1
|
Luo Z, Huang Y, Chen S, Zhang B, Huang H, Dabiri S, Chen Y, Zhang A, Andreas AR, Li S. Delivery of PARP inhibitors through 2HG-incorporated liposomes for synergistically targeting DNA repair in cancer. Cancer Lett 2024; 604:217268. [PMID: 39321912 DOI: 10.1016/j.canlet.2024.217268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
PARP inhibitors (PARPi) benefit only a small subset of patients with DNA homologous recombination (HR) defects. In addition, long-term administration of a PARPi can lead to the development of drug resistance. 2-Hydroxyglutarate (2HG) has long been known as an oncometabolite but is capable of inducing an HR defect, which makes tumor cells exquisitely sensitive to PARPi. To facilitate the translation of this discovery to the treatment of both HR-deficient and HR-proficient tumors, a liposomal formulation was developed for codelivery of 2HG and veliparib, a PARPi. A sequential loading protocol was developed such that the initial loading of 2HG into liposomes greatly facilitated the subsequent, pH gradient-driven remote loading of veliparib. The liposomes co-loaded with veliparib and 2HG exhibited favorable stability, slow kinetics of drug release, and targeted delivery to the tumor. Furthermore, the veliparib/2HG liposomes demonstrated enhanced anti-tumor activity in both PARPi-resistant BRCA mutant cancer and BRCA wildtype cancer by synergistically enhancing the defect in DNA repair. Moreover, combination of veliparib and 2HG via liposomal co-delivery also augmented the function of cytotoxic T cells by activating the STING pathway and downregulating PD-L1 expression via 2HG-induced hypermethylation.
Collapse
Affiliation(s)
- Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Shangyu Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Bei Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Sheida Dabiri
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Anju Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Alexis R Andreas
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Iacoban CG, Ramaglia A, Severino M, Tortora D, Resaz M, Parodi C, Piccardo A, Rossi A. Advanced imaging techniques and non-invasive biomarkers in pediatric brain tumors: state of the art. Neuroradiology 2024:10.1007/s00234-024-03476-y. [PMID: 39382639 DOI: 10.1007/s00234-024-03476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
In the pediatric age group, brain neoplasms are the second most common tumor category after leukemia, with an annual incidence of 6.13 per 100,000. Conventional MRI sequences, complemented by CT whenever necessary, are fundamental for the initial diagnosis and surgical planning as well as for post-operative evaluations, assessment of response to treatment, and surveillance; however, they have limitations, especially concerning histopathologic or biomolecular phenotyping and grading. In recent years, several advanced MRI sequences, including diffusion-weighted imaging, diffusion tensor imaging, arterial spin labelling (ASL) perfusion, and MR spectroscopy, have emerged as a powerful aid to diagnosis as well as prognostication; furthermore, other techniques such as diffusion kurtosis, amide proton transfer imaging, and MR elastography are being translated from the research environment to clinical practice. Molecular imaging, especially PET with amino-acid tracers, complement MRI in several aspects, including biopsy targeting and outcome prediction. Finally, radiomics with radiogenomics are opening entirely new perspectives for a quantitative approach aiming at identifying biomarkers that can be used for personalized, precision management strategies.
Collapse
Affiliation(s)
| | - Antonia Ramaglia
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Mariasavina Severino
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Domenico Tortora
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Martina Resaz
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Costanza Parodi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Arnoldo Piccardo
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Genoa, Italy
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147, Genoa, Italy.
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy.
| |
Collapse
|
3
|
Yadav N, Mohanty A, V A, Tiwari V. Fractal dimension and lacunarity measures of glioma subcomponents are discriminative of the grade of gliomas and IDH status. NMR IN BIOMEDICINE 2024:e5272. [PMID: 39367752 DOI: 10.1002/nbm.5272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
Since the overall glioma mass and its subcomponents-enhancing region (malignant part of the tumor), non-enhancing (less aggressive tumor cells), necrotic core (dead cells), and edema (water deposition)-are complex and irregular structures, non-Euclidean geometric measures such as fractal dimension (FD or "fractality") and lacunarity are needed to quantify their structural complexity. Fractality measures the extent of structural irregularity, while lacunarity measures the spatial distribution or gaps. The complex geometric patterns of the glioma subcomponents may be closely associated with the grade and molecular landscape. Therefore, we measured FD and lacunarity in the glioma subcomponents and developed machine learning models to discriminate between tumor grades and isocitrate dehydrogenase (IDH) gene status. 3D fractal dimension (FD3D) and lacunarity (Lac3D) were measured for the enhancing, non-enhancing plus necrotic core, and edema-subcomponents using preoperative structural-MRI obtained from the The Cancer Genome Atlas (TCGA) and University of California San Francisco Preoperative Diffuse Glioma MRI (UCSF-PDGM) glioma cohorts. The FD3D and Lac3D measures of the tumor-subcomponents were then compared across glioma grades (HGGs: high-grade gliomas vs. LGGs: low-grade gliomas) and IDH status (mutant vs. wild type). Using these measures, machine learning platforms discriminative of glioma grade and IDH status were developed. Kaplan-Meier survival analysis was used to assess the prognostic significance of FD3D and Lac3D measurements. HGG exhibited significantly higher fractality and lower lacunarity in the enhancing subcomponent, along with lower fractality in the non-enhancing subcomponent compared to LGG. This suggests that a highly irregular and complex geometry in the enhancing-subcomponent is a characteristic feature of HGGs. A comparison of FD3D and Lac3D between IDH-wild type and IDH-mutant gliomas revealed that mutant gliomas had ~2.5-fold lower FD3D in the enhancing-subcomponent and higher FD3D with lower Lac3D in the non-enhancing subcomponent, indicating a less complex and smooth enhancing subcomponent, and a more continuous non-enhancing subcomponent as features of IDH-mutant gliomas. Supervised ML models using FD3D from both the enhancing and non-enhancing subcomponents together demonstrated high-sensitivity in discriminating glioma grades (~97.9%) and IDH status (~94.4%). A combined fractal estimation of the enhancing and non-enhancing subcomponents using MR images could serve as a non-invasive, precise, and quantitative measure for discriminating glioma grade and IDH status. The combination of 2-hydroxyglutarate-magnetic resonance spectroscopy (2HG-MRS) with FD3D and Lac3D quantification may be established as a robust imaging signature for glioma subtyping.
Collapse
Affiliation(s)
- Neha Yadav
- Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Ankit Mohanty
- Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Aswin V
- Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Vivek Tiwari
- Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| |
Collapse
|
4
|
Davies-Jenkins CW, Zöllner HJ, Simicic D, Alcicek S, Edden RA, Oeltzschner G. Data-driven determination of 1H-MRS basis set composition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612503. [PMID: 39314430 PMCID: PMC11419043 DOI: 10.1101/2024.09.11.612503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Purpose Metabolite amplitude estimates derived from linear combination modeling of MR spectra depend upon the precise list of constituent metabolite basis functions used (the "basis set"). The absence of clear consensus on the "ideal" composition or objective criteria to determine the suitability of a particular basis set contributes to the poor reproducibility of MRS. In this proof-of-concept study, we demonstrate a novel, data-driven approach for deciding the basis-set composition using Bayesian information criteria (BIC). Methods We have developed an algorithm that iteratively adds metabolites to the basis set using iterative modeling, informed by BIC scores. We investigated two quantitative "stopping conditions", referred to as max-BIC and zero-amplitude, and whether to optimize the selection of basis set on a per-spectrum basis or at the group level. The algorithm was tested using two groups of synthetic in-vivo-like spectra representing healthy brain and tumor spectra, respectively, and the derived basis sets (and metabolite amplitude estimates) were compared to the ground truth. Results All derived basis sets correctly identified high-concentration metabolites and provided reasonable fits of the spectra. At the single-spectrum level, the two stopping conditions derived the underlying basis set with 77-87% accuracy. When optimizing across a group, basis set determination accuracy improved to 84-92%. Conclusion Data-driven determination of the basis set composition is feasible. With refinement, this approach could provide a valuable data-driven way to derive or refine basis sets, reducing the operator bias of MRS analyses, enhancing the objectivity of quantitative analyses, and increasing the clinical viability of MRS.
Collapse
Affiliation(s)
- Christopher W. Davies-Jenkins
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Helge J. Zöllner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Dunja Simicic
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Seyma Alcicek
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
| | - Richard A.E. Edden
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
5
|
Zhou M, Nie Z, Zhao J, Xiao Y, Hong X, Wang Y, Dong C, Lin AP, Lei Z. Optimization and validation of echo times of point-resolved spectroscopy for cystathionine detection in gliomas. Cancer Imaging 2024; 24:118. [PMID: 39223589 PMCID: PMC11367870 DOI: 10.1186/s40644-024-00764-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Cystathionine accumulates selectively in 1p/19q-codeleted gliomas, and can serve as a possible noninvasive biomarker. This study aims to optimize the echo time (TE) of point-resolved spectroscopy (PRESS) for cystathionine detection in gliomas, and evaluate the diagnostic accuracy of PRESS for 1p/19q-codeletion identification. METHODS The TE of PRESS was optimized with numerical and phantom analysis to better resolve cystathionine from the overlapping aspartate multiplets. The optimized and 97 ms TE PRESS were then applied to 84 prospectively enrolled patients suspected of glioma or glioma recurrence to examine the influence of aspartate on cystathionine quantification by fitting the spectra with and without aspartate. The diagnostic performance of PRESS for 1p/19q-codeleted gliomas were assessed. RESULTS The TE of PRESS was optimized as (TE1, TE2) = (17 ms, 28 ms). The spectral pattern of cystathionine and aspartate were consistent between calculation and phantom. The mean concentrations of cystathionine in vivo fitting without aspartate were significantly higher than those fitting with full basis-set for 97 ms TE PRESS (1.97 ± 2.01 mM vs. 1.55 ± 1.95 mM, p < 0.01), but not significantly different for 45 ms method (0.801 ± 1.217 mM and 0.796 ± 1.217 mM, p = 0.494). The cystathionine concentrations of 45 ms approach was better correlated with those of edited MRS than 97 ms counterparts (r = 0.68 vs. 0.49, both p < 0.01). The sensitivity and specificity for discriminating 1p/19q-codeleted gliomas were 66.7% and 73.7% for 45 ms method, and 44.4% and 52.5% for 97 ms method, respectively. CONCLUSION The 45 ms TE PRESS yields more precise cystathionine estimates than the 97 ms method, and is anticipated to facilitate noninvasive diagnosis of 1p/19q-codeleted gliomas, and treatment response monitoring in those patients. Medium diagnostic performance of PRESS for 1p/19q-codeleted gliomas were observed, and warrants further investigations.
Collapse
Affiliation(s)
- Min Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuang Nie
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Zhao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yao Xiao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaohua Hong
- Tumor Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuhui Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Alexander P Lin
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ziqiao Lei
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Baboli M, Wang F, Dong Z, Dietrich J, Uhlmann EJ, Batchelor TT, Cahill DP, Andronesi OC. Absolute Metabolite Quantification in Individuals with Glioma and Healthy Individuals Using Whole-Brain Three-dimensional MR Spectroscopic and Echo-planar Time-resolved Imaging. Radiology 2024; 312:e232401. [PMID: 39315894 PMCID: PMC11449233 DOI: 10.1148/radiol.232401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND MR spectroscopic imaging (MRSI) can be used to quantify an extended brain metabolic profile but is confounded by changes in tissue water levels due to disease. PURPOSE To develop a fast absolute quantification method for metabolite concentrations combining whole-brain MRSI with echo-planar time-resolved imaging (EPTI) relaxometry in individuals with glioma and healthy individuals. MATERIALS AND METHODS In this prospective study performed from August 2022 to August 2023, using internal water as concentration reference, the MRSI-EPTI quantification method was compared with the conventional method using population-average literature relaxation values. Healthy participants and participants with mutant IDH1 gliomas underwent imaging at 3 T with a 32-channel coil. Real-time navigated adiabatic spiral three-dimensional MRSI scans were acquired in approximately 8 minutes and reconstructed with a super-resolution pipeline to obtain brain metabolic images at 2.4-mm isotropic resolution. High-spatial-resolution multiparametric EPTI was performed in 3 minutes, with 1-mm isotropic resolution, to correct the relaxation and proton density of the water reference signal. Bland-Altman analysis and the Wilcoxon signed rank test were used to compare absolute quantifications from the proposed and conventional methods. RESULTS Six healthy participants (four male; mean age, 37 years ± 11 [SD]) and nine participants with glioma (six male; mean age, 41 years ± 15; one with wild-type IDH1 and eight with mutant IDH1) were included. In healthy participants, there was good agreement (+4% bias) between metabolic concentrations derived using the two methods, with a CI of plus or minus 26%. In participants with glioma, there was large disagreement between the two methods (+39% bias) and a CI of plus or minus 55%. The proposed quantification method improved tumor contrast-to-noise ratio (median values) for total N-acetyl-aspartate (EPTI: 0.541 [95% CI: 0.217, 0.910]; conventional: 0.484 [95% CI: 0.199, 0.823]), total choline (EPTI: 1.053 [95% CI: 0.681, 1.713]; conventional: 0.940 [95% CI: 0.617, 1.295]), and total creatine (EPTI: 0.745 [95% CI: 0.628, 0.909]; conventional: 0.553 [95% CI: 0.444, 0.828]) (P = .03 for all). CONCLUSION The whole-brain MRSI-EPTI method provided fast absolute quantification of metabolic concentrations with individual-specific corrections at 2.4-mm isotropic resolution, yielding concentrations closer to the true value in disease than the conventional literature-based corrections. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Mehran Baboli
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Fuyixue Wang
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Zijing Dong
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Jorg Dietrich
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Erik J Uhlmann
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Tracy T Batchelor
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Daniel P Cahill
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Ovidiu C Andronesi
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| |
Collapse
|
7
|
Alcicek S, Pilatus U, Manzhurtsev A, Weber KJ, Ronellenfitsch MW, Steinbach JP, Hattingen E, Wenger KJ. Amino acid metabolism in glioma: in vivo MR-spectroscopic detection of alanine as a potential biomarker of poor survival in glioma patients. J Neurooncol 2024:10.1007/s11060-024-04803-2. [PMID: 39192067 DOI: 10.1007/s11060-024-04803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
PURPOSE Reprogramming of amino acid metabolism is relevant for initiating and fueling tumor formation and growth. Therefore, there has been growing interest in anticancer therapies targeting amino acid metabolism. While developing personalized therapeutic approaches to glioma, in vivo proton magnetic resonance spectroscopy (MRS) is a valuable tool for non-invasive monitoring of tumor metabolism. Here, we evaluated MRS-detected brain amino acids and myo-inositol as potential diagnostic and prognostic biomarkers in glioma. METHOD We measured alanine, glycine, glutamate, glutamine, and myo-inositol in 38 patients with MRI-suspected glioma using short and long echo-time single-voxel PRESS MRS sequences. The detectability of alanine, glycine, and myo-inositol and the (glutamate + glutamine)/total creatine ratio were evaluated against the patients' IDH mutation status, CNS WHO grade, and overall survival. RESULTS While the detection of alanine and non-detection of myo-inositol significantly correlated with IDH wildtype (p = 0.0008, p = 0.007, respectively) and WHO grade 4 (p = 0.01, p = 0.04, respectively), glycine detection was not significantly associated with either. The ratio of (glutamate + glutamine)/total creatine was significantly higher in WHO grade 4 than in 2 and 3. We found that the overall survival was significantly shorter in glioma patients with alanine detection (p = 0.00002). CONCLUSION Focusing on amino acids in MRS can improve its diagnostic and prognostic value in glioma. Alanine, which is visible at long TE even in the presence of lipids, could be a relevant indicator for overall survival.
