cycle and TR, showing the optimal TR decreases with the RF duty cycle but plateaus beyond 60-80 %. The accuracy of the solution was validated with both numerical simulations and CEST MRI experiments on a dual pH creatine gel phantom. The desired equilibrium CEST effect was further reconstructed with the QUASS algorithm from the optimized CEST MRI scan. In summary, our study establishes a workflow for CEST MRI scan optimization and postprocessing analysis, providing a framework to boost both the sensitivity of CEST MRI scans and the accuracy of CEST quantification. This approach holds promise for future in vivo validation and translation.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Non-Human-Primate Imaging Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
2
|
Yin C, Hu P, Qin L, Wang Z, Zhao H. The Current Status and Future Directions on Nanoparticles for Tumor Molecular Imaging. Int J Nanomedicine 2024; 19:9549-9574. [PMID: 39296941 PMCID: PMC11409933 DOI: 10.2147/ijn.s484206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Molecular imaging is an advanced technology that utilizes specific probes or markers in conjunction with cutting-edge imaging techniques to observe and analyze the localization, distribution, activity, and interactions of biomolecules within living organisms. Tumor molecular imaging, by enabling the visualization and quantification of molecular characteristics of tumor cells, facilitates a deeper and more comprehensive understanding of tumors, providing valuable insights for early diagnosis, treatment monitoring, and cancer biology research. However, the image quality of molecular imaging still requires improvement, and nanotechnology has significantly propelled the advancement of molecular imaging. Currently, nanoparticle-based tumor molecular imaging technologies encompass radionuclide imaging, fluorescence imaging, magnetic resonance imaging, ultrasound imaging, photoacoustic imaging, and multimodal imaging, among others. As our understanding of the tumor microenvironment deepens, the design of nanoparticle probes for tumor molecular imaging has also evolved, offering new perspectives and expanding the applications of tumor molecular imaging. Beyond diagnostics, there is a marked trend towards integrated diagnosis and therapy, with image-guided treatment playing a pivotal role. This includes image-guided surgery, photodynamic therapy, and chemodynamic therapy. Despite continuous advancements and innovative developments in molecular imaging, many of these remain in the experimental stage and require breakthroughs before they can be fully integrated into clinical practice.
Collapse
Affiliation(s)
- Caiyun Yin
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, People's Republic of China
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Peiyun Hu
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, People's Republic of China
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Lijing Qin
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Zhicheng Wang
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People's Republic of China
| | - Hongguang Zhao
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
3
|
Gammaraccio F, Villano D, Irrera P, Anemone AA, Carella A, Corrado A, Longo DL. Development and Validation of Four Different Methods to Improve MRI-CEST Tumor pH Mapping in Presence of Fat. J Imaging 2024; 10:166. [PMID: 39057737 PMCID: PMC11277679 DOI: 10.3390/jimaging10070166] [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: 05/08/2024] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
CEST-MRI is an emerging imaging technique suitable for various in vivo applications, including the quantification of tumor acidosis. Traditionally, CEST contrast is calculated by asymmetry analysis, but the presence of fat signals leads to wrong contrast quantification and hence to inaccurate pH measurements. In this study, we investigated four post-processing approaches to overcome fat signal influences and enable correct CEST contrast calculations and tumor pH measurements using iopamidol. The proposed methods involve replacing the Z-spectrum region affected by fat peaks by (i) using a linear interpolation of the fat frequencies, (ii) applying water pool Lorentzian fitting, (iii) considering only the positive part of the Z-spectrum, or (iv) calculating a correction factor for the ratiometric value. In vitro and in vivo studies demonstrated the possibility of using these approaches to calculate CEST contrast and then to measure tumor pH, even in the presence of moderate to high fat fraction values. However, only the method based on the water pool Lorentzian fitting produced highly accurate results in terms of pH measurement in tumor-bearing mice with low and high fat contents.
Collapse
Affiliation(s)
- Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Pietro Irrera
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Annasofia A. Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| |
Collapse
|
4
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
5
|
Feng Q, Bennett Z, Grichuk A, Pantoja R, Huang T, Faubert B, Huang G, Chen M, DeBerardinis RJ, Sumer BD, Gao J. Severely polarized extracellular acidity around tumour cells. Nat Biomed Eng 2024; 8:787-799. [PMID: 38438799 DOI: 10.1038/s41551-024-01178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024]
Abstract
Extracellular pH impacts many molecular, cellular and physiological processes, and hence is tightly regulated. Yet, in tumours, dysregulated cancer cell metabolism and poor vascular perfusion cause the tumour microenvironment to become acidic. Here by leveraging fluorescent pH nanoprobes with a transistor-like activation profile at a pH of 5.3, we show that, in cancer cells, hydronium ions are excreted into a small extracellular region. Such severely polarized acidity (pH <5.3) is primarily caused by the directional co-export of protons and lactate, as we show for a diverse panel of cancer cell types via the genetic knockout or inhibition of monocarboxylate transporters, and also via nanoprobe activation in multiple tumour models in mice. We also observed that such spot acidification in ex vivo stained snap-frozen human squamous cell carcinoma tissue correlated with the expression of monocarboxylate transporters and with the exclusion of cytotoxic T cells. Severely spatially polarized tumour acidity could be leveraged for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Biomedical Engineering, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zachary Bennett
- Department of Biomedical Engineering, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anthony Grichuk
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Raymundo Pantoja
- Department of Biomedical Engineering, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tongyi Huang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brandon Faubert
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gang Huang
- Department of Biomedical Engineering, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralph J DeBerardinis
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Baran D Sumer
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jinming Gao
- Department of Biomedical Engineering, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
6
|
Fabian MS, Rajput JR, Schüre JR, Weinmüller S, Mennecke A, Möhle TA, Rampp S, Schmidt M, Dörfler A, Zaiss M. Comprehensive 7 T CEST: A clinical MRI protocol covering multiple exchange rate regimes. NMR IN BIOMEDICINE 2024; 37:e5096. [PMID: 38343093 DOI: 10.1002/nbm.5096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 04/04/2024]
Abstract
Chemical exchange saturation transfer (CEST) is a magnetic resonance (MR) imaging method providing molecular image contrasts based on indirect detection of low concentrated solutes. Previous CEST studies focused predominantly on the imaging of single CEST exchange regimes (e.g., slow, intermediate or fast exchanging groups). In this work, we aim to establish a so-called comprehensive CEST protocol for 7 T, covering the different exchange regimes by three saturation B1 amplitude regimes: low, intermediate and high. We used the results of previous publications and our own simulations in pulseq-CEST to produce a 7 T CEST protocol that has sensitivity to these three B1 regimes. With postprocessing optimization (simultaneous mapping of water shift and B1, B0-fitting, multiple interleaved mode saturation B1 correction, neural network employment (deepCEST) and analytical input feature reduction), we are able to shorten our initially 40 min protocol to 15 min and generate six CEST contrast maps simultaneously. With this protocol, we measured four healthy subjects and one patient with a brain tumor. We established a comprehensive CEST protocol for clinical 7 T MRI, covering three different B1 amplitude regimes. We were able to reduce the acquisition time significantly by more than 50%, while still maintaining decent image quality and contrast in healthy subjects and one patient with a tumor. Our protocol paves the way to perform comprehensive CEST studies in clinical scan times for hypothesis generation regarding molecular properties of certain pathologies, for example, ischemic stroke or high-grade brain tumours.
Collapse
Affiliation(s)
- Moritz Simon Fabian
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Junaid Rasool Rajput
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jan-Rüdiger Schüre
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Weinmüller
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Angelika Mennecke
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Alexius Möhle
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Rampp
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Schmidt
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Dörfler
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Moritz Zaiss
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
- High-field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
7
|
Mishra SK, Santana JG, Mihailovic J, Hyder F, Coman D. Transmembrane pH gradient imaging in rodent glioma models. NMR IN BIOMEDICINE 2024; 37:e5102. [PMID: 38263680 PMCID: PMC10987279 DOI: 10.1002/nbm.5102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/28/2023] [Accepted: 12/16/2023] [Indexed: 01/25/2024]
Abstract
A unique feature of the tumor microenvironment is extracellular acidosis in relation to intracellular milieu. Metabolic reprogramming in tumors results in overproduction of H+ ions (and lactate), which are extruded from the cells to support tumor survival and progression. As a result, the transmembrane pH gradient (ΔpH), representing the difference between intracellular pH (pHi) and extracellular pH (pHe), is posited to be larger in tumors compared with normal tissue. Controlling the transmembrane pH difference has promise as a potential therapeutic target in cancer as it plays an important role in regulating drug delivery into cells. The current study shows successful development of an MRI/MRSI-based technique that provides ΔpH imaging at submillimeter resolution. We applied this technique to image ΔpH in rat brains with RG2 and U87 gliomas, as well as in mouse brains with GL261 gliomas. pHi was measured with Amine and Amide Concentration-Independent Detection (AACID), while pHe was measured with Biosensor Imaging of Redundant Deviation in Shifts (BIRDS). The results indicate that pHi was slightly higher in tumors (7.40-7.43 in rats, 7.39-7.47 in mice) compared with normal brain (7.30-7.38 in rats, 7.32-7.36 in mice), while pHe was significantly lower in tumors (6.62-6.76 in rats, 6.74-6.84 in mice) compared with normal tissue (7.17-7.22 in rats, 7.20-7.21 in mice). As a result, ΔpH was higher in tumors (0.64-0.81 in rats, 0.62-0.65 in mice) compared with normal brain (0.13-0.16 in rats, 0.13-0.16 in mice). This work establishes an MRI/MRSI-based platform for ΔpH imaging at submillimeter resolution in gliomas.
