51
|
In vivo hypoxia characterization using blood oxygen level dependent magnetic resonance imaging in a preclinical glioblastoma mouse model. Magn Reson Imaging 2020; 76:52-60. [PMID: 33220448 DOI: 10.1016/j.mri.2020.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/23/2020] [Accepted: 11/12/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE Hypoxia measurements can provide crucial information regarding tumor aggressiveness, however current preclinical approaches are limited. Blood oxygen level dependent (BOLD) Magnetic Resonance Imaging (MRI) has the potential to continuously monitor tumor pathophysiology (including hypoxia). The aim of this preliminary work was to develop and evaluate BOLD MRI followed by post-image analysis to identify regions of hypoxia in a murine glioblastoma (GBM) model. METHODS A murine orthotopic GBM model (GL261-luc2) was used and independent images were generated from multiple slices in four different mice. Image slices were randomized and split into training and validation cohorts. A 7 T MRI was used to acquire anatomical images using a fast-spin-echo (FSE) T2-weighted sequence. BOLD images were taken with a T2*-weighted gradient echo (GRE) and an oxygen challenge. Thirteen images were evaluated in a training cohort to develop the MRI sequence and optimize post-image analysis. An in-house MATLAB code was used to evaluate MR images and generate hypoxia maps for a range of thresholding and ΔT2* values, which were compared against respective pimonidazole sections to optimize image processing parameters. The remaining (n = 6) images were used as a validation group. Following imaging, mice were injected with pimonidazole and collected for immunohistochemistry (IHC). A test of correlation (Pearson's coefficient) and agreement (Bland-Altman plot) were conducted to evaluate the respective MRI slices and pimonidazole IHC sections. RESULTS For the training cohort, the optimized parameters of "thresholding" (20 ≤ T2* ≤ 35 ms) and ΔT2* (±4 ms) yielded a Pearson's correlation of 0.697. These parameters were applied to the validation cohort confirming a strong Pearson's correlation (0.749) when comparing the respective analyzed MR and pimonidazole images. CONCLUSION Our preliminary study supports the hypothesis that BOLD MRI is correlated with pimonidazole measurements of hypoxia in an orthotopic GBM mouse model. This technique has further potential to monitor hypoxia during tumor development and therapy.
Collapse
|
52
|
Swartz HM, Flood AB, Schaner PE, Halpern H, Williams BB, Pogue BW, Gallez B, Vaupel P. How best to interpret measures of levels of oxygen in tissues to make them effective clinical tools for care of patients with cancer and other oxygen-dependent pathologies. Physiol Rep 2020; 8:e14541. [PMID: 32786045 PMCID: PMC7422807 DOI: 10.14814/phy2.14541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
It is well understood that the level of molecular oxygen (O2 ) in tissue is a very important factor impacting both physiology and pathological processes as well as responsiveness to some treatments. Data on O2 in tissue could be effectively utilized to enhance precision medicine. However, the nature of the data that can be obtained using existing clinically applicable techniques is often misunderstood, and this can confound the effective use of the information. Attempts to make clinical measurements of O2 in tissues will inevitably provide data that are aggregated over time and space and therefore will not fully represent the inherent heterogeneity of O2 in tissues. Additionally, the nature of existing techniques to measure O2 may result in uneven sampling of the volume of interest and therefore may not provide accurate information on the "average" O2 in the measured volume. By recognizing the potential limitations of the O2 measurements, one can focus on the important and useful information that can be obtained from these techniques. The most valuable clinical characterizations of oxygen are likely to be derived from a series of measurements that provide data about factors that can change levels of O2 , which then can be exploited both diagnostically and therapeutically. The clinical utility of such data ultimately needs to be verified by careful studies of outcomes related to the measured changes in levels of O2 .
Collapse
Affiliation(s)
- Harold M Swartz
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Ann Barry Flood
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
| | - Philip E Schaner
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Howard Halpern
- Department Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Benjamin B Williams
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Bernard Gallez
- Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Peter Vaupel
- Department Radiation Oncology, University Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Center Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
53
|
Gehrung M, Tomaszewski M, McIntyre D, Disselhorst J, Bohndiek S. Co-registration of optoacoustic tomography and magnetic resonance imaging data from murine tumour models. PHOTOACOUSTICS 2020; 18:100147. [PMID: 32042588 PMCID: PMC6997898 DOI: 10.1016/j.pacs.2019.100147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/26/2019] [Accepted: 11/08/2019] [Indexed: 05/11/2023]
Abstract
As optoacoustic tomography (OT) emerges as a mainstream pre-clinical imaging modality, understanding the relationship between optoacoustic and other imaging biomarkers in the context of the underlying tissue biology becomes vitally important. Complementary insight into tumour vasculature and hypoxia can be gained using OT alongside magnetic resonance imaging (MRI)-based techniques. To evaluate the relationship between these metrics and the relative performance of the two modalities in assessment of tumour physiology, co-registration of their output imaging data is required. Unfortunately, this poses a significant challenge due to differences in animal positioning during imaging. Here, we present an integrated framework for registration of OT and MR image data in mice. Our framework combines a novel MR animal holder, to improve animal positioning during imaging, and a landmark-based software co-registration algorithm. We demonstrate that our protocol significantly improves registration of both body and tumour contours between these modalities, enabling more precise multi-modal tumour characterisation.
Collapse
Affiliation(s)
- Marcel Gehrung
- Department of Physics, University of Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
- Werner Siemens Imaging Center, Preclinical Imaging and Radiopharmacy, University of Tuebingen, Germany
| | - Michal Tomaszewski
- Department of Physics, University of Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | | | - Jonathan Disselhorst
- Werner Siemens Imaging Center, Preclinical Imaging and Radiopharmacy, University of Tuebingen, Germany
| | - Sarah Bohndiek
- Department of Physics, University of Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| |
Collapse
|
54
|
Yang Z, Li L, Jin AJ, Huang W, Chen X. Rational design of semiconducting polymer brushes as cancer theranostics. MATERIALS HORIZONS 2020; 7:1474-1494. [PMID: 33777400 PMCID: PMC7990392 DOI: 10.1039/d0mh00012d] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Photonic theranostics (PTs) generally contain optical agents for the optical sensing of biomolecules and therapeutic components for converting light into heat or chemical energy. Semiconducting polymer nanoparticles (SPNs) as advanced PTs possessing good biocompatibility, stable photophysical properties, and sensitive and tunable optical responses from the ultraviolet to near-infrared (NIR) II window (300-1700 nm) have recently aroused great interest. Although semiconducting polymers (SPs) with various building blocks have been synthesized and developed to meet the demands of biophotonic applications, most of the SPNs were made by a nanoprecipitation method that used amphiphilic surfactants to encapsulate SPs. Such binary SP micelles usually exhibit weakened photophysical properties of SPs and undergo dissociation in vivo. SP brushes (SPBs) are products of functional post-modification of SP backbones, which endows unique features to SPNs (e.g. enhanced optical properties and multiple chemical reaction sites for the conjunction of organic/inorganic imaging agents and therapeutics). Furthermore, the SPB-based SPNs can be highly stable due to supramolecular self-assembly and/or chemical crosslinking. In this review, we highlight the recent progress in the development of SPBs for advanced theranostics.
Collapse
Affiliation(s)
- Zhen Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ling Li
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Albert J. Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Wei Huang
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi’an 710072, Shaanxi, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
55
|
Qian J, Yu X, Li B, Fei Z, Huang X, Luo P, Zhang L, Zhang Z, Lou J, Wang H. In vivo Monitoring of Oxygen Levels in Human Brain Tumor Between Fractionated Radiotherapy Using Oxygen-enhanced MR Imaging. Curr Med Imaging 2020; 16:427-432. [PMID: 32410542 DOI: 10.2174/1573405614666180925144814] [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: 06/22/2018] [Revised: 08/19/2018] [Accepted: 09/11/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND It was known that the response of tumor cells to radiation is closely related to tissue oxygen level and fractionated radiotherapy allows reoxygenation of hypoxic tumor cells. Non-invasive mapping of tissue oxygen level may hold great importance in clinic. OBJECTIVE The aim of this study is to evaluate the role of oxygen-enhanced MR imaging in the detection of tissue oxygen levels between fractionated radiotherapy. METHODS A cohort of 10 patients with brain metastasis was recruited. Quantitative oxygen enhanced MR imaging was performed prior to, 30 minutes and 22 hours after first fractionated radiotherapy. RESULTS The ΔR1 (the difference of longitudinal relaxivity between 100% oxygen breathing and air breathing) increased in the ipsilateral tumor site and normal tissue by 242% and 152%, respectively, 30 minutes after first fractionated radiation compared to pre-radiation levels. Significant recovery of ΔR1 in the contralateral normal tissue (p < 0.05) was observed 22 hours compared to 30 minutes after radiation levels. CONCLUSION R1-based oxygen-enhanced MR imaging may provide a sensitive endogenous marker for oxygen changes in the brain tissue between fractionated radiotherapy.
Collapse
Affiliation(s)
- Junchao Qian
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiang Yu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Bingbing Li
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Zhenle Fei
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Xiang Huang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Peng Luo
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Liwei Zhang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Zhiming Zhang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Jianjun Lou
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Hongzhi Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
56
|
Waschkies CF, Pfiffner FK, Heuberger DM, Schneider MA, Tian Y, Wolint P, Calcagni M, Giovanoli P, Buschmann J. Tumor grafts grown on the chicken chorioallantoic membrane are distinctively characterized by MRI under functional gas challenge. Sci Rep 2020; 10:7505. [PMID: 32371865 PMCID: PMC7200801 DOI: 10.1038/s41598-020-64290-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/14/2020] [Indexed: 11/09/2022] Open
Abstract
Recently, a tumor model based on the chorioallantoic membrane (CAM) was characterized structurally with Magnetic Resonance Imaging (MRI). Yet, capability of MRI to assess vascular functional reserve and potential of oxygenation-sensitive MRI remain largely unexplored in this model. For this purpose, we compared MC-38 colon and A549 lung adenocarcinoma cell grafts grown on the CAM, using quantitative T1 and T2* MRI readouts as imaging markers. These are associated with vascular functionality and oxygenation status when compared between periods of air and carbogen exposure. Our data show that in A549 lung adenocarcinoma cell grafts T2* values increased significantly upon carbogen exposure (p < 0.004, Wilcoxon test; no change in T1), while MC-38 grafts displayed no changes in T1 and T2*), indicating that the grafts differ in their vascular response. Heterogeneity with regard to T1 and T2* distribution within the grafts was noted. MC-38 grafts displayed larger T1 and T2* in the graft centre, while in A549 they were distributed more towards the graft surface. Finally, qualitative assessment of gadolinium-enhancement suggests that A549 grafts display more prominent enhancement compared to MC-38 grafts. Furthermore, MC-38 grafts had 65% larger volumes than A549 grafts. Histology revealed distinct underlying phenotypes of the two tumor grafts, pertaining to the proliferative status (Ki-67) and cellularity (H&E). In sum, a functional gas challenge with carbogen is feasible through gas exchange on the CAM, and it affects MRI signals associated with vascular reactivity and oxygenation status of the tumor graft planted on the CAM. Different grafts based on A549 lung adenocarcinoma and MC-38 colon carcinoma cell lines, respectively, display distinct phenotypes that can be distinguished and characterized non-invasively in ovo using MRI in the living chicken embryo.