Collapse
Affiliation(s)
- Seyma Alcicek
- Goethe University Frankfurt, University Hospital, Institute of Neuroradiology, Schleusenweg 2-16, 60528, Frankfurt/Main, Germany.
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany.
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany.
| | - Ulrich Pilatus
- Goethe University Frankfurt, University Hospital, Institute of Neuroradiology, Schleusenweg 2-16, 60528, Frankfurt/Main, Germany
| | - Andrei Manzhurtsev
- Goethe University Frankfurt, University Hospital, Institute of Neuroradiology, Schleusenweg 2-16, 60528, Frankfurt/Main, Germany
| | - Katharina J Weber
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany
- Goethe University Frankfurt, University Hospital, Institute of Neurology (Edinger-Institute), Frankfurt/Main, Germany
| | - Michael W Ronellenfitsch
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt/Main, Germany
| | - Joachim P Steinbach
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt/Main, Germany
| | - Elke Hattingen
- Goethe University Frankfurt, University Hospital, Institute of Neuroradiology, Schleusenweg 2-16, 60528, Frankfurt/Main, Germany
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany
| | - Katharina J Wenger
- Goethe University Frankfurt, University Hospital, Institute of Neuroradiology, Schleusenweg 2-16, 60528, Frankfurt/Main, Germany
- University Cancer Center Frankfurt (UCT), Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- German Cancer Research Center (DKFZ) Heidelberg, German Cancer Consortium (DKTK), Partner Site, Frankfurt/Mainz, Germany
| |
Collapse
|
8
|
Schiff D. Low-Grade Gliomas: A New Mutation, New Targeted Therapy, and Many Questions. Neurology 2024; 103:e209688. [PMID: 39008801 DOI: 10.1212/wnl.0000000000209688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
The discovery in 2008 that many adult gliomas harbor a hitherto unknown mutation in the metabolic gene isocitrate dehydrogenase (IDH) initiated revolutionary advances in our understanding of the biology, and correspondingly our classification, of gliomas. IDH mutations are found in most nonglioblastoma adult gliomas and portend a better prognosis. Massive efforts have unraveled many of the pleiotropic cellular effects of these mutations and spawned several lines of investigation to target the effect to therapeutic benefit. In this article are reviewed the implications of the IDH mutation in gliomas, in particular focusing on recent studies that have culminated in a rare positive phase 3 trial in these generally refractory tumors.
Collapse
Affiliation(s)
- David Schiff
- From the Departments of Neurology, Neurological Surgery, and Medicine, University of Virginia Health System
| |
Collapse
|
9
|
Richardson TE, Walker JM, Hambardzumyan D, Brem S, Hatanpaa KJ, Viapiano MS, Pai B, Umphlett M, Becher OJ, Snuderl M, McBrayer SK, Abdullah KG, Tsankova NM. Genetic and epigenetic instability as an underlying driver of progression and aggressive behavior in IDH-mutant astrocytoma. Acta Neuropathol 2024; 148:5. [PMID: 39012509 PMCID: PMC11252228 DOI: 10.1007/s00401-024-02761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024]
Abstract
In recent years, the classification of adult-type diffuse gliomas has undergone a revolution, wherein specific molecular features now represent defining diagnostic criteria of IDH-wild-type glioblastomas, IDH-mutant astrocytomas, and IDH-mutant 1p/19q-codeleted oligodendrogliomas. With the introduction of the 2021 WHO CNS classification, additional molecular alterations are now integrated into the grading of these tumors, given equal weight to traditional histologic features. However, there remains a great deal of heterogeneity in patient outcome even within these established tumor subclassifications that is unexplained by currently codified molecular alterations, particularly in the IDH-mutant astrocytoma category. There is also significant intercellular genetic and epigenetic heterogeneity and plasticity with resulting phenotypic heterogeneity, making these tumors remarkably adaptable and robust, and presenting a significant barrier to the design of effective therapeutics. Herein, we review the mechanisms and consequences of genetic and epigenetic instability, including chromosomal instability (CIN), microsatellite instability (MSI)/mismatch repair (MMR) deficits, and epigenetic instability, in the underlying biology, tumorigenesis, and progression of IDH-mutant astrocytomas. We also discuss the contribution of recent high-resolution transcriptomics studies toward defining tumor heterogeneity with single-cell resolution. While intratumoral heterogeneity is a well-known feature of diffuse gliomas, the contribution of these various processes has only recently been considered as a potential driver of tumor aggressiveness. CIN has an independent, adverse effect on patient survival, similar to the effect of histologic grade and homozygous CDKN2A deletion, while MMR mutation is only associated with poor overall survival in univariate analysis but is highly correlated with higher histologic/molecular grade and other aggressive features. These forms of genomic instability, which may significantly affect the natural progression of these tumors, response to therapy, and ultimately clinical outcome for patients, are potentially measurable features which could aid in diagnosis, grading, prognosis, and development of personalized therapeutics.
Collapse
Affiliation(s)
- Timothy E Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA.
| | - Jamie M Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
- Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kimmo J Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mariano S Viapiano
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Balagopal Pai
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Melissa Umphlett
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
| | - Oren J Becher
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health, New York, NY, 10016, USA
| | - Samuel K McBrayer
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Nadejda M Tsankova
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
10
|
Lane AN, Higashi RM, Fan TWM. Challenges of Spatially Resolved Metabolism in Cancer Research. Metabolites 2024; 14:383. [PMID: 39057706 PMCID: PMC11278851 DOI: 10.3390/metabo14070383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Stable isotope-resolved metabolomics comprises a critical set of technologies that can be applied to a wide variety of systems, from isolated cells to whole organisms, to define metabolic pathway usage and responses to perturbations such as drugs or mutations, as well as providing the basis for flux analysis. As the diversity of stable isotope-enriched compounds is very high, and with newer approaches to multiplexing, the coverage of metabolism is now very extensive. However, as the complexity of the model increases, including more kinds of interacting cell types and interorgan communication, the analytical complexity also increases. Further, as studies move further into spatially resolved biology, new technical problems have to be overcome owing to the small number of analytes present in the confines of a single cell or cell compartment. Here, we review the overall goals and solutions made possible by stable isotope tracing and their applications to models of increasing complexity. Finally, we discuss progress and outstanding difficulties in high-resolution spatially resolved tracer-based metabolic studies.
Collapse
Affiliation(s)
- Andrew N. Lane
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, USA; (R.M.H.); (T.W.-M.F.)
| | | | | |
Collapse
|
11
|
Galldiks N, Lohmann P, Friedrich M, Werner JM, Stetter I, Wollring MM, Ceccon G, Stegmayr C, Krause S, Fink GR, Law I, Langen KJ, Tonn JC. PET imaging of gliomas: Status quo and quo vadis? Neuro Oncol 2024:noae078. [PMID: 38970818 DOI: 10.1093/neuonc/noae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024] Open
Abstract
PET imaging, particularly using amino acid tracers, has become a valuable adjunct to anatomical MRI in the clinical management of patients with glioma. Collaborative international efforts have led to the development of clinical and technical guidelines for PET imaging in gliomas. The increasing readiness of statutory health insurance agencies, especially in European countries, to reimburse amino acid PET underscores its growing importance in clinical practice. Integrating artificial intelligence and radiomics in PET imaging of patients with glioma may significantly improve tumor detection, segmentation, and response assessment. Efforts are ongoing to facilitate the clinical translation of these techniques. Considerable progress in computer technology developments (eg quantum computers) may be helpful to accelerate these efforts. Next-generation PET scanners, such as long-axial field-of-view PET/CT scanners, have improved image quality and body coverage and therefore expanded the spectrum of indications for PET imaging in Neuro-Oncology (eg PET imaging of the whole spine). Encouraging results of clinical trials in patients with glioma have prompted the development of PET tracers directing therapeutically relevant targets (eg the mutant isocitrate dehydrogenase) for novel anticancer agents in gliomas to improve response assessment. In addition, the success of theranostics for the treatment of extracranial neoplasms such as neuroendocrine tumors and prostate cancer has currently prompted efforts to translate this approach to patients with glioma. These advancements highlight the evolving role of PET imaging in Neuro-Oncology, offering insights into tumor biology and treatment response, thereby informing personalized patient care. Nevertheless, these innovations warrant further validation in the near future.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Michel Friedrich
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Jan-Michael Werner
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Isabelle Stetter
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Michael M Wollring
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Sandra Krause
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
12
|
Galldiks N, Kaufmann TJ, Vollmuth P, Lohmann P, Smits M, Veronesi MC, Langen KJ, Rudà R, Albert NL, Hattingen E, Law I, Hutterer M, Soffietti R, Vogelbaum MA, Wen PY, Weller M, Tonn JC. Challenges, limitations, and pitfalls of PET and advanced MRI in patients with brain tumors: A report of the PET/RANO group. Neuro Oncol 2024; 26:1181-1194. [PMID: 38466087 PMCID: PMC11226881 DOI: 10.1093/neuonc/noae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Indexed: 03/12/2024] Open
Abstract
Brain tumor diagnostics have significantly evolved with the use of positron emission tomography (PET) and advanced magnetic resonance imaging (MRI) techniques. In addition to anatomical MRI, these modalities may provide valuable information for several clinical applications such as differential diagnosis, delineation of tumor extent, prognostication, differentiation between tumor relapse and treatment-related changes, and the evaluation of response to anticancer therapy. In particular, joint recommendations of the Response Assessment in Neuro-Oncology (RANO) Group, the European Association of Neuro-oncology, and major European and American Nuclear Medicine societies highlighted that the additional clinical value of radiolabeled amino acids compared to anatomical MRI alone is outstanding and that its widespread clinical use should be supported. For advanced MRI and its steadily increasing use in clinical practice, the Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition provided more recently an updated acquisition protocol for the widely used dynamic susceptibility contrast perfusion MRI. Besides amino acid PET and perfusion MRI, other PET tracers and advanced MRI techniques (e.g. MR spectroscopy) are of considerable clinical interest and are increasingly integrated into everyday clinical practice. Nevertheless, these modalities have shortcomings which should be considered in clinical routine. This comprehensive review provides an overview of potential challenges, limitations, and pitfalls associated with PET imaging and advanced MRI techniques in patients with gliomas or brain metastases. Despite these issues, PET imaging and advanced MRI techniques continue to play an indispensable role in brain tumor management. Acknowledging and mitigating these challenges through interdisciplinary collaboration, standardized protocols, and continuous innovation will further enhance the utility of these modalities in guiding optimal patient care.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | | | - Philipp Vollmuth
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Marion Smits
- Department of Radiology and Nuclear Medicine and Brain Tumour Center, Erasmus MC, Rotterdam, The Netherlands
| | - Michael C Veronesi
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilians-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Hattingen
- Goethe University, Department of Neuroradiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Markus Hutterer
- Department of Neurology with Acute Geriatrics, Saint John of God Hospital, Linz, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Michael A Vogelbaum
- Department of Neuro-Oncology and Neurosurgery, Moffit Cancer Center, Tampa, Florida, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, and University Hospital of Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Joerg-Christian Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
13
|
Iqbal Z, Albuquerque K, Chan KL. Magnetic Resonance Spectroscopy for Cervical Cancer: Review and Potential Prognostic Applications. Cancers (Basel) 2024; 16:2141. [PMID: 38893260 PMCID: PMC11171343 DOI: 10.3390/cancers16112141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
This review article investigates the utilization of MRS in the setting of cervical cancer. A variety of different techniques have been used in this space including single-voxel techniques such as point-resolved spectroscopy (PRESS) and stimulated echo acquisition mode spectroscopy (STEAM). Furthermore, the experimental parameters for these acquisitions including field strength, repetition times (TR), and echo times (TE) vary greatly. This study critically examines eleven MRS studies that focus on cervical cancer. Out of the eleven studies, ten studies utilized PRESS acquisition, while the remaining study used STEAM acquisition. These studies generally showed that the choline signal is altered in cervical cancer (4/11 studies), the lipid signal is generally increased in cervical cancer or the lipid distribution is changed (5/11 studies), and that diffusion-weighted imaging (DWI) can quantitatively detect lower apparent diffusion coefficient (ADC) values in cervical cancer (2/11 studies). Two studies also investigated the role of MRS for monitoring treatment response and demonstrated mixed results regarding choline signal, and one of these studies showed increased lipid signal for non-responders. There are several new MRS technologies that have yet to be implemented for cervical cancer including advanced spectroscopic imaging and artificial intelligence, and those technologies are also discussed in the article.
