1
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [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: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
2
|
Jalloh M, Kankam SB. Harnessing imaging biomarkers for glioblastoma metastasis diagnosis: a correspondence. J Neurooncol 2024; 167:365-367. [PMID: 38393522 DOI: 10.1007/s11060-024-04606-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Affiliation(s)
- Mohamed Jalloh
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Samuel Berchi Kankam
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
- Harvard T.H. Chan School of Public Health, Harvard University, Cambridge, USA.
| |
Collapse
|
3
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [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: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
4
|
Stumpo V, Guida L, Bellomo J, Van Niftrik CHB, Sebök M, Berhouma M, Bink A, Weller M, Kulcsar Z, Regli L, Fierstra J. Hemodynamic Imaging in Cerebral Diffuse Glioma-Part B: Molecular Correlates, Treatment Effect Monitoring, Prognosis, and Future Directions. Cancers (Basel) 2022; 14:1342. [PMID: 35267650 PMCID: PMC8909110 DOI: 10.3390/cancers14051342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
Gliomas, and glioblastoma in particular, exhibit an extensive intra- and inter-tumoral molecular heterogeneity which represents complex biological features correlating to the efficacy of treatment response and survival. From a neuroimaging point of view, these specific molecular and histopathological features may be used to yield imaging biomarkers as surrogates for distinct tumor genotypes and phenotypes. The development of comprehensive glioma imaging markers has potential for improved glioma characterization that would assist in the clinical work-up of preoperative treatment planning and treatment effect monitoring. In particular, the differentiation of tumor recurrence or true progression from pseudoprogression, pseudoresponse, and radiation-induced necrosis can still not reliably be made through standard neuroimaging only. Given the abundant vascular and hemodynamic alterations present in diffuse glioma, advanced hemodynamic imaging approaches constitute an attractive area of clinical imaging development. In this context, the inclusion of objective measurable glioma imaging features may have the potential to enhance the individualized care of diffuse glioma patients, better informing of standard-of-care treatment efficacy and of novel therapies, such as the immunotherapies that are currently increasingly investigated. In Part B of this two-review series, we assess the available evidence pertaining to hemodynamic imaging for molecular feature prediction, in particular focusing on isocitrate dehydrogenase (IDH) mutation status, MGMT promoter methylation, 1p19q codeletion, and EGFR alterations. The results for the differentiation of tumor progression/recurrence from treatment effects have also been the focus of active research and are presented together with the prognostic correlations identified by advanced hemodynamic imaging studies. Finally, the state-of-the-art concepts and advancements of hemodynamic imaging modalities are reviewed together with the advantages derived from the implementation of radiomics and machine learning analyses pipelines.
Collapse
Affiliation(s)
- Vittorio Stumpo
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Lelio Guida
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Jacopo Bellomo
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Christiaan Hendrik Bas Van Niftrik
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Martina Sebök
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Moncef Berhouma
- Department of Neurosurgical Oncology and Vascular Neurosurgery, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon, 69500 Lyon, France;
| | - Andrea Bink
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neuroradiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Michael Weller
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neurology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Zsolt Kulcsar
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neuroradiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Jorn Fierstra
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| |
Collapse
|
5
|
Corr F, Grimm D, Saß B, Pojskić M, Bartsch JW, Carl B, Nimsky C, Bopp MHA. Radiogenomic Predictors of Recurrence in Glioblastoma—A Systematic Review. J Pers Med 2022; 12:jpm12030402. [PMID: 35330402 PMCID: PMC8952807 DOI: 10.3390/jpm12030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma, as the most aggressive brain tumor, is associated with a poor prognosis and outcome. To optimize prognosis and clinical therapy decisions, there is an urgent need to stratify patients with increased risk for recurrent tumors and low therapeutic success to optimize individual treatment. Radiogenomics establishes a link between radiological and pathological information. This review provides a state-of-the-art picture illustrating the latest developments in the use of radiogenomic markers regarding prognosis and their potential for monitoring recurrence. Databases PubMed, Google Scholar, and Cochrane Library were searched. Inclusion criteria were defined as diagnosis of glioblastoma with histopathological and radiological follow-up. Out of 321 reviewed articles, 43 articles met these inclusion criteria. Included studies were analyzed for the frequency of radiological and molecular tumor markers whereby radiogenomic associations were analyzed. Six main associations were described: radiogenomic prognosis, MGMT status, IDH, EGFR status, molecular subgroups, and tumor location. Prospective studies analyzing prognostic features of glioblastoma together with radiological features are lacking. By reviewing the progress in the development of radiogenomic markers, we provide insights into the potential efficacy of such an approach for clinical routine use eventually enabling early identification of glioblastoma recurrence and therefore supporting a further personalized monitoring and treatment strategy.