Collapse
Affiliation(s)
- Sandeep Kumar Mishra
- Yale University, Department of Radiology & Biomedical Imaging, New Haven, CT 06510, USA
| | | | - Jelena Mihailovic
- Yale University, Department of Radiology & Biomedical Imaging, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Yale University, Department of Radiology & Biomedical Imaging, New Haven, CT 06510, USA
- Yale University, Department of Biomedical Engineering, New Haven, CT 06510, USA
| | - Daniel Coman
- Yale University, Department of Radiology & Biomedical Imaging, New Haven, CT 06510, USA
- Yale University, Department of Biomedical Engineering, New Haven, CT 06510, USA
| |
Collapse
|
8
|
Ding J, He L, Yang L, Cheng L, Zhao Z, Luo B, Jia Y. Novel Nanoprobe with Combined Ultrasonography/Chemical Exchange Saturation Transfer Magnetic Resonance Imaging for Precise Diagnosis of Tumors. Pharmaceutics 2023; 15:2693. [PMID: 38140034 PMCID: PMC10747786 DOI: 10.3390/pharmaceutics15122693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Given that cancer mortality is usually due to a late diagnosis, early detection is crucial to improve the patient's results and prevent cancer-related death. Imaging technology based on novel nanomaterials has attracted much attention for early-stage cancer diagnosis. In this study, a new block copolymer, poly(ethylene glycol)-poly(l-lactide) diblock copolymer (PEG-PLLA), was synthesized by the ring-opening polymerization method and thoroughly characterized using Fourier transform infrared spectroscopy (FT-IR), proton nuclear magnetic resonance spectroscopy (H-NMR), X-ray diffraction (XRD), and thermogravimetric analysis (TGA). The obtained PEG-PLLA was used to prepare nanoparticles encapsulated with perfluoropentane and salicylic acid by the emulsion-solvent evaporation method, resulting in a new dual-mode nano-image probe (PEG-PLLA@SA·PFP). The zeta potential and mean diameter of the obtained nanoparticles were measured using dynamic light scattering (DLS) with a Malvern Zetersizer Nano. The in vitro biocompatibility of the PEG-PLLA nanoparticles was evaluated with cell migration, hemolysis, and cytotoxicity assays. Ultrasonic imaging was performed using an ultrasonic imaging apparatus, and chemical exchange saturation transfer (CEST) MRI was conducted on a 7.0 T animal scanner. The results of IR and NMR confirmed that the PEG-PLLA was successfully synthesized. The particle size and negative charge of the nanoparticles were 223.8 ± 2.5 nm and -39.6 ± 1.9 mV, respectively. The polydispersity of the diameter was 0.153 ± 0.020. These nanoparticles possessed good stability at 4 °C for about one month. The results of cytotoxicity, cell migration, and hemolysis assays showed that the carrier material was biocompatible. Finally, PEG-PLLA nanoparticles were able to significantly enhance the imaging effect of tumors by the irradiation of ultrasound and saturation by a radiofrequency pulse, respectively. In conclusion, these nanoparticles exhibit promising dual-mode capabilities for US/CEST MR imaging.
Collapse
Affiliation(s)
- Jieqiong Ding
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (J.D.); (L.H.); (L.C.)
| | - Liu He
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (J.D.); (L.H.); (L.C.)
| | - Lin Yang
- Department of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China;
| | - Liyuan Cheng
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (J.D.); (L.H.); (L.C.)
| | - Zhiwei Zhao
- Department of Radiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning 437100, China;
| | - Binhua Luo
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (J.D.); (L.H.); (L.C.)
| | - Yanlong Jia
- Department of Radiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
| |
Collapse
|
9
|
Dhakan C, Anemone A, Ventura V, Carella A, Corrado A, Pirotta E, Villano D, Romdhane F, Gammaraccio F, Aime S, Longo DL. Assessing the Therapeutic Efficacy of Proton Transport Inhibitors in a Triple-Negative Breast Cancer Murine Model with Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer Tumor pH Imaging. Metabolites 2023; 13:1161. [PMID: 37999256 PMCID: PMC10673543 DOI: 10.3390/metabo13111161] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Proton transporters play a key role in maintaining the acidic tumor microenvironment; hence, their inhibition has been proposed as a new therapeutic treatment, although few methods can accurately assess their effect in vivo. In this study, we investigated whether MRI-CEST (Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer) tumor pH imaging can be a useful tool to evaluate in vivo the therapeutic efficacy of several Proton Pump Inhibitors (PPIs) in breast cancer. Cell viability and extracellular pH assays were carried out in breast cancer cells cultured at physiological pH (7.4) or acid-adapted (pH of 6.5 and 6.8) following the exposure to inhibitors of V-ATPase (Lansoprazole, Esomeprazole) or NHE1 (Amiloride, Cariporide) at several concentrations. Next, triple-negative breast cancer 4T1 tumor-bearing mice were treated with Lansoprazole or Amiloride and MRI-CEST tumor pH imaging was utilized to assess the in vivo efficacy. Only Lansoprazole induced, in addition to breast cancer cell toxicity, a significant inhibition of proton extrusion. A significant reduction in tumor volume, prolonged survival, and increase in extracellular tumor pH after 1 and 2 weeks were observed after Lansoprazole treatment, whereas no significant changes were detected upon Amiloride treatment. Our results suggested that MRI-CEST tumor pH imaging can monitor the therapeutic efficacy of PPIs in breast cancer murine models.
Collapse
Affiliation(s)
- Chetan Dhakan
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Annasofia Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Vittoria Ventura
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Elisa Pirotta
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Feriel Romdhane
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Silvio Aime
- IRCCS SynLAB SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
10
|
Sun PZ. Numerical simulation-based assessment of pH-sensitive chemical exchange saturation transfer MRI quantification accuracy across field strengths. NMR IN BIOMEDICINE 2023; 36:e5000. [PMID: 37401645 PMCID: PMC11990165 DOI: 10.1002/nbm.5000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/27/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI detects dilute labile protons via their exchange with bulk water, conferring pH sensitivity. Based on published exchange and relaxation properties, a 19-pool simulation was used to model the brain pH-dependent CEST effect and assess the accuracy of quantitative CEST (qCEST) analysis across magnetic field strengths under typical scan conditions. First, the optimal B1 amplitude was determined by maximizing pH-sensitive amide proton transfer (APT) contrast under the equilibrium condition. Apparent and quasi-steady-state (QUASS) CEST effects were then derived under the optimal B1 amplitude as functions of pH, RF saturation duration, relaxation delay, Ernst flip angle, and field strength. Finally, CEST effects, particularly the APT signal, were isolated with spinlock model-based Z-spectral fitting to evaluate the accuracy and consistency of CEST quantification. Our data showed that QUASS reconstruction significantly improved the consistency between simulated and equilibrium Z-spectra. The residual difference between QUASS and equilibrium CEST Z-spectra was, on average, 30 times less than that of the apparent CEST Z-spectra across field strengths, saturation, and repetition times. Also, the spinlock fitting of the QUASS CEST effect significantly reduced the residual errors 9-fold. Furthermore, the isolated APT amplitude from QUASS reconstruction was consistent and higher than the apparent CEST analysis under nonequilibrium conditions. To summarize, this study confirmed that QUASS reconstruction facilitates accurate determination of the CEST system under different scan protocols across field strengths, with the potential to help standardize CEST quantification.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Primate Imaging Center, Emory National Primate Research Center, Emory University, Atlanta, GA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
11
|
Nagar D, Vladimirov N, Farrar CT, Perlman O. Dynamic and rapid deep synthesis of chemical exchange saturation transfer and semisolid magnetization transfer MRI signals. Sci Rep 2023; 13:18291. [PMID: 37880343 PMCID: PMC10600114 DOI: 10.1038/s41598-023-45548-8] [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: 05/30/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
Model-driven analysis of biophysical phenomena is gaining increased attention and utility for medical imaging applications. In magnetic resonance imaging (MRI), the availability of well-established models for describing the relations between the nuclear magnetization, tissue properties, and the externally applied magnetic fields has enabled the prediction of image contrast and served as a powerful tool for designing the imaging protocols that are now routinely used in the clinic. Recently, various advanced imaging techniques have relied on these models for image reconstruction, quantitative tissue parameter extraction, and automatic optimization of acquisition protocols. In molecular MRI, however, the increased complexity of the imaging scenario, where the signals from various chemical compounds and multiple proton pools must be accounted for, results in exceedingly long model simulation times, severely hindering the progress of this approach and its dissemination for various clinical applications. Here, we show that a deep-learning-based system can capture the nonlinear relations embedded in the molecular MRI Bloch-McConnell model, enabling a rapid and accurate generation of biologically realistic synthetic data. The applicability of this simulated data for in-silico, in-vitro, and in-vivo imaging applications is then demonstrated for chemical exchange saturation transfer (CEST) and semisolid macromolecule magnetization transfer (MT) analysis and quantification. The proposed approach yielded 63-99% acceleration in data synthesis time while retaining excellent agreement with the ground truth (Pearson's r > 0.99, p < 0.0001, normalized root mean square error < 3%).
Collapse
Affiliation(s)
- Dinor Nagar
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Christian T Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
12
|
Igarashi T, Kim H, Sun PZ. Detection of tissue pH with quantitative chemical exchange saturation transfer magnetic resonance imaging. NMR IN BIOMEDICINE 2023; 36:e4711. [PMID: 35141979 PMCID: PMC10249910 DOI: 10.1002/nbm.4711] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 05/12/2023]
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) has emerged as a novel means for sensitive detection of dilute labile protons and chemical exchange rates. By sensitizing to pH-dependent chemical exchange, CEST MRI has shown promising results in monitoring tissue statuses such as pH changes in disorders like acute stroke, tumor, and acute kidney injury. This article briefly reviews the basic principles for CEST imaging and quantitative measures, from the simplistic asymmetry analysis to multipool Lorentzian decoupling and quasi-steady-state reconstruction. In particular, the advantages and limitations of commonly used quantitative approaches for CEST applications are discussed.
Collapse
Affiliation(s)
- Takahiro Igarashi
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Hahnsung Kim
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Phillip Zhe Sun
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| |
Collapse
|
13
|
Perlman O, Farrar CT, Heo HY. MR fingerprinting for semisolid magnetization transfer and chemical exchange saturation transfer quantification. NMR IN BIOMEDICINE 2023; 36:e4710. [PMID: 35141967 PMCID: PMC9808671 DOI: 10.1002/nbm.4710] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/18/2022] [Accepted: 02/04/2022] [Indexed: 05/11/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI has positioned itself as a promising contrast mechanism, capable of providing molecular information at sufficient resolution and amplified sensitivity. However, it has not yet become a routinely employed clinical technique, due to a variety of confounding factors affecting its contrast-weighted image interpretation and the inherently long scan time. CEST MR fingerprinting (MRF) is a novel approach for addressing these challenges, allowing simultaneous quantitation of several proton exchange parameters using rapid acquisition schemes. Recently, a number of deep-learning algorithms have been developed to further boost the performance and speed of CEST and semi-solid macromolecule magnetization transfer (MT) MRF. This review article describes the fundamental theory behind semisolid MT/CEST-MRF and its main applications. It then details supervised and unsupervised learning approaches for MRF image reconstruction and describes artificial intelligence (AI)-based pipelines for protocol optimization. Finally, practical considerations are discussed, and future perspectives are given, accompanied by basic demonstration code and data.