Collapse
Affiliation(s)
- Conny F Waschkies
- Center for Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | | | - Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Marcel A Schneider
- Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Yinghua Tian
- Visceral and Transplant Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Petra Wolint
- Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Calcagni
- Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Pietro Giovanoli
- Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Johanna Buschmann
- Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
57
|
Richardson PJ, Ottaviani S, Prelle A, Stebbing J, Casalini G, Corbellino M. CNS penetration of potential anti-COVID-19 drugs. J Neurol 2020; 267:1880-1882. [PMID: 32361836 PMCID: PMC7195609 DOI: 10.1007/s00415-020-09866-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/27/2022]
Affiliation(s)
| | - Silvia Ottaviani
- Department of Surgery and Cancer, Imperial College, London, W12 0NN, UK.
| | - Alessandro Prelle
- Department of Neurology and Stroke Unit, ASST Ovest Milanese, Milan, Italy
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College, London, W12 0NN, UK
| | - Giacomo Casalini
- III Division of Infectious Diseases, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Mario Corbellino
- III Division of Infectious Diseases, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
58
|
Bluemke E, Bulte D, Bertrand A, George B, Cooke R, Chu KY, Durrant L, Goh V, Jacobs C, Ng SM, Strauss VY, Hawkins MA, Muirhead R. Oxygen-enhanced MRI MOLLI T1 mapping during chemoradiotherapy in anal squamous cell carcinoma. Clin Transl Radiat Oncol 2020; 22:44-49. [PMID: 32211520 PMCID: PMC7082428 DOI: 10.1016/j.ctro.2020.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/01/2020] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxygen-enhanced magnetic resonance imaging (MRI) and T1-mapping was used to explore its effectiveness as a prognostic imaging biomarker for chemoradiotherapy outcome in anal squamous cell carcinoma. MATERIALS AND METHODS T2-weighted, T1 mapping, and oxygen-enhanced T1 maps were acquired before and after 8-10 fractions of chemoradiotherapy and examined whether the oxygen-enhanced MRI response relates to clinical outcome. Patient response to treatment was assessed 3 months following completion of chemoradiotherapy. A mean T1 was extracted from manually segmented tumour regions of interest and a paired two-tailed t-test was used to compare changes across the patient population. Regions of subcutaneous fat and muscle tissue were examined as control ROIs. RESULTS There was a significant increase in T1 of the tumour ROIs across patients following the 8-10 fractions of chemoradiotherapy (paired t-test, p < 0.001, n = 7). At baseline, prior to receiving chemoradiotherapy, there were no significant changes in T1 across patients from breathing oxygen (n = 9). In the post-chemoRT scans (8-10 fractions), there was a significant decrease in T1 of the tumour ROIs across patients when breathing 100% oxygen (paired t-test, p < 0.001, n = 8). Out of the 12 patients from which we successfully acquired a visit 1 T1-map, only 1 patient did not respond to treatment, therefore, we cannot correlate these results with clinical outcome. CONCLUSIONS These clinical data demonstrate feasibility and potential for T1-mapping and oxygen enhanced T1-mapping to indicate perfusion or treatment response in tumours of this nature. These data show promise for future work with a larger cohort containing more non-responders, which would allow us to relate these measurements to clinical outcome.
Collapse
Affiliation(s)
- Emma Bluemke
- Institute of Biomedical Engineering, University of Oxford, UK
| | - Daniel Bulte
- Institute of Biomedical Engineering, University of Oxford, UK
| | - Ambre Bertrand
- Institute of Biomedical Engineering, University of Oxford, UK
| | - Ben George
- Institute of Biomedical Engineering, University of Oxford, UK
| | - Rosie Cooke
- Radiotherapy Department, Oxford University Hospitals NHS Foundation Trust, UK
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | - Kwun-Ye Chu
- Radiotherapy Department, Oxford University Hospitals NHS Foundation Trust, UK
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | - Lisa Durrant
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | - Vicky Goh
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | - Clare Jacobs
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, UK
| | - Stasya M. Ng
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | | | | | - Rebecca Muirhead
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, UK
| |
Collapse
|
59
|
Toward noninvasive quantification of adipose tissue oxygenation with MRI. Int J Obes (Lond) 2020; 44:1776-1783. [PMID: 32231252 DOI: 10.1038/s41366-020-0567-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Molecular oxygen (O2) plays a key role in normal and pathological adipose tissue function, yet technologies to measure its role in adipose tissue function are limited. O2 is paramagnetic and, in principle, directly influences the magnetic resonance (MR) 1H longitudinal relaxation rate constant of lipids, R1; thus, we hypothesize that MR imaging (MRI) can directly measure adipose O2 via a simple measure of R1. METHODS R1 was measured in a 4.7T preclinical MRI system at discrete oxygen partial pressure (pO2) levels. These measures were made in vitro in an idealized system and in vivo in subcutaneous and visceral white adipose of rodents. pO2 was determined using an invasive fiber-optic oxygen monitor. From the MRI and fiber optic data we determined the "relaxivity" of O2 in lipid, a critical parameter in converting the MRI-based R1 measurement into pO2. We used breathing gas challenge to estimate the changes in lipid pO2 (ΔpO2). RESULTS The relaxivity of O2 in lipid was determined to be 1.7·10-3 ± 4·10-4 mmHg-1s-1 at 4.7T and 37 °C, and was consistent between in vitro and in vivo adipose tissue. There was a strong, significant correlation between MRI- and gold standard OxyLite-based measurements of lipid ΔpO2 for in vivo visceral and subcutaneous fat depots in rodents. CONCLUSION This study lays the foundation for a direct, noninvasive measure of adipose pO2 using MRI and will allow for noninvasive measurement of O2 flux in adipose tissue. The proposed approach would be of particular importance in the interrogation of the pathogenesis of type 2 diabetes, where it has been suggested that adipose tissue hypoxia is an independent driver of insulin resistance pathway.
Collapse
|
60
|
Li Y, Chen X, Zhou Z, Li Q, Westover KD, Wang M, Liu J, Zhang S, Zhang J, Xu B, Wei X. Dynamic surveillance of tamoxifen-resistance in ER-positive breast cancer by CAIX-targeted ultrasound imaging. Cancer Med 2020; 9:2414-2426. [PMID: 32048471 PMCID: PMC7131861 DOI: 10.1002/cam4.2878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/03/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Tamoxifen‐based hormone therapy is central for the treatment of estrogen receptor positive (ER+) breast cancer. However, the acquired tamoxifen resistance, typically co‐exists with hypoxia, remains a major challenge. We aimed to develop a non‐invasive, targeted ultrasound imaging approach to dynamically monitory of tamoxifen resistance. After we assessed acquired tamoxifen resistance in 235 breast cancer patients and a list of breast cancer cell lines, we developed poly(lactic‐co‐glycolic acid)‐poly(ethylene glycol)‐carbonic anhydrase IX mono antibody nanobubbles (PLGA‐PEG‐mAbCAIX NBs) to detect hypoxic breast cancer cells upon exposure of tamoxifen in nude mice. We demonstrate that carbonic anhydrase IX (CAIX) expression is associated with breast cancer local recurrence and tamoxifen resistance both in clinical and cellular models. We find that CAIX overexpression increases tamoxifen tolerance in MCF‐7 cells and predicts early tamoxifen resistance along with an oscillating pattern in intracellular ATP level in vitro. PLGA‐PEG‐mAbCAIX NBs are able to dynamically detect tamoxifen‐induced hypoxia and tamoxifen resistance in vivo. CAIX‐conjugated NBs with noninvasive ultrasound imaging is powerful for dynamically monitoring hypoxic microenvironment in ER+ breast cancer with tamoxifen resistance.
Collapse
Affiliation(s)
- Ying Li
- Breast Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoyu Chen
- Department of Diagnostic and Therapeutic Ultrasonography, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - ZhiWei Zhou
- Department of Radiation Oncology and Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Qing Li
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, China
| | - Kenneth D Westover
- Department of Radiation Oncology and Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Meng Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Junjun Liu
- Breast Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Sheng Zhang
- Department of Diagnostic and Therapeutic Ultrasonography, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jin Zhang
- Breast Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bo Xu
- Breast Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
61
|
Cao‐Pham T, Tran‐Ly‐Binh A, Heyerick A, Fillée C, Joudiou N, Gallez B, Jordan BF. Combined endogenous MR biomarkers to assess changes in tumor oxygenation induced by an allosteric effector of hemoglobin. NMR IN BIOMEDICINE 2020; 33:e4181. [PMID: 31762121 PMCID: PMC7003919 DOI: 10.1002/nbm.4181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
Hypoxia is a crucial factor in cancer therapy, determining prognosis and the effectiveness of treatment. Although efforts are being made to develop methods for assessing tumor hypoxia, no markers of hypoxia are currently used in routine clinical practice. Recently, we showed that the combined endogenous MR biomarkers, R1 and R2 *, which are sensitive to [dissolved O2 ] and [dHb], respectively, were able to detect changes in tumor oxygenation induced by a hyperoxic breathing challenge. In this study, we further validated the ability of the combined MR biomarkers to assess the change in tumor oxygenation induced by an allosteric effector of hemoglobin, myo-inositol trispyrophosphate (ITPP), on rat tumor models. ITPP induced an increase in tumor pO2 , as observed using L-band electron paramagnetic resonance oximetry, as well as an increase in both R1 and R2 * MR parameters. The increase in R1 indicated an increase in [O2 ], whereas the increase in R2 * resulted from an increase in O2 release from blood, inducing an increase in [dHb]. The impact of ITPP was then evaluated on factors that can influence tumor oxygenation, including tumor perfusion, saturation rate of hemoglobin, blood pH and oxygen consumption rate (OCR). ITPP decreased blood [HbO2 ] and significantly increased blood acidity, which is also a factor that right-shifts the oxygen dissociation curve. No change in tumor perfusion was observed after ITPP treatment. Interestingly, ITPP decreased OCR in both tumor cell lines. In conclusion, ITPP increased tumor pO2 via a combined mechanism involving a decrease in OCR and an allosteric effect on hemoglobin that was further enhanced by a decrease in blood pH. MR biomarkers could assess the change in tumor oxygenation induced by ITPP. At the intra-tumoral level, a majority of tumor voxels were responsive to ITPP treatment in both of the models studied.
Collapse
Affiliation(s)
- Thanh‐Trang Cao‐Pham
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research GroupUniversité catholique de LouvainBrusselsBelgium
| | - An Tran‐Ly‐Binh
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research GroupUniversité catholique de LouvainBrusselsBelgium
| | | | - Catherine Fillée
- Institut de Recherche Expérimentale et Clinique (IREC), UCLouvainUniversite catholique de LouvainBrusselsBelgium
| | - Nicolas Joudiou
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research GroupUniversité catholique de LouvainBrusselsBelgium
| | - Bernard Gallez
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research GroupUniversité catholique de LouvainBrusselsBelgium
| | - Bénédicte F. Jordan
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research GroupUniversité catholique de LouvainBrusselsBelgium
| |
Collapse
|
62
|
Chin S, Eccles CL, McWilliam A, Chuter R, Walker E, Whitehurst P, Berresford J, Van Herk M, Hoskin PJ, Choudhury A. Magnetic resonance-guided radiation therapy: A review. J Med Imaging Radiat Oncol 2020; 64:163-177. [PMID: 31646742 DOI: 10.1111/1754-9485.12968] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022]
Abstract
Magnetic resonance-guided radiation therapy (MRgRT) is a promising approach to improving clinical outcomes for patients treated with radiation therapy. The roles of image guidance, adaptive planning and magnetic resonance imaging in radiation therapy have been increasing over the last two decades. Technical advances have led to the feasible combination of magnetic resonance imaging and radiation therapy technologies, leading to improved soft-tissue visualisation, assessment of inter- and intrafraction motion, motion management, online adaptive radiation therapy and the incorporation of functional information into treatment. MRgRT can potentially transform radiation oncology by improving tumour control and quality of life after radiation therapy and increasing convenience of treatment by shortening treatment courses for patients. Multiple groups have developed clinical implementations of MRgRT predominantly in the abdomen and pelvis, with patients having been treated since 2014. While studies of MRgRT have primarily been dosimetric so far, an increasing number of trials are underway examining the potential clinical benefits of MRgRT, with coordinated efforts to rigorously evaluate the benefits of the promising technology. This review discusses the current implementations, studies, potential benefits and challenges of MRgRT.
Collapse
Affiliation(s)
- Stephen Chin
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Cynthia L Eccles
- Department of Radiotherapy, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Alan McWilliam
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Robert Chuter
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Emma Walker
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Philip Whitehurst
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Joseph Berresford
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Marcel Van Herk
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Peter J Hoskin
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
63
|
Taylor E, Zhou J, Lindsay P, Foltz W, Cheung M, Siddiqui I, Hosni A, Amir AE, Kim J, Hill RP, Jaffray DA, Hedley DW. Quantifying Reoxygenation in Pancreatic Cancer During Stereotactic Body Radiotherapy. Sci Rep 2020; 10:1638. [PMID: 32005829 PMCID: PMC6994660 DOI: 10.1038/s41598-019-57364-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 02/05/2023] Open
Abstract
Hypoxia, the state of low oxygenation that often arises in solid tumours due to their high metabolism and irregular vasculature, is a major contributor to the resistance of tumours to radiation therapy (RT) and other treatments. Conventional RT extends treatment over several weeks or more, and nominally allows time for oxygen levels to increase ("reoxygenation") as cancer cells are killed by RT, mitigating the impact of hypoxia. Recent advances in RT have led to an increase in the use stereotactic body radiotherapy (SBRT), which delivers high doses in five or fewer fractions. For cancers such as pancreatic adenocarcinoma for which hypoxia varies significantly between patients, SBRT might not be optimal, depending on the extent to which reoxygenation occurs during its short duration. We used fluoro-5-deoxy-α-D-arabinofuranosyl)-2-nitroimidazole positron-emission tomography (FAZA-PET) imaging to quantify hypoxia before and after 5-fraction SBRT delivered to patient-derived pancreatic cancer xenografts orthotopically implanted in mice. An imaging technique using only the pre-treatment FAZA-PET scan and repeat dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) scans throughout treatment was able to predict the change in hypoxia. Our results support the further testing of this technique for imaging of reoxygenation in the clinic.
