1
|
Shrestha B, Stern NB, Zhou A, Dunn A, Porter T. Current trends in the characterization and monitoring of vascular response to cancer therapy. Cancer Imaging 2024; 24:143. [PMID: 39438891 PMCID: PMC11515715 DOI: 10.1186/s40644-024-00767-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/26/2024] [Indexed: 10/25/2024] Open
Abstract
Tumor vascular physiology is an important determinant of disease progression as well as the therapeutic outcome of cancer treatment. Angiogenesis or the lack of it provides crucial information about the tumor's blood supply and therefore can be used as an index for cancer growth and progression. While standalone anti-angiogenic therapy demonstrated limited therapeutic benefits, its combination with chemotherapeutic agents improved the overall survival of cancer patients. This could be attributed to the effect of vascular normalization, a dynamic process that temporarily reverts abnormal vasculature to the normal phenotype maximizing the delivery and intratumor distribution of chemotherapeutic agents. Longitudinal monitoring of vascular changes following antiangiogenic therapy can indicate an optimal window for drug administration and estimate the potential outcome of treatment. This review primarily focuses on the status of various imaging modalities used for the longitudinal characterization of vascular changes before and after anti-angiogenic therapies and their clinical prospects.
Collapse
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Noah B Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Annie Zhou
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Tyrone Porter
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
2
|
Martín-Noguerol T, Santos-Armentia E, Ramos A, Luna A. An update on susceptibility-weighted imaging in brain gliomas. Eur Radiol 2024; 34:6763-6775. [PMID: 38581609 DOI: 10.1007/s00330-024-10703-w] [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: 11/18/2023] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 04/08/2024]
Abstract
Susceptibility-weighted imaging (SWI) has become a standard component of most brain MRI protocols. While traditionally used for detecting and characterising brain hemorrhages typically associated with stroke or trauma, SWI has also shown promising results in glioma assessment. Numerous studies have highlighted SWI's role in differentiating gliomas from other brain lesions, such as primary central nervous system lymphomas or metastases. Additionally, SWI aids radiologists in non-invasively grading gliomas and predicting their phenotypic profiles. Various researchers have suggested incorporating SWI as an adjunct sequence for predicting treatment response and for post-treatment monitoring. A significant focus of these studies is on the detection of intratumoural susceptibility signals (ITSSs) in gliomas, which are indicative of microhemorrhages and vessels within the tumour. The quantity, distribution, and characteristics of these ITSSs can provide radiologists with more precise information for evaluating and characterising gliomas. Furthermore, the potential benefits and added value of performing SWI after the administration of gadolinium-based contrast agents (GBCAs) have been explored. This review offers a comprehensive, educational, and practical overview of the potential applications and future directions of SWI in the context of glioma assessment. CLINICAL RELEVANCE STATEMENT: SWI has proven effective in evaluating gliomas, especially through assessing intratumoural susceptibility signal changes, and is becoming a promising, easily integrated tool in MRI protocols for both pre- and post-treatment assessments. KEY POINTS: • Susceptibility-weighted imaging is the most sensitive sequence for detecting blood and calcium inside brain lesions. • This sequence, acquired with and without gadolinium, helps with glioma diagnosis, characterisation, and grading through the detection of intratumoural susceptibility signals. • There are ongoing challenges that must be faced to clarify the role of susceptibility-weighted imaging for glioma assessment.
Collapse
Affiliation(s)
| | | | - Ana Ramos
- Department of Neuroradiology, University Hospital, 12 de Octubre, Madrid, Spain
| | - Antonio Luna
- MRI Unit, Radiology Department, HT Medica, Carmelo Torres 2, 23007, Jaén, Spain
| |
Collapse
|
3
|
Kuang RZ, Wang J, Wang YC, Tang XP. Effects of Apatinib combined with Temozolomide on levels of sPD-1 and sPD-L1 in patients with drug-resistant recurrent glioblastoma. Clinics (Sao Paulo) 2024; 79:100376. [PMID: 38733690 PMCID: PMC11103373 DOI: 10.1016/j.clinsp.2024.100376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/15/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVE This study aimed to explore the effects of Apatinib combined with Temozolomide (TMZ) on the levels of Soluble PD-1 (sPD-1) and Soluble Programmed Death-1 Ligand (sPD-L1) in patients with drug-resistant recurrent Glioblastoma (GB). STUDY DESIGN A total of 69 patients with recurrent GB from September 2020 to March 2022 were recruited and assigned to the control group (n = 34) and observation group (n = 35) according to different treatment options after tumor recurrence. The control group was treated with TMZ, and the observation group was treated with Apatinib combined with TMZ. Levels of sPD-1 and spd-l1, clinical efficacy, survival time and adverse reactions were observed and compared between the two groups. RESULTS General data including gender, age, body mass index, and combined diseases indicated no statistical significance between groups (p > 0.05). Before the intervention, sPD-1 and sPD-L1 levels were not significantly different in the two groups (p > 0.05). After interventions, levels of PD-1 and sPD-L1 levels decreased significantly (p < 0.05). The objective remission rate and clinical benefit rate of the observation group were higher and overall survival and progression-free survival were longer than those of the control group (p < 0.05). No significant difference was observed in major adverse reactions among patients (p > 0.05). CONCLUSIONS Apatinib combined with TMZ is safe and effective in the treatment of recurrent GB. The combined application of the two can reduce the levels of sPD-1 and sPD-L1, which has important clinical application value.