Collapse
Affiliation(s)
- Or Perlman
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Knutsson L, Xu X, van Zijl PCM, Chan KWY. Imaging of sugar-based contrast agents using their hydroxyl proton exchange properties. NMR IN BIOMEDICINE 2023; 36:e4784. [PMID: 35665547 PMCID: PMC9719573 DOI: 10.1002/nbm.4784] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 05/13/2023]
Abstract
The ability of CEST MRI to detect the presence of millimolar concentrations of non-metallic contrast agents has made it possible to study, non-invasively, important biological molecules such as proteins and sugars, as well as drugs already approved for clinical use. Here, we review efforts to use sugar and sugar polymers as exogenous contrast agents, which is possible based on the exchange of their hydroxyl protons with water protons. While this capability has raised early enthusiasm, for instance about the possibility of imaging D-glucose metabolism with MRI in a way analogous to PET, experience over the past decade has shown that this is not trivial. On the other hand, many studies have confirmed the possibility of imaging a large variety of sugar analogues, each with potentially interesting applications to assess tissue physiology. Some promising applications are the study of (i) sugar delivery and transport to assess blood-brain barrier integrity and (ii) sugar uptake by cells for their characterization (e.g., cancer versus healthy), as well as (iii) clearance of sugars to assess tissue drainage-for instance, through the glymphatic system. To judge these opportunities and their challenges, especially in the clinic, it is necessary to understand the technical aspects of detecting the presence of rapidly exchanging protons through the water signal in MRI, especially as a function of magnetic field strength. We expect that novel approaches in terms of MRI detection (both saturation transfer and relaxation based), MRI data analysis, and sugar design will push this young field forward in the next decade.
Collapse
Affiliation(s)
- Linda Knutsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, US
| | - Xiang Xu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Peter CM van Zijl
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, US
| | - Kannie WY Chan
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong
- City University of Hong Kong Shenzhen Institute, Shenzhen, China
| |
Collapse
|
15
|
Jardim-Perassi BV, Irrera P, Lau JYC, Budzevich M, Whelan CJ, Abrahams D, Ruiz E, Ibrahim-Hashim A, Damgaci Erturk S, Longo DL, Pilon-Thomas SA, Gillies RJ. Intraperitoneal Delivery of Iopamidol to Assess Extracellular pH of Orthotopic Pancreatic Tumor Model by CEST-MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2023; 2023:1944970. [PMID: 36704211 PMCID: PMC9836819 DOI: 10.1155/2023/1944970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/05/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023]
Abstract
The extracellular pH (pHe) of solid tumors is often acidic, as a consequence of the Warburg effect, and an altered metabolic state is often associated with malignancy. It has been shown that acidosis can promote tumor progression; thus, many therapeutic strategies have been adopted against tumor metabolism; one of these involves alkalinization therapies to raise tumor pH to inhibit tumor progression, improve immune surveillance, and overcome resistance to chemotherapies. Chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) is a noninvasive technique that can measure pH in vivo using pH-sensitive contrast agents. Iopamidol, an iodinated contrast agent, clinically used for computed tomography (CT), contains amide group protons with pH-dependent exchange rates that can reveal the pHe of the tumor microenvironment. In this study, we optimized intraperitoneal (IP) delivery of iopamidol to facilitate longitudinal assessments of orthotopic pancreatic tumor pHe by CEST-MRI. Following IV-infusion and IP-bolus injections, we compared the two protocols for assessing tumor pH. Time-resolved CT imaging was used to evaluate the uptake of iopamidol in the tumor, revealing that IP-bolus delivered a high amount of contrast agent 40 min postinjection, which was similar to the amounts reached with the IV-infusion protocol. As expected, both IP and IV injection protocols produced comparable measurements of tumor pHe, showing no statistically significant difference between groups (p=0.16). In addition, we showed the ability to conduct longitudinal monitoring of tumor pHe using CEST-MRI with the IP injection protocol, revealing a statistically significant increase in tumor pHe following bicarbonate administration (p < 0.001). In conclusion, this study shows the capability to measure pHe using an IP delivery of iopamidol into orthotopic pancreatic tumors, which is important to conduct longitudinal studies.
Collapse
Affiliation(s)
| | - Pietro Irrera
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Justin Y. C. Lau
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mikalai Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Christopher J. Whelan
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Biological Sciences, University of Illinois, Chicago, IL, USA
| | | | - Epifanio Ruiz
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Arig Ibrahim-Hashim
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sultan Damgaci Erturk
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dario Livio Longo
- Institute of Biostructures and Bioimages (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Shari A. Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert J. Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
16
|
Freidel L, Li S, Choffart A, Kuebler L, Martins AF. Imaging Techniques in Pharmacological Precision Medicine. Handb Exp Pharmacol 2023; 280:213-235. [PMID: 36907970 DOI: 10.1007/164_2023_641] [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] [Indexed: 03/14/2023]
Abstract
Biomedical imaging is a powerful tool for medical diagnostics and personalized medicines. Examples of commonly used imaging modalities include Positron Emission Tomography (PET), Ultrasound (US), Single Photon Emission Computed Tomography (SPECT), and hybrid imaging. By combining these modalities, scientists can gain a comprehensive view and better understand physiology and pathology at the preclinical, clinical, and multiscale levels. This can aid in the accuracy of medical diagnoses and treatment decisions. Moreover, biomedical imaging allows for evaluating the metabolic, functional, and structural details of living tissues. This can be particularly useful for the early diagnosis of diseases such as cancer and for the application of personalized medicines. In the case of hybrid imaging, two or more modalities are combined to produce a high-resolution image with enhanced sensitivity and specificity. This can significantly improve the accuracy of diagnosis and offer more detailed treatment plans. In this book chapter, we showcase how continued advancements in biomedical imaging technology can potentially revolutionize medical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Lucas Freidel
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Sixing Li
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Anais Choffart
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Laura Kuebler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - André F Martins
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany.
- German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Xiao P, Huang J, Han X, Cheu JWS, Liu Y, Law LH, Lai JHC, Li J, Park SW, Wong CCL, Lam RHW, Chan KWY. Monitor Tumor pHe and Response Longitudinally during Treatment Using CEST MRI-Detectable Alginate Microbeads. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54401-54410. [PMID: 36448714 PMCID: PMC9756293 DOI: 10.1021/acsami.2c10493] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/05/2022] [Indexed: 06/17/2023]
Abstract
Imaging pHe of the tumor microenvironment has paramount importance for characterizing aggressive, invasive tumors, as well as therapeutic responses. Here, a robust approach to image pH changes in the tumor microenvironment longitudinally and during sodium bicarbonate treatment was reported. The pH-sensing microbeads were designed and prepared based on materials approved for clinical use, i.e., alginate microbead-containing computed tomography (CT) contrast-agent (iopamidol)-loaded liposomes (Iop-lipobeads). This Iop-lipobead prepared using a customized microfluidic device generated a CEST contrast of 10.6% at 4.2 ppm at pH 7.0, which was stable for 20 days in vitro. The CEST contrast decreased by 11.8% when the pH decreased from 7.0 to 6.5 in vitro. Optimized Iop-lipobeads next to tumors showed a significant increase of 19.7 ± 6.1% (p < 0.01) in CEST contrast at 4.2 ppm during the first 3 days of treatment and decreased to 15.2 ± 4.8% when treatment stopped. Notably, percentage changes in Iop-lipobeads were higher than that of amide CEST (11.7% and 9.1%) in tumors during and after treatment. These findings demonstrated that the Iop-lipobead could provide an independent and sensitive assessment of the pHe changes for a noninvasive and longitudinal monitoring of the treatment effects using multiple CEST contrast.
Collapse
Affiliation(s)
- Peng Xiao
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jianpan Huang
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Xiongqi Han
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jacinth W. S. Cheu
- Department
of Pathology, Li Ka Shing Faculty of Medicine,
The University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Lok Hin Law
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Joseph H. C. Lai
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jiyu Li
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Se Weon Park
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Carmen C. L. Wong
- Department
of Pathology, Li Ka Shing Faculty of Medicine,
The University of Hong Kong, Hong Kong, China
- State
Key Laboratory of Liver Research, The University
of Hong Kong, Hong Kong, China
| | - Raymond H. W. Lam
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Kannie W. Y. Chan
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
- City
University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Russell
H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Tung
Biomedical
Sciences Centre, City University of Hong
Kong, Hong Kong, China
- Hong
Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| |
Collapse
|
18
|
Zhou J, Zaiss M, Knutsson L, Sun PZ, Ahn SS, Aime S, Bachert P, Blakeley JO, Cai K, Chappell MA, Chen M, Gochberg DF, Goerke S, Heo HY, Jiang S, Jin T, Kim SG, Laterra J, Paech D, Pagel MD, Park JE, Reddy R, Sakata A, Sartoretti-Schefer S, Sherry AD, Smith SA, Stanisz GJ, Sundgren PC, Togao O, Vandsburger M, Wen Z, Wu Y, Zhang Y, Zhu W, Zu Z, van Zijl PCM. Review and consensus recommendations on clinical APT-weighted imaging approaches at 3T: Application to brain tumors. Magn Reson Med 2022; 88:546-574. [PMID: 35452155 PMCID: PMC9321891 DOI: 10.1002/mrm.29241] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/16/2022]
Abstract
Amide proton transfer-weighted (APTw) MR imaging shows promise as a biomarker of brain tumor status. Currently used APTw MRI pulse sequences and protocols vary substantially among different institutes, and there are no agreed-on standards in the imaging community. Therefore, the results acquired from different research centers are difficult to compare, which hampers uniform clinical application and interpretation. This paper reviews current clinical APTw imaging approaches and provides a rationale for optimized APTw brain tumor imaging at 3 T, including specific recommendations for pulse sequences, acquisition protocols, and data processing methods. We expect that these consensus recommendations will become the first broadly accepted guidelines for APTw imaging of brain tumors on 3 T MRI systems from different vendors. This will allow more medical centers to use the same or comparable APTw MRI techniques for the detection, characterization, and monitoring of brain tumors, enabling multi-center trials in larger patient cohorts and, ultimately, routine clinical use.