Collapse
Affiliation(s)
- Felix Corr
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
- Correspondence:
| | - Dustin Grimm
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
| | - Benjamin Saß
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Jörg W. Bartsch
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Barbara Carl
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Department of Neurosurgery, Helios Dr. Horst Schmidt Kliniken, Ludwig-Erhard-Strasse 100, 65199 Wiesbaden, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Miriam H. A. Bopp
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| |
Collapse
|
6
|
Prostate Cancer Radiogenomics-From Imaging to Molecular Characterization. Int J Mol Sci 2021; 22:ijms22189971. [PMID: 34576134 PMCID: PMC8465891 DOI: 10.3390/ijms22189971] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Radiomics and genomics represent two of the most promising fields of cancer research, designed to improve the risk stratification and disease management of patients with prostate cancer (PCa). Radiomics involves a conversion of imaging derivate quantitative features using manual or automated algorithms, enhancing existing data through mathematical analysis. This could increase the clinical value in PCa management. To extract features from imaging methods such as magnetic resonance imaging (MRI), the empiric nature of the analysis using machine learning and artificial intelligence could help make the best clinical decisions. Genomics information can be explained or decoded by radiomics. The development of methodologies can create more-efficient predictive models and can better characterize the molecular features of PCa. Additionally, the identification of new imaging biomarkers can overcome the known heterogeneity of PCa, by non-invasive radiological assessment of the whole specific organ. In the future, the validation of recent findings, in large, randomized cohorts of PCa patients, can establish the role of radiogenomics. Briefly, we aimed to review the current literature of highly quantitative and qualitative results from well-designed studies for the diagnoses, treatment, and follow-up of prostate cancer, based on radiomics, genomics and radiogenomics research.
Collapse
|
7
|
Kinoshita M, Kanemura Y, Narita Y, Kishima H. Reverse Engineering Glioma Radiomics to Conventional Neuroimaging. Neurol Med Chir (Tokyo) 2021; 61:505-514. [PMID: 34373429 PMCID: PMC8443974 DOI: 10.2176/nmc.ra.2021-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel radiological research field pursuing comprehensive quantitative image, namely “Radiomics,” gained traction along with the advancement of computational technology and artificial intelligence. This novel concept for analyzing medical images brought extensive interest to the neuro-oncology and neuroradiology research community to build a diagnostic workflow to detect clinically relevant genetic alteration of gliomas noninvasively. Although quite a few promising results were published regarding MRI-based diagnosis of isocitrate dehydrogenase (IDH) mutation in gliomas, it has become clear that an ample amount of effort is still needed to render this technology clinically applicable. At the same time, many significant insights were discovered through this research project, some of which could be “reverse engineered” to improve conventional non-radiomic MR image acquisition. In this review article, the authors aim to discuss the recent advancements and encountering issues of radiomics, how we can apply the knowledge provided by radiomics to standard clinical images, and further expected technological advances in the realm of radiomics and glioma.
Collapse
Affiliation(s)
- Manabu Kinoshita
- Department of Neurosurgery, Asahikawa Medical University.,Department of Neurosurgery, Osaka University Graduate School of Medicine.,Department of Neurosurgery, Osaka International Cancer Institute
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine
| |
Collapse
|
8
|
Wang H, Xue J, Qu T, Bernstein K, Chen T, Barbee D, Silverman JS, Kondziolka D. Predicting local failure of brain metastases after stereotactic radiosurgery with radiomics on planning MR images and dose maps. Med Phys 2021; 48:5522-5530. [PMID: 34287940 DOI: 10.1002/mp.15110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/10/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Stereotactic radiosurgery (SRS) has become an important modality in the treatment of brain metastases. The purpose of this study is to investigate the potential of radiomic features from planning magnetic resonance (MR) images and dose maps to predict local failure after SRS for brain metastases. MATERIALS/METHODS Twenty-eight patients who received Gamma Knife (GK) radiosurgery for brain metastases were retrospectively reviewed in this IRB-approved study. 179 irradiated tumors included 42 that locally failed within one-year follow-up. Using SRS tumor volumes, radiomic features were calculated on T1-weighted contrast-enhanced MR images acquired for treatment planning and planned dose maps. 125 radiomic features regarding tumor shape, dose distribution, MR intensities and textures were extracted for each tumor. Logistic regression with automatic feature selection was built to predict tumor progression from local control after SRS. Feature selection and model evaluation using receiver operating characteristic (ROC) curves were performed in a nested cross validation (CV) scheme. The associations between selected radiomic features and treatment outcomes were statistically assessed by univariate analysis. RESULTS The logistic model with feature selection achieved ROC AUC of 0.82 ± 0.09 on 5-fold CV, providing 83% sensitivity and 70% specificity for predicting local failure. A total of 10 radiomic features including 1 shape feature, 6 MR images and 3 dose distribution features were selected. These features were significantly associated with treatment outcomes (p < 0.05). The model was validated on independent holdout data with an AUC of 0.78. CONCLUSIONS Radiomic features from planning MR images and dose maps provided prognostic information in SRS for brain metastases. A model built on the radiomic features shows promise for early prediction of tumor local failure after treatment, potentially aiding in personalized care for brain metastases.