Collapse
Affiliation(s)
- Edward Taylor
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Jitao Zhou
- Department of Abdominal Oncology, Cancer Center and Laboratory of Signal Transduction and Molecular Targeting Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Patricia Lindsay
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Warren Foltz
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - May Cheung
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Iram Siddiqui
- Department of Pathology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Ahmed El Amir
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - John Kim
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Richard P Hill
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David A Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David W Hedley
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
64
|
Agarwal S, Gulaka PK, Rastogi U, Kodibagkar VD. More bullets for PISTOL: linear and cyclic siloxane reporter probes for quantitative 1H MR oximetry. Sci Rep 2020; 10:1399. [PMID: 31996701 PMCID: PMC6989524 DOI: 10.1038/s41598-020-57889-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Tissue oximetry can assist in diagnosis and prognosis of many diseases and enable personalized therapy. Previously, we reported the ability of hexamethyldisiloxane (HMDSO) for accurate measurements of tissue oxygen tension (pO2) using Proton Imaging of Siloxanes to map Tissue Oxygenation Levels (PISTOL) magnetic resonance imaging. Here we report the feasibility of several commercially available linear and cyclic siloxanes (molecular weight 162–410 g/mol) as PISTOL-based oxygen reporters by characterizing their calibration constants. Further, field and temperature dependence of pO2 calibration curves of HMDSO, octamethyltrisiloxane (OMTSO) and polydimethylsiloxane (PDMSO) were also studied. The spin-lattice relaxation rate R1 of all siloxanes studied here exhibited a linear relationship with oxygenation (R1 = A′ + B′*pO2) at all temperatures and field strengths evaluated here. The sensitivity index η( = B′/A′) decreased with increasing molecular weight with values ranged from 4.7 × 10−3–11.6 × 10−3 torr−1 at 4.7 T. No substantial change in the anoxic relaxation rate and a slight decrease in pO2 sensitivity was observed at higher magnetic fields of 7 T and 9.4 T for HMDSO and OMTSO. Temperature dependence of calibration curves for HMDSO, OMTSO and PDMSO was small and simulated errors in pO2 measurement were 1–2 torr/°C. In summary, we have demonstrated the feasibility of various linear and cyclic siloxanes as pO2-reporters for PISTOL-based oximetry.
Collapse
Affiliation(s)
- Shubhangi Agarwal
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85295, USA
| | - Praveen K Gulaka
- Department of Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Ujjawal Rastogi
- Department of Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85295, USA.
| |
Collapse
|
65
|
Thompson E, Smart S, Kinchesh P, Bulte D, Stride E. Magnetic resonance imaging of oxygen microbubbles. Healthc Technol Lett 2019; 6:138-142. [PMID: 31832209 PMCID: PMC6849496 DOI: 10.1049/htl.2018.5058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 04/12/2019] [Accepted: 05/23/2019] [Indexed: 12/26/2022] Open
Abstract
Oxygen loaded microbubbles are being investigated as a means of reducing tumour hypoxia in order to improve response to cancer therapy. To optimise this approach, it is desirable to be able to measure changes in tissue oxygenation in real-time during treatment. In this study, the feasibility of using magnetic resonance imaging (MRI) for this purpose was investigated. Longitudinal relaxation time (T1) measurements were made in simple hydrogel phantoms containing two different concentrations of oxygen microbubbles. T1 was found to be unaffected by the presence of oxygen microbubbles at either concentration. Upon application of ultrasound to destroy the microbubbles, however, a statistically significant reduction in T1 was seen for the higher microbubble concentration. Further work is needed to assess the influence of physiological conditions upon the measurements, but these preliminary results suggest that MRI could provide a method for quantifying the changes in tissue oxygenation produced by microbubbles during therapy.
Collapse
Affiliation(s)
- Elinor Thompson
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Sean Smart
- Radiobiology Research Institute, Department of Oncology, University of Oxford, OX3 7DQ, UK
| | - Paul Kinchesh
- Radiobiology Research Institute, Department of Oncology, University of Oxford, OX3 7DQ, UK
| | - Daniel Bulte
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| |
Collapse
|
66
|
Fiordelisi MF, Cavaliere C, Auletta L, Basso L, Salvatore M. Magnetic Resonance Imaging for Translational Research in Oncology. J Clin Med 2019; 8:jcm8111883. [PMID: 31698697 PMCID: PMC6912299 DOI: 10.3390/jcm8111883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The translation of results from the preclinical to the clinical setting is often anything other than straightforward. Indeed, ideas and even very intriguing results obtained at all levels of preclinical research, i.e., in vitro, on animal models, or even in clinical trials, often require much effort to validate, and sometimes, even useful data are lost or are demonstrated to be inapplicable in the clinic. In vivo, small-animal, preclinical imaging uses almost the same technologies in terms of hardware and software settings as for human patients, and hence, might result in a more rapid translation. In this perspective, magnetic resonance imaging might be the most translatable technique, since only in rare cases does it require the use of contrast agents, and when not, sequences developed in the lab can be readily applied to patients, thanks to their non-invasiveness. The wide range of sequences can give much useful information on the anatomy and pathophysiology of oncologic lesions in different body districts. This review aims to underline the versatility of this imaging technique and its various approaches, reporting the latest preclinical studies on thyroid, breast, and prostate cancers, both on small laboratory animals and on human patients, according to our previous and ongoing research lines.
Collapse
|
67
|
Hormuth DA, Sorace AG, Virostko J, Abramson RG, Bhujwalla ZM, Enriquez-Navas P, Gillies R, Hazle JD, Mason RP, Quarles CC, Weis JA, Whisenant JG, Xu J, Yankeelov TE. Translating preclinical MRI methods to clinical oncology. J Magn Reson Imaging 2019; 50:1377-1392. [PMID: 30925001 PMCID: PMC6766430 DOI: 10.1002/jmri.26731] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
The complexity of modern in vivo magnetic resonance imaging (MRI) methods in oncology has dramatically changed in the last 10 years. The field has long since moved passed its (unparalleled) ability to form images with exquisite soft-tissue contrast and morphology, allowing for the enhanced identification of primary tumors and metastatic disease. Currently, it is not uncommon to acquire images related to blood flow, cellularity, and macromolecular content in the clinical setting. The acquisition of images related to metabolism, hypoxia, pH, and tissue stiffness are also becoming common. All of these techniques have had some component of their invention, development, refinement, validation, and initial applications in the preclinical setting using in vivo animal models of cancer. In this review, we discuss the genesis of quantitative MRI methods that have been successfully translated from preclinical research and developed into clinical applications. These include methods that interrogate perfusion, diffusion, pH, hypoxia, macromolecular content, and tissue mechanical properties for improving detection, staging, and response monitoring of cancer. For each of these techniques, we summarize the 1) underlying biological mechanism(s); 2) preclinical applications; 3) available repeatability and reproducibility data; 4) clinical applications; and 5) limitations of the technique. We conclude with a discussion of lessons learned from translating MRI methods from the preclinical to clinical setting, and a presentation of four fundamental problems in cancer imaging that, if solved, would result in a profound improvement in the lives of oncology patients. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;50:1377-1392.
Collapse
Affiliation(s)
- David A. Hormuth
- Institute for Computational Engineering and Sciences,Livestrong Cancer Institutes, The University of Texas at Austin
| | - Anna G. Sorace
- Department of Biomedical Engineering, The University of Texas at Austin,Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| | - John Virostko
- Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| | - Richard G. Abramson
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center
| | | | - Pedro Enriquez-Navas
- Departments of Cancer Imaging and Metabolism, Cancer Physiology, The Moffitt Cancer Center
| | - Robert Gillies
- Departments of Cancer Imaging and Metabolism, Cancer Physiology, The Moffitt Cancer Center
| | - John D. Hazle
- Imaging Physics, The University of Texas M.D. Anderson Cancer Center
| | - Ralph P. Mason
- Department of Radiology, The University of Texas Southwestern Medical Center
| | - C. Chad Quarles
- Department of NeuroImaging Research, The Barrow Neurological Institute
| | - Jared A. Weis
- Department of Biomedical Engineering Wake Forest School of Medicine
| | | | - Junzhong Xu
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center,Institute of Imaging Science, Vanderbilt University Medical Center
| | - Thomas E. Yankeelov
- Institute for Computational Engineering and Sciences,Department of Biomedical Engineering, The University of Texas at Austin,Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| |
Collapse
|
68
|
Jiang K, Tang H, Mishra PK, Macura SI, Lerman LO. Measurement of murine kidney functional biomarkers using DCE-MRI: A multi-slice TRICKS technique and semi-automated image processing algorithm. Magn Reson Imaging 2019; 63:226-234. [PMID: 31442558 DOI: 10.1016/j.mri.2019.08.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 11/24/2022]
Abstract
PURPOSE To propose a rapid multi-slice T1 measurement method using time-resolved imaging of contrast kinetics (TRICKS) and a semi-automated image processing algorithm for comprehensive assessment murine kidney function using dynamic contrast-enhanced MRI (DCE-MRI). METHODS A multi-slice TRICKS sampling scheme was implemented in an established rapid T1 measurement method. A semi-automated image-processing scheme employing basic image processing techniques and machine learning was developed to facilitate image analysis. Reliability of the multi-slice technique in measuring renal perfusion and glomerular filtration rate (GFR) was tested in normal mice (n = 7 for both techniques) by comparing to the validated single-slice technique. Utility of this method was demonstrated on mice after either sham surgery (n = 7) or induction of unilateral renal artery stenosis (RAS, n = 8). Renal functional parameters were extracted using a validated bi-compartment model. RESULTS The TRICKS sampling scheme achieved an acceleration factor of 2.7, allowing imaging of eight axial slices at 1.23 s/scan. With the aid of the semi-automated scheme, image analysis required under 15-min for both kidneys per mouse. The multi-slice technique yielded renal perfusion and GFR values comparable to the single-slice technique. Model-fitted renal parameters successfully differentiated control and stenotic mouse kidneys, including renal perfusion (706.5 ± 164.0 vs. 375.9 ± 277.9 mL/100 g/min, P = 0.002), blood flow (1.6 ± 0.4 vs. 0.7 ± 0.7 mL/min, P < 0.001), and GFR (142.9 ± 17.9 vs. 58.0 ± 42.8 μL/min, P < 0.001). CONCLUSION The multi-slice TRICKS-based DCE-MRI technique, with a semi-automated image processing scheme, allows rapid and comprehensive measurement of murine kidney function.
Collapse
Affiliation(s)
- Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Prasanna K Mishra
- Division of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Slobodan I Macura
- Division of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
69
|
Development of multifunctional Overhauser-enhanced magnetic resonance imaging for concurrent in vivo mapping of tumor interstitial oxygenation, acidosis and inorganic phosphate concentration. Sci Rep 2019; 9:12093. [PMID: 31431629 PMCID: PMC6702349 DOI: 10.1038/s41598-019-48524-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor oxygenation (pO2), acidosis (pH) and interstitial inorganic phosphate concentration (Pi) are important parameters of the malignant behavior of cancer. A noninvasive procedure that enables visualization of these parameters may provide unique information about mechanisms of tumor pathophysiology and provide clues to new treatment targets. In this research, we present a multiparametric imaging method allowing for concurrent mapping of pH, spin probe concentration, pO2, and Pi using a single contrast agent and Overhauser-enhanced magnetic resonance imaging technique. The developed approach was applied to concurrent multifunctional imaging in phantom samples and in vivo in a mouse model of breast cancer. Tumor tissues showed higher heterogeneity of the distributions of the parameters compared with normal mammary gland and demonstrated the areas of significant acidosis, hypoxia, and elevated Pi content.