Collapse
Affiliation(s)
- Ren Zhao Kuang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Jun Wang
- Department of Orthopedics and Cosmetology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Yuan Chuan Wang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Xiao Ping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China.
| |
Collapse
|
4
|
Arias-Ramos N, Vieira C, Pérez-Carro R, López-Larrubia P. Integrative Magnetic Resonance Imaging and Metabolomic Characterization of a Glioblastoma Rat Model. Brain Sci 2024; 14:409. [PMID: 38790388 PMCID: PMC11118082 DOI: 10.3390/brainsci14050409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Glioblastoma (GBM) stands as the most prevalent and lethal malignant brain tumor, characterized by its highly infiltrative nature. This study aimed to identify additional MRI and metabolomic biomarkers of GBM and its impact on healthy tissue using an advanced-stage C6 glioma rat model. Wistar rats underwent a stereotactic injection of C6 cells (GBM group, n = 10) or cell medium (sham group, n = 4). A multiparametric MRI, including anatomical T2W and T1W images, relaxometry maps (T2, T2*, and T1), the magnetization transfer ratio (MTR), and diffusion tensor imaging (DTI), was performed. Additionally, ex vivo magnetic resonance spectroscopy (MRS) HRMAS spectra were acquired. The MRI analysis revealed significant differences in the T2 maps, T1 maps, MTR, and mean diffusivity parameters between the GBM tumor and the rest of the studied regions, which were the contralateral areas of the GBM rats and both regions of the sham rats (the ipsilateral and contralateral). The ex vivo spectra revealed markers of neuronal loss, apoptosis, and higher glucose uptake by the tumor. Notably, the myo-inositol and phosphocholine levels were elevated in both the tumor and the contralateral regions of the GBM rats compared to the sham rats, suggesting the effects of the tumor on the healthy tissue. The MRI parameters related to inflammation, cellularity, and tissue integrity, along with MRS-detected metabolites, serve as potential biomarkers for the tumor evolution, treatment response, and impact on healthy tissue. These techniques can be potent tools for evaluating new drugs and treatment targets.
Collapse
Affiliation(s)
| | | | | | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (N.A.-R.)
| |
Collapse
|
5
|
Deng S, Zhu Y. Prediction of Glioma Grade by Tumor Heterogeneity Radiomic Analysis Based on Multiparametric MRI. INT J COMPUT INT SYS 2023. [DOI: 10.1007/s44196-023-00230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
AbstractPredicting glioma grade plays a pivotal role in treatment and prognosis. However, several current methods for grading depend on the characteristics of the whole tumor. Predicting grade by analyzing tumor subregions has not been thoroughly investigated, which aims to improve the prediction performance. To predict glioma grade via analysis of tumor heterogeneity with features extracted from tumor subregions, it is mainly divided into four magnetic resonance imaging (MRI) sequences, including T2-weighted (T2), fluid-attenuated inversion recovery (FLAIR), pre-gadolinium T1-weighted (T1), and post-gadolinium T1-weighted methods. This study included the data of 97 patients with glioblastomas and 42 patients with low-grade gliomas before surgery. Three subregions, including enhanced tumor (ET), non-enhanced tumor, and peritumoral edema, were obtained based on segmentation labels generated by the GLISTRBoost algorithm. One hundred radiomic features were extracted from each subregion. Feature selection was performed using the cross-validated recursive feature elimination with a support vector machine (SVM) algorithm. SVM classifiers with grid search were established to predict glioma grade based on unparametric and multiparametric MRI. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of the classifiers, and the performance of the subregions was compared with the results of the whole tumor. In uniparametric analysis, the features from the ET subregion yielded a higher AUC value of 0.8697, 0.8474, and 0.8474 than those of the whole tumor of FLAIR, T1, and T2. In multiparametric analysis, the ET subregion achieved the best performance (AUC = 0.8755), which was higher than the uniparametric results. Radiomic features from the tumor subregion can potentially be used as clinical markers to improve the predictive accuracy of glioma grades.