Collapse
Affiliation(s)
- Hesheng Wang
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Jinyu Xue
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Tanxia Qu
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Kenneth Bernstein
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Ting Chen
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - David Barbee
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Joshua S Silverman
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Douglas Kondziolka
- Department of Radiation Oncology, NYU Langone Medical Center, New York University, New York, New York, USA.,Department of Neurosurgery, NYU Langone Medical Center, New York University, New York, New York, USA
| |
Collapse
|
9
|
Zhang B, Zhao Z, Huang Y, Mao H, Zou M, Wang C, Yu G, Zhang M. Correlation between quantitative perfusion histogram parameters of DCE-MRI and PTEN, P-Akt and m-TOR in different pathological types of lung cancer. BMC Med Imaging 2021; 21:73. [PMID: 33865336 PMCID: PMC8052821 DOI: 10.1186/s12880-021-00604-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 04/07/2021] [Indexed: 01/01/2023] Open
Abstract
Background To explore if the quantitative perfusion histogram parameters of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) correlates with the expression of PTEN, P-Akt and m-TOR protein in lung cancer. Methods Thirty‐three patients with 33 lesions who had been diagnosed with lung cancer were enrolled in this study. They were divided into three groups: squamous cell carcinoma (SCC, 15 cases), adenocarcinoma (AC, 12 cases) and small cell lung cancer (SCLC, 6 cases). Preoperative imaging (conventional imaging and DCE-MRI) was performed on all patients. The Exchange model was used to measure the phar- macokinetic parameters, including Ktrans, Vp, Kep, Ve and Fp, and then the histogram parameters meanvalue, skewness, kurtosis, uniformity, energy, entropy, quantile of above five parameters were analyzed. The expression of PTEN, P-Akt and m-TOR were assessed by immunohistochemistry. Spearman correlation analysis was used to compare the correlation between the quantitative perfusion histogram parameters and the expression of PTEN, P-Akt and m-TOR in different pathological subtypes of lung cancer. Results The expression of m-TOR (P = 0.013) and P-Akt (P = 0.002) in AC was significantly higher than those in SCC. Vp (uniformity) in SCC group, Ktrans (uniformity), Ve (kurtosis, Q10, Q25) in AC group, Fp (skewness, kurtosis, energy), Ve (Q75, Q90, Q95) in SCLC group was positively correlated with PTEN, and Fp (entropy) in the SCLC group was negatively correlated with PTEN (P < 0.05); Kep (Q5, Q10) in the SCLC group was positively correlated with P-Akt, and Kep (energy) in the SCLC group was negatively correlated with P-Akt (P < 0.05); Kep (Q5) in SCC group and Vp (meanvalue, Q75, Q90, Q95) in SCLC group was positively correlated with m-TOR, and Ve (meanvalue) in SCC group was negatively correlated with m-TOR (P < 0.05). Conclusions The quantitative perfusion histogram parameters of DCE-MRI was correlated with the expression of PTEN, P-Akt and m-TOR in different pathological types of lung cancer, which may be used to indirectly evaluate the activation status of PI3K/Akt/mTOR signal pathway gene in lung cancer, and provide important reference for clinical treatment.
Collapse
Affiliation(s)
- Bingqian Zhang
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), No. 568, North Zhongxing Road, Yuecheng District, Shaoxing City, 312000, Zhejiang Province, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), No. 568, North Zhongxing Road, Yuecheng District, Shaoxing City, 312000, Zhejiang Province, China.
| | - Ya'nan Huang
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), No. 568, North Zhongxing Road, Yuecheng District, Shaoxing City, 312000, Zhejiang Province, China
| | - Haijia Mao
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), No. 568, North Zhongxing Road, Yuecheng District, Shaoxing City, 312000, Zhejiang Province, China
| | - Mingyue Zou
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), No. 568, North Zhongxing Road, Yuecheng District, Shaoxing City, 312000, Zhejiang Province, China
| | - Cheng Wang
- Department of Pathology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), Shaoxing, 312000, China
| | - Guangmao Yu
- Cardiothoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School), Shaoxing, 312000, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
10
|
Magnetic Resonance Features of Lower-grade Gliomas in Prediction of the Reverse Phase Protein A. J Comput Assist Tomogr 2021; 45:300-307. [PMID: 33512852 DOI: 10.1097/rct.0000000000001132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The Cancer Genome Atlas Research Network identified 4 novel protein expression-defined subgroups in patients with lower-grade gliomas (LGGs). The RPPA3 subtype had high levels of Epidermal Growth Factor Receptor and Human epidermal growth factor receptor-2, further increasing the chances for targeted therapy. In this study, we aimed to explore the relationships between magnetic resonance features and reverse phase protein array (RPPA) subtypes (R1-R4). METHODS Survival estimates for the Cancer Genome Atlas cohort were generated using the Kaplan-Meier method and time-dependent receiver operating characteristic curves. A total of 153 patients with LGG with brain magnetic resonance imaging from The Cancer Imaging Archive were retrospectively analyzed. Least absolute shrinkage and selection operator algorithm was used to reduce the feature dimensions of the RPPA3 subtype. RESULTS A total of 51 (33.3%) RPPA1 subtype, 42 (27.4) RPPA2 subtype, 19 (12.4%) RPPA3 subtype, and 38 (24.8%) RPPA4 subtype were identified. On multivariate logistic regression analysis, subventricular zone involvement [odds ratio (OR), 0.370; P = 0.006; 95% confidence interval (CI), 0.181-0.757) was associated with RPPA1 subtype [area under the curve (AUC), 0.598]. Volume of 60 cm3 or greater (OR, 5.174; P < 0.001; 95% CI, 2.182-12.267) was associated with RPPA2 subtype (AUC, 0.684). Proportion contrast-enhanced tumor greater than 5% (OR, 4.722; P = 0.010; 95% CI, 1.456-15.317), extranodular growth (OR, 5.524; P = 0.010; 95% CI, 1.509-20.215), and L/CS ratio equal to or greater than median (OR, 0.132; P = 0.003; 95% CI, 0.035-0.500) were associated with RPPA3 subtype (AUC, 0.825). Proportion contrast-enhanced tumor greater than 5% (OR, 0.206; P = 0.005; 95% CI, 0.068-0.625) was associated with RPPA4 subtype (AUC, 0.638). For the prediction of RPPA3 subtype, the nomogram showed good discrimination, with an AUC of 0.825 (95% CI, 0.711-0.939) and was well calibrated. The RPPA3 subtype was associated with shortest mean overall survival (RPPA3 subtype vs other: 613 vs 873 days; P < 0.05). The time-dependent receiver operating characteristic curves for the RPPA3 subtype was 0.72 (95% CI, 0.60-0.84) for survival at 1 year. Decision curve analysis indicated that prediction for the RPPA3 model was clinically useful. CONCLUSIONS The RPPA3 subtype is an unfavorable prognostic biomarker for overall survival in patients with LGG. Radiogenomics analysis of magnetic resonance features can predict the RPPA subtype preoperatively and may be of clinical value in tailoring the management strategies in patients with LGG.
Collapse
|
11
|
Correlation between dynamic susceptibility contrast perfusion MRI and genomic alterations in glioblastoma. Neuroradiology 2021; 63:1801-1810. [PMID: 33738509 DOI: 10.1007/s00234-021-02674-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/07/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE To determine if dynamic susceptibility contrast perfusion MR imaging (DSC-pMRI) can predict significant genomic alterations in glioblastoma (GB). METHODS A total of 47 patients with treatment-naive GB (M/F: 23/24, mean age: 54 years, age range: 20-90 years) having DSC-pMRI with leakage correction and genomic analysis were reviewed. Mean relative cerebral blood volume (rCBV), maximum rCBV, relative percent signal recovery (rPSR), and relative peak height (rPH) were derived from T2* signal intensity-time curves by ROI analysis. Major genomic alterations of IDH1-132H, MGMT, p53, EGFR, ATRX, and PTEN status were correlated with DSC-pMRI-derived GB parameters. Statistical analysis was performed utilizing the independent-samples t-test, ROC (receiver operating characteristic) curve analysis, and multivariable stepwise regression model. RESULTS rCBVmean and rCBVmax were significantly different in relation to the IDH1, MGMT, p53, and PTEN mutation status (all p < 0.05). The rPH of the p53 mutation-positive GBs (mean 5.8 ± 2.8) was significantly higher than those of the p53 mutation-negative GBs (mean 4.0 ± 1.5) (p = 0.022). Multivariable stepwise regression analysis revealed that the presence of IDH-1 mutation (B = - 2.81, p = 0.005) was associated with decreased rCBVmean; PTEN mutation (B = - 1.21, p = 0.003) and MGMT methylation (B = - 1.47, p = 0.038) were associated with decreased rCBVmax; and ATRX loss (B = - 1.05, p = 0.008) was associated with decreased rPH. CONCLUSION Significant associations were identified between DSC-pMRI-derived parameters and major genomic alterations, including IDH-1 mutation, MGMT methylation, ATRX loss, and PTEN mutation status in GB.
Collapse
|
12
|
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, Zhu H, Shui P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front Oncol 2021; 10:570465. [PMID: 33575207 PMCID: PMC7870863 DOI: 10.3389/fonc.2020.570465] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Ren
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ziwei Chen
- Department of Nephrology, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
Reuter G, Moïse M, Roll W, Martin D, Lombard A, Scholtes F, Stummer W, Suero Molina E. Conventional and advanced imaging throughout the cycle of care of gliomas. Neurosurg Rev 2021; 44:2493-2509. [PMID: 33411093 DOI: 10.1007/s10143-020-01448-3] [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: 08/03/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
Although imaging of gliomas has evolved tremendously over the last decades, published techniques and protocols are not always implemented into clinical practice. Furthermore, most of the published literature focuses on specific timepoints in glioma management. This article reviews the current literature on conventional and advanced imaging techniques and chronologically outlines their practical relevance for the clinical management of gliomas throughout the cycle of care. Relevant articles were located through the Pubmed/Medline database and included in this review. Interpretation of conventional and advanced imaging techniques is crucial along the entire process of glioma care, from diagnosis to follow-up. In addition to the described currently existing techniques, we expect deep learning or machine learning approaches to assist each step of glioma management through tumor segmentation, radiogenomics, prognostication, and characterization of pseudoprogression. Thorough knowledge of the specific performance, possibilities, and limitations of each imaging modality is key for their adequate use in glioma management.