Collapse
|
70
|
Brown E, Brunker J, Bohndiek SE. Photoacoustic imaging as a tool to probe the tumour microenvironment. Dis Model Mech 2019; 12:dmm039636. [PMID: 31337635 PMCID: PMC6679374 DOI: 10.1242/dmm.039636] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The tumour microenvironment (TME) is a complex cellular ecosystem subjected to chemical and physical signals that play a role in shaping tumour heterogeneity, invasion and metastasis. Studying the roles of the TME in cancer progression would strongly benefit from non-invasive visualisation of the tumour as a whole organ in vivo, both preclinically in mouse models of the disease, as well as in patient tumours. Although imaging techniques exist that can probe different facets of the TME, they face several limitations, including limited spatial resolution, extended scan times and poor specificity from confounding signals. Photoacoustic imaging (PAI) is an emerging modality, currently in clinical trials, that has the potential to overcome these limitations. Here, we review the biological properties of the TME and potential of existing imaging methods that have been developed to analyse these properties non-invasively. We then introduce PAI and explore the preclinical and clinical evidence that support its use in probing multiple features of the TME simultaneously, including blood vessel architecture, blood oxygenation, acidity, extracellular matrix deposition, lipid concentration and immune cell infiltration. Finally, we highlight the future prospects and outstanding challenges in the application of PAI as a tool in cancer research and as part of a clinical oncologist's arsenal.
Collapse
Affiliation(s)
- Emma Brown
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Joanna Brunker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
71
|
Salem A, Little RA, Latif A, Featherstone AK, Babur M, Peset I, Cheung S, Watson Y, Tessyman V, Mistry H, Ashton G, Behan C, Matthews JC, Asselin MC, Bristow RG, Jackson A, Parker GJM, Faivre-Finn C, Williams KJ, O'Connor JPB. Oxygen-enhanced MRI Is Feasible, Repeatable, and Detects Radiotherapy-induced Change in Hypoxia in Xenograft Models and in Patients with Non-small Cell Lung Cancer. Clin Cancer Res 2019; 25:3818-3829. [PMID: 31053599 DOI: 10.1158/1078-0432.ccr-18-3932] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/04/2019] [Accepted: 03/14/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Hypoxia is associated with poor prognosis and is predictive of poor response to cancer treatments, including radiotherapy. Developing noninvasive biomarkers that both detect hypoxia prior to treatment and track change in tumor hypoxia following treatment is required urgently. EXPERIMENTAL DESIGN We evaluated the ability of oxygen-enhanced MRI (OE-MRI) to map and quantify therapy-induced changes in tumor hypoxia by measuring oxygen-refractory signals in perfused tissue (perfused Oxy-R). Clinical first-in-human study in patients with non-small cell lung cancer (NSCLC) was performed alongside preclinical experiments in two xenograft tumors (Calu6 NSCLC model and U87 glioma model). RESULTS MRI perfused Oxy-R tumor fraction measurement of hypoxia was validated with ex vivo tissue pathology in both xenograft models. Calu6 and U87 experiments showed that MRI perfused Oxy-R tumor volume was reduced relative to control following single fraction 10-Gy radiation and fractionated chemoradiotherapy (P < 0.001) due to both improved perfusion and reduced oxygen consumption rate. Next, evaluation of 23 patients with NSCLC showed that OE-MRI was clinically feasible and that tumor perfused Oxy-R volume is repeatable [interclass correlation coefficient: 0.961 (95% CI, 0.858-0.990); coefficient of variation: 25.880%]. Group-wise perfused Oxy-R volume was reduced at 14 days following start of radiotherapy (P = 0.015). OE-MRI detected between-subject variation in hypoxia modification in both xenograft and patient tumors. CONCLUSIONS These findings support applying OE-MRI biomarkers to monitor hypoxia modification, to stratify patients in clinical trials of hypoxia-modifying therapies, to identify patients with hypoxic tumors that may fail treatment with immunotherapy, and to guide adaptive radiotherapy by mapping regional hypoxia.
Collapse
Affiliation(s)
- Ahmed Salem
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie Hospital NHS Trust, Manchester, United Kingdom
| | - Ross A Little
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Ayşe Latif
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Adam K Featherstone
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Muhammad Babur
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Isabel Peset
- Imaging and Flow Cytometry, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Susan Cheung
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Yvonne Watson
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Victoria Tessyman
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Hitesh Mistry
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Garry Ashton
- Histology, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Caron Behan
- Histology, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Julian C Matthews
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Marie-Claude Asselin
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Robert G Bristow
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie Hospital NHS Trust, Manchester, United Kingdom
| | - Alan Jackson
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Geoff J M Parker
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, United Kingdom
- Bioxydyn Limited, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie Hospital NHS Trust, Manchester, United Kingdom
| | - Kaye J Williams
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - James P B O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom. James.O'
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, United Kingdom
| |
Collapse
|
72
|
Yang DM, Arai TJ, Campbell JW, Gerberich JL, Zhou H, Mason RP. Oxygen-sensitive MRI assessment of tumor response to hypoxic gas breathing challenge. NMR IN BIOMEDICINE 2019; 32:e4101. [PMID: 31062902 PMCID: PMC6581571 DOI: 10.1002/nbm.4101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/16/2019] [Accepted: 03/08/2019] [Indexed: 06/09/2023]
Abstract
Oxygen-sensitive MRI has been extensively used to investigate tumor oxygenation based on the response (R2 * and/or R1 ) to a gas breathing challenge. Most studies have reported response to hyperoxic gas indicating potential biomarkers of hypoxia. Few studies have examined hypoxic gas breathing and we have now evaluated acute dynamic changes in rat breast tumors. Rats bearing syngeneic subcutaneous (n = 15) or orthotopic (n = 7) 13762NF breast tumors were exposed to a 16% O2 gas breathing challenge and monitored using blood oxygen level dependent (BOLD) R2 * and tissue oxygen level dependent (TOLD) T1 -weighted measurements at 4.7 T. As a control, we used a traditional hyperoxic gas breathing challenge with 100% O2 on a subset of the subcutaneous tumor bearing rats (n = 6). Tumor subregions identified as responsive on the basis of R2 * dynamics coincided with the viable tumor area as judged by subsequent H&E staining. As expected, R2 * decreased and T1 -weighted signal increased in response to 100% O2 breathing challenge. Meanwhile, 16% O2 breathing elicited an increase in R2 *, but divergent response (increase or decrease) in T1 -weighted signal. The T1 -weighted signal increase may signify a dominating BOLD effect triggered by 16% O2 in the relatively more hypoxic tumors, whereby the influence of increased paramagnetic deoxyhemoglobin outweighs decreased pO2 . The results emphasize the importance of combined BOLD and TOLD measurements for the correct interpretation of tumor oxygenation properties.
Collapse
Affiliation(s)
- Donghan M Yang
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tatsuya J Arai
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - James W Campbell
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Heling Zhou
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ralph P Mason
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
73
|
Gérard M, Corroyer-Dulmont A, Lesueur P, Collet S, Chérel M, Bourgeois M, Stefan D, Limkin EJ, Perrio C, Guillamo JS, Dubray B, Bernaudin M, Thariat J, Valable S. Hypoxia Imaging and Adaptive Radiotherapy: A State-of-the-Art Approach in the Management of Glioma. Front Med (Lausanne) 2019; 6:117. [PMID: 31249831 PMCID: PMC6582242 DOI: 10.3389/fmed.2019.00117] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/13/2019] [Indexed: 01/31/2023] Open
Abstract
Severe hypoxia [oxygen partial pressure (pO2) below 5–10 mmHg] is more frequent in glioblastoma multiforme (GBM) compared to lower-grade gliomas. Seminal studies in the 1950s demonstrated that hypoxia was associated with increased resistance to low–linear energy transfer (LET) ionizing radiation. In experimental conditions, the total radiation dose has to be multiplied by a factor of 3 to achieve the same cell lethality in anoxic situations. The presence of hypoxia in human tumors is assumed to contribute to treatment failures after radiotherapy (RT) in cancer patients. Therefore, a logical way to overcome hypoxia-induced radioresistance would be to deliver substantially higher doses of RT in hypoxic volumes delineated on pre-treatment imaging as biological target volumes (BTVs). Such an approach faces various fundamental, technical, and clinical challenges. The present review addresses several technical points related to the delineation of hypoxic zones, which include: spatial accuracy, quantitative vs. relative threshold, variations of hypoxia levels during RT, and availability of hypoxia tracers. The feasibility of hypoxia imaging as an assessment tool for early tumor response to RT and for predicting long-term outcomes is discussed. Hypoxia imaging for RT dose painting is likewise examined. As for the radiation oncologist's point of view, hypoxia maps should be converted into dose-distribution objectives for RT planning. Taking into account the physics and the radiobiology of various irradiation beams, preliminary in silico studies are required to investigate the feasibility of dose escalation in terms of normal tissue tolerance before clinical trials are undertaken.
Collapse
Affiliation(s)
- Michael Gérard
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | | | - Paul Lesueur
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Solène Collet
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiophysics, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Michel Chérel
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Mickael Bourgeois
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Dinu Stefan
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Elaine Johanna Limkin
- Department of Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Cécile Perrio
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Jean-Sébastien Guillamo
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Neurology, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Bernard Dubray
- Département de Radiothérapie et de Physique Médicale, Laboratoire QuantIF-LITIS [EA 4108], Centre de Lutte Contre le Cancer Henri Becquerel, Université de Normandie, Rouen, France
| | - Myriam Bernaudin
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Juliette Thariat
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Samuel Valable
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| |
Collapse
|
74
|
Ron A, Deán-Ben XL, Gottschalk S, Razansky D. Volumetric Optoacoustic Imaging Unveils High-Resolution Patterns of Acute and Cyclic Hypoxia in a Murine Model of Breast Cancer. Cancer Res 2019; 79:4767-4775. [PMID: 31097477 DOI: 10.1158/0008-5472.can-18-3769] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/03/2019] [Accepted: 05/10/2019] [Indexed: 11/16/2022]
Abstract
Mapping tumor heterogeneity and hypoxia within a living intact organism is essential for understanding the processes involved in cancer progression and assessing long-term responses to therapies. Efficient investigations into tumor hypoxia mechanisms have been hindered by the lack of intravital imaging tools capable of multiparametric probing of entire solid tumors with high spatial and temporal resolution. Here, we exploit volumetric multispectral optoacoustic tomography (vMSOT) for accurate, label-free delineation of tumor heterogeneity and dynamic oxygenation behavior. Mice bearing orthotopic MDA-MB-231 breast cancer xenografts were imaged noninvasively during rest and oxygen stress challenge, attaining time-lapse three-dimensional oxygenation maps across entire tumors with 100 μm spatial resolution. Volumetric quantification of the hypoxic fraction rendered values of 3.9% to 21.2%, whereas the oxygen saturation (sO2) rate declined at 1.7% to 2.3% per mm in all tumors when approaching their core. Three distinct functional areas (the rim, hypoxic, and normoxic cores) were clearly discernible based on spatial sO2 profiles and responses to oxygen challenge. Notably, although sO2 readings were responsive to the challenge, deoxyhemoglobin (HbR) trends exhibited little to no variations in all mice. Dynamic analysis further revealed the presence of cyclic hypoxia patterns with a 21% average discrepancy between cyclic fractions assessed via sO2 (42.2% ± 17.3%) and HbR fluctuations (63% ± 14.1%) within the hypoxic core. These findings corroborate the strong potential of vMSOT for advancing preclinical imaging of cancer and informing clinical decisions on therapeutic interventions. SIGNIFICANCE: vMSOT provides quantitative measures of volumetric hypoxic fraction and cyclic hypoxia in a label-free and noninvasive manner, providing new readouts to aid tumor staging and treatment decision making. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/18/4767/F1.large.jpg.See related commentary by Klibanov and Hu, p. 4577.