Collapse
|
6
|
Waqar M, Van Houdt PJ, Hessen E, Li KL, Zhu X, Jackson A, Iqbal M, O’Connor J, Djoukhadar I, van der Heide UA, Coope DJ, Borst GR. Visualising spatial heterogeneity in glioblastoma using imaging habitats. Front Oncol 2022; 12:1037896. [PMID: 36505856 PMCID: PMC9731157 DOI: 10.3389/fonc.2022.1037896] [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: 09/06/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma is a high-grade aggressive neoplasm characterised by significant intra-tumoral spatial heterogeneity. Personalising therapy for this tumour requires non-invasive tools to visualise its heterogeneity to monitor treatment response on a regional level. To date, efforts to characterise glioblastoma's imaging features and heterogeneity have focussed on individual imaging biomarkers, or high-throughput radiomic approaches that consider a vast number of imaging variables across the tumour as a whole. Habitat imaging is a novel approach to cancer imaging that identifies tumour regions or 'habitats' based on shared imaging characteristics, usually defined using multiple imaging biomarkers. Habitat imaging reflects the evolution of imaging biomarkers and offers spatially preserved assessment of tumour physiological processes such perfusion and cellularity. This allows for regional assessment of treatment response to facilitate personalised therapy. In this review, we explore different methodologies to derive imaging habitats in glioblastoma, strategies to overcome its technical challenges, contrast experiences to other cancers, and describe potential clinical applications.
Collapse
Affiliation(s)
- Mueez Waqar
- Department of Neurosurgery, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Petra J. Van Houdt
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Eline Hessen
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ka-Loh Li
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Xiaoping Zhu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Alan Jackson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Neuroradiology, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Mudassar Iqbal
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - James O’Connor
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Radiology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Ibrahim Djoukhadar
- Department of Neuroradiology, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Uulke A. van der Heide
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - David J. Coope
- Department of Neurosurgery, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Gerben R. Borst
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
7
|
Li H, Kong Z, Xiang Y, Zheng R, Liu S. The role of PET/CT in radiotherapy for nasopharyngeal carcinoma. Front Oncol 2022; 12:1017758. [PMID: 36338692 PMCID: PMC9634754 DOI: 10.3389/fonc.2022.1017758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/06/2022] [Indexed: 05/29/2024] Open
Abstract
Radiotherapy has already been developed as the standard of care for patients with nasopharyngeal carcinoma (NPC), and precision staging, target volume delineation, prognosis prediction, and post-treatment surveillance are essential in the management of NPC. Positron emission tomography/computed tomography (PET/CT) is increasingly recognized as an imaging modality to guide precision radiotherapy in these areas. The feasibility and efficacy of 18F-FDG PET/CT have been confirmed in tumor diagnosis, treatment planning, prognosis, surveillance, and assessment. Coupled with the capability of revealing tumor metabolic information, 18F-FDG PET/CT is more accurate in identifying primary lesions and metastases of NPC than other conventional imaging methods including CT and MRI and shows the independently diagnostic and prognostic value for radiotherapy. However, 18F-FDG has limitations due to its physiological distribution in brain tissue and increasing uptake in post-radiation inflammation. Novel PET radiotracers including FAPI, NaF, CHO, and FLT are explored as alternatives with potential superiority for radiotherapy in NPC. In this review, we summarized the evolving role of PET/CT in the management of radiotherapy in NPC patients, aiming to facilitate precision radiotherapy from a molecular imaging aspect.