Collapse
Affiliation(s)
- Zohaib Iqbal
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Kevin Albuquerque
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Kimberly L. Chan
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| |
Collapse
|
14
|
Nayarisseri A, Bandaru S, Khan A, Sharma K, Bhrdwaj A, Kaur M, Ghosh D, Chopra I, Panicker A, Kumar A, Saravanan P, Belapurkar P, Mendonça Junior FJB, Singh SK. Epigenetic dysregulation in cancers by isocitrate dehydrogenase 2 (IDH2). ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:223-253. [PMID: 38960475 DOI: 10.1016/bs.apcsb.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Recent advances in genome-wide studies have revealed numerous epigenetic regulations brought about by genes involved in cellular metabolism. Isocitrate dehydrogenase (IDH), an essential enzyme, that converts isocitrate into -ketoglutarate (KG) predominantly in the tricarboxylic acid (TCA) cycle, has gained particular importance due to its cardinal role in the metabolic pathway in cells. IDH1, IDH2, and IDH3 are the three isomeric IDH enzymes that have been shown to regulate cellular metabolism. Of particular importance, IDH2 genes are associated with several cancers, including gliomas, oligodendroglioma, and astrocytomas. These mutations lead to the production of oncometabolite D-2-hydroxyglutarate (D-2-HG), which accumulates in cells promoting tumor growth. The enhanced levels of D-2-HG competitively inhibit α-KG dependent enzymes, inhibiting cell TCA cycle, upregulating the cell growth and survival relevant HIF-1α pathway, promoting DNA hypermethylation related epigenetic activity, all of which synergistically contribute to carcinogenesis. The present review discusses epigenetic mechanisms inIDH2 regulation in cells and further its clinical implications.
Collapse
Affiliation(s)
- Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Bioinformatics Research Laboratory, LeGene Biosciences Pvt Ltd, Indore, Madhya Pradesh, India.
| | - Srinivas Bandaru
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Department of Biotechnology, Koneru Lakshmaiah Educational Foundation (KLEF), Green Fields, Vaddeswaram, Andhra Pradesh, India
| | - Arshiya Khan
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Khushboo Sharma
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Anushka Bhrdwaj
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Manmeet Kaur
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Dipannita Ghosh
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Ishita Chopra
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Aravind Panicker
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Abhishek Kumar
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India; Department of Biosciences, Acropolis Institute, Indore, Madhya Pradesh, India
| | - Priyadevi Saravanan
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Pranoti Belapurkar
- Department of Biosciences, Acropolis Institute, Indore, Madhya Pradesh, India
| | | | - Sanjeev Kumar Singh
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
15
|
Martinez Luque E, Liu Z, Sung D, Goldberg RM, Agarwal R, Bhattacharya A, Ahmed NS, Allen JW, Fleischer CC. An Update on MR Spectroscopy in Cancer Management: Advances in Instrumentation, Acquisition, and Analysis. Radiol Imaging Cancer 2024; 6:e230101. [PMID: 38578207 PMCID: PMC11148681 DOI: 10.1148/rycan.230101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 04/06/2024]
Abstract
MR spectroscopy (MRS) is a noninvasive imaging method enabling chemical and molecular profiling of tissues in a localized, multiplexed, and nonionizing manner. As metabolic reprogramming is a hallmark of cancer, MRS provides valuable metabolic and molecular information for cancer diagnosis, prognosis, treatment monitoring, and patient management. This review provides an update on the use of MRS for clinical cancer management. The first section includes an overview of the principles of MRS, current methods, and conventional metabolites of interest. The remainder of the review is focused on three key areas: advances in instrumentation, specifically ultrahigh-field-strength MRI scanners and hybrid systems; emerging methods for acquisition, including deuterium imaging, hyperpolarized carbon 13 MRI and MRS, chemical exchange saturation transfer, diffusion-weighted MRS, MR fingerprinting, and fast acquisition; and analysis aided by artificial intelligence. The review concludes with future recommendations to facilitate routine use of MRS in cancer management. Keywords: MR Spectroscopy, Spectroscopic Imaging, Molecular Imaging in Oncology, Metabolic Reprogramming, Clinical Cancer Management © RSNA, 2024.
Collapse
Affiliation(s)
- Eva Martinez Luque
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Zexuan Liu
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Dongsuk Sung
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Rachel M. Goldberg
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Rishab Agarwal
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Aditya Bhattacharya
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Nadine S. Ahmed
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Jason W. Allen
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Candace C. Fleischer
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| |
Collapse
|
16
|
Dowdy T, Larion M. Resolving Challenges in Detection and Quantification of D-2-hydroxyglutarate and L-2-hydroxyglutarate via LC/MS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591335. [PMID: 38903117 PMCID: PMC11188093 DOI: 10.1101/2024.04.26.591335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
D-2-Hydroxyglutarate and L-2-Hydroxyglutarate (D-2HG/L-2HG) are typically metabolites of non-specific enzymatic reactions that are kept in check by the housekeeping enzymes, D-2HG /L-2HG dehydrogenase (D-2HGDH/L-2HGDH). In certain disease states, such as D-2HG or L-2HG aciduria and cancers, accumulation of these biomarkers interferes with oxoglutarate-dependent enzymes that regulate bioenergetic metabolism, histone methylation, post-translational modification, protein expression and others. D-2HG has a complex role in tumorigenesis that drives metabolomics investigations. Meanwhile, L-2HG is produced by non-specific action of malate dehydrogenase and lactate dehydrogenase under acidic or hypoxic environments. Characterization of divergent effects of D-2HG/L-2HG on the activity of specific enzymes in diseased metabolism depends on their accurate quantification via mass spectrometry. Despite advancements in high-resolution quadrupole time-of-flight mass spectrometry (HR-QTOF-MS), challenges are typically encountered when attempting to resolve of isobaric and isomeric metabolites such as D-2HG/L-2HG for quantitative analysis. Herein, available D-2HG/L-2HG derivatization and liquid chromatography (LC) MS quantification methods were examined. The outcome led to the development of a robust, high-throughput HR-QTOF-LC/MS approach that permits concomitant quantification of the D-2HG and L-2HG enantiomers with the benefit to quantify the dysregulation of other intermediates within interconnecting pathways. Calibration curve was obtained over the linear range of 0.8-104 nmol/mL with r 2 ≥ 0.995 for each enantiomer. The LC/MS-based assay had an overall precision with intra-day CV % ≤ 8.0 and inter-day CV % ≤ 6.3 across the quality control level for commercial standard and pooled biological samples; relative error % ≤ 2.7 for accuracy; and resolution, R s = 1.6 between 2HG enantiomers (m/z 147.030), D-2HG and L-2HG (at retention time of 5.82 min and 4.75 min, respectively) following chiral derivatization with diacetyl-L-tartaric anhydride (DATAN). Our methodology was applied to disease relevant samples to illustrate the implications of proper enantioselective quantification of both D-2HG and L-2HG. The stability of the method allows scaling to large cohorts of clinical samples in the future.
Collapse
|
17
|
Bauer J, Raum HN, Kugel H, Müther M, Mannil M, Heindel W. 2-Hydroxyglutarate as an MR spectroscopic predictor of an IDH mutation in gliomas. ROFO-FORTSCHR RONTG 2024. [PMID: 38648790 DOI: 10.1055/a-2285-4923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The mutated enzyme isocitrate dehydrogenase (IDH) 1 and 2 has been detected in various tumor entities such as gliomas and can convert α-ketoglutarate into the oncometabolite 2-hydroxyglutarate (2-HG). This neuro-oncologically significant metabolic product can be detected by MR spectroscopy and is therefore suitable for noninvasive glioma classification and therapy monitoring.This paper provides an up-to-date overview of the methodology and relevance of 1H-MR spectroscopy (MRS) in the oncological primary and follow-up diagnosis of gliomas. The possibilities and limitations of this MR spectroscopic examination are evaluated on the basis of the available literature.By detecting 2-HG, MRS can in principle offer a noninvasive alternative to immunohistological analysis thus avoiding surgical intervention in some cases. However, in addition to an adapted and optimized examination protocol, the individual measurement conditions in the examination region are of decisive importance. Due to the inherently small signal of 2-HG, unfavorable measurement conditions can influence the reliability of detection. · MR spectroscopy enables the non-invasive detection of 2-hydroxyglutarate.. · The measurement of this metabolite allows the detection of an IDH mutation in gliomas.. · The choice of MR examination method is particularly important.. · Detection reliability is influenced by glioma size, necrotic tissue and the existing measurement conditions.. · Bauer J, Raum HN, Kugel H et al. 2-Hydroxyglutarate as an MR spectroscopic predictor of an IDH mutation in gliomas. Fortschr Röntgenstr 2024; DOI 10.1055/a-2285-4923.
Collapse
Affiliation(s)
- Jochen Bauer
- Clinic for Radiology, University of Münster and University Hospital Münster, Münster, Germany
| | - Heiner N Raum
- Clinic for Radiology, University of Münster and University Hospital Münster, Münster, Germany
| | - Harald Kugel
- Clinic for Radiology, University of Münster and University Hospital Münster, Münster, Germany
| | - Michael Müther
- Department of Neurosurgery, University of Münster and University Hospital Münster, Münster, Germany
| | - Manoj Mannil
- Clinic for Radiology, University of Münster and University Hospital Münster, Münster, Germany
- Institute for Diagnostic and Interventional Radiology, Caritas Hospital Bad Mergentheim, Bad Mergentheim, Germany
| | - Walter Heindel
- Clinic for Radiology, University of Münster and University Hospital Münster, Münster, Germany
| |
Collapse
|
18
|
Valerius AR, Webb MJ, Hammad N, Sener U, Malani R. Cerebrospinal Fluid Liquid Biopsies in the Evaluation of Adult Gliomas. Curr Oncol Rep 2024; 26:377-390. [PMID: 38488990 DOI: 10.1007/s11912-024-01517-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE OF REVIEW This review aims to discuss recent research regarding the biomolecules explored in liquid biopsies and their potential clinical uses for adult-type diffuse gliomas. RECENT FINDINGS Evaluation of tumor biomolecules via cerebrospinal fluid (CSF) is an emerging technology in neuro-oncology. Studies to date have already identified various circulating tumor DNA, extracellular vesicle, micro-messenger RNA and protein biomarkers of interest. These biomarkers show potential to assist in multiple avenues of central nervous system (CNS) tumor evaluation, including tumor differentiation and diagnosis, treatment selection, response assessment, detection of tumor progression, and prognosis. In addition, CSF liquid biopsies have the potential to better characterize tumor heterogeneity compared to conventional tissue collection and CNS imaging. Current imaging modalities are not sufficient to establish a definitive glioma diagnosis and repeated tissue sampling via conventional biopsy is risky, therefore, there is a great need to improve non-invasive and minimally invasive sampling methods. CSF liquid biopsies represent a promising, minimally invasive adjunct to current approaches which can provide diagnostic and prognostic information as well as aid in response assessment.
Collapse
Affiliation(s)
| | - Mason J Webb
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Nouran Hammad
- Jordan University of Science and Technology School of Medicine, Irbid, Jordan
| | - Ugur Sener
- Department of Neurology, Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Rachna Malani
- University of UT - Huntsman Cancer Institute (Department of Neurosurgery), Salt Lake City, UT, USA
| |
Collapse
|
19
|
Belle R, Saraç H, Salah E, Bhushan B, Szykowska A, Roper G, Tumber A, Kriaucionis S, Burgess-Brown N, Schofield CJ, Brown T, Kawamura A. Focused Screening Identifies Different Sensitivities of Human TET Oxygenases to the Oncometabolite 2-Hydroxyglutarate. J Med Chem 2024; 67:4525-4540. [PMID: 38294854 PMCID: PMC10983004 DOI: 10.1021/acs.jmedchem.3c01820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/10/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024]
Abstract
Ten-eleven translocation enzymes (TETs) are Fe(II)/2-oxoglutarate (2OG) oxygenases that catalyze the sequential oxidation of 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine in eukaryotic DNA. Despite their roles in epigenetic regulation, there is a lack of reported TET inhibitors. The extent to which 2OG oxygenase inhibitors, including clinically used inhibitors and oncometabolites, modulate DNA modifications via TETs has been unclear. Here, we report studies on human TET1-3 inhibition by a set of 2OG oxygenase-focused inhibitors, employing both enzyme-based and cellular assays. Most inhibitors manifested similar potencies for TET1-3 and caused increases in cellular 5hmC levels. (R)-2-Hydroxyglutarate, an oncometabolite elevated in isocitrate dehydrogenase mutant cancer cells, showed different degrees of inhibition, with TET1 being less potently inhibited than TET3 and TET2, potentially reflecting the proposed role of TET2 mutations in tumorigenesis. The results highlight the tractability of TETs as drug targets and provide starting points for selective inhibitor design.