Collapse
Affiliation(s)
- Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Moritz Zaiss
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Linda Knutsson
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Medical Radiation Physics, Lund University, Lund, Sweden.,F.M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Phillip Zhe Sun
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Peter Bachert
- Department of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany.,Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michael A Chappell
- Mental Health and Clinical Neurosciences and Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Min Chen
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Daniel F Gochberg
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Physics, Vanderbilt University, Nashville, Tennessee, USA
| | - Steffen Goerke
- Department of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Jin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science and Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - John Laterra
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Daniel Paech
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany.,Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Mark D Pagel
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Ravinder Reddy
- Center for Advance Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akihiko Sakata
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - A Dean Sherry
- Advanced Imaging Research Center and Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas, USA
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Pia C Sundgren
- Department of Diagnostic Radiology/Clinical Sciences Lund, Lund University, Lund, Sweden.,Lund University Bioimaging Center, Lund University, Lund, Sweden.,Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Osamu Togao
- Department of Molecular Imaging and Diagnosis, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yi Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Peter C M van Zijl
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
20
|
Radiometal-Based PET/MRI Contrast Agents for Sensing Tumor Extracellular pH. BIOSENSORS 2022; 12:bios12020134. [PMID: 35200394 PMCID: PMC8870419 DOI: 10.3390/bios12020134] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/03/2023]
Abstract
Acidosis is a useful biomarker for tumor diagnoses and for evaluating early response to anti-cancer treatments. Despite these useful applications, there are few methods for non-invasively measuring tumor extracellular pH, and none are routinely used in clinics. Responsive MRI contrast agents have been developed, and they undergo a change in MRI signal with pH. However, these signal changes are concentration-dependent, and it is difficult to accurately measure the concentration of an MRI contrast agent in vivo. PET/MRI provides a unique opportunity to overcome this concentration dependence issue by using the PET component to report on the concentration of the pH-responsive MRI agent. Herein, we synthesized PET/MRI co-agents based on the design of a pH-dependent MRI agent, and we have correlated pH with the r1 relaxivity of the MRI co-agent. We have also developed a procedure that uses PET radioactivity measurements and MRI R1 relaxation rate measurements to determine the r1 relaxivity of the MRI co-agent, which can then be used to estimate pH. This simultaneous PET/MRI procedure accurately measured pH in solution, with a precision that depended on the concentration of the MRI co-agent. We used our procedure to measure extracellular pH in a subcutaneous flank model of MIA PaCa-2 pancreatic cancer. Although the PET co-agents were stable in serum, post-imaging studies showed evidence that the PET co-agents were degraded in vivo. These results showed that tumor acidosis can be evaluated with simultaneous PET/MRI, although improvements are needed to more precisely measure MRI R1 relaxation rates, and ensure the in vivo stability of the agents.
Collapse
|
21
|
Huang J, Chen Z, Park SW, Lai JHC, Chan KWY. Molecular Imaging of Brain Tumors and Drug Delivery Using CEST MRI: Promises and Challenges. Pharmaceutics 2022; 14:451. [PMID: 35214183 PMCID: PMC8880023 DOI: 10.3390/pharmaceutics14020451] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/10/2022] Open
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) detects molecules in their natural forms in a sensitive and non-invasive manner. This makes it a robust approach to assess brain tumors and related molecular alterations using endogenous molecules, such as proteins/peptides, and drugs approved for clinical use. In this review, we will discuss the promises of CEST MRI in the identification of tumors, tumor grading, detecting molecular alterations related to isocitrate dehydrogenase (IDH) and O-6-methylguanine-DNA methyltransferase (MGMT), assessment of treatment effects, and using multiple contrasts of CEST to develop theranostic approaches for cancer treatments. Promising applications include (i) using the CEST contrast of amide protons of proteins/peptides to detect brain tumors, such as glioblastoma multiforme (GBM) and low-grade gliomas; (ii) using multiple CEST contrasts for tumor stratification, and (iii) evaluation of the efficacy of drug delivery without the need of metallic or radioactive labels. These promising applications have raised enthusiasm, however, the use of CEST MRI is not trivial. CEST contrast depends on the pulse sequences, saturation parameters, methods used to analyze the CEST spectrum (i.e., Z-spectrum), and, importantly, how to interpret changes in CEST contrast and related molecular alterations in the brain. Emerging pulse sequence designs and data analysis approaches, including those assisted with deep learning, have enhanced the capability of CEST MRI in detecting molecules in brain tumors. CEST has become a specific marker for tumor grading and has the potential for prognosis and theranostics in brain tumors. With increasing understanding of the technical aspects and associated molecular alterations detected by CEST MRI, this young field is expected to have wide clinical applications in the near future.
Collapse
Affiliation(s)
- Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Se-Weon Park
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Joseph H. C. Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Tung Biomedical Science Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
22
|
Kombala CJ, Kotrotsou A, Schuler FW, de la Cerda J, Ma JC, Zhang S, Pagel MD. Development of a Nanoscale Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Contrast Agent That Measures pH. ACS NANO 2021; 15:20678-20688. [PMID: 34870957 PMCID: PMC11847439 DOI: 10.1021/acsnano.1c10107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
AcidoCEST MRI can measure the extracellular pH (pHe) of the tumor microenvironment in mouse models of human cancers and in patients who have cancer. However, chemical exchange saturation transfer (CEST) is an insensitive magnetic resonance imaging (MRI) contrast mechanism, requiring a high concentration of small-molecule agent to be delivered to the tumor. Herein, we developed a nanoscale CEST agent that can measure pH using acidoCEST MRI, which may decrease the requirement for high delivery concentrations of agent. We also developed a monomer agent for comparison to the polymer. After optimizing CEST experimental conditions, we determined that the polymer agent could be used during acidoCEST MRI studies at 125-fold and 488-fold lower concentration than the monomer agent and iopamidol, respectively. We also determined that both agents can measure pH with negligible dependence on temperature. However, pH measurements with both agents were dependent on concentration, which may be due to concentration-dependent changes in hydrogen bonding and/or steric hindrance. We performed in vivo acidoCEST MRI studies using the three agents to study a xenograft MDA-MB-231 model of mammary carcinoma. The tumor pHe measurements were 6.33 ± 0.12, 6.70 ± 0.15, and 6.85 ± 0.15 units with iopamidol, the monomer agent, and polymer agent, respectively. The higher pHe measurements with the monomer and polymer agents were attributed to the concentration dependence of these agents. This study demonstrated that nanoscale agents have merit for CEST MRI studies, but consideration should be given to the dependence of CEST contrast on the concentration of these agents.
Collapse
Affiliation(s)
- Chathuri J Kombala
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Aikaterini Kotrotsou
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - F William Schuler
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Jorge de la Cerda
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Jacqueline C Ma
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Shu Zhang
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Mark D Pagel
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| |
Collapse
|
23
|
Kombala CJ, Lokugama SD, Kotrotsou A, Li T, Pollard AC, Pagel MD. Simultaneous Evaluations of pH and Enzyme Activity with a CEST MRI Contrast Agent. ACS Sens 2021; 6:4535-4544. [PMID: 34856102 PMCID: PMC11936461 DOI: 10.1021/acssensors.1c02408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The extracellular tumor microenvironment of many solid tumors has high acidosis and high protease activity. Simultaneously assessing both characteristics may improve diagnostic evaluations of aggressive tumors and the effects of anticancer treatments. Noninvasive imaging methods have previously been developed that measure extracellular pH or can detect enzyme activity using chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI). Herein, we developed a single-hybrid CEST agent that can simultaneously measure pH and evaluate protease activity using a combination of dual-power acidoCEST MRI and catalyCEST MRI. Our agent showed CEST signals at 9.2 ppm from a salicylic acid moiety and at 5.0 ppm from an aryl amide. The CEST signal at 9.2 ppm could be measured after selective saturation was applied at 1 and 4 μT, and these measurements could be used with a ratiometric analysis to determine pH. The CEST signal at 5.0 ppm from the aryl amide disappeared after the agent was treated with cathepsin B, while the CEST signal at 9.2 ppm remained, indicating that the agent could detect protease activity through the amide bond cleavage. Michaelis-Menten kinetics studies with catalyCEST MRI demonstrated that the binding affinity (as shown with the Michaelis constant KM), the catalytic turnover rate (kcat), and catalytic efficiency (kcat/KM) were each higher for cathepsin B at lower pH. The kcat rates measured with catalyCEST MRI were lower than the comparable rates measured with liquid chromatography-mass spectrometry (LC-MS), which reflected a limitation of inherently noisy and relatively insensitive CEST MRI analyses. Although this level of precision limited catalyCEST MRI to semiquantitative evaluations, these semiquantitative assessments of high and low protease activity still had value by demonstrating that high acidosis and high protease activity can be used as synergistic, multiparametric biomarkers.
Collapse
Affiliation(s)
- Chathuri J Kombala
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Sanjaya D Lokugama
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Aikaterini Kotrotsou
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Tianzhe Li
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Alyssa C Pollard
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Chemistry, Rice University, Houston, Texas 77251, United States
| | - Mark D Pagel
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| |
Collapse
|
24
|
Wei H, Frey AM, Jasanoff A. Molecular fMRI of neurochemical signaling. J Neurosci Methods 2021; 364:109372. [PMID: 34597714 DOI: 10.1016/j.jneumeth.2021.109372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Magnetic resonance imaging (MRI) is the most widely applied technique for brain-wide measurement of neural function in humans and animals. In conventional functional MRI (fMRI), brain signaling is detected indirectly, via localized activity-dependent changes in regional blood flow, oxygenation, and volume, to which MRI contrast can be readily sensitized. Although such hemodynamic fMRI methods are powerful tools for analysis of brain activity, they lack specificity for the many molecules and cell types that play functionally distinct roles in neural processing. A suite of techniques collectively known to as "molecular fMRI," addresses this limitation by permitting MRI-based detection of specific molecular processes in deep brain tissue. This review discusses how molecular fMRI is coming to be used in the study of neurochemical dynamics that mediate intercellular communication in the brain. Neurochemical molecular fMRI is a potentially powerful approach for mechanistic analysis of brain-wide function, but the techniques are still in early stages of development. Here we provide an overview of the major advances and results that have been achieved to date, as well as directions for further development.