Collapse
Affiliation(s)
- Hongjia Li
- Department of Nuclear Medicine/PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziren Kong
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongbo Xiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Zheng
- Department of Nuclear Medicine/PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoyan Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Chen L, Tong F, Peng L, Huang Y, Yin P, Feng Y, Cheng S, Wang J, Dong X. Efficacy and safety of recombinant human endostatin combined with whole-brain radiation therapy in patients with brain metastases from non-small cell lung cancer. Radiother Oncol 2022; 174:44-51. [PMID: 35788355 DOI: 10.1016/j.radonc.2022.06.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/11/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Brain metastasis (BM) is the leading cause of poor prognosis in non-small cell lung cancer (NSCLC) patients. To date, whole-brain radiation therapy (WBRT) is a standard treatment for patients with multiple BMs, while its effectiveness is currently unsatisfactory. This study aimed to investigate the effects of Rh-endostatin combined with WBRT on NSCLC patients with BMs. MATERIALS AND METHODS A total of 43 patients with BM were randomly divided into two groups. The Rh-endostatin combination group (n=19) received Rh-endostatin combined with WBRT, and the radiation group (n=24) received WBRT only. The primary endpoint of the study was progression-free survival (PFS), and the secondary endpoints were intracranial progression free survival (iPFS), overall survival (OS), objective response rate (ORR), and changes in the cerebral blood volume (CBV) and cerebral blood flow (CBF). RESULTS Median PFS (mPFS) was 8.1 months in the Rh-endostatin combination group and 4.9 months in the radiation group (95%CI 0.2612-0.9583, p=0·0428). Besides, the median iPFS was 11.6 months in the Rh-endostatin combination group and 4.8 months in the radiation group (95%CI 0.2530-0.9504, p=0·0437). OS was 14.2 months in the Rh-endostatin combination group and 6.4 months in the radiation group (95%CI 0.2508-1.026, p=0·0688). CBV and CBF in the Rh-endostatin combination group were better improved than that in the radiation group, which indicated that Rh-endostatin might improve local blood supply and microcirculation. CONCLUSION Rh-endostatin showed better survival and improved cerebral perfusion parameters, which may provide further insights into the application of Rh-endostatin for NSCLC patients with BMs.
Collapse
Affiliation(s)
- Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Fang Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ling Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yu Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yue Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Shishi Cheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
9
|
Li AY, Iv M. Conventional and Advanced Imaging Techniques in Post-treatment Glioma Imaging. FRONTIERS IN RADIOLOGY 2022; 2:883293. [PMID: 37492665 PMCID: PMC10365131 DOI: 10.3389/fradi.2022.883293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 07/27/2023]
Abstract
Despite decades of advancement in the diagnosis and therapy of gliomas, the most malignant primary brain tumors, the overall survival rate is still dismal, and their post-treatment imaging appearance remains very challenging to interpret. Since the limitations of conventional magnetic resonance imaging (MRI) in the distinction between recurrence and treatment effect have been recognized, a variety of advanced MR and functional imaging techniques including diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), perfusion-weighted imaging (PWI), MR spectroscopy (MRS), as well as a variety of radiotracers for single photon emission computed tomography (SPECT) and positron emission tomography (PET) have been investigated for this indication along with voxel-based and more quantitative analytical methods in recent years. Machine learning and radiomics approaches in recent years have shown promise in distinguishing between recurrence and treatment effect as well as improving prognostication in a malignancy with a very short life expectancy. This review provides a comprehensive overview of the conventional and advanced imaging techniques with the potential to differentiate recurrence from treatment effect and includes updates in the state-of-the-art in advanced imaging with a brief overview of emerging experimental techniques. A series of representative cases are provided to illustrate the synthesis of conventional and advanced imaging with the clinical context which informs the radiologic evaluation of gliomas in the post-treatment setting.
Collapse
Affiliation(s)
- Anna Y. Li
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael Iv
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
10
|
Gonçalves SI, Simões RV, Shemesh N. Short TE downfield magnetic resonance spectroscopy in a mouse model of brain glioma. Magn Reson Med 2022; 88:524-536. [PMID: 35315536 DOI: 10.1002/mrm.29243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Enhanced cell proliferation in tumors can be associated with altered metabolic profiles and dramatic microenvironmental changes. Downfield magnetic resonance spectroscopy (MRS) has received increasing attention due to its ability to report on labile resonances of molecules not easily detected in upfield 1 H MRS. Image-selected-in-vivo-spectroscopy-relaxation enhanced MRS (iRE-MRS) was recently introduced for acquiring short echo-time (TE) spectra. Here, iRE-MRS was used to investigate in-vivo downfield spectra in glioma-bearing mice. METHODS Experiments were performed in vivo in an immunocompetent glioma mouse model at 9.4 T using a cryogenic coil. iRE-MRS spectra were acquired in N = 6 glioma-bearing mice (voxel size = 2.23 mm3 ) and N = 6 control mice. Spectra were modeled by a sum of Lorentzian peaks simulating known downfield resonances, and differences between controls and tumors were quantified using relative peak areas. RESULTS Short TE tumor spectra exhibited large qualitative differences compared to control spectra. Most peaks appeared modulated, with strong attenuation of NAA (∼7.82, 7.86 ppm) and changes in relative peak areas between 6.75 and 8.49 ppm. Peak areas tended to be smaller for DF6.83 , DF7.60 , DF8.18 and NAA; and larger for DF7.95 and DF8.24 . Differences were also detected in signals resonating above 8.5 ppm, assumed to arise from NAD+. CONCLUSIONS In-vivo downfield 1 H iRE-MRS of mouse glioma revealed differences between controls and tumor bearing mice, including in metabolites which are not easily detectable in the more commonly investigated upfield spectrum. These findings motivate future downfield MRS investigations exploring pH and exchange contributions to these differences.