Collapse
Affiliation(s)
- Roman Belle
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Chemistry
− School of Natural and Environmental Sciences, Bedson Building, Newcastle University, NE1 7RU Newcastle upon Tyne, United Kingdom
| | - Hilal Saraç
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Chemistry
− School of Natural and Environmental Sciences, Bedson Building, Newcastle University, NE1 7RU Newcastle upon Tyne, United Kingdom
- Radcliffe
Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human
Genetics, Roosevelt Drive, OX3 7BN Oxford, United Kingdom
| | - Eidarus Salah
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Centre
for Medicines Discovery, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, OX3 7DQ Oxford, United Kingdom
| | - Bhaskar Bhushan
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Radcliffe
Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human
Genetics, Roosevelt Drive, OX3 7BN Oxford, United Kingdom
| | - Aleksandra Szykowska
- Centre
for Medicines Discovery, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, OX3 7DQ Oxford, United Kingdom
| | - Grace Roper
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Chemistry
− School of Natural and Environmental Sciences, Bedson Building, Newcastle University, NE1 7RU Newcastle upon Tyne, United Kingdom
| | - Anthony Tumber
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Ineos Oxford
Institute for Antimicrobial Research, University
of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
| | - Skirmantas Kriaucionis
- Ludwig
Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, OX3 7DQ Oxford, United Kingdom
| | - Nicola Burgess-Brown
- Centre
for Medicines Discovery, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, OX3 7DQ Oxford, United Kingdom
| | - Christopher J. Schofield
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Ineos Oxford
Institute for Antimicrobial Research, University
of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
| | - Tom Brown
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
| | - Akane Kawamura
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, OX1 3TA Oxford, United Kingdom
- Chemistry
− School of Natural and Environmental Sciences, Bedson Building, Newcastle University, NE1 7RU Newcastle upon Tyne, United Kingdom
- Radcliffe
Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human
Genetics, Roosevelt Drive, OX3 7BN Oxford, United Kingdom
| |
Collapse
|
20
|
Sawlani V, Jen JP, Patel M, Jain M, Haq H, Ughratdar I, Wykes V, Nagaraju S, Watts C, Pohl U. Multiparametric MRI and T2/FLAIR mismatch complements the World Health Organization 2021 classification for the diagnosis of IDH-mutant 1p/19q non-co-deleted/ATRX-mutant astrocytoma. Clin Radiol 2024; 79:197-204. [PMID: 38101998 DOI: 10.1016/j.crad.2023.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/14/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
AIM To investigate whether T2-weighted imaging-fluid-attenuated inversion recovery (T2/FLAIR) mismatch, T2∗ dynamic susceptibility contrast (DSC) perfusion, and magnetic resonance spectroscopy (MRS) correlated with the histological diagnosis and grading of IDH (isocitrate dehydrogenase)-mutant, 1p/19q non-co-deleted/ATRX (alpha-thalassemia mental retardation X-linked)-mutant astrocytoma. MATERIALS Imaging of 101 IDH-mutant diffuse glioma cases of histological grades 2-3 (2019-2021) were analysed retrospectively by two neuroradiologists blinded to the molecular diagnosis. T2/FLAIR mismatch sign is used for radio-phenotyping, and pre-biopsy multiparametric MRI images were assessed for grading purposes. Cut-off values pre-determined for radiologically high-grade lesions were relative cerebral blood volume (rCBV) ≥2, choline/creatine ratio (Cho/Cr) ≥1.5 (30 ms echo time [TE]), Cho/Cr ≥1.8 (135 ms TE). RESULTS Sixteen of the 101 cases showed T2/FLAIR mismatch, all of which were histogenetically confirmed IDH-mutant 1p/19q non-co-deleted/ATRX mutant astrocytomas; 50% were grade 3 (8/16) and 50% grade 2 (8/16). None showed contrast enhancement. Nine of the 16 had adequate multiparametric MRI for analysis. Any positive value by combining rCBV ≥2 with Cho/Cr ≥1.5 (30 ms TE) or Cho/Cr ≥1.8 (135 ms TE) predicted grade 3 histology with sensitivity, specificity, positive predictive value, negative predictive value, and accuracy of 100%. CONCLUSION The T2/FLAIR mismatch sign detected diffuse astrocytomas with 100% specificity. When combined with high Cho/Cr and raised rCBV, this predicted histological grading with high accuracy. The future direction for imaging should explore a similar integrated layered approach of 2021 classification of central nervous system (CNS) tumours combining radio-phenotyping and grading from structural and multiparametric imaging.
Collapse
Affiliation(s)
- V Sawlani
- Department of Neuroradiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK; Department of Imaging, Neurosurgery and Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK.
| | - J P Jen
- Department of Neuroradiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - M Patel
- Department of Neuroradiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK; Department of Imaging, Neurosurgery and Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - M Jain
- Department of Neuroradiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - H Haq
- Department of Neuroradiology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - I Ughratdar
- Department of Imaging, Neurosurgery and Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK; Department of Neurosurgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - V Wykes
- Department of Imaging, Neurosurgery and Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK; Department of Neurosurgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - S Nagaraju
- Department of Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - C Watts
- Department of Imaging, Neurosurgery and Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK; Department of Neurosurgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - U Pohl
- Department of Neuropathology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS FT, Birmingham, UK
| |
Collapse
|
21
|
Moore JE, Robison RK, Hu J, Sengupta ST, Mahdi OS, Anderson AW, Luo LY, Mohler AC, Merrell RT, Choi C. Optimization of the flip angles of narrow-band editing pulses in J-difference edited MRS of lactate at 3T. Magn Reson Med 2024; 91:886-895. [PMID: 38010083 PMCID: PMC10929535 DOI: 10.1002/mrm.29933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Application of highly selective editing RF pulses provides a means of minimizing co-editing of contaminants in J-difference MRS (MEGA), but it causes reduction in editing yield. We examined the flip angles (FAs) of narrow-band editing pulses to maximize the lactate edited signal with minimal co-editing of threonine. METHODS The effect of editing-pulse FA on the editing performance was examined, with numerical and phantom analyses, for bandwidths of 17.6-300 Hz in MEGA-PRESS editing of lactate at 3T. The FA and envelope of 46 ms Gaussian editing pulses were tailored to maximize the lactate edited signal at 1.3 ppm and minimize co-editing of threonine. The optimized editing-pulse FA MEGA scheme was tested in brain tumor patients. RESULTS Simulation and phantom data indicated that the optimum FA of MEGA editing pulses is progressively larger than 180° as the editing-pulse bandwidth decreases. For 46 ms long 17.6 Hz bandwidth Gaussian pulses and other given sequence parameters, the lactate edited signal was maximum at the first and second editing-pulse FAs of 241° and 249°, respectively. The edit-on and difference-edited lactate peak areas of the optimized FA MEGA were greater by 43% and 25% compared to the 180°-FA MEGA, respectively. In-vivo data confirmed the simulation and phantom results. The lesions of the brain tumor patients showed elevated lactate and physiological levels of threonine. CONCLUSION The lactate MEGA editing yield is significantly increased with editing-pulse FA much larger than 180° when the editing-pulse bandwidth is comparable to the lactate quartet frequency width.
Collapse
Affiliation(s)
- Jason E. Moore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ryan K. Robison
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Philips, Nashville, TN, USA
| | - Jie Hu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saikat T. Sengupta
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Olaimatu S. Mahdi
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam W. Anderson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Leo Y. Luo
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander C. Mohler
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ryan T. Merrell
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Changho Choi
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
22
|
Kino S, Kanamori M, Shimoda Y, Niizuma K, Endo H, Matsuura Y. Distinguishing IDH mutation status in gliomas using FTIR-ATR spectra of peripheral blood plasma indicating clear traces of protein amyloid aggregation. BMC Cancer 2024; 24:222. [PMID: 38365669 PMCID: PMC10870484 DOI: 10.1186/s12885-024-11970-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Glioma is a primary brain tumor and the assessment of its molecular profile in a minimally invasive manner is important in determining treatment strategies. Among the molecular abnormalities of gliomas, mutations in the isocitrate dehydrogenase (IDH) gene are strong predictors of treatment sensitivity and prognosis. In this study, we attempted to non-invasively diagnose glioma development and the presence of IDH mutations using multivariate analysis of the plasma mid-infrared absorption spectra for a comprehensive and sensitive view of changes in blood components associated with the disease and genetic mutations. These component changes are discussed in terms of absorption wavenumbers that contribute to differentiation. METHODS Plasma samples were collected at our institutes from 84 patients with glioma (13 oligodendrogliomas, 17 IDH-mutant astrocytoma, 7 IDH wild-type diffuse glioma, and 47 glioblastomas) before treatment initiation and 72 healthy participants. FTIR-ATR spectra were obtained for each plasma sample, and PLS discriminant analysis was performed using the absorbance of each wavenumber in the fingerprint region of biomolecules as the explanatory variable. This data was used to distinguish patients with glioma from healthy participants and diagnose the presence of IDH mutations. RESULTS The derived classification algorithm distinguished the patients with glioma from healthy participants with 83% accuracy (area under the curve (AUC) in receiver operating characteristic (ROC) = 0.908) and diagnosed the presence of IDH mutation with 75% accuracy (AUC = 0.752 in ROC) in cross-validation using 30% of the total test data. The characteristic changes in the absorption spectra suggest an increase in the ratio of β-sheet structures in the conformational composition of blood proteins of patients with glioma. Furthermore, these changes were more pronounced in patients with IDH-mutant gliomas. CONCLUSIONS The plasma infrared absorption spectra could be used to diagnose gliomas and the presence of IDH mutations in gliomas with a high degree of accuracy. The spectral shape of the protein absorption band showed that the ratio of β-sheet structures in blood proteins was significantly higher in patients with glioma than in healthy participants, and protein aggregation was a distinct feature in patients with glioma with IDH mutations.
Collapse
Affiliation(s)
- Saiko Kino
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-05, Aza-Aoba, Aramaki, Aoba, Sendai City, 980-8579, Miyagi Prefecture, Japan
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Yoshiteru Shimoda
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Seiryo 2-1, Aoba, Sendai City, 980-8575, Miyagi Prefecture, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, 980-8575 Seiryo 2-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Yuji Matsuura
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-05, Aza-Aoba, Aramaki, Aoba, Sendai City, 980-8579, Miyagi Prefecture, Japan.
| |
Collapse
|
23
|
Sabeghi P, Zarand P, Zargham S, Golestany B, Shariat A, Chang M, Yang E, Rajagopalan P, Phung DC, Gholamrezanezhad A. Advances in Neuro-Oncological Imaging: An Update on Diagnostic Approach to Brain Tumors. Cancers (Basel) 2024; 16:576. [PMID: 38339327 PMCID: PMC10854543 DOI: 10.3390/cancers16030576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
This study delineates the pivotal role of imaging within the field of neurology, emphasizing its significance in the diagnosis, prognostication, and evaluation of treatment responses for central nervous system (CNS) tumors. A comprehensive understanding of both the capabilities and limitations inherent in emerging imaging technologies is imperative for delivering a heightened level of personalized care to individuals with neuro-oncological conditions. Ongoing research in neuro-oncological imaging endeavors to rectify some limitations of radiological modalities, aiming to augment accuracy and efficacy in the management of brain tumors. This review is dedicated to the comparison and critical examination of the latest advancements in diverse imaging modalities employed in neuro-oncology. The objective is to investigate their respective impacts on diagnosis, cancer staging, prognosis, and post-treatment monitoring. By providing a comprehensive analysis of these modalities, this review aims to contribute to the collective knowledge in the field, fostering an informed approach to neuro-oncological care. In conclusion, the outlook for neuro-oncological imaging appears promising, and sustained exploration in this domain is anticipated to yield further breakthroughs, ultimately enhancing outcomes for individuals grappling with CNS tumors.
Collapse
Affiliation(s)
- Paniz Sabeghi
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Paniz Zarand
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Sina Zargham
- Department of Basic Science, California Northstate University College of Medicine, 9700 West Taron Drive, Elk Grove, CA 95757, USA;
| | - Batis Golestany
- Division of Biomedical Sciences, Riverside School of Medicine, University of California, 900 University Ave., Riverside, CA 92521, USA;
| | - Arya Shariat
- Kaiser Permanente Los Angeles Medical Center, 4867 W Sunset Blvd, Los Angeles, CA 90027, USA;
| | - Myles Chang
- Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, CA 90089, USA;
| | - Evan Yang
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Priya Rajagopalan
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Daniel Chang Phung
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| |
Collapse
|
24
|
Autry AW, Vaziri S, Gordon JW, Chen HY, Kim Y, Dang D, LaFontaine M, Noeske R, Bok R, Villanueva-Meyer JE, Clarke JL, Oberheim Bush NA, Chang SM, Xu D, Lupo JM, Larson PEZ, Vigneron DB, Li Y. Advanced Hyperpolarized 13C Metabolic Imaging Protocol for Patients with Gliomas: A Comprehensive Multimodal MRI Approach. Cancers (Basel) 2024; 16:354. [PMID: 38254844 PMCID: PMC10814348 DOI: 10.3390/cancers16020354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
This study aimed to implement a multimodal 1H/HP-13C imaging protocol to augment the serial monitoring of patients with glioma, while simultaneously pursuing methods for improving the robustness of HP-13C metabolic data. A total of 100 1H/HP [1-13C]-pyruvate MR examinations (104 HP-13C datasets) were acquired from 42 patients according to the comprehensive multimodal glioma imaging protocol. Serial data coverage, accuracy of frequency reference, and acquisition delay were evaluated using a mixed-effects model to account for multiple exams per patient. Serial atlas-based HP-13C MRI demonstrated consistency in volumetric coverage measured by inter-exam dice coefficients (0.977 ± 0.008, mean ± SD; four patients/11 exams). The atlas-derived prescription provided significantly improved data quality compared to manually prescribed acquisitions (n = 26/78; p = 0.04). The water-based method for referencing [1-13C]-pyruvate center frequency significantly reduced off-resonance excitation relative to the coil-embedded [13C]-urea phantom (4.1 ± 3.7 Hz vs. 9.9 ± 10.7 Hz; p = 0.0007). Significantly improved capture of tracer inflow was achieved with the 2-s versus 5-s HP-13C MRI acquisition delay (p = 0.007). This study demonstrated the implementation of a comprehensive multimodal 1H/HP-13C MR protocol emphasizing the monitoring of steady-state/dynamic metabolism in patients with glioma.
Collapse
Affiliation(s)
- Adam W. Autry
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Sana Vaziri
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jeremy W. Gordon
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Hsin-Yu Chen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yaewon Kim
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Duy Dang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Marisa LaFontaine
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | | | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Javier E. Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer L. Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nancy Ann Oberheim Bush
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Susan M. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Duan Xu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Peder E. Z. Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel B. Vigneron
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
25
|
Bhalla M, Mittal R, Kumar M, Bhatia R, Kushwah AS. Metabolomics: A Tool to Envisage Biomarkers in Clinical Interpretation of Cancer. Curr Drug Res Rev 2024; 16:333-348. [PMID: 37702236 DOI: 10.2174/2589977516666230912120412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/22/2023] [Accepted: 07/20/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Cancer is amongst the most dreadful ailments of modern times, and its impact continuously worsens global health systems. Early diagnosis and suitable therapeutic agents are the prime keys to managing this disease. Metabolomics deals with the complete profiling of cells and physiological phenomena in their organelles, thus helping in keen knowledge of the pathological status of the disease. It has been proven to be one of the best strategies in the early screening of cancer. OBJECTIVE This review has covered the recent updates on the promising role of metabolomics in the identification of significant biochemical markers in cancer-prone individuals that could lead to the identification of cancer in the early stages. METHODS The literature was collected through various databases, like Scopus, PubMed, and Google Scholar, with stress laid on the last ten years' publications. CONCLUSION It was assessed in this review that early recognition of cancerous growth could be achieved via complete metabolic profiling in association with transcriptomics and proteomics. The outcomes are rooted in various clinical studies that anticipated various biomarkers like tryptophan, phenylalanine, lactates, and different metabolic pathways associated with the Warburg effect. This metabolite imaging has been a fundamental step for the target acquisition, evaluation of predictive cancer biomarkers for early detection, and outlooks into cancer therapy along with critical evaluation. Significant efforts should be made to make this technique most reliable and easy.