Collapse
Affiliation(s)
- He Wei
- Department of Biological Engineering, Massachusetts Institute of Technology, United States
| | - Abigail M Frey
- Department of Chemical Engineering, Massachusetts Institute of Technology, United States
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, United States; Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, United States; Department of Nuclear Science & Engineering, Massachusetts Institute of Technology, United States.
| |
Collapse
|
25
|
Gao T, Zou C, Li Y, Jiang Z, Tang X, Song X. A Brief History and Future Prospects of CEST MRI in Clinical Non-Brain Tumor Imaging. Int J Mol Sci 2021; 22:11559. [PMID: 34768990 PMCID: PMC8584005 DOI: 10.3390/ijms222111559] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/12/2021] [Accepted: 10/23/2021] [Indexed: 02/08/2023] Open
Abstract
Chemical exchange saturation transfer (CEST) MRI is a promising molecular imaging tool which allows the specific detection of metabolites that contain exchangeable amide, amine, and hydroxyl protons. Decades of development have progressed CEST imaging from an initial concept to a clinical imaging tool that is used to assess tumor metabolism. The first translation efforts involved brain imaging, but this has now progressed to imaging other body tissues. In this review, we summarize studies using CEST MRI to image a range of tumor types, including breast cancer, pelvic tumors, digestive tumors, and lung cancer. Approximately two thirds of the published studies involved breast or pelvic tumors which are sites that are less affected by body motion. Most studies conclude that CEST shows good potential for the differentiation of malignant from benign lesions with a number of reports now extending to compare different histological classifications along with the effects of anti-cancer treatments. Despite CEST being a unique 'label-free' approach with a higher sensitivity than MR spectroscopy, there are still some obstacles for implementing its clinical use. Future research is now focused on overcoming these challenges. Vigorous ongoing development and further clinical trials are expected to see CEST technology become more widely implemented as a mainstream imaging technology.
Collapse
Affiliation(s)
- Tianxin Gao
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Chuyue Zou
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Yifan Li
- Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Zhenqi Jiang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Xiaoying Tang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Xiaolei Song
- Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
26
|
Impact of Inhibition of the Mitochondrial Pyruvate Carrier on the Tumor Extracellular pH as Measured by CEST-MRI. Cancers (Basel) 2021; 13:cancers13174278. [PMID: 34503089 PMCID: PMC8428345 DOI: 10.3390/cancers13174278] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/22/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
(1) Background: The acidosis of the tumor micro-environment may have profound impact on cancer progression and on the efficacy of treatments. In the present study, we evaluated the impact of a treatment with UK-5099, a mitochondrial pyruvate carrier (MPC) inhibitor on tumor extracellular pH (pHe); (2) Methods: glucose consumption, lactate secretion and extracellular acidification rate (ECAR) were measured in vitro after exposure of cervix cancer SiHa cells and breast cancer 4T1 cells to UK-5099 (10 µM). Mice bearing the 4T1 tumor model were treated daily during four days with UK-5099 (3 mg/kg). The pHe was evaluated in vivo using either chemical exchange saturation transfer (CEST)-MRI with iopamidol as pHe reporter probe or 31P-NMR spectroscopy with 3-aminopropylphosphonate (3-APP). MR protocols were applied before and after 4 days of treatment; (3) Results: glucose consumption, lactate release and ECAR were increased in both cell lines after UK-5099 exposure. CEST-MRI showed a significant decrease in tumor pHe of 0.22 units in UK-5099-treated mice while there was no change over time for mice treated with the vehicle. Parametric images showed a large heterogeneity in response with 16% of voxels shifting to pHe values under 7.0. In contrast, 31P-NMR spectroscopy was unable to detect any significant variation in pHe; (4) Conclusions: MPC inhibition led to a moderate acidification of the extracellular medium in vivo. CEST-MRI provided high resolution parametric images (0.44 µL/voxel) of pHe highlighting the heterogeneity of response within the tumor when exposed to UK-5099.
Collapse
|
27
|
Minhas AS, Sharkey J, Randtke EA, Murray P, Wilm B, Pagel MD, Poptani H. Measuring Kidney Perfusion, pH, and Renal Clearance Consecutively Using MRI and Multispectral Optoacoustic Tomography. Mol Imaging Biol 2021; 22:494-503. [PMID: 31529408 PMCID: PMC7250811 DOI: 10.1007/s11307-019-01429-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Purpose: To establish multi-modal imaging for the assessment of kidney pH, perfusion, and clearance rate using magnetic resonance imaging (MRI) and multispectral optoacoustic tomography (MSOT) in healthy mice. Kidney pH and perfusion values were measured on a pixel-by-pixel basis using the MRI acidoCEST and FAIR-EPI methods. Kidney filtration rate was measured by analyzing the renal clearance rate of IRdye 800 using MSOT. To test the effect of one imaging method on the other, a set of 3 animals were imaged with MSOT followed by MRI, and a second set of 3 animals were imaged with MRI followed by MSOT. In a subsequent study, the reproducibility of pH, perfusion, and renal clearance measurements were tested by imaging 4 animals twice, separated by 4 days. The contrast agents used for acidoCEST based pH measurements influenced the results of MSOT. Specifically, the exponential decay time from the kidney cortex, as measured by MSOT, was significantly altered when MRI was performed prior to MSOT. However, no significant difference in the cortex to pelvis area under the curve (AUC) was noted. When the order of experiments was reversed, no significant differences were noted in the pH or perfusion values. Reproducibility measurements demonstrated similar pH and cortex to pelvis AUC; however, perfusion values were significantly different with the cortex values being higher and the pelvic values being lower in the second imaging time. We demonstrate that using a combination of MRI and MSOT, physiological measurements of pH, blood flow, and clearance rates can be measured in the mouse kidney in the same imaging session.
Collapse
Affiliation(s)
- Atul S Minhas
- Center for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, Merseyside, UK.,School of Engineering, Macquarie University, Sydney, NSW, Australia
| | - Jack Sharkey
- Center for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, Merseyside, UK
| | - Edward A Randtke
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Patricia Murray
- Center for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, Merseyside, UK
| | - Bettina Wilm
- Center for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, Merseyside, UK
| | | | - Harish Poptani
- Center for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, Merseyside, UK.
| |
Collapse
|
28
|
Hajjo R, Sabbah DA, Bardaweel SK, Tropsha A. Identification of Tumor-Specific MRI Biomarkers Using Machine Learning (ML). Diagnostics (Basel) 2021; 11:742. [PMID: 33919342 PMCID: PMC8143297 DOI: 10.3390/diagnostics11050742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
The identification of reliable and non-invasive oncology biomarkers remains a main priority in healthcare. There are only a few biomarkers that have been approved as diagnostic for cancer. The most frequently used cancer biomarkers are derived from either biological materials or imaging data. Most cancer biomarkers suffer from a lack of high specificity. However, the latest advancements in machine learning (ML) and artificial intelligence (AI) have enabled the identification of highly predictive, disease-specific biomarkers. Such biomarkers can be used to diagnose cancer patients, to predict cancer prognosis, or even to predict treatment efficacy. Herein, we provide a summary of the current status of developing and applying Magnetic resonance imaging (MRI) biomarkers in cancer care. We focus on all aspects of MRI biomarkers, starting from MRI data collection, preprocessing and machine learning methods, and ending with summarizing the types of existing biomarkers and their clinical applications in different cancer types.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan;
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA;
- National Center for Epidemics and Communicable Disease Control, Amman 11118, Jordan
| | - Dima A. Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan;
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan;
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
29
|
Sun PZ. Quasi-steady-state CEST (QUASS CEST) solution improves the accuracy of CEST quantification: QUASS CEST MRI-based omega plot analysis. Magn Reson Med 2021; 86:765-776. [PMID: 33749052 DOI: 10.1002/mrm.28744] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE CEST MRI omega plot quantifies the labile proton fraction ratio (fr ) and exchange rate (ksw ), yet it assumes long RF saturation time (Ts) and relaxation delay (Td). Our study aimed to test if a quasi-steady-state (QUASS) CEST analysis that accounts for the effect of finite Ts and Td could improve the accuracy of CEST MRI quantification. METHODS We modeled the MRI signal evolution using a typical CEST EPI sequence. The signal relaxes toward its thermal equilibrium following the bulk water relaxation rate during Td, and then toward its CEST steady state following the spin-lock relaxation rate during Ts from which the QUASS CEST effect is derived. Both fr and ksw were solved from simulated conventional apparent CEST and QUASS CEST MRI. We also performed MRI experiments from a Cr-gel phantom under serially varied Ts and Td times from 1.5 to 7.5 s. RESULTS Simulation showed that, although ksw could be slightly overestimated (3%-15%) for the range of Ts and Td, fr could be substantially underestimated by as much as 67%. In contrast, the QUASS solution provided accurate ksw and fr determination within 2%. The CEST MRI experiments confirmed that the QUASS solution enabled robust quantification of ksw and fr , superior over the omega plot analysis based on the conventional apparent CEST MRI measurements. CONCLUSIONS The QUASS CEST MRI algorithm corrects the effect of finite Ts and Td times on CEST measurements, thereby allowing robust and accurate CEST quantification.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
30
|
Bo S, Sedaghat F, Pavuluri K, Rowe SP, Cohen A, Kates M, McMahon MT. Dynamic Contrast Enhanced-MR CEST Urography: An Emerging Tool in the Diagnosis and Management of Upper Urinary Tract Obstruction. Tomography 2021; 7:80-94. [PMID: 33801533 PMCID: PMC8103243 DOI: 10.3390/tomography7010008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/16/2021] [Indexed: 02/04/2023] Open
Abstract
Upper urinary tract obstructions (UTOs) are blockages that inhibit the flow of urine through its normal course, leading to impaired kidney function. Imaging plays a significant role in the initial diagnosis of UTO, with anatomic imaging (primarily ultrasound (US) and non-contrast computed tomography (CT)) serving as screening tools for the detection of the dilation of the urinary collecting systems (i.e., hydronephrosis). Whether hydronephrosis represents UTO or a non-obstructive process is determined by functional imaging (typically nuclear medicine renal scintigraphy). If these exams reveal evidence of UTO but no discernable source, multiphase contrast enhanced CT urography and/or dynamic contrast enhanced MR urography (DCE-MRU) may be performed to delineate a cause. These are often performed in conjunction with direct ureteroscopic evaluation. While contrast-enhanced CT currently predominates, it can induce renal injury due to contrast induced nephropathy (CIN), subject patients to ionizing radiation and is limited in quantifying renal function (traditionally assessed by renal scintigraphy) and establishing the extent to which hydronephrosis is due to functional obstruction. Traditional MRI is similarly limited in its ability to quantify function. DCE-MRU presents concerns regarding nephrogenic systemic fibrosis (NSF), although decreased with newer gadolinium-based contrast agents, and regarding cumulative gadolinium deposition in the basal ganglia. DCE-MR CEST urography is a promising alternative, employing new MRI contrast agents and imaging schemes and allowing for concurrent assessment of renal anatomy and functional parameters. In this review we highlight clinical challenges in the diagnosis and management of UTO, identify key advances in imaging agents and techniques for DCE-MR CEST urography and provide perspective on how this technique may evolve in clinical importance.
Collapse
Affiliation(s)
- Shaowei Bo
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA; (S.B.); (F.S.); (K.P.); (S.P.R.)
| | - Farzad Sedaghat
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA; (S.B.); (F.S.); (K.P.); (S.P.R.)
| | - KowsalyaDevi Pavuluri
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA; (S.B.); (F.S.); (K.P.); (S.P.R.)