Collapse
Affiliation(s)
- Avihai Ron
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany.,Faculty of Medicine, Technical University of Munich, Munich, Germany
| | - Xosé Luís Deán-Ben
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany.,Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering ETH Zurich, Zurich, Switzerland
| | - Sven Gottschalk
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
| | - Daniel Razansky
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany. .,Faculty of Medicine, Technical University of Munich, Munich, Germany.,Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering ETH Zurich, Zurich, Switzerland
| |
Collapse
|
75
|
Zhou H, Chiguru S, Hallac RR, Yang D, Hao G, Peschke P, Mason RP. Examining correlations of oxygen sensitive MRI (BOLD/TOLD) with [ 18F]FMISO PET in rat prostate tumors. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2019; 9:156-167. [PMID: 31139498 PMCID: PMC6526364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/25/2019] [Indexed: 06/09/2023]
Abstract
Hypoxia is regarded as a potential prognostic biomarker for tumor aggressiveness, progression, and response to therapy. The radiotracer 18F-fluoromisonidazole ([18F]FMISO) has been used with positron emission tomography (PET) to reveal tumor hypoxia. Meanwhile, blood oxygen level dependent (BOLD) MRI and tissue oxygen level dependent (TOLD) MRI offer insight into oxygenation based on endogenous signals without the need for radiolabels. Here, we compared BOLD and TOLD MRI with [18F]FMISO uptake using Dunning prostate R3327-AT1 tumor bearing rats. BOLD and TOLD MRI were acquired with respect to an oxygen gas breathing challenge. The following day, dynamic PET was performed up to 90 minutes following IV injection of [18F]FMISO. Tumors showed distinct heterogeneity based on each technique. Correlations were observed between magnitude of mean BOLD or TOLD MRI signal responses to oxygen-breathing challenge and initial distribution of [18F]FMISO. Correlations were observed for whole tumor as well on a regional basis with stronger correlations in the well perfused tumor periphery indicating the strong influence of perfused vasculature. After 90 minutes most correlations with signal intensity became quite weak, but correlations were observed between hypoxic fraction based on FMISO and fractions of tumor showing BOLD or TOLD response in a subset of tumors. This emphasizes the importance of considering regional heterogeneity and responsive fractions, as opposed to simple magnitudes of responses. Although the data represent a small cohort of tumors they present direct correlations between oxygen sensitive MRI and PET hypoxia reporter agents in the same tumors, indicating the potential utility of further investigations.
Collapse
Affiliation(s)
- Heling Zhou
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
| | - Srinivas Chiguru
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
| | - Rami R Hallac
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
- AIM Center, Children’s HealthDallas, TX, United States
| | - Donghan Yang
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
| | - Guiyang Hao
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
| | - Peter Peschke
- Medical Physics in Radiation Oncology, German Cancer Research CenterHeidelberg, Germany
| | - Ralph P Mason
- Department of Radiology, UT Southwestern Medical CenterDallas, TX 75390-9058, USA
| |
Collapse
|
76
|
O'Connor JPB, Robinson SP, Waterton JC. Imaging tumour hypoxia with oxygen-enhanced MRI and BOLD MRI. Br J Radiol 2019; 92:20180642. [PMID: 30272998 PMCID: PMC6540855 DOI: 10.1259/bjr.20180642] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is known to be a poor prognostic indicator for nearly all solid tumours and also is predictive of treatment failure for radiotherapy, chemotherapy, surgery and targeted therapies. Imaging has potential to identify, spatially map and quantify tumour hypoxia prior to therapy, as well as track changes in hypoxia on treatment. At present no hypoxia imaging methods are available for routine clinical use. Research has largely focused on positron emission tomography (PET)-based techniques, but there is gathering evidence that MRI techniques may provide a practical and more readily translational alternative. In this review we focus on the potential for imaging hypoxia by measuring changes in longitudinal relaxation [R1; termed oxygen-enhanced MRI or tumour oxygenation level dependent (TOLD) MRI] and effective transverse relaxation [R2*; termed blood oxygenation level dependent (BOLD) MRI], induced by inhalation of either 100% oxygen or the radiosensitising hyperoxic gas carbogen. We explain the scientific principles behind oxygen-enhanced MRI and BOLD and discuss significant studies and their limitations. All imaging biomarkers require rigorous validation in order to translate into clinical use and the steps required to further develop oxygen-enhanced MRI and BOLD MRI into decision-making tools are discussed.
Collapse
Affiliation(s)
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | |
Collapse
|
77
|
Vaidya T, Agrawal A, Mahajan S, Thakur MH, Mahajan A. The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part II). Mol Diagn Ther 2019; 23:27-51. [PMID: 30387041 DOI: 10.1007/s40291-018-0367-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present era of precision medicine sees "cancer" as a consequence of molecular derangements occurring at the commencement of the disease process, with morphological changes happening much later in the process of tumourigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound (US) and magnetic resonance imaging (MRI) play an integral role in the detection of disease at the macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumourigenesis. MFI has the potential to play a key role in heralding the transition from the concept of "one-size-fits-all" treatment to "precision medicine". Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called "radiogenomics", which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning and artificial intelligence, the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, the majority of these require validation before clinical translation is possible. In this two part review, we discuss the systematic collaboration across structural, anatomical and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
Affiliation(s)
- Tanvi Vaidya
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Parel, Mumbai, Maharashtra, 400 012, India
| | - Shivani Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - M H Thakur
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Abhishek Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
78
|
Zhou H, Belzile O, Zhang Z, Wagner J, Ahn C, Richardson JA, Saha D, Brekken RA, Mason RP. The effect of flow on blood oxygen level dependent (R * 2 ) MRI of orthotopic lung tumors. Magn Reson Med 2019; 81:3787-3797. [PMID: 30697815 DOI: 10.1002/mrm.27661] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/23/2022]
Abstract
PURPOSE Blood oxygen level dependent (BOLD) MRI based on R 2 * measurements can provide insights into tumor vascular oxygenation. However, measurements are susceptible to blood flow, which may vary accompanying a hyperoxic gas challenge. We investigated flow sensitivity by comparing R 2 * measurements with and without flow suppression (fs) in 2 orthotopic lung xenograft tumor models. METHODS H460 (n = 20) and A549 (n = 20) human lung tumor xenografts were induced by surgical implantation of cancer cells in the right lung of nude rats. MRI was performed at 4.7T after tumors reached 5 to 8 mm in diameter. A multiecho gradient echo MRI sequence was acquired with and without spatial saturation bands on each side of the imaging plane to evaluate the effect of flow on R 2 * . fs and non-fs R 2 * MRI measurements were interleaved during an oxygen breathing challenge (from air to 100% O2 ). T 2 * -weighted signal intensity changes (ΔSI(%)) and R 2 * measurements were obtained for regions of interest and on a voxel-by-voxel basis and discrepancies quantified with Bland-Altman analysis. RESULTS Flow suppression affected ΔSI(%) and R 2 * measurements in each tumor model. Average discrepancy and limits of agreement from Bland-Altman analyses revealed greater flow-related bias in A549 than H460. CONCLUSION The effect of flow on R 2 * , and hence BOLD, was tumor model dependent with measurements being more sensitive in well-perfused A549 tumors.
Collapse
Affiliation(s)
- Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Olivier Belzile
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhang Zhang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jo Wagner
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chul Ahn
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James A Richardson
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
79
|
Emerging Functional Imaging Biomarkers of Tumour Responses to Radiotherapy. Cancers (Basel) 2019; 11:cancers11020131. [PMID: 30678055 PMCID: PMC6407112 DOI: 10.3390/cancers11020131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/11/2022] Open
Abstract
Tumour responses to radiotherapy are currently primarily assessed by changes in size. Imaging permits non-invasive, whole-body assessment of tumour burden and guides treatment options for most tumours. However, in most tumours, changes in size are slow to manifest and can sometimes be difficult to interpret or misleading, potentially leading to prolonged durations of ineffective treatment and delays in changing therapy. Functional imaging techniques that monitor biological processes have the potential to detect tumour responses to treatment earlier and refine treatment options based on tumour biology rather than solely on size and staging. By considering the biological effects of radiotherapy, this review focusses on emerging functional imaging techniques with the potential to augment morphological imaging and serve as biomarkers of early response to radiotherapy.
Collapse
|
80
|
Spiegelberg L, Houben R, Niemans R, de Ruysscher D, Yaromina A, Theys J, Guise CP, Smaill JB, Patterson AV, Lambin P, Dubois LJ. Hypoxia-activated prodrugs and (lack of) clinical progress: The need for hypoxia-based biomarker patient selection in phase III clinical trials. Clin Transl Radiat Oncol 2019; 15:62-69. [PMID: 30734002 PMCID: PMC6357685 DOI: 10.1016/j.ctro.2019.01.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 01/07/2023] Open
Abstract
Hypoxia-activated prodrugs have yielded promising results up to phase II trials. Implementation of hypoxia-activated prodrugs in the clinic has not been successful. Phase III clinical trials lack patient stratification based on tumor hypoxia status. Stratification will decrease the number of patients needed and increase success. Improvements in hypoxia-activated prodrug design can also increase success rates.
Hypoxia-activated prodrugs (HAPs) are designed to specifically target the hypoxic cells of tumors, which are an important cause of treatment resistance to conventional therapies. Despite promising preclinical and clinical phase I and II results, the most important of which are described in this review, the implementation of hypoxia-activated prodrugs in the clinic has, so far, not been successful. The lack of stratification of patients based on tumor hypoxia status, which can vary widely, is sufficient to account for the failure of phase III trials. To fully exploit the potential of hypoxia-activated prodrugs, hypoxia stratification of patients is needed. Here, we propose a biomarker-stratified enriched Phase III study design in which only biomarker-positive (i.e. hypoxia-positive) patients are randomized between standard treatment and the combination of standard treatment with a hypoxia-activated prodrug. This implies the necessity of a Phase II study in which the biomarker or a combination of biomarkers will be evaluated. The total number of patients needed for both clinical studies will be far lower than in currently used randomize-all designs. In addition, we elaborate on the improvements in HAP design that are feasible to increase the treatment success rates.
Collapse
Affiliation(s)
- Linda Spiegelberg
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ruud Houben
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Raymon Niemans
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Dirk de Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ala Yaromina
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Theys
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Christopher P Guise
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Jeffrey B Smaill
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Adam V Patterson
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Philippe Lambin
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ludwig J Dubois
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
81
|
Serša I, Bajd F, Savarin M, Jesenko T, Čemažar M, Serša G. Multiparametric High-Resolution MRI as a Tool for Mapping of Hypoxic Level in Tumors. Technol Cancer Res Treat 2019; 17:1533033818797066. [PMID: 30176769 PMCID: PMC6122235 DOI: 10.1177/1533033818797066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hypoxia is a condition, common to most malignant tumors, where oxygen tension in the tissue is below the physiological level. Among consequences of tumor hypoxia is also altered cancer cell metabolism that contributes to cancer therapy resistance. Therefore, precise assessment of tumor hypoxia is important for monitoring the tumor treatment progression. In this study, we propose a simple model for prediction of hypoxic level in tumors based on multiparametric magnetic resonance imaging. The study was performed on B16F1 murine melanoma tumors ex vivo that were first magnetic resonance scanned and then analyzed for hypoxic level using hypoxia-inducable factor 1-alpha antibody staining. Each tumor was analyzed in identical sections and in identical regions of interest for pairs of hypoxic level and magnetic resonance values (apparent diffusion coefficient and T2). This was followed by correlation analysis between hypoxic level and respective magnetic resonance values. A moderate correlation was found between hypoxic level and apparent diffusion coefficient (ρ = 0.56, P < .00001) and lower between hypoxic level and T2 (ρ = 0.38, P < .00001). The data were analyzed further to obtain simple predictive models based on the multiple linear regression analysis of the measured hypoxic level (dependent variable) and apparent diffusion coefficient and T2 (independent variables). Among the hypoxic level models, the most efficient was the 3-parameter model given by relation (HL = kADCADC + kT2T2 + b), where kADC = 26%/µm2/ms, kT2 = 0.8%/ms, and b = −32%. The model can be used for calculation of the predicted hypoxic level map based on magnetic resonance–measured apparent diffusion coefficient and T2 maps. Similar prediction models, based on tumor apparent diffusion coefficient and T2 maps, can be done also for other tumor types in vivo and can therefore help in assessment of tumor treatment as well as to better understand the role of hypoxia in cancer progression.