Collapse
Affiliation(s)
- Medha Bhalla
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Ropar, 140111, India
| | - Roopal Mittal
- Department of Pharmacology, IKG Punjab Technical University, Jalandhar, 144601, India
- Department of Pharmacology, R.K.S.D. College of Pharmacy, Kaithal, 136027, India
| | - Manish Kumar
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry, Indo Soviet Friendship College of Pharmacy, Moga, 142001, India
| | - Ajay Singh Kushwah
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Ropar, 140111, India
| |
Collapse
|
26
|
Hou S, Kang Z, Liu Y, Lü C, Wang X, Wang Q, Ma C, Xu P, Gao C. An enzymic l-2-hydroxyglutarate biosensor based on l-2-hydroxyglutarate dehydrogenase from Azoarcus olearius. Biosens Bioelectron 2024; 243:115740. [PMID: 37862756 DOI: 10.1016/j.bios.2023.115740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
l-2-Hydroxyglutarate (l-2-HG) is a critical signaling and immune metabolite but its excessive accumulation can lead to l-2-hydroxyglutaric aciduria, renal cancer, and other diseases. Development of efficient and high-throughput methods for selective l-2-HG detection is urgently required. In this study, l-2-HG dehydrogenase in Azoarcus olearius BH72 (AoL2HGDH) was screened from ten homologs and identified as an enzyme with high specificity and activity toward l-2-HG dehydrogenation. Then, an enzymatic assay-based l-2-HG-sensing fluorescent reporter, EaLHGFR which consists of AoL2HGDH and resazurin, was developed for the detection of l-2-HG. The response magnitude and limit of detection of EaLHGFR were systematically optimized using a single-factor screening strategy. The optimal biosensor EaLHGFR-2 exhibited a response magnitude of 2189.25 ± 26.89% and a limit of detection of 0.042 μM. It can accurately detect the concentration of l-2-HG in bacterial and cellular samples as well as human body fluids. Considering its desirable properties, EaLHGFR-2 may be a promising alternative for quantitation of l-2-HG in biological samples.
Collapse
Affiliation(s)
- Shuang Hou
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Zhaoqi Kang
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Yidong Liu
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Chuanjuan Lü
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Xia Wang
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Qian Wang
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, People's Republic of China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, People's Republic of China.
| |
Collapse
|
27
|
Paech D, Weckesser N, Franke VL, Breitling J, Görke S, Deike-Hofmann K, Wick A, Scherer M, Unterberg A, Wick W, Bendszus M, Bachert P, Ladd ME, Schlemmer HP, Korzowski A. Whole-Brain Intracellular pH Mapping of Gliomas Using High-Resolution 31P MR Spectroscopic Imaging at 7.0 T. Radiol Imaging Cancer 2024; 6:e220127. [PMID: 38133553 PMCID: PMC10825708 DOI: 10.1148/rycan.220127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023]
Abstract
Malignant tumors commonly exhibit a reversed pH gradient compared with normal tissue, with a more acidic extracellular pH and an alkaline intracellular pH (pHi). In this prospective study, pHi values in gliomas were quantified using high-resolution phosphorous 31 (31P) spectroscopic MRI at 7.0 T and were used to correlate pHi alterations with histopathologic findings. A total of 12 participants (mean age, 58 years ± 18 [SD]; seven male, five female) with histopathologically proven, newly diagnosed glioma were included between September 2018 and November 2019. The 31P spectroscopic MRI scans were acquired using a double-resonant 31P/1H phased-array head coil together with a three-dimensional (3D) 31P chemical shift imaging sequence (5.7-mL voxel volume) performed with a 7.0-T whole-body system. The 3D volumetric segmentations were performed for the whole-tumor volumes (WTVs); tumor subcompartments of necrosis, gadolinium enhancement, and nonenhancing T2 (NCE T2) hyperintensity; and normal-appearing white matter (NAWM), and pHi values were compared. Spearman correlation was used to assess association between pHi and the proliferation index Ki-67. For all study participants, mean pHi values were higher in the WTV (7.057 ± 0.024) compared with NAWM (7.006 ± 0.012; P < .001). In eight participants with high-grade gliomas, pHi was increased in all tumor subcompartments (necrosis, 7.075 ± 0.033; gadolinium enhancement, 7.075 ± 0.024; NCE T2 hyperintensity, 7.043 ± 0.015) compared with NAWM (7.004 ± 0.014; all P < .01). The pHi values of WTV positively correlated with Ki-67 (R2 = 0.74, r = 0.78, P = .001). In conclusion, 31P spectroscopic MRI at 7.0 T enabled high-resolution quantification of pHi in gliomas, with pHi alteration associated with the Ki-67 proliferation index, and may aid in diagnosis and treatment monitoring. Keywords: 31P MRSI, pH, Glioma, Glioblastoma, Ultra-High-Field MRI, Imaging Biomarker, 7 Tesla Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
| | | | - Vanessa L. Franke
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes Breitling
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Steffen Görke
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Katerina Deike-Hofmann
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Antje Wick
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Moritz Scherer
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Bachert
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Mark E. Ladd
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Heinz-Peter Schlemmer
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Korzowski
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
28
|
Maekawa M. Analysis of Metabolic Changes in Endogenous Metabolites and Diagnostic Biomarkers for Various Diseases Using Liquid Chromatography and Mass Spectrometry. Biol Pharm Bull 2024; 47:1087-1105. [PMID: 38825462 DOI: 10.1248/bpb.b24-00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Analysis of endogenous metabolites in various diseases is useful for searching diagnostic biomarkers and elucidating the molecular mechanisms of pathophysiology. The author and collaborators have developed some LC/tandem mass spectrometry (LC/MS/MS) methods for metabolites and applied them to disease-related samples. First, we identified urinary conjugated cholesterol metabolites and serum N-palmitoyl-O-phosphocholine serine as useful biomarkers for Niemann-Pick disease type C (NPC). For the purpose of intraoperative diagnosis of glioma patients, we developed the LC/MS/MS analysis methods for 2-hydroxyglutaric acid or cystine and found that they could be good differential biomarkers. For renal cell carcinoma, we searched for various biomarkers for early diagnosis, malignancy evaluation and recurrence prediction by global metabolome analysis and targeted LC/MS/MS analysis. In pathological analysis, we developed a simultaneous LC/MS/MS analysis method for 13 steroid hormones and applied it to NPC cells, we found 6 types of reductions in NPC model cells. For non-alcoholic steatohepatitis (NASH), model mice were prepared with special diet and plasma bile acids were measured, and as a result, hydrophilic bile acids were significantly increased. In addition, we developed an LC/MS/MS method for 17 sterols and analyzed liver cholesterol metabolites and found a decrease in phytosterols and cholesterol synthetic markers and an increase in non-enzymatic oxidative sterols in the pre-onset stage of NASH. We will continue to challenge themselves to add value to clinical practice based on cutting-edge analytical chemistry methodology.
Collapse
|
29
|
Guillevin R, Naudin M, Fayolle P, Giraud C, Le Guillou X, Thomas C, Herpe G, Miranville A, Fernandez-Maloigne C, Pellerin L, Guillevin C. Diagnostic and Therapeutic Issues in Glioma Using Imaging Data: The Challenge of Numerical Twinning. J Clin Med 2023; 12:7706. [PMID: 38137775 PMCID: PMC10744312 DOI: 10.3390/jcm12247706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Glial tumors represent the leading etiology of primary brain tumors. Their particularities lie in (i) their location in a highly functional organ that is difficult to access surgically, including for biopsy, and (ii) their rapid, anisotropic mode of extension, notably via the fiber bundles of the white matter, which further limits the possibilities of resection. The use of mathematical tools enables the development of numerical models representative of the oncotype, genotype, evolution, and therapeutic response of lesions. The significant development of digital technologies linked to high-resolution NMR exploration, coupled with the possibilities offered by AI, means that we can envisage the creation of digital twins of tumors and their host organs, thus reducing the use of physical sampling.
Collapse
Affiliation(s)
- Rémy Guillevin
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Mathieu Naudin
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Pierre Fayolle
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Clément Giraud
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Xavier Le Guillou
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
- Department of Genetic, University Hospital Center of Poitiers, 86000 Poitiers, France
| | - Clément Thomas
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Guillaume Herpe
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | - Alain Miranville
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| | | | - Luc Pellerin
- IRMETIST Laboratory, INSERM U1313, University of Poitiers and University Hospital Center of Poitiers, 86000 Poitiers, France
| | - Carole Guillevin
- Department of Imaging, University Hospital Center of Poitiers, 86000 Poitiers, France
- Labcom I3M, University of Poitiers, 86000 Poitiers, France
- DACTIM-MIS Team, Laboratoire de Mathématiques Appliquées LMA, CNRS UMR 7348, 86021 Poitiers, France
| |
Collapse
|
30
|
Sanvito F, Kaufmann TJ, Cloughesy TF, Wen PY, Ellingson BM. Standardized brain tumor imaging protocols for clinical trials: current recommendations and tips for integration. FRONTIERS IN RADIOLOGY 2023; 3:1267615. [PMID: 38152383 PMCID: PMC10751345 DOI: 10.3389/fradi.2023.1267615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023]
Abstract
Standardized MRI acquisition protocols are crucial for reducing the measurement and interpretation variability associated with response assessment in brain tumor clinical trials. The main challenge is that standardized protocols should ensure high image quality while maximizing the number of institutions meeting the acquisition requirements. In recent years, extensive effort has been made by consensus groups to propose different "ideal" and "minimum requirements" brain tumor imaging protocols (BTIPs) for gliomas, brain metastases (BM), and primary central nervous system lymphomas (PCSNL). In clinical practice, BTIPs for clinical trials can be easily integrated with additional MRI sequences that may be desired for clinical patient management at individual sites. In this review, we summarize the general concepts behind the choice and timing of sequences included in the current recommended BTIPs, we provide a comparative overview, and discuss tips and caveats to integrate additional clinical or research sequences while preserving the recommended BTIPs. Finally, we also reflect on potential future directions for brain tumor imaging in clinical trials.
Collapse
Affiliation(s)
- Francesco Sanvito
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Timothy F. Cloughesy
- UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA, United States
| | - Benjamin M. Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
31
|
Mehjardi NZ, Kessler J, Sanin AY, Picard D, Westhoff P, Nickel AC, Uhlmann C, Shi W, Steiger HJ, Remke M, Fischer I, Vordermark D, Croner RS, Kahlert UD. The development of a hiPSC-based platform to identify tissue-dependencies of IDH1 R132H. Cell Death Discov 2023; 9:452. [PMID: 38086797 PMCID: PMC10716401 DOI: 10.1038/s41420-023-01747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 07/29/2024] Open
Abstract
The application of patient-derived (PD) in vitro tumor models represents the classical strategy for clinical translational oncology research. Using these cellular heterogeneous cultures for the isolation of cancer stem cells (CSCs), suggested to be the main driver for disease malignancy, relies on the use of surrogate biomarkers or is based on CSC-enriching culture conditions. However, the ability of those strategies to exclusively and efficiently enrich for CSC pool has been questioned. Here we present an alternative in vitro CSC model based on the oncogenic transformation of single clone-derived human induced pluripotent stem cells (hiPSC). Hotspot mutations in the DNA encoding for the R132 codon of the enzyme isocitrate dehydrogenase 1 (IDH1) and codon R175 of p53 are commonly occurring molecular features of different tumors and were selected for our transformation strategy. By choosing p53 mutant glial tumors as our model disease, we show that in vitro therapy discovery tests on IDH1-engineered synthetic CSCs (sCSCs) can identify kinases-targeting chemotherapeutics that preferentially target tumor cells expressing corresponding genetic alteration. In contrast, neural stem cells (NSCs) derived from the IDH1R132H overexpressing hiPSCs increase their resistance to the tested interventions indicating glial-to-neural tissue-dependent differences of IDH1R132H. Taken together, we provide proof for the potential of our sCSC technology as a potent addition to biomarker-driven drug development projects or studies on tumor therapy resistance. Moreover, follow-up projects such as comparing in vitro drug sensitivity profiles of hiPSC-derived tissue progenitors of different lineages, might help to understand a variety of tissue-related functions of IDH1 mutations.
Collapse
Affiliation(s)
- N Z Mehjardi
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - J Kessler
- Clinic for Radiation Therapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - A Y Sanin
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - D Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - P Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science, Heinrich Heine University, Düsseldorf, Germany
| | - Ann-Christin Nickel
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
- Diaceutics PLC, Düsseldorf, Germany
| | - C Uhlmann
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
- Charles River, Wuppertal, Germany
| | - W Shi
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - H J Steiger
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - M Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Pediatric Oncology and Hematology, University Hospital Saarbrücken, Saarbrücken, Germany
| | - I Fischer
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - D Vordermark
- Clinic for Radiation Therapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - R S Croner
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - U D Kahlert
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany.
| |
Collapse
|
32
|
Godoy LFDS, Paes VR, Ayres AS, Bandeira GA, Moreno RA, Hirata FDCC, Silva FAB, Nascimento F, Campos Neto GDC, Gentil AF, Lucato LT, Amaro Junior E, Young RJ, Malheiros SMF. Advances in diffuse glial tumors diagnosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:1134-1145. [PMID: 38157879 PMCID: PMC10756793 DOI: 10.1055/s-0043-1777729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/27/2023] [Indexed: 01/03/2024]
Abstract
In recent decades, there have been significant advances in the diagnosis of diffuse gliomas, driven by the integration of novel technologies. These advancements have deepened our understanding of tumor oncogenesis, enabling a more refined stratification of the biological behavior of these neoplasms. This progress culminated in the fifth edition of the WHO classification of central nervous system (CNS) tumors in 2021. This comprehensive review article aims to elucidate these advances within a multidisciplinary framework, contextualized within the backdrop of the new classification. This article will explore morphologic pathology and molecular/genetics techniques (immunohistochemistry, genetic sequencing, and methylation profiling), which are pivotal in diagnosis, besides the correlation of structural neuroimaging radiophenotypes to pathology and genetics. It briefly reviews the usefulness of tractography and functional neuroimaging in surgical planning. Additionally, the article addresses the value of other functional imaging techniques such as perfusion MRI, spectroscopy, and nuclear medicine in distinguishing tumor progression from treatment-related changes. Furthermore, it discusses the advantages of evolving diagnostic techniques in classifying these tumors, as well as their limitations in terms of availability and utilization. Moreover, the expanding domains of data processing, artificial intelligence, radiomics, and radiogenomics hold great promise and may soon exert a substantial influence on glioma diagnosis. These innovative technologies have the potential to revolutionize our approach to these tumors. Ultimately, this review underscores the fundamental importance of multidisciplinary collaboration in employing recent diagnostic advancements, thereby hoping to translate them into improved quality of life and extended survival for glioma patients.