Collapse
Affiliation(s)
- Gilles Reuter
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium. .,GIGA-CRC In-vivo Imaging Center, ULiege, Liège, Belgium.
| | - Martin Moïse
- Department of Radiology, University Hospital of Liège, Liège, Belgium
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Didier Martin
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Arnaud Lombard
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Félix Scholtes
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium.,Department of Neuroanatomy, University of Liège, Liège, Belgium
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| |
Collapse
|
14
|
Kang H, Chen P, Guo H, Zhang L, Tan Y, Xiao H, Yang A, Fang J, Zhang W. Vessel Size Imaging is Associated with IDH Mutation and Patient Survival in Diffuse Lower-Grade Glioma. Cancer Manag Res 2020; 12:9801-9811. [PMID: 33116839 PMCID: PMC7550213 DOI: 10.2147/cmar.s266533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/07/2020] [Indexed: 11/23/2022] Open
Abstract
Background Patients with isocitrate dehydrogenase (IDH) mutant gliomas have better survival and appear to be more sensitive to chemotherapy than their IDH wild-type counterparts. We attempted to assess the correlations of vessel size imaging (VSI) values with IDH mutation status and patient survival in diffuse lower-grade glioma (LGG). Methods We enrolled 60 patients with diffuse LGGs, among which 43 had IDH-mutant tumors. All patients underwent VSI examination and VSI values for active tumors were calculated. Receiver operating characteristic (ROC) curves were established to evaluate the detection efficiency. Logistic regression was employed to determine the ability of variables to discriminate IDH mutational status. Kaplan–Meier survival analysis and Cox proportional hazards models were utilized to estimate the correlations of VSI values and other risk factors with patient survival. Results We observed that VSI values were lower in IDH-mutant LGGs than IDH wild-type LGGs. The VSImax and VSImean values had AUC values of 0.7305 and 0.7401, respectively, in distinguishing IDH-mutant LGGs from IDH wild-type LGGs. Logistic regression showed that VSImean values, age and tumor location were associated with IDH-mutant status, and the formula integrating the three factors had an AUC value of 0.7798 when distinguishing IDH-mutant LGGs from IDH wild-type LGGs. Moreover, LGG patients with high VSI values exhibited worse survival rates than those with low VSI values for both progression-free survival (PFS) and overall survival (OS). Multivariate Cox proportional hazards regression analysis suggested that IDH mutation status, VSImean values and multiple lesions or lobes were risk factors for PFS of LGG patients. Conclusion VSI value is associated with IDH genotype and maybe an independent predictor of the survival of patients with LGGs.
Collapse
Affiliation(s)
- Houyi Kang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Peng Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Hong Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Letian Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Yong Tan
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ao Yang
- Department of Traffic Injury Research Office, Daping Hospital, Army Medical Center of PLA, Chongqing, People's Republic of China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| |
Collapse
|
15
|
Park JE, Kim HS, Park SY, Nam SJ, Chun SM, Jo Y, Kim JH. Prediction of Core Signaling Pathway by Using Diffusion- and Perfusion-based MRI Radiomics and Next-generation Sequencing in Isocitrate Dehydrogenase Wild-type Glioblastoma. Radiology 2019; 294:388-397. [PMID: 31845844 DOI: 10.1148/radiol.2019190913] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Next-generation sequencing (NGS) enables highly sensitive cancer genomics analysis, but its clinical implications for therapeutic options from imaging-based prediction have been limited. Purpose To predict core signaling pathways in isocitrate dehydrogenase (IDH) wild-type glioblastoma by using diffusion and perfusion MRI radiomics and NGS. Materials and Methods The radiogenomics model was developed by using retrospective patients with glioma who underwent NGS and anatomic, diffusion-, and perfusion-weighted imaging between March 2017 and February 2019. For testing model performance in predicting core signaling pathway, patients with IDH wild-type glioblastoma from a retrospective analysis from a registry (ClinicalTrials.gov NCT02619890) were evaluated. Radiogenomic feature selection was performed by using t tests, least absolute shrinkage and selection operator penalization, and random forest. Combining radiogenomic features, age, and location, the performance of predicting receptor tyrosine kinase (RTK), tumor protein p53 (P53), and retinoblastoma 1 pathways was evaluated by using the area under the receiver operating characteristic curve (AUC). Results There were 120 patients (52 years ± 13 [standard deviation]; 61 women) who were evaluated. Eighty-five patients (51 years ± 13; 43 men) were in the training set and 35 patients with IDH wild-type glioblastoma (56 years ± 12; 19 women) were in the validation set. Radiogenomics model identified 71 features in the RTK, 17 features in P53, and 35 features in the retinoblastoma pathway. The combined model showed better performance than anatomic imaging-based prediction in the RTK (P = .03) and retinoblastoma (P = .03) and perfusion imaging-based prediction in the P53 pathway (P = .04) in the training set. AUC values of the combined model for the prediction of core signaling pathways were 0.88 (95% confidence interval [CI]: 0.74, 1) for RTK, 0.76 (95% CI: 0.59, 0.92) for P53, and 0.81 (95% CI: 0.64, 0.97) for retinoblastoma in the validation set. Conclusion A diffusion- and perfusion-weighted MRI radiomics model can help characterize core signaling pathways and potentially guide targeted therapy for isocitrate dehydrogenase wild-type glioblastoma. © RSNA, 2019 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ji Eun Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Ho Sung Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Seo Young Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Soo Jung Nam