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA; (S.B.); (F.S.); (K.P.); (S.P.R.)
- The James Buchanan Brady Urological Institute, Department of Urology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (A.C.); (M.K.)
| | - Andrew Cohen
- The James Buchanan Brady Urological Institute, Department of Urology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (A.C.); (M.K.)
| | - Max Kates
- The James Buchanan Brady Urological Institute, Department of Urology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (A.C.); (M.K.)
| | - Michael T. McMahon
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA; (S.B.); (F.S.); (K.P.); (S.P.R.)
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
31
|
Dai Z, Kalra S, Mah D, Seres P, Sun H, Wu R, Wilman AH. Amide signal intensities may be reduced in the motor cortex and the corticospinal tract of ALS patients. Eur Radiol 2021; 31:1401-1409. [PMID: 32909054 DOI: 10.1007/s00330-020-07243-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The aim of the study is to assess amide concentration changes in ALS patients compared with healthy controls by using quantitative amide proton transfer (APT) and multiparameter magnetic resonance imaging, and testing its correlation with clinical scores. METHODS Sixteen ALS patients and sixteen healthy controls were recruited as part of the Canadian ALS Neuroimaging Consortium, and multimodal magnetic resonance imaging was performed at 3 T, including APT and diffusion imaging. Lorentz fitting was used to quantify the amide effect. Clinical disability was evaluated using the revised ALS functional rating scale (ALSFRS-R), and its correlation with image characteristics was assessed. The diagnostic performance of different imaging parameters was evaluated with receiver operating characteristic analysis. RESULTS Our results showed that the amide peak was significantly different between the motor cortex and other gray matter territories within the brain of ALS patients (p < 0.001). Compared with controls, amide signal intensities in ALS were significantly reduced in the motor cortex (p < 0.001) and corticospinal tract (p = 0.046), while abnormalities were not detected using routine imaging methods. There was no significant correlation between amide and ALSFRS-R score. The diagnostic accuracy of the amide peak was superior to that of diffusion imaging. CONCLUSIONS This study demonstrated changes of amide signal intensities in the motor cortex and corticospinal tract of ALS patients. KEY POINTS • The neurodegenerative disease amyotrophic lateral sclerosis (ALS) has a lack of objective imaging indicators for diagnosis and assessment. • Analysis of amide proton transfer imaging revealed changes in the motor cortex and corticospinal tract of ALS patients that were not visible on standard magnetic resonance imaging. • The diagnostic accuracy of the amide peak was superior to that of diffusion imaging.
Collapse
Affiliation(s)
- Zhuozhi Dai
- Department of Radiology, 2nd Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada
| | - Sanjay Kalra
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Dennell Mah
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Peter Seres
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada
| | - Hongfu Sun
- School of Information Technology and Electrical Engineering, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Renhua Wu
- Department of Radiology, 2nd Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
| | - Alan H Wilman
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada.
| |
Collapse
|
32
|
Sun PZ. Quasi-steady state chemical exchange saturation transfer (QUASS CEST) analysis-correction of the finite relaxation delay and saturation time for robust CEST measurement. Magn Reson Med 2021; 85:3281-3289. [PMID: 33486816 DOI: 10.1002/mrm.28653] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE CEST provides a MR contrast mechanism sensitizing to the exchange between dilute labile and bulk water protons. However, the CEST effect depends on the RF saturation duration and relaxation delay, which need to be long to reach its steady state. Our study aims to estimate the QUAsi-Steady State (QUASS) CEST signal from experiments with shorter saturation and relaxation delay times. METHODS The evolution of the CEST signal was modeled as a function of the bulk water longitudinal relaxation rate during the relaxation delay (Td) and spin-lock relaxation rate during the RF saturation (Ts), from which the QUASS CEST effect is solved. Numeric simulations were programmed to compare the apparent CEST and QUASS CEST effects as a function of Ts and Td times. We also performed CEST MRI experiments from a creatine-gel pH phantom under serially varied Ts and Td times. RESULTS The numeric simulation showed that although the apparent CEST effect depends on Td and Ts, the QUASS CEST solution has little dependence. Phantom results showed that the routine CEST pH contrast could be described by a nonlinear regression model (ie, Δ C E S T R = Δ C E S T R eq app 1 - e - R 1 ρ app · t ). We had Δ C E S T R eq app = 3.90 ± 0.03 % (P < 5e-8) and R 1 ρ app = 0.62 ± 0.02 s - 1 (P < 5e-6). For the QUASS CEST analysis, we modeled the pH contrast as Δ C E S T R = Δ C E S T R eq QUASS + s · t , using a linear regression model. We had Δ C E S T R eq QUASS = 3.63 ± 0.01 % (P < 5e-9) and s = - 0.02 ± 0.00 % / s (P < 0.01), the slope of which is minimal. CONCLUSIONS The QUASS CEST algorithm provides a post-processing solution that facilitates robust CEST measurement.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
33
|
Berry DB, Englund EK, Chen S, Frank LR, Ward SR. Medical imaging of tissue engineering and regenerative medicine constructs. Biomater Sci 2021; 9:301-314. [PMID: 32776044 PMCID: PMC8262082 DOI: 10.1039/d0bm00705f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Advancement of tissue engineering and regenerative medicine (TERM) strategies to replicate tissue structure and function has led to the need for noninvasive assessment of key outcome measures of a construct's state, biocompatibility, and function. Histology based approaches are traditionally used in pre-clinical animal experiments, but are not always feasible or practical if a TERM construct is going to be tested for human use. In order to transition these therapies from benchtop to bedside, rigorously validated imaging techniques must be utilized that are sensitive to key outcome measures that fulfill the FDA standards for TERM construct evaluation. This review discusses key outcome measures for TERM constructs and various clinical- and research-based imaging techniques that can be used to assess them. Potential applications and limitations of these techniques are discussed, as well as resources for the processing, analysis, and interpretation of biomedical images.
Collapse
Affiliation(s)
- David B Berry
- Departments of NanoEngineering, University of California, San Diego, USA.
| | | | | | | | | |
Collapse
|
34
|
Liu G, van Zijl PC. CEST (Chemical Exchange Saturation Transfer) MR Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Wilk B, Wisenberg G, Dharmakumar R, Thiessen JD, Goldhawk DE, Prato FS. Hybrid PET/MR imaging in myocardial inflammation post-myocardial infarction. J Nucl Cardiol 2020; 27:2083-2099. [PMID: 31797321 PMCID: PMC7391987 DOI: 10.1007/s12350-019-01973-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 01/24/2023]
Abstract
Hybrid PET/MR imaging is an emerging imaging modality combining positron emission tomography (PET) and magnetic resonance imaging (MRI) in the same system. Since the introduction of clinical PET/MRI in 2011, it has had some impact (e.g., imaging the components of inflammation in myocardial infarction), but its role could be much greater. Many opportunities remain unexplored and will be highlighted in this review. The inflammatory process post-myocardial infarction has many facets at a cellular level which may affect the outcome of the patient, specifically the effects on adverse left ventricular remodeling, and ultimately prognosis. The goal of inflammation imaging is to track the process non-invasively and quantitatively to determine the best therapeutic options for intervention and to monitor those therapies. While PET and MRI, acquired separately, can image aspects of inflammation, hybrid PET/MRI has the potential to advance imaging of myocardial inflammation. This review contains a description of hybrid PET/MRI, its application to inflammation imaging in myocardial infarction and the challenges, constraints, and opportunities in designing data collection protocols. Finally, this review explores opportunities in PET/MRI: improved registration, partial volume correction, machine learning, new approaches in the development of PET and MRI pulse sequences, and the use of novel injection strategies.
Collapse
Affiliation(s)
- B Wilk
- Department of Medical Imaging, Western University, London, Canada.
- Lawson Health Research Institute, London, Canada.
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada.
| | - G Wisenberg
- Department of Medical Imaging, Western University, London, Canada
- MyHealth Centre, Arva, Canada
| | - R Dharmakumar
- Biomedical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - J D Thiessen
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| | - D E Goldhawk
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| | - F S Prato
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| |
Collapse
|
36
|
Shin SH, Wendland MF, Vandsburger MH. Delayed urea differential enhancement CEST (dudeCEST)-MRI with T 1 correction for monitoring renal urea handling. Magn Reson Med 2020; 85:2791-2804. [PMID: 33180343 DOI: 10.1002/mrm.28583] [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] [Received: 05/29/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE We demonstrate a method of delayed urea differential enhancement CEST for probing urea recycling action of the kidney using expanded multi-pool Lorentzian fitting and apparent exchange-dependent relaxation compensation. METHODS T1 correction of urea CEST contrast by apparent exchange-dependent relaxation was tested in phantoms. Nine mice were scanned at 7 Tesla following intraperitoneal injection of 2M 150 μL urea, and later saline. T1 maps and Z-spectra were acquired before and 20 and 40 min postinjection. Z-spectra were fit to a 7-pool Lorentzian model for CEST quantification and compared to urea assay of kidney homogenate. Renal injury was induced by aristolochic acid in 7 mice, and the same scan protocol was performed. RESULTS Apparent exchange-dependent relaxation corrected for variable T1 times in phantoms. Urea CEST contrast at +1 ppm increased significantly at both time points following urea injection in the inner medulla and papilla. When normalizing the postinjection urea CEST contrast to the corresponding baseline value, both urea and saline injection resulted in identical fold changes in urea CEST contrast. Urea assay of kidney homogenate showed a significant correlation to both apparent exchange-dependent relaxation (R2 = 0.4687, P = .0017) and non-T1 -corrected Lorentzian amplitudes (R2 = 0.4964, P = .0011). Renal injury resulted in increased T1 time in the cortex and reduced CEST contrast change upon urea and saline infusion. CONCLUSION Delayed urea enhancement following infusion can provide insight into renal urea handling. In addition, changes in CEST contrast at 1.0 ppm following saline infusion may provide insight into renal function.