Collapse
Affiliation(s)
- Luis Filipe de Souza Godoy
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Vitor Ribeiro Paes
- Hospital Israelita Albert Einstein, Laboratório de Patologia Cirúrgica, São Paulo SP, Brazil.
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Patologia, São Paulo SP, Brazil.
| | - Aline Sgnolf Ayres
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Gabriela Alencar Bandeira
- Instituto do Câncer do Estado de São Paulo, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Raquel Andrade Moreno
- Instituto do Câncer do Estado de São Paulo, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Rede D'Or São Luiz, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | | | | | - Felipe Nascimento
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | | | - Andre Felix Gentil
- Hospital Israelita Albert Einstein, Departamento de Neurocirurgia, São Paulo SP, Brazil.
| | - Leandro Tavares Lucato
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Grupo Fleury, São Paulo SP, Brazil.
| | - Edson Amaro Junior
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Robert J. Young
- Memorial Sloan-Kettering Cancer Center, Neuroradiology Service, New York, New York, United States.
| | | |
Collapse
|
33
|
Özütemiz C, White M, Elvendahl W, Eryaman Y, Marjańska M, Metzger GJ, Patriat R, Kulesa J, Harel N, Watanabe Y, Grant A, Genovese G, Cayci Z. Use of a Commercial 7-T MRI Scanner for Clinical Brain Imaging: Indications, Protocols, Challenges, and Solutions-A Single-Center Experience. AJR Am J Roentgenol 2023; 221:788-804. [PMID: 37377363 PMCID: PMC10825876 DOI: 10.2214/ajr.23.29342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The first commercially available 7-T MRI scanner (Magnetom Terra) was approved by the FDA in 2017 for clinical imaging of the brain and knee. After initial protocol development and sequence optimization efforts in volunteers, the 7-T system, in combination with an FDA-approved 1-channel transmit/32-channel receive array head coil, can now be routinely used for clinical brain MRI examinations. The ultrahigh field strength of 7-T MRI has the advantages of improved spatial resolution, increased SNR, and increased CNR but also introduces an array of new technical challenges. The purpose of this article is to describe an institutional experience with the use of the commercially available 7-T MRI scanner for routine clinical brain imaging. Specific clinical indications for which 7-T MRI may be useful for brain imaging include brain tumor evaluation with possible perfusion imaging and/or spectroscopy, radiotherapy planning; evaluation of multiple sclerosis and other demyelinating diseases, evaluation of Parkinson disease and guidance of deep brain stimulator placement, high-detail intracranial MRA and vessel wall imaging, evaluation of pituitary pathology, and evaluation of epilepsy. Detailed protocols, including sequence parameters, for these various indications are presented, and implementation challenges (including artifacts, safety, and side effects) and potential solutions are explored.
Collapse
Affiliation(s)
- Can Özütemiz
- Department of Radiology, University of Minnesota, 420 Delaware St SE, MMC 292, Minneapolis, MN 55455
| | - Matthew White
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
- Center for Clinical Imaging Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Wendy Elvendahl
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
- Center for Clinical Imaging Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Yigitcan Eryaman
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Małgorzata Marjańska
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Gregory J Metzger
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Rémi Patriat
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Jeramy Kulesa
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Noam Harel
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Yoichi Watanabe
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN
| | - Andrea Grant
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Guglielmo Genovese
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Zuzan Cayci
- Department of Radiology, University of Minnesota, 420 Delaware St SE, MMC 292, Minneapolis, MN 55455
- Center for Clinical Imaging Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
34
|
Chardin D, Jing L, Chazal-Ngo-Mai M, Guigonis JM, Rigau V, Goze C, Duffau H, Virolle T, Pourcher T, Burel-Vandenbos F. Identification of Metabolomic Markers in Frozen or Formalin-Fixed and Paraffin-Embedded Samples of Diffuse Glioma from Adults. Int J Mol Sci 2023; 24:16697. [PMID: 38069019 PMCID: PMC10705927 DOI: 10.3390/ijms242316697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
The aim of this study was to identify metabolomic signatures associated with the gliomagenesis pathway (IDH-mutant or IDH-wt) and tumor grade of diffuse gliomas (DGs) according to the 2021 WHO classification on frozen samples and to evaluate the diagnostic performances of these signatures in tumor samples that are formalin-fixed and paraffin-embedded (FFPE). An untargeted metabolomic study was performed using liquid chromatography/mass spectrometry on a cohort of 213 DG samples. Logistic regression with LASSO penalization was used on the frozen samples to build classification models in order to identify IDH-mutant vs. IDH-wildtype DG and high-grade vs low-grade DG samples. 2-Hydroxyglutarate (2HG) was a metabolite of interest to predict IDH mutational status and aminoadipic acid (AAA) and guanidinoacetic acid (GAA) were significantly associated with grade. The diagnostic performances of the models were 82.6% AUC, 70.6% sensitivity and 80.4% specificity for 2HG to predict IDH status and 84.7% AUC, 78.1% sensitivity and 73.4% specificity for AAA and GAA to predict grade from FFPE samples. Thus, this study showed that AAA and GAA are two novel metabolites of interest in DG and that metabolomic data can be useful in the classification of DG, both in frozen and FFPE samples.
Collapse
Affiliation(s)
- David Chardin
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Cote d’Azur (UCA), 06000 Nice, France; (D.C.); (L.J.); (J.-M.G.); (T.P.)
- Service de Médecine Nucléaire, Centre Antoine Lacassagne, Université Cote d’Azur, 06000 Nice, France
| | - Lun Jing
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Cote d’Azur (UCA), 06000 Nice, France; (D.C.); (L.J.); (J.-M.G.); (T.P.)
| | | | - Jean-Marie Guigonis
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Cote d’Azur (UCA), 06000 Nice, France; (D.C.); (L.J.); (J.-M.G.); (T.P.)
| | - Valérie Rigau
- Department of Pathology and Oncobiology, Institute for Neurosciences of Montpellier, INSERM U1051, University Hospital of Montpellier, 34000 Montpellier, France;
| | - Catherine Goze
- Laboratory of Solid Tumors Biology, Institute for Neurosciences of Montpellier, INSERM U1051, University Hospital of Montpellier, 34000 Montpellier, France;
| | - Hugues Duffau
- Neurosurgery Department, Institute for Neurosciences of Montpellier, INSERM U1051, University Hospital of Montpellier, 34000 Montpellier, France;
| | - Thierry Virolle
- Team INSERM “Cancer Stem Cell Plasticity and Functional Intra-Tumor Heterogeneity”, Institut de Biologie Valrose, Université Côte D’Azur, CNRS, INSERM, 06000 Nice, France;
| | - Thierry Pourcher
- Laboratory Transporter in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Cote d’Azur (UCA), 06000 Nice, France; (D.C.); (L.J.); (J.-M.G.); (T.P.)
| | - Fanny Burel-Vandenbos
- Department of Pathology, University Hospital of Nice, 06000 Nice, France;
- Laboratory “Cancer Stem Cell Plasticity and Functional Intra-Tumor Heterogeneity”, UMR CNRS 7277-UMR INSERM 1091, Institute of Biology Valrose, University Côte d’Azur, 06000 Nice, France
| |
Collapse
|
35
|
Ioannidis GS, Pigott LE, Iv M, Surlan-Popovic K, Wintermark M, Bisdas S, Marias K. Investigating the value of radiomics stemming from DSC quantitative biomarkers in IDH mutation prediction in gliomas. Front Neurol 2023; 14:1249452. [PMID: 38046592 PMCID: PMC10690367 DOI: 10.3389/fneur.2023.1249452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Objective This study aims to assess the value of biomarker based radiomics to predict IDH mutation in gliomas. The patient cohort consists of 160 patients histopathologicaly proven of primary glioma (WHO grades 2-4) from 3 different centers. Methods To quantify the DSC perfusion signal two different mathematical modeling methods were used (Gamma fitting, leakage correction algorithms) considering the assumptions about the compartments contributing in the blood flow between the extra- and intra vascular space. Results The Mean slope of increase (MSI) and the K1 parameter of the bidirectional exchange model exhibited the highest performance with (ACC 74.3% AUROC 74.2%) and (ACC 75% AUROC 70.5%) respectively. Conclusion The proposed framework on DSC-MRI radiogenomics in gliomas has the potential of becoming a reliable diagnostic support tool exploiting the mathematical modeling of the DSC signal to characterize IDH mutation status through a more reproducible and standardized signal analysis scheme for facilitating clinical translation.
Collapse
Affiliation(s)
- Georgios S. Ioannidis
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology—Hellas (FORTH), Heraklion, Greece
| | - Laura Elin Pigott
- Institute of Health and Social Care, London South Bank University, London, United Kingdom
- Faculty of Brain Science, Queen Square Institute of Neurology, University College London, London, United Kingdom
- Lysholm Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery University College London, London, United Kingdom
| | - Michael Iv
- Department of Radiology, Division of Neuroimaging and Neurointervention, Stanford University, Stanford, CA, United States
| | - Katarina Surlan-Popovic
- Department of Radiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Neuroradiology, University Medical Centre, Ljubljana, Slovenia
| | - Max Wintermark
- Department of Radiology, Division of Neuroimaging and Neurointervention, Stanford University, Stanford, CA, United States
| | - Sotirios Bisdas
- Department of Brain Repair and Rehabilitation, Queen Square Institute of Neurology, UCL, London, United Kingdom
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom
| | - Kostas Marias
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology—Hellas (FORTH), Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, Heraklion, Greece
| |
Collapse
|
36
|
McAfee D, Moyer M, Queen J, Mortazavi A, Boddeti U, Bachani M, Zaghloul K, Ksendzovsky A. Differential metabolic alterations in IDH1 mutant vs. wildtype glioma cells promote epileptogenesis through distinctive mechanisms. Front Cell Neurosci 2023; 17:1288918. [PMID: 38026690 PMCID: PMC10680369 DOI: 10.3389/fncel.2023.1288918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Glioma-related epilepsy (GRE) is a hallmark clinical presentation of gliomas with significant impacts on patient quality of life. The current standard of care for seizure management is comprised of anti-seizure medications (ASMs) and surgical resection. Seizures in glioma patients are often drug-resistant and can often recur after surgery despite total tumor resection. Therefore, current research is focused on the pro-epileptic pathological changes occurring in tumor cells and the peritumoral environment. One important contribution to seizures in GRE patients is metabolic reprogramming in tumor and surrounding cells. This is most evident by the significantly heightened seizure rate in patients with isocitrate dehydrogenase mutated (IDHmut) tumors compared to patients with IDH wildtype (IDHwt) gliomas. To gain further insight into glioma metabolism in epileptogenesis, this review compares the metabolic changes inherent to IDHmut vs. IDHwt tumors and describes the pro-epileptic effects these changes have on both the tumor cells and the peritumoral environment. Understanding alterations in glioma metabolism can help to uncover novel therapeutic interventions for seizure management in GRE patients.
Collapse
Affiliation(s)
- Darrian McAfee
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mitchell Moyer
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jaden Queen
- The College of Arts and Sciences, Cornell University, Ithaca, NY, United States
| | - Armin Mortazavi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Ujwal Boddeti
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Muzna Bachani
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kareem Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, United States
| | - Alexander Ksendzovsky
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
37
|
Sollmann N, Zhang H, Kloth C, Zimmer C, Wiestler B, Rosskopf J, Kreiser K, Schmitz B, Beer M, Krieg SM. Modern preoperative imaging and functional mapping in patients with intracranial glioma. ROFO-FORTSCHR RONTG 2023; 195:989-1000. [PMID: 37224867 DOI: 10.1055/a-2083-8717] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Magnetic resonance imaging (MRI) in therapy-naïve intracranial glioma is paramount for neuro-oncological diagnostics, and it provides images that are helpful for surgery planning and intraoperative guidance during tumor resection, including assessment of the involvement of functionally eloquent brain structures. This study reviews emerging MRI techniques to depict structural information, diffusion characteristics, perfusion alterations, and metabolism changes for advanced neuro-oncological imaging. In addition, it reflects current methods to map brain function close to a tumor, including functional MRI and navigated transcranial magnetic stimulation with derived function-based tractography of subcortical white matter pathways. We conclude that modern preoperative MRI in neuro-oncology offers a multitude of possibilities tailored to clinical needs, and advancements in scanner technology (e. g., parallel imaging for acceleration of acquisitions) make multi-sequence protocols increasingly feasible. Specifically, advanced MRI using a multi-sequence protocol enables noninvasive, image-based tumor grading and phenotyping in patients with glioma. Furthermore, the add-on use of preoperatively acquired MRI data in combination with functional mapping and tractography facilitates risk stratification and helps to avoid perioperative functional decline by providing individual information about the spatial location of functionally eloquent tissue in relation to the tumor mass. KEY POINTS:: · Advanced preoperative MRI allows for image-based tumor grading and phenotyping in glioma.. · Multi-sequence MRI protocols nowadays make it possible to assess various tumor characteristics (incl. perfusion, diffusion, and metabolism).. · Presurgical MRI in glioma is increasingly combined with functional mapping to identify and enclose individual functional areas.. · Advancements in scanner technology (e. g., parallel imaging) facilitate increasing application of dedicated multi-sequence imaging protocols.. CITATION FORMAT: · Sollmann N, Zhang H, Kloth C et al. Modern preoperative imaging and functional mapping in patients with intracranial glioma. Fortschr Röntgenstr 2023; 195: 989 - 1000.