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Sung-Min Chun
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Youngheun Jo
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Jeong Hoon Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| |
Collapse
|
16
|
Xing Z, Zhang H, She D, Lin Y, Zhou X, Zeng Z, Cao D. IDH genotypes differentiation in glioblastomas using DWI and DSC-PWI in the enhancing and peri-enhancing region. Acta Radiol 2019; 60:1663-1672. [PMID: 31084193 DOI: 10.1177/0284185119842288] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH) mutation has diagnostic and prognostic values in glioblastomas. Peritumoral invasion of glioma cells is a cardinal feature of glioblastomas. PURPOSE To evaluate the contribution of DWI and DSC-PWI in the enhancing and peri-enhancing region for discriminating glioblastomas IDH genotypes, and the diagnostic values of combining two techniques in the peri-enhancing region compared with those in the enhancing region. MATERIAL AND METHODS We retrospectively reviewed the conventional MRI (cMRI), DWI and DSC-PWI obtained from 10 patients with IDH-mutated (IDH-m) glioblastomas and 65 patients with IDH wild-type (IDH-w) glioblastomas. Features of cMRI, relative minimum ADC in the enhancing region (rADCmin-t) and peri-enhancing area (rADCmin-p), and relative maximum CBV values in the enhancing region (rCBVmax-t) and peri-enhancing region (rCBVmax-p) were compared between two groups. Receiver operating characteristic curves and logistic regression analysis were used to assess diagnostic performance. RESULTS IDH-m glioblastomas tended to present in frontal lobes and younger patients. The rADCmin-t (P = 0.042) were significantly lower in IDH-w than IDH-m. Both rCBVmax-t and rCBVmax-p showed significant differences between two subgroups (all P < 0.001). The optimal cutoff values in prediction of IDH-m were >0.98 for rADCmin-t, <7.27 for rCBVmax-t, and < 0.97 for rCBVmax-p. Multivariate logistic regression revealed that the combination of rADCmin-t and rCBVmax-t yielded the highest sensitivity and specificity. CONCLUSION The rCBVmax-t or rCBVmax-p may serve as preferable and comparable imaging biomarkers for evaluation of glioblastomas IDH status. The combination of rADCmin-t and rCBVmax-t may yield the maximum predictive power for differentiating IDH status.
Collapse
Affiliation(s)
- Zhen Xing
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Hua Zhang
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Dejun She
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Yu Lin
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiaofang Zhou
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Zheng Zeng
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Dairong Cao
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
17
|
Lin H, Xu Y, Chen L, Na P, Li W. Multiparametric and multiregional diffusion features help predict molecule information, grade and survival in lower-grade gliomas: a feasibility study. Br J Radiol 2019; 92:20190324. [PMID: 31386559 DOI: 10.1259/bjr.20190324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE This study was to investigate the relationship of diffusion features with molecule information, and then predict grade and survival in lower-grade gliomas. METHODS 65 patients with primary lower-grade gliomas (WHO Grade II & III) who underwent conventional MRI and diffusion tensor imaging were retrospectively studied. The tumor region was automatically segmented into contrast-enhancing tumor, non-enhancing tumor, edematous and necrotic volumes. Diffusion features, including fractional anisotropy (FA), axial diffusivity, radial diffusivity and apparent diffusion coefficient (ADC), were extracted from each volume using histogram analysis. To estimate molecule biomarkers and predict clinical characteristics of grade and survival, support vector machine, generalized linear model, logistic regression and Cox regression were performed on the related features. RESULTS The diffusion features in non-enhancing tumor volume showed differences between isocitrate dehydrogenase mutant and wild-type gliomas. And the mean accuracy of support vector machine classifiers was 0.79. Ki-67 labeling index was correlated with these features, which were combined to significantly estimate Ki-67 expression level (r = 0.657, p < 0.001). These features also showed differences between Grade II and III gliomas. A combination of them for grade classification resulted in an area under the curve of 0.914 (0.857-0.971). Mean FA and fifth percentile of ADC were independently associated with overall survival, with lower FA and higher ADC showing better survival outcome. CONCLUSION In lower-grade gliomas, multiparametric and multiregional diffusion features could help predict molecule information, histological grade and survival. ADVANCES IN KNOWLEDGE The multi parametric diffusion features in non-enhancing tumor were associated with molecule information, grade and survival in lower-grade gliomas.