Collapse
Affiliation(s)
- Soo Hyun Shin
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Michael F Wendland
- Berkeley Preclinical Imaging Core (BPIC), University of California, Berkeley, Berkeley, California, USA
| | - Moriel H Vandsburger
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
37
|
Guo C, Wu J, Rosenberg JT, Roussel T, Cai S, Cai C. Fast chemical exchange saturation transfer imaging based on PROPELLER acquisition and deep neural network reconstruction. Magn Reson Med 2020; 84:3192-3205. [PMID: 32602965 DOI: 10.1002/mrm.28376] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE To develop a method for fast chemical exchange saturation transfer (CEST) imaging. METHODS The periodically rotated overlapping parallel lines enhanced reconstruction (PROPELLER) sampling scheme was introduced to shorten the acquisition time. Deep neural network was employed to reconstruct CEST contrast images. Numerical simulation and experiments on a creatine phantom, hen egg, and in vivo tumor rat brain were performed to test the feasibility of this method. RESULTS The results from numerical simulation and experiments show that there is no significant difference between reference images and CEST-PROPELLER reconstructed images under an acceleration factor of 8. CONCLUSION Although the deep neural network is trained entirely on synthesized data, it works well on reconstructing experimental data. The proof of concept study demonstrates that the combination of the PROPELLER sampling scheme and the deep neural network enables considerable acceleration of saturated image acquisition and may find applications in CEST MRI.
Collapse
Affiliation(s)
- Chenlu Guo
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jian Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jens T Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | - Tangi Roussel
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Congbo Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| |
Collapse
|
38
|
Abstract
OBJECTIVES The goal of this study was to demonstrate feasibility of measuring extracellular pH in cartilage and meniscus using acidoCEST technique with a 3-dimensional ultrashort echo time readout (acidoCEST-UTE) magnetic resonance imaging (MRI). MATERIALS AND METHODS Magnetization transfer ratio asymmetry, radiofrequency (RF) power mismatch, and relative saturation transfer were evaluated in liquid phantoms for iopromide, iopamidol, and iohexol over a pH range of 6.2 to 7.8, at various agent concentrations, temperatures, and buffer concentrations. Tissue phantoms containing cartilage and meniscus were evaluated with the same considerations for iopamidol and iohexol. Phantoms were imaged with the acidoCEST-UTE MRI sequence at 3 T. Correlation coefficients and coefficients of variations were calculated. Paired Wilcoxon rank-sum tests were used to evaluate for statistically significant differences. RESULTS The RF power mismatch and relative saturation transfer analyses of liquid phantoms showed iopamidol and iohexol to be the most promising agents for this study. Both these agents appeared to be concentration independent and feasible for use with or without buffer and at physiologic temperature over a pH range of 6.2 to 7.8. Ultimately, RF power mismatch fitting of iohexol showed the strongest correlation coefficients between cartilage, meniscus, and fluid. In addition, ratiometric values for iohexol are similar among liquid as well as different tissue types. CONCLUSIONS Measuring extracellular pH in cartilage and meniscus using acidoCEST-UTE MRI is feasible.
Collapse
|
39
|
Tang Y, Xiao G, Shen Z, Zhuang C, Xie Y, Zhang X, Yang Z, Guan J, Shen Y, Chen Y, Lai L, Chen Y, Chen S, Dai Z, Wang R, Wu R. Noninvasive Detection of Extracellular pH in Human Benign and Malignant Liver Tumors Using CEST MRI. Front Oncol 2020; 10:578985. [PMID: 33224880 PMCID: PMC7667286 DOI: 10.3389/fonc.2020.578985] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE In this study, we aimed to use 3T magnetic resonance imaging (MRI), which is clinically available, to determine the extracellular pH (pHe) of liver tumors and prospectively evaluate the ability of chemical exchange saturation transfer (CEST) MRI to distinguish between benign and malignant liver tumors. METHODS Different radiofrequency irradiation schemes were assessed for ioversol-based pH measurements at 3T. CEST effects were quantified in vitro using the asymmetric magnetization transfer ratio (MTRasym) at 4.3 ppm from the corrected Z spectrum. Generalized ratiometric analysis was conducted by rationing resolved ioversol CEST effects at 4.3 ppm at a flip angle of 60 and 350°. Fifteen patients recently diagnosed with hepatic carcinoma and five patients diagnosed with hepatic hemangioma [1 male; mean age, 48.6 (range, 37-59) years] were assessed. RESULTS By conducting dual-power CEST MRI, the pH of solutions was determined to be 6.0-7.2 at 3T in vitro. In vivo, ioversol signal intensities in the tumor region showed that the extracellular pH in hepatic carcinoma was acidic(mean ± standard deviation, 6.66 ± 0.19), whereas the extracellular pH was more physiologically neutral in hemangioma (mean ± standard deviation, 7.34 ± 0.09).The lesion size was similar between CEST pH MRI and T2-weighted imaging. CONCLUSION dual-power CEST MRI can detect extracellular pH in human liver tumors and can provide molecular-level diagnostic tools for differentiating benign and malignant liver tumors at 3T.
Collapse
Affiliation(s)
- Yanyan Tang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Department of Medical Imaging, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Gang Xiao
- Department of Mathematics and Statistics, Hanshan Normal University, Chaozhou, China
| | | | - Caiyu Zhuang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yudan Xie
- Department of General Surgery, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xiaolei Zhang
- College of Air Traffic Management, Civil Aviation Flight University of China, Guanghan, China
| | - Zhongxian Yang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Jitian Guan
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yuanyu Shen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yanzi Chen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Lihua Lai
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yuanfeng Chen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Shuo Chen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhuozhi Dai
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Runrun Wang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Renhua Wu
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Provincial Key Laboratory of Medical Molecular Imaging, Shantou, China
- *Correspondence: Renhua Wu,
| |
Collapse
|
40
|
Extracellular acidosis differentiates pancreatitis and pancreatic cancer in mouse models using acidoCEST MRI. Neoplasia 2019; 21:1085-1090. [PMID: 31734629 PMCID: PMC6888716 DOI: 10.1016/j.neo.2019.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 01/07/2023] Open
Abstract
Differentiating pancreatitis from pancreatic cancer would improve diagnostic specificity, and prognosticating pancreatitis that progresses to pancreatic cancer would also improve diagnoses of pancreas pathology. The high glycolytic metabolism of pancreatic cancer can cause tumor acidosis, and different levels of pancreatitis may also have different levels of acidosis, so that extracellular acidosis may be a diagnostic biomarker for these pathologies. AcidoCEST MRI can noninvasively measure extracellular pH (pHe) in the pancreas and pancreatic tissue. We used acidoCEST MRI to measure pHe in a KC model treated with caerulein, which causes pancreatitis followed by development of pancreatic cancer. We also evaluated the KC model treated with PBS, and wild-type mice treated with caerulein or PBS as controls. The caerulein-treated KC cohort had lower pHe of 6.85–6.92 before and during the first 48 h after initiating treatment, relative to a pHe of 6.92 to 7.05 pHe units for the other cohorts. The pHe of the caerulein-treated KC cohort decreased to 6.79 units at 5 weeks when pancreatic tumors were detected with anatomical MRI, and sustained a pHe of 6.75 units at the 8-week time point. Histopathology was used to evaluate and validate the presence of tumors and inflammation in each cohort. These results showed that acidoCEST MRI can differentiate pancreatic cancer from pancreatitis in this mouse model, but does not appear to differentiate pancreatitis that progresses to pancreatic cancer vs. pancreatitis that does not progress to cancer.
Collapse
|
41
|
Goldenberg JM, Pagel MD. Assessments of tumor metabolism with CEST MRI. NMR IN BIOMEDICINE 2019; 32:e3943. [PMID: 29938857 PMCID: PMC7377947 DOI: 10.1002/nbm.3943] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 05/06/2023]
Abstract
Chemical exchange saturation transfer (CEST) is a relatively new contrast mechanism for MRI. CEST MRI exploits a specific MR frequency (chemical shift) of a molecule while generating an image with good spatial resolution using standard MRI techniques, combining the specificity of MRS with the spatial resolution of MRI. Many CEST MRI acquisition methods have been developed to improve analyses of tumor metabolism. GluCEST, CrCEST, and LATEST can map glutamate, creatine, and lactate, which are important metabolites involved in tumor metabolism. GlucoCEST MRI tracks the pharmacokinetics of glucose transport and cell internalization within tumors. CatalyCEST MRI detects enzyme catalysis that changes a substrate CEST agent. AcidoCEST MRI measures extracellular pH of the tumor microenvironment by exploiting a ratio of two pH-dependent CEST signals. This review describes each technique, the technical issues involved with CEST MRI and each specific technique, and the merits and challenges associated with applying each CEST MRI technique to study tumor metabolism.
Collapse
Affiliation(s)
- Joshua M. Goldenberg
- Department of Pharmaceutical Sciences, The University of Arizona, Tucson, AZ, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark D. Pagel
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
42
|
Lindeman LR, Jones KM, High RA, Howison CM, Shubitz LF, Pagel MD. Differentiating lung cancer and infection based on measurements of extracellular pH with acidoCEST MRI. Sci Rep 2019; 9:13002. [PMID: 31506562 PMCID: PMC6736855 DOI: 10.1038/s41598-019-49514-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/27/2019] [Indexed: 01/17/2023] Open
Abstract
Lung cancer diagnosis via imaging may be confounded by the presence of indolent infectious nodules in imaging studies. This issue is pervasive in the southwestern US where coccidioidomycosis (Valley Fever) is endemic. AcidoCEST MRI is a noninvasive imaging method that quantifies the extracellular pH (pHe) of tissues in vivo, allowing tumor acidosis to be used as a diagnostic biomarker. Using murine models of lung adenocarcinoma and coccidoidomycosis, we found that average lesion pHe differed significantly between tumors and granulomas. Our study shows that acidoCEST MRI is a promising tool for improving the specificity of lung cancer diagnosis.
Collapse
Affiliation(s)
- Leila R Lindeman
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Kyle M Jones
- Bioengineering Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Rachel A High
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | | | - Lisa F Shubitz
- Valley Fever Center for Excellence, University of Arizona, Tucson, AZ, USA
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
deSouza NM, Achten E, Alberich-Bayarri A, Bamberg F, Boellaard R, Clément O, Fournier L, Gallagher F, Golay X, Heussel CP, Jackson EF, Manniesing R, Mayerhofer ME, Neri E, O'Connor J, Oguz KK, Persson A, Smits M, van Beek EJR, Zech CJ. Validated imaging biomarkers as decision-making tools in clinical trials and routine practice: current status and recommendations from the EIBALL* subcommittee of the European Society of Radiology (ESR). Insights Imaging 2019; 10:87. [PMID: 31468205 PMCID: PMC6715762 DOI: 10.1186/s13244-019-0764-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Observer-driven pattern recognition is the standard for interpretation of medical images. To achieve global parity in interpretation, semi-quantitative scoring systems have been developed based on observer assessments; these are widely used in scoring coronary artery disease, the arthritides and neurological conditions and for indicating the likelihood of malignancy. However, in an era of machine learning and artificial intelligence, it is increasingly desirable that we extract quantitative biomarkers from medical images that inform on disease detection, characterisation, monitoring and assessment of response to treatment. Quantitation has the potential to provide objective decision-support tools in the management pathway of patients. Despite this, the quantitative potential of imaging remains under-exploited because of variability of the measurement, lack of harmonised systems for data acquisition and analysis, and crucially, a paucity of evidence on how such quantitation potentially affects clinical decision-making and patient outcome. This article reviews the current evidence for the use of semi-quantitative and quantitative biomarkers in clinical settings at various stages of the disease pathway including diagnosis, staging and prognosis, as well as predicting and detecting treatment response. It critically appraises current practice and sets out recommendations for using imaging objectively to drive patient management decisions.