Collapse
Affiliation(s)
- Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, United States
| | - Haosu Zhang
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Christopher Kloth
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Johannes Rosskopf
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Section of Neuroradiology, Bezirkskrankenhaus Günzburg, Günzburg, Germany
| | - Kornelia Kreiser
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Department of Radiology and Neuroradiology, Universitäts- und Rehabilitationskliniken Ulm, Ulm, Germany
| | - Bernd Schmitz
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Section of Neuroradiology, Bezirkskrankenhaus Günzburg, Günzburg, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Sandro M Krieg
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| |
Collapse
|
38
|
Mendes Serrão E, Klug M, Moloney BM, Jhaveri A, Lo Gullo R, Pinker K, Luker G, Haider MA, Shinagare AB, Liu X. Current Status of Cancer Genomics and Imaging Phenotypes: What Radiologists Need to Know. Radiol Imaging Cancer 2023; 5:e220153. [PMID: 37921555 DOI: 10.1148/rycan.220153] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology. Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
- Eva Mendes Serrão
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Maximiliano Klug
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Brian M Moloney
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Aaditeya Jhaveri
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Roberto Lo Gullo
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Katja Pinker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Gary Luker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Masoom A Haider
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Atul B Shinagare
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Xiaoyang Liu
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| |
Collapse
|
39
|
Kas A, Rozenblum L, Pyatigorskaya N. Clinical Value of Hybrid PET/MR Imaging: Brain Imaging Using PET/MR Imaging. Magn Reson Imaging Clin N Am 2023; 31:591-604. [PMID: 37741643 DOI: 10.1016/j.mric.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Hybrid PET/MR imaging offers a unique opportunity to acquire MR imaging and PET information during a single imaging session. PET/MR imaging has numerous advantages, including enhanced diagnostic accuracy, improved disease characterization, and better treatment planning and monitoring. It enables the immediate integration of anatomic, functional, and metabolic imaging information, allowing for personalized characterization and monitoring of neurologic diseases. This review presents recent advances in PET/MR imaging and highlights advantages in clinical practice for neuro-oncology, epilepsy, and neurodegenerative disorders. PET/MR imaging provides valuable information about brain tumor metabolism, perfusion, and anatomic features, aiding in accurate delineation, treatment response assessment, and prognostication.
Collapse
Affiliation(s)
- Aurélie Kas
- Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris F-75006, France.
| | - Laura Rozenblum
- Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris F-75006, France
| | - Nadya Pyatigorskaya
- Neuroradiology Department, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, UMR S 1127, CNRS UMR 722, Institut du Cerveau, Paris, France
| |
Collapse
|
40
|
Masui K, Mischel PS. Metabolic and epigenetic reprogramming in the pathogenesis of glioblastoma: Toward the establishment of "metabolism-based pathology". Pathol Int 2023; 73:533-541. [PMID: 37755062 DOI: 10.1111/pin.13379] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/03/2023] [Indexed: 09/28/2023]
Abstract
Molecular genetic approaches are now mandatory for cancer diagnostics, especially for brain tumors. Genotype-based diagnosis has predominated over the phenotype-based approach, with its prognostic and predictive powers. However, comprehensive genetic testing would be difficult to perform in the clinical setting, and translational research is required to histologically decipher the peculiar biology of cancer. Of interest, recent studies have demonstrated discrete links between oncogenotypes and the resultant metabolic phenotypes, revealing cancer metabolism as a promising histologic surrogate to reveal specific characteristics of each cancer type and indicate the best way to manage cancer patients. Here, we provide an overview of our research progress to work on cancer metabolism, with a particular focus on the genomically well-characterized malignant tumor glioblastoma. With the use of clinically relevant animal models and human tissue, we found that metabolic reprogramming plays a major role in the aggressive cancer biology by conferring therapeutic resistance to cancer cells and rewiring their epigenomic landscapes. We further discuss our future endeavor to establish "metabolism-based pathology" on how the basic knowledge of cancer metabolism could be leveraged to improve the management of patients by linking cancer cell genotype, epigenotype, and phenotype through metabolic reprogramming.
Collapse
Affiliation(s)
- Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Paul S Mischel
- Department of Pathology, Stanford University, Stanford, California, USA
- Department of Neurosurgery, Stanford University, Stanford, California, USA
- Sarafan ChEM-H, Stanford University, Stanford, California, USA
| |
Collapse
|
41
|
Lohmeier J, Radbruch H, Brenner W, Hamm B, Tietze A, Makowski MR. Predictive IDH Genotyping Based on the Evaluation of Spatial Metabolic Heterogeneity by Compartmental Uptake Characteristics in Preoperative Glioma Using 18F-FET PET. J Nucl Med 2023; 64:1683-1689. [PMID: 37652542 PMCID: PMC10626372 DOI: 10.2967/jnumed.123.265642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/13/2023] [Indexed: 09/02/2023] Open
Abstract
Molecular markers are of increasing importance for classifying, treating, and determining the prognosis for central nervous system tumors. Isocitrate dehydrogenase (IDH) is a critical regulator of glucose and amino acid metabolism. Our objective was to investigate metabolic reprogramming of glioma using compartmental uptake (CU) characteristics in O-(2-18F-fluoroethyl)-l-tyrosine (FET) PET and to evaluate its diagnostic potential for IDH genotyping. Methods: Between 2017 and 2022, patients with confirmed glioma were preoperatively investigated using static 18F-FET PET. Metabolic tumor volume (MTV), MTV for 60%-100% uptake (MTV60), and T2-weighted and contrast-enhancing lesion volumes were automatically segmented using U-Net neural architecture and isocontouring. Volume intersections were determined using the Dice coefficient. Uptake characteristics were determined for metabolically defined compartments (central [80%-100%] and peripheral [60%-75%] areas of 18F-FET uptake). CU ratio was defined as the fraction between the peripheral and central compartments. Mean target-to-background ratio was calculated. Comparisons were performed using parametric and nonparametric tests. Receiver-operating-characteristic curves, regression, and correlation were used for statistical analysis. Results: In total, 52 participants (male, 27, female, 25; mean age ± SD, 51 ± 16 y) were evaluated. MTV60 was greater and distinct from contrast-enhancing lesion volume (P = 0.046). IDH-mutated tumors presented a greater volumetric CU ratio and SUV CU ratio than IDH wild-type tumors (P < 0.05). Volumetric CU ratio determined IDH genotype with excellent diagnostic performance (area under the curve [AUC], 0.88; P < 0.001) at more than 5.49 (sensitivity, 86%, specificity, 90%), because IDH-mutated tumors presented a greater peripheral metabolic compartment than IDH wild-type tumors (P = 0.045). MTV60 and MTV were not suitable for IDH classification (P > 0.05). SUV CU ratio (AUC, 0.72; P = 0.005) and target-to-background ratio (AUC, 0.68; P = 0.016) achieved modest diagnostic performance-inferior to the volumetric CU ratio. Furthermore, the classification of loss of heterozygosity of chromosomes 1p and 19q (AUC, 0.75; P = 0.019), MGMT promoter methylation (AUC, 0.70; P = 0.011), and ATRX loss (AUC, 0.73; P = 0.004) by amino acid PET was evaluated. Conclusion: We proposed parametric 18F-FET PET as a noninvasive metabolic biomarker for the evaluation of CU characteristics, which differentiated IDH genotype with excellent diagnostic performance, establishing a critical association between spatial metabolic heterogeneity, mitochondrial tricarboxylic acid cycle, and genomic features with critical implications for clinical management and the diagnostic workup of patients with central nervous system cancer.
Collapse
Affiliation(s)
- Johannes Lohmeier
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany;
| | - Helena Radbruch
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Tietze
- Institute of Neuroradiology, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - Marcus R Makowski
- Department of Radiology, Technical University Munich, Munich, Germany
| |
Collapse
|
42
|
Wang Y, Fushimi Y, Arakawa Y, Shimizu Y, Sano K, Sakata A, Nakajima S, Okuchi S, Hinoda T, Oshima S, Otani S, Ishimori T, Tanji M, Mineharu Y, Yoshida K, Nakamoto Y. Evaluation of isocitrate dehydrogenase mutation in 2021 world health organization classification grade 3 and 4 glioma adult-type diffuse gliomas with 18F-fluoromisonidazole PET. Jpn J Radiol 2023; 41:1255-1264. [PMID: 37219717 PMCID: PMC10613590 DOI: 10.1007/s11604-023-01450-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE This study aimed to investigate the uptake characteristics of 18F-fluoromisonidazole (FMISO), in mutant-type isocitrate dehydrogenase (IDH-mutant, grade 3 and 4) and wild-type IDH (IDH-wildtype, grade 4) 2021 WHO classification adult-type diffuse gliomas. MATERIALS AND METHODS Patients with grade 3 and 4 adult-type diffuse gliomas (n = 35) were included in this prospective study. After registering 18F-FMISO PET and MR images, standardized uptake value (SUV) and apparent diffusion coefficient (ADC) were evaluated in hyperintense areas on fluid-attenuated inversion recovery (FLAIR) imaging (HIA), and in contrast-enhanced tumors (CET) by manually placing 3D volumes of interest. Relative SUVmax (rSUVmax) and SUVmean (rSUVmean), 10th percentile of ADC (ADC10pct), mean ADC (ADCmean) were measured in HIA and CET, respectively. RESULTS rSUVmean in HIA and rSUVmean in CET were significantly higher in IDH-wildtype than in IDH-mutant (P = 0.0496 and 0.03, respectively). The combination of FMISO rSUVmean in HIA and ADC10pct in CET, that of rSUVmax and ADC10pct in CET, that of rSUVmean in HIA and ADCmean in CET, were able to differentiate IDH-mutant from IDH-wildtype (AUC 0.80). When confined to astrocytic tumors except for oligodendroglioma, rSUVmax, rSUVmean in HIA and rSUVmean in CET were higher for IDH-wildtype than for IDH-mutant, but not significantly (P = 0.23, 0.13 and 0.14, respectively). The combination of FMISO rSUVmean in HIA and ADC10pct in CET was able to differentiate IDH-mutant (AUC 0.81). CONCLUSION PET using 18F-FMISO and ADC might provide a valuable tool for differentiating between IDH mutation status of 2021 WHO classification grade 3 and 4 adult-type diffuse gliomas.
Collapse
Affiliation(s)
- Yang Wang
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Yoshiki Arakawa
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yoichi Shimizu
- Division of Clinical Radiology Service, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Kohei Sano
- Division of Clinical Radiology Service, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Akihiko Sakata
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Satoshi Nakajima
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sachi Okuchi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takuya Hinoda
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sonoko Oshima
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sayo Otani
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masahiro Tanji
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazumichi Yoshida
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
43
|
Scott AJ, Mittal A, Meghdadi B, Palavalasa S, Achreja A, O'Brien A, Kothari AU, Zhou W, Xu J, Lin A, Wilder-Romans K, Edwards DM, Wu Z, Feng J, Andren AC, Zhang L, Tarnal V, Redic KA, Qi N, Fischer J, Yang E, Regan MS, Stopka SA, Baquer G, Lawrence TS, Venneti S, Agar NYR, Lyssiotis CA, Al-Holou WN, Nagrath D, Wahl DR. Rewiring of cortical glucose metabolism fuels human brain cancer growth. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.24.23297489. [PMID: 37961582 PMCID: PMC10635194 DOI: 10.1101/2023.10.24.23297489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The brain avidly consumes glucose to fuel neurophysiology. Cancers of the brain, such as glioblastoma (GBM), lose aspects of normal biology and gain the ability to proliferate and invade healthy tissue. How brain cancers rewire glucose utilization to fuel these processes is poorly understood. Here we perform infusions of 13 C-labeled glucose into patients and mice with brain cancer to define the metabolic fates of glucose-derived carbon in tumor and cortex. By combining these measurements with quantitative metabolic flux analysis, we find that human cortex funnels glucose-derived carbons towards physiologic processes including TCA cycle oxidation and neurotransmitter synthesis. In contrast, brain cancers downregulate these physiologic processes, scavenge alternative carbon sources from the environment, and instead use glucose-derived carbons to produce molecules needed for proliferation and invasion. Targeting this metabolic rewiring in mice through dietary modulation selectively alters GBM metabolism and slows tumor growth. Significance This study is the first to directly measure biosynthetic flux in both glioma and cortical tissue in human brain cancer patients. Brain tumors rewire glucose carbon utilization away from oxidation and neurotransmitter production towards biosynthesis to fuel growth. Blocking these metabolic adaptations with dietary interventions slows brain cancer growth with minimal effects on cortical metabolism.
Collapse
|
44
|
Cao C, Zhang L, Sorensen MD, Reifenberger G, Kristensen BW, McIntyre TM, Lin F. D-2-hydroxyglutarate regulates human brain vascular endothelial cell proliferation and barrier function. J Neuropathol Exp Neurol 2023; 82:921-933. [PMID: 37740942 PMCID: PMC10588003 DOI: 10.1093/jnen/nlad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2023] Open
Abstract
Gain-of-function mutations in isocitrate dehydrogenase (IDH) genes result in excessive production of (D)-2-hydroxyglutarate (D-2HG) which intrinsically modifies tumor cell epigenetics and impacts surrounding noncancerous cells through nonepigenetic pathways. However, whether D-2HG has a paracrine effect on endothelial cells in the tumor microenvironment needs further clarification. We quantified microvessel density by immunohistochemistry using tissue sections from 60 high-grade astrocytic gliomas with or without IDH mutation. Microvessel density was found to be reduced in tumors carrying an IDH mutation. Ex vivo experiments showed that D-2HG inhibited endothelial cell migration, wound healing, and tube formation by suppressing cell proliferation but not viability, possibly through reduced activation of the mTOR/STAT3 pathway. Further, D-2HG reduced fluorescent dextran permeability and decreased paracellular T-cell transendothelial migration by augmenting expression of junctional proteins thereby collectively increasing endothelial barrier function. These results indicate that D-2HG may influence the tumor vascular microenvironment by reducing the intratumoral vasculature density and by inhibiting the transport of metabolites and extravasation of circulating cells into the astrocytoma microenvironment. These observations provide a rationale for combining IDH inhibition with antitumor immunological/angiogenic approaches and suggest a molecular basis for resistance to antiangiogenic drugs in patients whose tumors express a mutant IDH allele.