Collapse
Affiliation(s)
- Igor Serša
- 1 Jožef Stefan Institute, Ljubljana, Slovenia.,2 Institute of Physiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Franci Bajd
- 3 Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | | | - Tanja Jesenko
- 4 Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Maja Čemažar
- 4 Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Gregor Serša
- 4 Institute of Oncology Ljubljana, Ljubljana, Slovenia.,5 Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
82
|
Clinical and Pre-clinical Methods for Quantifying Tumor Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:19-41. [PMID: 31201714 DOI: 10.1007/978-3-030-12734-3_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, a prevalent characteristic of most solid malignant tumors, contributes to diminished therapeutic responses and more aggressive phenotypes. The term hypoxia has two definitions. One definition would be a physiologic state where the oxygen partial pressure is below the normal physiologic range. For most normal tissues, the normal physiologic range is between 10 and 20 mmHg. Hypoxic regions develop when there is an imbalance between oxygen supply and demand. The impact of hypoxia on cancer therapeutics is significant: hypoxic tissue is 3× less radiosensitive than normoxic tissue, the impaired blood flow found in hypoxic tumor regions influences chemotherapy delivery, and the immune system is dependent on oxygen for functionality. Despite the clinical implications of hypoxia, there is not a universal, ideal method for quantifying hypoxia, particularly cycling hypoxia because of its complexity and heterogeneity across tumor types and individuals. Most standard imaging techniques can be modified and applied to measuring hypoxia and quantifying its effects; however, the benefits and challenges of each imaging modality makes imaging hypoxia case-dependent. In this chapter, a comprehensive overview of the preclinical and clinical methods for quantifying hypoxia is presented along with the advantages and disadvantages of each.
Collapse
|
83
|
Suh JY, Cho G, Song Y, Woo DC, Choi YS, Ryu EK, Park BW, Shim WH, Kim YR, Kim JK. Hyperoxia-Induced ΔR 1. Stroke 2018; 49:3012-3019. [PMID: 30571431 DOI: 10.1161/strokeaha.118.021469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Acceleration of longitudinal relaxation under hyperoxic challenge (ie, hyperoxia-induced ΔR1) indicates oxygen accumulation and reflects baseline tissue oxygenation. We evaluated the feasibility of hyperoxia-induced ΔR1 for evaluating cerebral oxygenation status and degree of ischemic damage in stroke. Methods- In 24-hour transient stroke rat models (n=13), hyperoxia-induced ΔR1, ischemic severity (apparent diffusion coefficient [ADC]), vasogenic edema (R2), total and microvascular blood volume (superparamagnetic iron oxide-driven ΔR2* and ΔR2, respectively), and glucose metabolism activity (18F-fluorodeoxyglucose uptake on positron emission tomography) were measured. The distribution of these parameters according to hyperoxia-induced ΔR1 was analyzed. The partial pressure of tissue oxygen change during hyperoxic challenge was measured using fiberoptic tissue oximetry. In 4-hour stroke models (n=6), ADC and hyperoxia-induced ΔR1 was analyzed with 2,3,5-triphenyltetrazolium chloride staining being a criterion of infarction. Results- Ischemic hemisphere showed significantly higher hyperoxia-induced ΔR1 than nonischemic brain in a pattern depending on ADC. During hyperoxic challenge, ischemic hemisphere demonstrated uncontrolled increase of partial pressure of tissue oxygen, whereas contralateral hemisphere rapidly plateaued. Ischemic hemisphere also demonstrated significant correlation between hyperoxia-induced ΔR1 and R2. Hyperoxia-induced ΔR1 showed a significant negative correlation with 18F-fluorodeoxyglucose uptake. The ADC, R2, ΔR2, and 18F-fluorodeoxyglucose uptake showed a dichotomized distribution according to the hyperoxia-induced ΔR1 as their slopes and values were higher at low hyperoxia-induced ΔR1 (<50 ms-1) than at high ΔR1. In 4-hour stroke rats, the distribution of ADC according to the hyperoxia-induced ΔR1 was similar with 24-hour stroke rats. The hyperoxia-induced ΔR1 was greater in the infarct area (47±10 ms-1) than in peri-infarct area (16±4 ms-1; P<0.01). Conclusions- Hyperoxia-induced ΔR1 adequately indicates cerebral oxygenation and can be a feasible biomarker to classify the degree of ischemia-induced damage in neurovascular function and metabolism in stroke brain.
Collapse
Affiliation(s)
- Ji-Yeon Suh
- From the Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.-Y.S., D.-C.W., B.W.P., W.H.S., J.K.K.).,Bioimaging Research Team, Korea Basic Science Institute, Ochang Cheongwon, Chungbuk, Korea (J.-Y.S., G.C., Y.S., E.K.R.)
| | - Gyunggoo Cho
- Bioimaging Research Team, Korea Basic Science Institute, Ochang Cheongwon, Chungbuk, Korea (J.-Y.S., G.C., Y.S., E.K.R.)
| | - Youngkyu Song
- Bioimaging Research Team, Korea Basic Science Institute, Ochang Cheongwon, Chungbuk, Korea (J.-Y.S., G.C., Y.S., E.K.R.)
| | - Dong-Cheol Woo
- From the Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.-Y.S., D.-C.W., B.W.P., W.H.S., J.K.K.)
| | - Yoon Seok Choi
- Medical Research Institute, Gangneung Asan Hospital, Gangwon-do, South Korea (Y.S.C.)
| | - Eun Kyung Ryu
- Bioimaging Research Team, Korea Basic Science Institute, Ochang Cheongwon, Chungbuk, Korea (J.-Y.S., G.C., Y.S., E.K.R.)
| | - Bum Woo Park
- From the Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.-Y.S., D.-C.W., B.W.P., W.H.S., J.K.K.)
| | - Woo Hyun Shim
- From the Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.-Y.S., D.-C.W., B.W.P., W.H.S., J.K.K.)
| | - Young Ro Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown (Y.R.K.)
| | - Jeong Kon Kim
- From the Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.-Y.S., D.-C.W., B.W.P., W.H.S., J.K.K.)
| |
Collapse
|
84
|
The Challenges of Using MRI During Radiotherapy. Clin Oncol (R Coll Radiol) 2018; 30:680-685. [DOI: 10.1016/j.clon.2018.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/01/2018] [Accepted: 08/20/2018] [Indexed: 12/29/2022]
|
85
|
Datta A, Aznar MC, Dubec M, Parker GJM, O'Connor JPB. Delivering Functional Imaging on the MRI-Linac: Current Challenges and Potential Solutions. Clin Oncol (R Coll Radiol) 2018; 30:702-710. [PMID: 30224203 DOI: 10.1016/j.clon.2018.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Magnetic resonance imaging (MRI) is a highly versatile imaging modality that can be used to measure features of the tumour microenvironment including cell death, proliferation, metabolism, angiogenesis, and hypoxia. Mapping and quantifying these pathophysiological features has the potential to alter the use of adaptive radiotherapy planning. Although these methods are available for use on diagnostic machines, several challenges exist for implementing these functional MRI methods on the MRI-linear accelerators (linacs). This review considers these challenges and potential solutions.
Collapse
Affiliation(s)
- A Datta
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK
| | - M C Aznar
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - M Dubec
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Christie Medical Physics and Engineering, The Christie Hospital NHS Trust, Manchester, UK
| | - G J M Parker
- Bioxydyn Ltd, Manchester, UK; Centre for Imaging Sciences, University of Manchester, Manchester, UK
| | - J P B O'Connor
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
86
|
Moosvi F, Baker JH, Yung A, Kozlowski P, Minchinton AI, Reinsberg SA. Fast and sensitive dynamic oxygen‐enhanced MRI with a cycling gas challenge and independent component analysis. Magn Reson Med 2018; 81:2514-2525. [DOI: 10.1002/mrm.27584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Firas Moosvi
- Department of Physics & Astronomy University of British Columbia Vancouver Canada
| | - Jennifer H.E. Baker
- Radiation Biology Unit British Columbia Cancer Research Centre Vancouver Canada
| | - Andrew Yung
- UBC MRI Research Centre Life Sciences Centre Vancouver Canada
| | - Piotr Kozlowski
- UBC MRI Research Centre Life Sciences Centre Vancouver Canada
- Department of Radiology University of British Columbia Vancouver Canada
| | | | - Stefan A. Reinsberg
- Department of Physics & Astronomy University of British Columbia Vancouver Canada
| |
Collapse
|
87
|
Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomedicine 2018; 13:6049-6058. [PMID: 30323592 PMCID: PMC6177375 DOI: 10.2147/ijn.s140462] [Citation(s) in RCA: 390] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia exists to some degree in most solid tumors due to inadequate oxygen delivery of the abnormal vasculature which cannot meet the demands of the rapidly proliferating cancer cells. The levels of oxygenation within the same tumor are highly variable from one area to another and can change over time. Tumor hypoxia is an important impediment to effective cancer therapy. In radiotherapy, the primary mechanism is the creation of reactive oxygen species; hypoxic tumors are therefore radiation resistant. A number of chemotherapeutic drugs have been shown to be less effective when exposed to a hypoxic environment which can lead to further disease progression. Hypoxia is also a potent barrier to effective immunotherapy in cancer treatment. Because of the recognition of hypoxia as an important barrier to cancer treatment, a variety of approaches have been undertaken to overcome or reverse tumor hypoxia. Such approaches have included breathing hyperbaric oxygen, artificial hemoglobins, allosteric hemoglobin modifiers, hypoxia activated prodrugs and fluorocarbons (FCs). These approaches have largely failed due to limited efficacy and/or adverse side effects. Oxygen therapeutics, based on liquid FCs, can potentially increase the oxygen-carrying capacity of the blood to reverse tumor hypoxia. Currently, at least two drugs are in clinical trials to reverse tumor hypoxia; one of these is designed to improve permeability of oxygen into the tumor tissue and the other is based upon a low boiling point FC that transports higher amounts of oxygen per gram than previously tested FCs.
Collapse
|
88
|
Bonnitcha P, Grieve S, Figtree G. Clinical imaging of hypoxia: Current status and future directions. Free Radic Biol Med 2018; 126:296-312. [PMID: 30130569 DOI: 10.1016/j.freeradbiomed.2018.08.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/20/2022]
Abstract
Tissue hypoxia is a key feature of many important causes of morbidity and mortality. In pathologies such as stroke, peripheral vascular disease and ischaemic heart disease, hypoxia is largely a consequence of low blood flow induced ischaemia, hence perfusion imaging is often used as a surrogate for hypoxia to guide clinical diagnosis and treatment. Importantly, ischaemia and hypoxia are not synonymous conditions as it is not universally true that well perfused tissues are normoxic or that poorly perfused tissues are hypoxic. In pathologies such as cancer, for instance, perfusion imaging and oxygen concentration are less well correlated, and oxygen concentration is independently correlated to radiotherapy response and overall treatment outcomes. In addition, the progression of many diseases is intricately related to maladaptive responses to the hypoxia itself. Thus there is potentially great clinical and scientific utility in direct measurements of tissue oxygenation. Despite this, imaging assessment of hypoxia in patients is rarely performed in clinical settings. This review summarises some of the current methods used to clinically evaluate hypoxia, the barriers to the routine use of these methods and the newer agents and techniques being explored for the assessment of hypoxia in pathological processes.
Collapse
Affiliation(s)
- Paul Bonnitcha
- Northern and Central Clinical Schools, Faculty of Medicine, Sydney University, Sydney, NSW 2006, Australia; Chemical Pathology Department, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia.