Collapse
Affiliation(s)
- Nandita M deSouza
- Cancer Research UK Imaging Centre, The Institute of Cancer Research and The Royal Marsden Hospital, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | | | | | - Fabian Bamberg
- Department of Radiology, University of Freiburg, Freiburg im Breisgau, Germany
| | | | | | | | | | | | - Claus Peter Heussel
- Universitätsklinik Heidelberg, Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
| | - Edward F Jackson
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rashindra Manniesing
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525, GA, Nijmegen, The Netherlands
| | | | - Emanuele Neri
- Department of Translational Research, University of Pisa, Pisa, Italy
| | - James O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | | | - Marion Smits
- Department of Radiology and Nuclear Medicine (Ne-515), Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Edwin J R van Beek
- Edinburgh Imaging, Queen's Medical Research Institute, Edinburgh Bioquarter, 47 Little France Crescent, Edinburgh, UK
| | - Christoph J Zech
- University Hospital Basel, Radiology and Nuclear Medicine, University of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| |
Collapse
|
44
|
Spatiotemporal pH Heterogeneity as a Promoter of Cancer Progression and Therapeutic Resistance. Cancers (Basel) 2019; 11:cancers11071026. [PMID: 31330859 PMCID: PMC6678451 DOI: 10.3390/cancers11071026] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of pH in solid tumors is a hallmark of cancer. In recent years, the role of altered pH heterogeneity in space, between benign and aggressive tissues, between individual cancer cells, and between subcellular compartments, has been steadily elucidated. Changes in temporal pH-related processes on both fast and slow time scales, including altered kinetics of bicarbonate-CO2 exchange and its effects on pH buffering and gradual, progressive changes driven by changes in metabolism, are further implicated in phenotypic changes observed in cancers. These discoveries have been driven by advances in imaging technologies. This review provides an overview of intra- and extracellular pH alterations in time and space reflected in cancer cells, as well as the available technology to study pH spatiotemporal heterogeneity.
Collapse
|
45
|
Cai J, Wu J, Guo C, Cai S, Cai C. Ultrafast multi-slice chemical exchange saturation transfer imaging scheme based on segmented spatiotemporal encoding. Magn Reson Imaging 2019; 60:122-129. [PMID: 30953697 DOI: 10.1016/j.mri.2019.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/25/2022]
Abstract
Chemical exchange saturation transfer (CEST) imaging is an important magnetic resonance molecular imaging technology. However, long acquisition time limits its clinical application, especially when multi-slice CEST imaging is needed. Though single-shot EPI can be used to accelerate CEST imaging, images are often distorted under inhomogeneous magnetic fields. In this work, we propose a new method called CEST-SeSPEN for ultrafast multi-slice CEST imaging based on segmented spatiotemporally encoded (SeSPEN) MRI. Experiments were performed on creatine phantom and hen egg. The results show that CEST-SeSPEN can provide good CEST contrast images. Its acquisition time is much shorter than other multi-slice CEST methods currently available. It may be used in challenging situation where high temporal resolution and robustness to field inhomogeneity are vital.
Collapse
Affiliation(s)
- Jizhou Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jian Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Chenlu Guo
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China.
| | - Congbo Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China.
| |
Collapse
|
46
|
Zhou J, Heo HY, Knutsson L, van Zijl PCM, Jiang S. APT-weighted MRI: Techniques, current neuro applications, and challenging issues. J Magn Reson Imaging 2019; 50:347-364. [PMID: 30663162 DOI: 10.1002/jmri.26645] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/26/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Amide proton transfer-weighted (APTw) imaging is a molecular MRI technique that generates image contrast based predominantly on the amide protons in mobile cellular proteins and peptides that are endogenous in tissue. This technique, the most studied type of chemical exchange saturation transfer imaging, has been used successfully for imaging of protein content and pH, the latter being possible due to the strong dependence of the amide proton exchange rate on pH. In this article we briefly review the basic principles and recent technical advances of APTw imaging, which is showing promise clinically, especially for characterizing brain tumors and distinguishing recurrent tumor from treatment effects. Early applications of this approach to stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and traumatic brain injury are also illustrated. Finally, we outline the technical challenges for clinical APT-based imaging and discuss several controversies regarding the origin of APTw imaging signals in vivo. Level of Evidence: 3 Technical Efficacy Stage: 3 J. Magn. Reson. Imaging 2019;50:347-364.
Collapse
Affiliation(s)
- Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Linda Knutsson
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Peter C M van Zijl
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
47
|
|
48
|
Ferrauto G, Di Gregorio E, Auboiroux V, Petit M, Berger F, Aime S, Lahrech H. CEST-MRI for glioma pH quantification in mouse model: Validation by immunohistochemistry. NMR IN BIOMEDICINE 2018; 31:e4005. [PMID: 30256478 DOI: 10.1002/nbm.4005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 06/08/2023]
Abstract
In glioma, the acidification of the extracellular tumor microenvironment drives proliferation, angiogenesis, immunosuppression, invasion and chemoresistance. Therefore, quantification of glioma extracellular pH (pHe) is of crucial importance. This study is focused on the application of the YbHPDO3A (ytterbium 1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane) probe for in vivo glioma pHe quantification using chemical exchange saturation transfer (CEST)-MRI and its correlation with tumor metabolism assessed by immunohistochemistry. The U87 glioma mouse model was used (n = 18) and MRI performed at 4.7 T. CEST-MRI of YbHPDO3A solutions at different pH values showed two resolved CEST spectra at 71 ppm and 99 ppm, both sensitive to pH variations, allowing therefore calculation of the ratiometric curve for in vivo pH quantification. In vivo MRI sequences consisted of T2w for tumor localization, T2w * to assess YbHPDO3A biodistribution by exploiting its magnetic susceptibility effect and CEST for glioma pHe mapping. T2w * images show that YbHPDO3A extravasates in tumor in regions with damaged blood-brain barrier. The pHe is calculated only in these regions. Hematoxylin/eosin histology and Ki-67, CA-IX (carbonic anhydrase 9) and NHE-1 immunohistochemical staining were performed; their expression rates were compared with the in vivo pHe values. On the basis of the cell proliferation marker Ki-67, two groups were defined: one group with a lower mitotic index (MI% < 20% = mean value) and a mean pHe value of 7.00 (low-proliferation/high-pH group) and the other with MI% > 20% and an acidic pHe of 6.6 (high-proliferation/low-pH group). CA-IX and NHE-1 were over-expressed in the high-proliferation/low-pH group (CA-IX, 92 ± 7% versus 30 ± 13%; NHE-1, 84 ± 8% versus 35 ± 11%), indicating an acidic/hypoxic microenvironment. These immunohistochemical results are consistent with our pHe mapping (Pearson correlation coefficient > 0.70) and provide evidence for the feasibility of the CEST-MRI method with the YbHPDO3A probe for glioma pHe quantification at 4.7 T. Importantly, the YbHPDO3A probe has similar chemical and biological properties to the clinically approved MRI contrast agent GdHPDO3A. This makes the method promising for a clinical translation.
Collapse
Affiliation(s)
- Giuseppe Ferrauto
- Molecular Imaging Center, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Turin, Italy
| | - Enza Di Gregorio
- Molecular Imaging Center, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Turin, Italy
| | | | - Manuel Petit
- BrainTech Lab-INSERM U12O5-University of Grenoble Alpes, Grenoble, France
| | - François Berger
- BrainTech Lab-INSERM U12O5-University of Grenoble Alpes, Grenoble, France
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Turin, Italy
| | - Hana Lahrech
- BrainTech Lab-INSERM U12O5-University of Grenoble Alpes, Grenoble, France
| |
Collapse
|
49
|
Shazeeb MS, Corazzini R, Konowicz PA, Fogle R, Bangari DS, Johnson J, Ying X, Dhal PK. Assessment of in vivo degradation profiles of hyaluronic acid hydrogels using temporal evolution of chemical exchange saturation transfer (CEST) MRI. Biomaterials 2018; 178:326-338. [PMID: 29861090 DOI: 10.1016/j.biomaterials.2018.05.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/19/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022]
Abstract
Hyaluronic acid (HA) hydrogels have found a wide range of applications in biomedicine: regenerative medicine to drug delivery applications. In vivo quantitative assessment of these hydrogels using magnetic resonance imaging (MRI) provides an effective, accurate, safe, and non-invasive translational approach to assess the biodegradability of HA hydrogels. Chemical exchange saturation transfer (CEST) is an MRI contrast enhancement technique that overcomes the concentration limitation of other techniques like magnetic resonance spectroscopy (MRS) by detecting metabolites at up to two orders of magnitude or higher. In this study, HA hydrogels were synthesized based on different crosslinking agents and assessed using CEST MRI to investigate the in vivo degradation profiles of these gels in a mouse subcutaneous injection model over a three-month period. Nature of crosslinking agents was found to influence their degradation profiles. Since CEST MRI provides a unique chemical signature to visualize HA hydrogels, our studies proved that this technique could be used as a guide in the hydrogel optimization process for drug delivery and regenerative medicine applications.
Collapse
Affiliation(s)
| | - Rubina Corazzini
- Diabetes Research, Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02451, USA
| | - Paul A Konowicz
- Diabetes Research, Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02451, USA
| | - Robert Fogle
- Bioimaging Research, Sanofi Global R&D, 49 New York Avenue, Framingham, MA 01701, USA
| | - Dinesh S Bangari
- Pathology Research, Sanofi Global R&D, 5 Mountain Road, Framingham, MA 01701, USA
| | - Jennifer Johnson
- Pathology Research, Sanofi Global R&D, 5 Mountain Road, Framingham, MA 01701, USA
| | - Xiaoyou Ying
- Bioimaging Research, Sanofi Global R&D, 49 New York Avenue, Framingham, MA 01701, USA.
| | - Pradeep K Dhal
- Diabetes Research, Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02451, USA.
| |
Collapse
|