Collapse
Affiliation(s)
- Chun Cao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lingjun Zhang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mia D Sorensen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Guido Reifenberger
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Thomas M McIntyre
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Astarita G, Kelly RS, Lasky-Su J. Metabolomics and lipidomics strategies in modern drug discovery and development. Drug Discov Today 2023; 28:103751. [PMID: 37640150 PMCID: PMC10543515 DOI: 10.1016/j.drudis.2023.103751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Metabolomics and lipidomics have an increasingly pivotal role in drug discovery and development. In the context of drug discovery, monitoring changes in the levels or composition of metabolites and lipids relative to genetic variations yields functional insights, bolstering human genetics and (meta)genomic methodologies. This approach also sheds light on potential novel targets for therapeutic intervention. In the context of drug development, metabolite and lipid biomarkers contribute to enhanced success rates, promising a transformative impact on precision medicine. In this review, we deviate from analytical chemist-focused perspectives, offering an overview tailored to drug discovery. We provide introductory insight into state-of-the-art mass spectrometry (MS)-based metabolomics and lipidomics techniques utilized in drug discovery and development, drawing from the collective expertise of our research teams. We comprehensively outline the application of metabolomics and lipidomics in advancing drug discovery and development, spanning fundamental research, target identification, mechanisms of action, and the exploration of biomarkers.
Collapse
Affiliation(s)
- Giuseppe Astarita
- Georgetown University, Washington, DC, USA; Arkuda Therapeutics, Watertown, MA, USA.
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
de Godoy LL, Lim KC, Rajan A, Verma G, Hanaoka M, O’Rourke DM, Lee JYK, Desai A, Chawla S, Mohan S. Non-Invasive Assessment of Isocitrate Dehydrogenase-Mutant Gliomas Using Optimized Proton Magnetic Resonance Spectroscopy on a Routine Clinical 3-Tesla MRI. Cancers (Basel) 2023; 15:4453. [PMID: 37760422 PMCID: PMC10526791 DOI: 10.3390/cancers15184453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE The isocitrate dehydrogenase (IDH) mutation has become one of the most important prognostic biomarkers in glioma management, indicating better treatment response and prognosis. IDH mutations confer neomorphic activity leading to the conversion of alpha-ketoglutarate (α-KG) to 2-hydroxyglutarate (2HG). The purpose of this study was to investigate the clinical potential of proton MR spectroscopy (1H-MRS) in identifying IDH-mutant gliomas by detecting characteristic resonances of 2HG and its complex interplay with other clinically relevant metabolites. MATERIALS AND METHODS Thirty-two patients with suspected infiltrative glioma underwent a single-voxel (SVS, n = 17) and/or single-slice-multivoxel (1H-MRSI, n = 15) proton MR spectroscopy (1H-MRS) sequence with an optimized echo-time (97 ms) on 3T-MRI. Spectroscopy data were analyzed using the linear combination (LC) model. Cramér-Rao lower bound (CRLB) values of <40% were considered acceptable for detecting 2HG and <20% for other metabolites. Immunohistochemical analyses for determining IDH mutational status were subsequently performed from resected tumor specimens and findings were compared with the results from spectral data. Mann-Whitney and chi-squared tests were performed to ascertain differences in metabolite levels between IDH-mutant and IDH-wild-type gliomas. Receiver operating characteristic (ROC) curve analyses were also performed. RESULTS Data from eight cases were excluded due to poor spectral quality or non-tumor-related etiology, and final data analyses were performed from 24 cases. Of these cases, 9/12 (75%) were correctly identified as IDH-mutant or IDH-wildtype gliomas through SVS and 10/12 (83%) through 1H-MRSI with an overall concordance rate of 79% (19/24). The sensitivity, specificity, positive predictive value, and negative predictive value were 80%, 77%, 86%, and 70%, respectively. The metabolite 2HG was found to be significant in predicting IDH-mutant gliomas through the chi-squared test (p < 0.01). The IDH-mutant gliomas also had a significantly higher NAA/Cr ratio (1.20 ± 0.09 vs. 0.75 ± 0.12 p = 0.016) and lower Glx/Cr ratio (0.86 ± 0.078 vs. 1.88 ± 0.66; p = 0.029) than those with IDH wild-type gliomas. The areas under the ROC curves for NAA/Cr and Glx/Cr were 0.808 and 0.786, respectively. CONCLUSIONS Noninvasive optimized 1H-MRS may be useful in predicting IDH mutational status and 2HG may serve as a valuable diagnostic and prognostic biomarker in patients with gliomas.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (M.H.); (S.M.)
| | - Kheng Choon Lim
- Department of Neuroradiology, Singapore General Hospital, Singapore 169609, Singapore;
| | - Archith Rajan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (M.H.); (S.M.)
| | - Gaurav Verma
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Mauro Hanaoka
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (M.H.); (S.M.)
| | - Donald M. O’Rourke
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.O.); (J.Y.K.L.)
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19014, USA
| | - John Y. K. Lee
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.O.); (J.Y.K.L.)
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Arati Desai
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (M.H.); (S.M.)
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (M.H.); (S.M.)
| |
Collapse
|
47
|
Bangalore Yogananda CG, Wagner BC, Truong NCD, Holcomb JM, Reddy DD, Saadat N, Hatanpaa KJ, Patel TR, Fei B, Lee MD, Jain R, Bruce RJ, Pinho MC, Madhuranthakam AJ, Maldjian JA. MRI-Based Deep Learning Method for Classification of IDH Mutation Status. Bioengineering (Basel) 2023; 10:1045. [PMID: 37760146 PMCID: PMC10525372 DOI: 10.3390/bioengineering10091045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Isocitrate dehydrogenase (IDH) mutation status has emerged as an important prognostic marker in gliomas. This study sought to develop deep learning networks for non-invasive IDH classification using T2w MR images while comparing their performance to a multi-contrast network. Methods: Multi-contrast brain tumor MRI and genomic data were obtained from The Cancer Imaging Archive (TCIA) and The Erasmus Glioma Database (EGD). Two separate 2D networks were developed using nnU-Net, a T2w-image-only network (T2-net) and a multi-contrast network (MC-net). Each network was separately trained using TCIA (227 subjects) or TCIA + EGD data (683 subjects combined). The networks were trained to classify IDH mutation status and implement single-label tumor segmentation simultaneously. The trained networks were tested on over 1100 held-out datasets including 360 cases from UT Southwestern Medical Center, 136 cases from New York University, 175 cases from the University of Wisconsin-Madison, 456 cases from EGD (for the TCIA-trained network), and 495 cases from the University of California, San Francisco public database. A receiver operating characteristic curve (ROC) was drawn to calculate the AUC value to determine classifier performance. Results: T2-net trained on TCIA and TCIA + EGD datasets achieved an overall accuracy of 85.4% and 87.6% with AUCs of 0.86 and 0.89, respectively. MC-net trained on TCIA and TCIA + EGD datasets achieved an overall accuracy of 91.0% and 92.8% with AUCs of 0.94 and 0.96, respectively. We developed reliable, high-performing deep learning algorithms for IDH classification using both a T2-image-only and a multi-contrast approach. The networks were tested on more than 1100 subjects from diverse databases, making this the largest study on image-based IDH classification to date.
Collapse
Affiliation(s)
- Chandan Ganesh Bangalore Yogananda
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Benjamin C. Wagner
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Nghi C. D. Truong
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - James M. Holcomb
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Divya D. Reddy
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Niloufar Saadat
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Kimmo J. Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Toral R. Patel
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Baowei Fei
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Matthew D. Lee
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA; (M.D.L.); (R.J.)
| | - Rajan Jain
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA; (M.D.L.); (R.J.)
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Richard J. Bruce
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
| | - Marco C. Pinho
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Ananth J. Madhuranthakam
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| | - Joseph A. Maldjian
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.C.W.); (N.C.D.T.); (J.M.H.); (D.D.R.); (N.S.); (B.F.); (M.C.P.); (A.J.M.); (J.A.M.)
| |
Collapse
|
48
|
Lin AP. Cystathionine: A Spectroscopic Biomarker for 1p/19q Codeleted Gliomas. Radiology 2023; 308:e232100. [PMID: 37668521 DOI: 10.1148/radiol.232100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Affiliation(s)
- Alexander P Lin
- From the Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, BLI236C, Boston, MA 02115
| |
Collapse
|
49
|
Branzoli F, Liserre R, Deelchand DK, Poliani PL, Bielle F, Nichelli L, Sanson M, Lehéricy S, Marjańska M. Neurochemical Differences between 1p/19q Codeleted and Noncodeleted IDH-mutant Gliomas by in Vivo MR Spectroscopy. Radiology 2023; 308:e223255. [PMID: 37668523 PMCID: PMC10546286 DOI: 10.1148/radiol.223255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 09/06/2023]
Abstract
Background Noninvasive identification of glioma subtypes is important for optimizing treatment strategies. Purpose To compare the in vivo neurochemical profiles between isocitrate dehydrogenase (IDH) 1-mutant 1p/19q codeleted gliomas and their noncodeleted counterparts measured by MR spectroscopy at 3.0 T with a point-resolved spectroscopy (PRESS) sequence optimized for D-2-hydroxyglutarate (2HG) detection. Materials and Methods Adults with IDH1-mutant gliomas were retrospectively included for this study from two university hospitals (inclusion period: January 2015 to July 2016 and September 2019 to June 2021, respectively) based on availability of 1p/19q codeletion status and a PRESS acquisition optimized for 2HG detection (echo time, 97 msec) at 3.0 T before any treatment. Spectral analysis was performed using LCModel and a simulated basis set. Metabolite quantification was performed using the water signal as a reference and correcting for water and metabolite longitudinal and transverse relaxation time constants. Concentration ratios were computed using total creatine (tCr) and total choline. A two-tailed unpaired t test was used to compare metabolite concentrations obtained in codeleted versus noncodeleted gliomas, accounting for multiple comparisons. Results Thirty-one adults (mean age, 39 years ± 8 [SD]; 19 male) were included, and 19 metabolites were quantified. Cystathionine concentration was higher in codeleted (n = 13) than noncodeleted (n = 18) gliomas when quantification was performed using the water signal or tCr as references (2.33 mM ± 0.98 vs 0.93 mM ± 0.94, and 0.34 mM ± 0.14 vs 0.14 mM ± 0.14, respectively; both P < .001). The sensitivity and specificity of PRESS to detect codeletion by means of cystathionine quantification were 92% and 61%, respectively. Other metabolites did not show evidence of a difference between groups (P > .05). Conclusion Higher cystathionine levels were detected in IDH1-mutant 1p/19q codeleted gliomas than in their noncodeleted counterparts with use of a PRESS sequence optimized for 2HG detection. Of 19 metabolites quantified, only cystathionine showed evidence of a difference in concentration between groups. Clinical trial registry no. NCT01703962 © RSNA, 2023 See also the editorial by Lin in this issue.
Collapse
Affiliation(s)
- Francesca Branzoli
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Roberto Liserre
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Dinesh K. Deelchand
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Pietro Luigi Poliani
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Franck Bielle
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Lucia Nichelli
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Marc Sanson
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Stéphane Lehéricy
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Małgorzata Marjańska
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| |
Collapse
|
50
|
Robison RK, Haynes JR, Ganji SK, Nockowski CP, Kovacs Z, Pham W, Morgan VL, Smith SA, Thompson RC, Omary RA, Gore JC, Choi C. J-Difference editing (MEGA) of lactate in the human brain at 3T. Magn Reson Med 2023; 90:852-862. [PMID: 37154389 PMCID: PMC10901256 DOI: 10.1002/mrm.29693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE The need to detect and quantify brain lactate accurately by MRS has stimulated the development of editing sequences based on J coupling effects. In J-difference editing of lactate, threonine can be co-edited and it contaminates lactate estimates due to the spectral proximity of the coupling partners of their methyl protons. We therefore implemented narrow-band editing 180° pulses (E180) in MEGA-PRESS acquisitions to resolve separately the 1.3-ppm resonances of lactate and threonine. METHODS Two 45.3-ms rectangular E180 pulses, which had negligible effects 0.15-ppm away from the carrier frequency, were implemented in a MEGA-PRESS sequence with TE 139 ms. Three acquisitions were designed to selectively edit lactate and threonine, in which the E180 pulses were tuned to 4.1 ppm, 4.25 ppm, and a frequency far off resonance. Editing performance was validated with numerical analyses and acquisitions from phantoms. The narrow-band E180 MEGA and another MEGA-PRESS sequence with broad-band E180 pulses were evaluated in six healthy subjects. RESULTS The 45.3-ms E180 MEGA offered a difference-edited lactate signal with lower intensity and reduced contamination from threonine compared to the broad-band E180 MEGA. The 45.3 ms E180 pulse had MEGA editing effects over a frequency range larger than seen in the singlet-resonance inversion profile. Lactate and threonine in healthy brain were both estimated to be 0.4 ± 0.1 mM, with reference to N-acetylaspartate at 12 mM. CONCLUSION Narrow-band E180 MEGA editing minimizes threonine contamination of lactate spectra and may improve the ability to detect modest changes in lactate levels.
Collapse
Affiliation(s)
- Ryan K Robison
- Philips, Nashville, Tennessee, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin R Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sandeep K Ganji
- Philips, Rochester, Minnesota, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | - Charles P Nockowski
- Philips, Nashville, Tennessee, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zoltan Kovacs
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Victoria L Morgan
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Reid C Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Reed A Omary
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA
| | - Changho Choi
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|