| | - Stuart Grieve
- Sydney Translational Imaging Laboratory, Heart Research Institute, Charles Perkins Centre and Sydney Medical School, University of Sydney, NSW 2050, Australia
| | - Gemma Figtree
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia; Cardiology Department, Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia
| |
Collapse
|
89
|
Desmet CM, Tran LBA, Danhier P, Gallez B. Characterization of a clinically used charcoal suspension for in vivo EPR oximetry. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2018; 32:205-212. [DOI: 10.1007/s10334-018-0704-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/21/2018] [Accepted: 08/31/2018] [Indexed: 12/18/2022]
|
90
|
Little RA, Jamin Y, Boult JKR, Naish JH, Watson Y, Cheung S, Holliday KF, Lu H, McHugh DJ, Irlam J, West CML, Betts GN, Ashton G, Reynolds AR, Maddineni S, Clarke NW, Parker GJM, Waterton JC, Robinson SP, O’Connor JPB. Mapping Hypoxia in Renal Carcinoma with Oxygen-enhanced MRI: Comparison with Intrinsic Susceptibility MRI and Pathology. Radiology 2018; 288:739-747. [PMID: 29869970 PMCID: PMC6122194 DOI: 10.1148/radiol.2018171531] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/21/2017] [Indexed: 12/28/2022]
Abstract
Purpose To cross-validate T1-weighted oxygen-enhanced (OE) MRI measurements of tumor hypoxia with intrinsic susceptibility MRI measurements and to demonstrate the feasibility of translation of the technique for patients. Materials and Methods Preclinical studies in nine 786-0-R renal cell carcinoma (RCC) xenografts and prospective clinical studies in eight patients with RCC were performed. Longitudinal relaxation rate changes (∆R1) after 100% oxygen inhalation were quantified, reflecting the paramagnetic effect on tissue protons because of the presence of molecular oxygen. Native transverse relaxation rate (R2*) and oxygen-induced R2* change (∆R2*) were measured, reflecting presence of deoxygenated hemoglobin molecules. Median and voxel-wise values of ∆R1 were compared with values of R2* and ∆R2*. Tumor regions with dynamic contrast agent-enhanced MRI perfusion, refractory to signal change at OE MRI (referred to as perfused Oxy-R), were distinguished from perfused oxygen-enhancing (perfused Oxy-E) and nonperfused regions. R2* and ∆R2* values in each tumor subregion were compared by using one-way analysis of variance. Results Tumor-wise and voxel-wise ∆R1 and ∆R2* comparisons did not show correlative relationships. In xenografts, parcellation analysis revealed that perfused Oxy-R regions had faster native R2* (102.4 sec-1 vs 81.7 sec-1) and greater negative ∆R2* (-22.9 sec-1 vs -5.4 sec-1), compared with perfused Oxy-E and nonperfused subregions (all P < .001), respectively. Similar findings were present in human tumors (P < .001). Further, perfused Oxy-R helped identify tumor hypoxia, measured at pathologic analysis, in both xenografts (P = .002) and human tumors (P = .003). Conclusion Intrinsic susceptibility biomarkers provide cross validation of the OE MRI biomarker perfused Oxy-R. Consistent relationship to pathologic analyses was found in xenografts and human tumors, demonstrating biomarker translation. Published under a CC BY 4.0 license. Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ross A. Little
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Yann Jamin
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Jessica K. R. Boult
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Josephine H. Naish
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Yvonne Watson
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Susan Cheung
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Katherine F. Holliday
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Huiqi Lu
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Damien J. McHugh
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Joely Irlam
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Catharine M. L. West
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Guy N. Betts
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Garry Ashton
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | | | - Satish Maddineni
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Noel W. Clarke
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Geoff J. M. Parker
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - John C. Waterton
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - Simon P. Robinson
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| | - James P. B. O’Connor
- From the Centre for Imaging Sciences (R.A.L., J.H.N., Y.W., S.C.,
K.F.H., H.L., D.J.M., G.J.M.P., J.C.W.) and Division of Cancer Sciences (J.I.,
C.M.L.W., N.W.C., J.P.B.O.), University of Manchester, Manchester, England;
Division of Radiotherapy and Imaging, The Institute of Cancer Research, London,
England (Y.J., J.K.R.B., S.P.R.); Department of Pathology, Central Manchester
University Hospitals NHS Foundation Trust, Manchester, England (G.N.B.);
Department of Histology, CRUK Manchester Institute, Manchester, England (G.A.);
Tumour Biology Team, The Breast Cancer Now Toby Robins Research Centre, The
Institute of Cancer Research, London, England (A.R.R.); Department of Urology,
Salford Royal Hospitals NHS Foundation Trust, Salford, England (S.M., N.W.C.);
Bioxydyn Ltd, Manchester, England (G.J.M.P., J.C.W.); and Department of
Radiology, The Christie NHS Foundation Trust, Manchester, England
(J.P.B.O.)
| |
Collapse
|
91
|
Sugiura A, Rathmell JC. Metabolic Barriers to T Cell Function in Tumors. THE JOURNAL OF IMMUNOLOGY 2018; 200:400-407. [PMID: 29311381 DOI: 10.4049/jimmunol.1701041] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
The metabolic programs that drive T cell functions are exquisitely sensitive to cell intrinsic and extrinsic factors, allowing T cells to respond in a fine-tuned manner to a variety of immune challenges and conditions. However, many of the factors essential for effector T cell function are perturbed in the tumor microenvironment, where oncogenic mutations drive unrestrained cancer cell growth that leads to excess nutrient consumption, excess waste excretion, and insufficient oxygen delivery. This imposes metabolic constraints on infiltrating cells that result in dysfunction and loss of potential antitumor activity in both naturally occurring as well as tailored T cells introduced as part of immunotherapy. In this review, we highlight the metabolic properties that characterize tumor-infiltrating T cells, the barriers within the metabolic landscape of the tumor microenvironment, and the opportunities and challenges they present in development of new cancer therapeutics.
Collapse
Affiliation(s)
- Ayaka Sugiura
- Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt-Ingram Cancer Center, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey C Rathmell
- Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt-Ingram Cancer Center, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
92
|
Luan X, Guan Y, Liu H, Lu Q, Zhao M, Sun D, Lovell JF, Sun P, Chen H, Fang C. A Tumor Vascular-Targeted Interlocking Trimodal Nanosystem That Induces and Exploits Hypoxia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800034. [PMID: 30128230 PMCID: PMC6097144 DOI: 10.1002/advs.201800034] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/06/2018] [Indexed: 05/04/2023]
Abstract
Vascular-targeted photodynamic therapy (VTP) is a recently approved strategy for treating solid tumors. However, the exacerbated hypoxic stress makes tumor eradication challenging with such a single modality approach. Here, a new graphene oxide (GO)-based nanosystem for rationally designed, interlocking trimodal cancer therapy that enables VTP using photosensitizer verteporfin (VP) (1) with codelivery of banoxantrone dihydrochloride (AQ4N) (2), a hypoxia-activated prodrug (HAP), and HIF-1α siRNA (siHIF-1α) (3) is reported. The VTP-induced aggravated hypoxia is highly favorable for AQ4N activation into AQ4 (a topoisomerase II inhibitor) for chemotherapy. However, the hypoxia-induced HIF-1α acts as a "hidden brake," through downregulating CYP450 (the dominant HAP-activating reductases), to substantially hinder AQ4N activation. siHIF-1α is rationally adopted to suppress the HIF-1α expression upon hypoxia and further enhance AQ4N activation. This trimodal nanosystem significantly delays the growth of PC-3 tumors in vivo compared to the control nanoparticles carrying VP, AQ4N, or siHIF-1α alone or their pairwise combinations. This multimodal nanoparticle design presents, the first example exploiting VTP to actively induce hypoxia for enhanced HAP activation. It is also revealed that HAP activation is still insufficient under hypoxia due to the hidden downregulation of the HAP-activating reductases (CYP450), and this can be well overcome by GO nanoparticle-mediated siHIF-1α intervention.
Collapse
Affiliation(s)
- Xin Luan
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of MichiganAnn ArborMI48105USA
- Institute of Interdisciplinary Integrative Biomedical ResearchShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Ying‐Yun Guan
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Department of PharmacyRuijin HospitalSJTU‐SM, 197 Rui Jin Er RoadShanghai200025China
| | - Hai‐Jun Liu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| | - Qin Lu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| | - Mei Zhao
- Department of PharmacyShanghai University of Medicine & Health Sciences279 Zhouzhu RoadShanghai201318China
| | - Duxin Sun
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of MichiganAnn ArborMI48105USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Peng Sun
- Department of General SurgeryShanghai Tongren HospitalSJTU‐SM, 1111 Xianxia RoadShanghai200336China
| | - Hong‐Zhuan Chen
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Institute of Interdisciplinary Integrative Biomedical ResearchShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Chao Fang
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| |
Collapse
|
93
|
Baker LCJ, Sikka A, Price JM, Boult JKR, Lepicard EY, Box G, Jamin Y, Spinks TJ, Kramer-Marek G, Leach MO, Eccles SA, Box C, Robinson SP. Evaluating Imaging Biomarkers of Acquired Resistance to Targeted EGFR Therapy in Xenograft Models of Human Head and Neck Squamous Cell Carcinoma. Front Oncol 2018; 8:271. [PMID: 30083516 PMCID: PMC6064942 DOI: 10.3389/fonc.2018.00271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/02/2018] [Indexed: 01/18/2023] Open
Abstract
Background: Overexpression of EGFR is a negative prognostic factor in head and neck squamous cell carcinoma (HNSCC). Patients with HNSCC who respond to EGFR-targeted tyrosine kinase inhibitors (TKIs) eventually develop acquired resistance. Strategies to identify HNSCC patients likely to benefit from EGFR-targeted therapies, together with biomarkers of treatment response, would have clinical value. Methods: Functional MRI and 18F-FDG PET were used to visualize and quantify imaging biomarkers associated with drug response within size-matched EGFR TKI-resistant CAL 27 (CALR) and sensitive (CALS) HNSCC xenografts in vivo, and pathological correlates sought. Results: Intrinsic susceptibility, oxygen-enhanced and dynamic contrast-enhanced MRI revealed significantly slower baseline R 2 ∗ , lower hyperoxia-induced Δ R 2 ∗ and volume transfer constant Ktrans in the CALR tumors which were associated with significantly lower Hoechst 33342 uptake and greater pimonidazole-adduct formation. There was no difference in oxygen-induced ΔR1 or water diffusivity between the CALR and CALS xenografts. PET revealed significantly higher relative uptake of 18F-FDG in the CALR cohort, which was associated with significantly greater Glut-1 expression. Conclusions: CALR xenografts established from HNSCC cells resistant to EGFR TKIs are more hypoxic, poorly perfused and glycolytic than sensitive CALS tumors. MRI combined with PET can be used to non-invasively assess HNSCC response/resistance to EGFR inhibition.
Collapse
Affiliation(s)
- Lauren C. J. Baker
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Arti Sikka
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jonathan M. Price
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jessica K. R. Boult
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Elise Y. Lepicard
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Gary Box
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Yann Jamin
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Terry J. Spinks
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Gabriela Kramer-Marek
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Martin O. Leach
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Suzanne A. Eccles
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Carol Box
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Simon P. Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
94
|
MR-CBCT image-guided system for radiotherapy of orthotopic rat prostate tumors. PLoS One 2018; 13:e0198065. [PMID: 29847586 PMCID: PMC5976174 DOI: 10.1371/journal.pone.0198065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/14/2018] [Indexed: 01/20/2023] Open
Abstract
Multi-modality image-guided radiotherapy is the standard of care in contemporary cancer management; however, it is not common in preclinical settings due to both hardware and software limitations. Soft tissue lesions, such as orthotopic prostate tumors, are difficult to identify using cone beam computed tomography (CBCT) imaging alone. In this study, we characterized a research magnetic resonance (MR) scanner for preclinical studies and created a protocol for combined MR-CBCT image-guided small animal radiotherapy. Two in-house dual-modality, MR and CBCT compatible, phantoms were designed and manufactured using 3D printing technology. The phantoms were used for quality assurance tests and to facilitate end-to-end testing for combined preclinical MR and CBCT based treatment planning. MR and CBCT images of the phantoms were acquired utilizing a Varian 4.7 T scanner and XRad-225Cx irradiator, respectively. The geometry distortion was assessed by comparing MR images to phantom blueprints and CBCT. The corrected MR scans were co-registered with CBCT and subsequently used for treatment planning. The fidelity of 3D printed phantoms compared to the blueprint design yielded favorable agreement as verified with the CBCT measurements. The geometric distortion, which varied between -5% and 11% throughout the scanning volume, was substantially reduced to within 0.4% after correction. The distortion free MR images were co-registered with the corresponding CBCT images and imported into a commercial treatment planning software SmART Plan. The planning target volume (PTV) was on average 19% smaller when contoured on the corrected MR-CBCT images relative to raw images without distortion correction. An MR-CBCT based preclinical workflow was successfully designed and implemented for small animal radiotherapy. Combined MR-CBCT image-guided radiotherapy for preclinical research potentially delivers enhanced relevance to human radiotherapy for various disease sites. This novel protocol is wide-ranging and not limited to the orthotopic prostate tumor study presented in the study.