Collapse
Affiliation(s)
- Hai Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yanwen Xu
- Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Lei Chen
- Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Department of Neurosurgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Peng Na
- Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Weiping Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Department of Neurosurgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
18
|
Liao X, Cai B, Tian B, Luo Y, Song W, Li Y. Machine-learning based radiogenomics analysis of MRI features and metagenes in glioblastoma multiforme patients with different survival time. J Cell Mol Med 2019; 23:4375-4385. [PMID: 31001929 PMCID: PMC6533509 DOI: 10.1111/jcmm.14328] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/21/2019] [Indexed: 12/11/2022] Open
Abstract
Background This study aimed to examine multi‐dimensional MRI features’ predictability on survival outcome and associations with differentially expressed Genes (RNA Sequencing) in groups of glioblastoma multiforme (GBM) patients. Methods Radiomics features were extracted from segmented lesions of T2‐FLAIR MRI data of 137 GBM patients. Radiomics features include intensity, shape and textural features in seven classes were included in the analysis. Patients were divided into two groups depending on their survival time (shorter or longer than 1‐year survival). Four different machine learning algorithms were implemented to construct the prediction models. Features with top importance (importance >0.04) were selected to construct the prediction model using the model with the best performance. The interactions between image features and genomics were then analysed with Pearson's correlation analysis. Results The GBDT model with 72 features with highest importance had the highest accuracy of 0.81 on both short and long survival time classes, and the area under the curve (AUC) of the receiver operative characteristic (ROC) of the short and long survival time class were 0.79 and 0.81. Six metagenes showed significant interactive effect (P < 0.05), and Pearson's correlation analysis revealed that three of these metagenes (TIMP1,ROS1 EREG) showed moderate (0.3 < |r| < 0.5) or high correlation (|r| > 0.5) with image features. Conclusion Radiogenomics analysis shows that MRI features are predictive of survival outcomes, and image features are highly associated with selective metagenes. Radiogenomics analysis is a useful method for optimizing clinical diagnosis and selecting effective treatments.
Collapse
Affiliation(s)
- Xin Liao
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Bo Cai
- Department of Medical Imaging, The Third People's Hospital of Guizhou Province, Guiyang, Guizhou, China
| | - Bin Tian
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yilin Luo
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Wen Song
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yinglong Li
- Department of Interventional Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
19
|
Ozturk K, Soylu E, Tolunay S, Narter S, Hakyemez B. Dynamic Contrast-Enhanced T1-Weighted Perfusion Magnetic Resonance Imaging Identifies Glioblastoma Immunohistochemical Biomarkers via Tumoral and Peritumoral Approach: A Pilot Study. World Neurosurg 2019; 128:e195-e208. [PMID: 31003026 DOI: 10.1016/j.wneu.2019.04.089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We aimed to evaluate the usefulness of dynamic contrast-enhanced T1-weighted perfusion magnetic resonance imaging (DCE-pMRI) to predict certain immunohistochemical (IHC) biomarkers of glioblastoma (GB) in this pilot study. METHODS We retrospectively reviewed 36 patients (male/female, 25:11; mean age, 53 years; age range, 29-85 years) who had pretreatment DCE-pMRI with IHC analysis of their excised GBs. Regions of interest of the enhancing tumor (ER) and nonenhancing peritumoral region (NER) were used to calculate DCE-pMRI parameters of volume transfer constant, back flux constant, volume of the extravascular extracellular space, initial area under enhancement curve, and maximum slope. IHC biomarkers including Ki-67 labeling index, epidermal growth factor receptor (EGFR), oligodendrocyte transcription factor 2 (OLIG2), isocitrate dehydrogenase 1 (IDH1), and p53 mutation status were determined. The imaging metrics of GB with IHC markers were compared using the Kruskal-Wallis test and Spearman correlation analysis. RESULTS Among 30 patients with available IDH1 status, 14 patients (46.6%) had IDH1 mutation. EGFR amplification was present in 24/36 (66.6%) patients. Mean Ki-67 labeling index was 29% (range, 1.5%-80%). p53 mutation was present in 20/36 GBs (55%), whereas OLIG2 expression was positive in 29/36 GBs (80.5%). Various DCE-pMRI parameters gathered from the ER and NER were significantly correlated with IDH1 mutation, EGFR amplification, and OLIG2 expression (P < 0.05). Ki-67 labeling index showed a strong positive correlation with initial area under enhancement curve (r = 0.619; P < 0.001). CONCLUSIONS DCE-pMRI could determine surrogate IHC biomarkers in GB via tumoral and peritumoral approach, potential targets for individualized treatment protocols.