Collapse
|
95
|
Jarrett AM, Hormuth DA, Barnes SL, Feng X, Huang W, Yankeelov TE. Incorporating drug delivery into an imaging-driven, mechanics-coupled reaction diffusion model for predicting the response of breast cancer to neoadjuvant chemotherapy: theory and preliminary clinical results. Phys Med Biol 2018; 63:105015. [PMID: 29697054 PMCID: PMC5985823 DOI: 10.1088/1361-6560/aac040] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Clinical methods for assessing tumor response to therapy are largely rudimentary, monitoring only temporal changes in tumor size. Our goal is to predict the response of breast tumors to therapy using a mathematical model that utilizes magnetic resonance imaging (MRI) data obtained non-invasively from individual patients. We extended a previously established, mechanically coupled, reaction-diffusion model for predicting tumor response initialized with patient-specific diffusion weighted MRI (DW-MRI) data by including the effects of chemotherapy drug delivery, which is estimated using dynamic contrast-enhanced (DCE-) MRI data. The extended, drug incorporated, model is initialized using patient-specific DW-MRI and DCE-MRI data. Data sets from five breast cancer patients were used-obtained before, after one cycle, and at mid-point of neoadjuvant chemotherapy. The DCE-MRI data was used to estimate spatiotemporal variations in tumor perfusion with the extended Kety-Tofts model. The physiological parameters derived from DCE-MRI were used to model changes in delivery of therapy drugs within the tumor for incorporation in the extended model. We simulated the original model and the extended model in both 2D and 3D and compare the results for this five-patient cohort. Preliminary results show reductions in the error of model predicted tumor cellularity and size compared to the experimentally-measured results for the third MRI scan when therapy was incorporated. Comparing the two models for agreement between the predicted total cellularity and the calculated total cellularity (from the DW-MRI data) reveals an increased concordance correlation coefficient from 0.81 to 0.98 for the 2D analysis and 0.85 to 0.99 for the 3D analysis (p < 0.01 for each) when the extended model was used in place of the original model. This study demonstrates the plausibility of using DCE-MRI data as a means to estimate drug delivery on a patient-specific basis in predictive models and represents a step toward the goal of achieving individualized prediction of tumor response to therapy.
Collapse
Affiliation(s)
- Angela M. Jarrett
- Institute for Computational Engineering and Sciences, The University of Texas at Austin Austin, Texas USA
| | - David A. Hormuth
- Institute for Computational Engineering and Sciences, The University of Texas at Austin Austin, Texas USA
| | - Stephane L. Barnes
- Institute for Computational Engineering and Sciences, The University of Texas at Austin Austin, Texas USA
| | - Xinzeng Feng
- Institute for Computational Engineering and Sciences, The University of Texas at Austin Austin, Texas USA
| | - Wei Huang
- Advanced Imaging Research Center Oregon Health and Science University Portland, Oregon USA
| | - Thomas E. Yankeelov
- Institute for Computational Engineering and Sciences, The University of Texas at Austin Austin, Texas USA
- Livestrong Cancer Institutes, The University of Texas at Austin Austin, Texas USA
- Department of Biomedical Engineering, The University of Texas at Austin Austin, Texas USA
- Department of Oncology, The University of Texas at Austin Austin, Texas USA
- Department of Diagnostic Medicine, The University of Texas at Austin Austin, Texas USA
| |
Collapse
|
96
|
Featherstone AK, O'Connor JP, Little RA, Watson Y, Cheung S, Babur M, Williams KJ, Matthews JC, Parker GJ. Data-driven mapping of hypoxia-related tumor heterogeneity using DCE-MRI and OE-MRI. Magn Reson Med 2018; 79:2236-2245. [PMID: 28856728 PMCID: PMC5836865 DOI: 10.1002/mrm.26860] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 01/06/2023]
Abstract
PURPOSE Previous work has shown that combining dynamic contrast-enhanced (DCE)-MRI and oxygen-enhanced (OE)-MRI binary enhancement maps can identify tumor hypoxia. The current work proposes a novel, data-driven method for mapping tissue oxygenation and perfusion heterogeneity, based on clustering DCE/OE-MRI data. METHODS DCE-MRI and OE-MRI were performed on nine U87 (glioblastoma) and seven Calu6 (non-small cell lung cancer) murine xenograft tumors. Area under the curve and principal component analysis features were calculated and clustered separately using Gaussian mixture modelling. Evaluation metrics were calculated to determine the optimum feature set and cluster number. Outputs were quantitatively compared with a previous non data-driven approach. RESULTS The optimum method located six robustly identifiable clusters in the data, yielding tumor region maps with spatially contiguous regions in a rim-core structure, suggesting a biological basis. Mean within-cluster enhancement curves showed physiologically distinct, intuitive kinetics of enhancement. Regions of DCE/OE-MRI enhancement mismatch were located, and voxel categorization agreed well with the previous non data-driven approach (Cohen's kappa = 0.61, proportional agreement = 0.75). CONCLUSION The proposed method locates similar regions to the previous published method of binarization of DCE/OE-MRI enhancement, but renders a finer segmentation of intra-tumoral oxygenation and perfusion. This could aid in understanding the tumor microenvironment and its heterogeneity. Magn Reson Med 79:2236-2245, 2018. © 2017 The Authors Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- Adam K. Featherstone
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
| | - James P.B. O'Connor
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Division of Cancer StudiesThe University of ManchesterManchesterUK
- Department of RadiologyChristie NHS Foundation TrustManchesterUK
| | - Ross A. Little
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Yvonne Watson
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Sue Cheung
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Muhammad Babur
- Division of Pharmacy & OptometryThe University of ManchesterManchesterUK
| | - Kaye J. Williams
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Division of Pharmacy & OptometryThe University of ManchesterManchesterUK
| | - Julian C. Matthews
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
| | - Geoff J.M. Parker
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Bioxydyn LtdManchesterUK
| |
Collapse
|
97
|
Zhou H, Zhang Z, Denney R, Williams JS, Gerberich J, Stojadinovic S, Saha D, Shelton JM, Mason RP. Tumor physiological changes during hypofractionated stereotactic body radiation therapy assessed using multi-parametric magnetic resonance imaging. Oncotarget 2018; 8:37464-37477. [PMID: 28415581 PMCID: PMC5514922 DOI: 10.18632/oncotarget.16395] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 03/02/2017] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy is a primary treatment for non-resectable lung cancer and hypoxia is thought to influence tumor response. Hypoxia is expected to be particularly relevant to the evolving new radiation treatment scheme of hypofractionated stereotactic body radiation therapy (SBRT). As such, we sought to develop non-invasive tools to assess tumor pathophysiology and response to irradiation. We applied blood oxygen level dependent (BOLD) and tissue oxygen level dependent (TOLD) MRI, together with dynamic contrast enhanced (DCE) MRI to explore the longitudinal effects of SBRT on tumor oxygenation and vascular perfusion using A549 human lung cancer xenografts in a subcutaneous rat model. Intra-tumor heterogeneity was seen on multi-parametric maps, especially in BOLD, T2* and DCE. At baseline, most tumors showed a positive BOLD signal response (%ΔSI) and increased T2* in response to oxygen breathing challenge, indicating increased vascular oxygenation. Control tumors showed similar response 24 hours and 1 week later. Twenty-four hours after a single dose of 12 Gy, the irradiated tumors showed a significantly decreased T2* (-2.9±4.2 ms) and further decrease was observed (-4.0±6.0 ms) after 1 week, suggesting impaired vascular oxygenation. DCE revealed tumor heterogeneity, but showed minimal changes following irradiation. Rats were cured of the primary tumors by 3x12 Gy, providing long term survival, though with ultimate metastatic recurrence.
Collapse
Affiliation(s)
- Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Zhang Zhang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Rebecca Denney
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Jessica S Williams
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Jeni Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Strahinja Stojadinovic
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
98
|
Salem A, Asselin MC, Reymen B, Jackson A, Lambin P, West CML, O'Connor JPB, Faivre-Finn C. Targeting Hypoxia to Improve Non-Small Cell Lung Cancer Outcome. J Natl Cancer Inst 2018; 110:4096546. [PMID: 28922791 DOI: 10.1093/jnci/djx160] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
Oxygen deprivation (hypoxia) in non-small cell lung cancer (NSCLC) is an important factor in treatment resistance and poor survival. Hypoxia is an attractive therapeutic target, particularly in the context of radiotherapy, which is delivered to more than half of NSCLC patients. However, NSCLC hypoxia-targeted therapy trials have not yet translated into patient benefit. Recently, early termination of promising evofosfamide and tarloxotinib bromide studies due to futility highlighted the need for a paradigm shift in our approach to avoid disappointments in future trials. Radiotherapy dose painting strategies based on hypoxia imaging require careful refinement prior to clinical investigation. This review will summarize the role of hypoxia, highlight the potential of hypoxia as a therapeutic target, and outline past and ongoing hypoxia-targeted therapy trials in NSCLC. Evidence supporting radiotherapy dose painting based on hypoxia imaging will be critically appraised. Carefully selected hypoxia biomarkers suitable for integration within future NSCLC hypoxia-targeted therapy trials will be examined. Research gaps will be identified to guide future investigation. Although this review will focus on NSCLC hypoxia, more general discussions (eg, obstacles of hypoxia biomarker research and developing a framework for future hypoxia trials) are applicable to other tumor sites.
Collapse
Affiliation(s)
- Ahmed Salem
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marie-Claude Asselin
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Bart Reymen
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Alan Jackson
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Philippe Lambin
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Catharine M L West
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - James P B O'Connor
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Corinne Faivre-Finn
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
99
|
Zhou H, Arias-Ramos N, López-Larrubia P, Mason RP, Cerdán S, Pacheco-Torres J. Oxygenation Imaging by Nuclear Magnetic Resonance Methods. Methods Mol Biol 2018; 1718:297-313. [PMID: 29341016 DOI: 10.1007/978-1-4939-7531-0_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxygen monitoring is a topic of exhaustive research due to its central role in many biological processes, from energy metabolism to gene regulation. The ability to monitor in vivo the physiological distribution and the dynamics of oxygen from subcellular to macroscopic levels is a prerequisite to better understand the mechanisms associated with both normal and disease states (cancer, neurodegeneration, stroke, etc.). This chapter focuses on magnetic resonance imaging (MRI) based techniques to assess oxygenation in vivo. The first methodology uses injected fluorinated agents to provide quantitative pO2 measurements with high precision and suitable spatial and temporal resolution for many applications. The second method exploits changes in endogenous contrasts, i.e., deoxyhemoglobin and oxygen molecules through measurements of T 2* and T 1, in response to an intervention to qualitatively evaluate hypoxia and its potential modulation.
Collapse
Affiliation(s)
- Heling Zhou
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nuria Arias-Ramos
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Ralph P Mason
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Jesús Pacheco-Torres
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
100
|
Cao-Pham TT, Joudiou N, Van Hul M, Bouzin C, Cani PD, Gallez B, Jordan BF. Combined endogenous MR biomarkers to predict basal tumor oxygenation and response to hyperoxic challenge. NMR IN BIOMEDICINE 2017; 30:e3836. [PMID: 29024086 DOI: 10.1002/nbm.3836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/18/2017] [Accepted: 08/31/2017] [Indexed: 06/07/2023]
Abstract
Hypoxia is a common feature of solid tumors, which translates into increased angiogenesis, malignant phenotype cell selection, change in gene expression and greater resistance to radiotherapy and chemotherapy. Therefore, there is a need for markers of hypoxia to stratify patients, in order to personalize treatment to improve therapeutic outcome. However, no modality has yet been validated for the screening of hypoxia in routine clinical practice. Magnetic resonance imaging (MRI) R1 and R2 * relaxation parameters are sensitive to tissue oxygenation: R1 is sensitive to dissolved oxygen and R2 * is sensitive to intravascular deoxyhemoglobin content. Two rat tumor models with distinct levels of hypoxia, 9L-glioma and rhabdomyosarcoma, were imaged for R1 and R2 * under air and carbogen (95% O2 and 5% CO2 ) breathing conditions. It was observed that the basal tumor oxygenation level had an impact on the amplitude of response to carbogen in the vascular compartment (R2 *), but not in the tissue compartment (R1 ). In addition, the change in tissue oxygenation estimated by ΔR1 correlated with the change in vascular oxygenation estimated by ΔR2 *, which is consistent with an increase in oxygen supply generating an elevated tumor pO2 . At the intra-tumoral level, we identified four types of voxel to which a hypoxic feature was attributed (mild hypoxia, severe hypoxia, normoxia and vascular steal), depending on the carbogen-induced change in R1 and R2 * values for each voxel. The results showed that 9L-gliomas present more normoxic fractions, whereas rhabdomyosarcomas present more hypoxic fractions, which is in accordance with a previous study using 18 F-fluoroazomycin arabinoside (18 F-FAZA) and electron paramagnetic resonance (EPR) oximetry. The response of the combined endogenous MRI contrasts to carbogen challenge could be a useful tool to predict different tumor hypoxic fractions.
Collapse
Affiliation(s)
- Thanh-Trang Cao-Pham
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| | - Nicolas Joudiou
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| | - Matthias Van Hul
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Caroline Bouzin
- Université catholique de Louvain, IREC Imaging Platform, Brussels, Belgium
| | - Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Bernard Gallez
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| | - Bénédicte F Jordan
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| |
Collapse
|