Collapse
Affiliation(s)
- Kerem Ozturk
- Department of Radiology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Esra Soylu
- Department of Radiology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Sahsine Tolunay
- Department of Pathology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Selin Narter
- Department of Pathology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Bahattin Hakyemez
- Department of Radiology, Uludag University Faculty of Medicine, Bursa, Turkey.
| |
Collapse
|
20
|
Radiogenomics of rectal adenocarcinoma in the era of precision medicine: A pilot study of associations between qualitative and quantitative MRI imaging features and genetic mutations. Eur J Radiol 2019; 113:174-181. [PMID: 30927944 DOI: 10.1016/j.ejrad.2019.02.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/24/2019] [Accepted: 02/17/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate associations between genetic mutations and qualitative as well as quantitative features on MRI in rectal adenocarcinoma at primary staging. METHODS In this retrospective study, patients with rectal adenocarcinoma, genome sequencing, and pretreatment rectal MRI were included. Statistical analysis was performed to evaluate associations between qualitative features obtained from subjective evaluation of rectal MRI and gene mutations as well as between quantitative textural features and gene mutations. For the qualitative evaluation, Fisher's Exact test was used to analyze categorical associations and Wilcoxon Rank Sum test was used for continuous clinical variables. For the quantitative evaluation, we performed manual segmentation of T2-weighted images for radiomics-based quantitative image analysis. Thirty-four texture features consisting of first order intensity histogram-based features (n = 4), second order Haralick textures (n = 5), and Gabor-edge based Haralick textures were computed at two different orientations. Consensus clustering was performed with 34 computed texture features using the K-means algorithm with Euclidean distance between the texture features. The clusters resulting from the algorithm were then used to enumerate the prevalence of gene mutations in those clusters. RESULTS In 65 patients, 45 genes were mutated in more than 3/65 patients (5%) and were included in the statistical analysis. Regarding qualitative imaging features, on univariate analysis, tumor location was significantly associated with APC (p = 0.032) and RASA1 mutation (p = 0.032); CRM status was significantly associated with ATM mutation (p = 0.021); and lymph node metastasis was significantly associated with BRCA2 (p = 0.046) mutation. However, these associations were not significant after adjusting for multiple comparisons. Regarding quantitative imaging features, Cluster C1 had tumors with higher mean Gabor edge intensity compared with cluster C2 (θ = 0°, p = 0.018; θ = 45°, p = 0.047; θ = 90°, p = 0.037; cluster C3 (θ = 0°, p = 0.18; θ = 45°, p = 0.1; θ = 90°, p = 0.052), and cluster C4 (θ = 0°, p = 0.016; θ = 45°, p = 0.033; θ = 90°, p = 0.014) suggesting that the cluster C1 had tumors with more distinct edges or heterogeneous appearance compared with other clusters. CONCLUSIONS Although this preliminary study showed promising associations between quantitative features and genetic mutations, it did not show any correlation between qualitative features and genetic mutations. Further studies with larger sample size are warranted to validate our preliminary data.
Collapse
|
21
|
Abstract
The most aggressive brain malignancy, glioblastoma, accounts for 60-70% of all gliomas and is uniformly fatal. According to the molecular signature, glioblastoma is divided into four subtypes (proneural, neural, classical, and mesenchymal), each with its own genetic background. The Cancer Genome Atlas project provides information about the most common genetic changes in glioblastoma. They involve mutations in TP53, TERT, and PTEN, and amplifications in EFGR, PDGFRA, CDK4, CDK6, MDM2, and MDM4. Recently, epigenetics was used to demonstrate the oncogenic roles of miR-124, miR-137, and miR-128. The most important findings so far are mutations in IDH1/2 and MGMT promoter methylation, which are routinely used as predictive biomarkers in patient care. Current clinical treatment leaves patients with only a 10% chance for 5-year survival. Attempts to define the mutational profile of glioblastoma to identify clinically relevant changes have not yet yielded significant results. This can be attributed to inter- and intra-tumor heterogeneity that is present in most glioblastomas, as well as hypermutation that appears as a consequence of chemotherapy. The evolving field of radiogenomics aims to classify glioblastoma using a combination of magnetic resonance imaging and genomic information. In the era of genomic medicine, next-generation sequencing is extensively used in glioblastoma research because it can detect multiple changes in a single biological sample; its potential in detecting circulating cell-free DNA has been tested in cerebrospinal fluid and plasma, and it shows promise in the examination of the cellular content of extracellular vesicles as a potential source of biomarkers. Next-generation sequencing is making its way into glioblastoma diagnostics. Gene panels like GlioSeq, which includes the most commonly mutated genes, are currently being tested on snap frozen and formalin fixed paraffin embedded tissues. This new methodology is helping to define the "next generation of glioblastomas" - clinically defined and better understood, with greater potential to improve patient care. However, limitations of the necessary infrastructure, space for data storage, technical expertise, and data ownership need to be considered carefully.
Collapse
Affiliation(s)
- Ivana Jovčevska
- a Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|