Collapse
Affiliation(s)
| | - Rui V Simões
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
11
|
Moon HH, Park JE, Kim YH, Kim JH, Kim HS. Contrast enhancing pattern on pre-treatment MRI predicts response to anti-angiogenic treatment in recurrent glioblastoma: comparison of bevacizumab and temozolomide treatment. J Neurooncol 2022; 157:405-415. [PMID: 35275335 DOI: 10.1007/s11060-022-03980-2] [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: 01/06/2022] [Accepted: 03/04/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate the value of the contrast enhancing pattern on pre-treatment MRI for predicting the response to anti-angiogenic treatment in patients with IDH-wild type recurrent glioblastoma. METHODS This retrospective study enrolled 65 patients with IDH wild-type recurrent glioblastoma who received standard therapy and then received either bevacizumab (46 patients) or temozolomide (19 patients) as a secondary treatment. The contrast enhancing pattern on pre-treatment MRI was visually analyzed and dichotomized into contrast enhancing lesion (CEL) dominant and non-enhancing lesion (NEL) dominant types. Quantitative volumetric analysis was used to support the dichotomization. The Kaplan-Meier method and Cox proportional hazards regression analysis were used to stratify progression free survival (PFS) according to the treatment in the entire patients, CEL dominant group, and NEL dominant group. RESULTS In all patients, the PFS of those treated with bevacizumab was not significantly different from those treated with temozolomide (log-rank test, P = 0.96). When the contrast enhancing pattern was considered, bevacizumab was associated with longer PFS in the CEL dominant group (P = 0.031), whereas temozolomide showed longer PFS in the NEL dominant group (P = 0.022). Quantitative analysis revealed mean values for the proportion of solid-enhancing tumor of 13.7% for the CEL dominant group and 4.3% for the NEL dominant group. CONCLUSION Patients with the CEL dominant type showed a better treatment response to bevacizumab, whereas NEL dominant types showed a better response to temozolomide. The contrast enhancing pattern on pre-treatment MRI can be used to stratify patients with IDH wild-type recurrent glioblastoma according to the effect of anti-angiogenic treatment.
Collapse
Affiliation(s)
- Hye Hyeon Moon
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea.
| | - Young-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea
| |
Collapse
|
12
|
Bhaduri S, Lesbats C, Sharkey J, Kelly CL, Mukherjee S, Taylor A, Delikatny EJ, Kim SG, Poptani H. Assessing Tumour Haemodynamic Heterogeneity and Response to Choline Kinase Inhibition Using Clustered Dynamic Contrast Enhanced MRI Parameters in Rodent Models of Glioblastoma. Cancers (Basel) 2022; 14:cancers14051223. [PMID: 35267531 PMCID: PMC8909848 DOI: 10.3390/cancers14051223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/04/2022] Open
Abstract
To investigate the utility of DCE-MRI derived pharmacokinetic parameters in evaluating tumour haemodynamic heterogeneity and treatment response in rodent models of glioblastoma, imaging was performed on intracranial F98 and GL261 glioblastoma bearing rodents. Clustering of the DCE-MRI-based parametric maps (using Tofts, extended Tofts, shutter speed, two-compartment, and the second generation shutter speed models) was performed using a hierarchical clustering algorithm, resulting in areas with poor fit (reflecting necrosis), low, medium, and high valued pixels representing parameters Ktrans, ve, Kep, vp, τi and Fp. There was a significant increase in the number of necrotic pixels with increasing tumour volume and a significant correlation between ve and tumour volume suggesting increased extracellular volume in larger tumours. In terms of therapeutic response in F98 rat GBMs, a sustained decrease in permeability and perfusion and a reduced cell density was observed during treatment with JAS239 based on Ktrans, Fp and ve as compared to control animals. No significant differences in these parameters were found for the GL261 tumour, indicating that this model may be less sensitive to JAS239 treatment regarding changes in vascular parameters. This study demonstrates that region-based clustered pharmacokinetic parameters derived from DCE-MRI may be useful in assessing tumour haemodynamic heterogeneity with the potential for assessing therapeutic response.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Arthur Taylor
- Department of Molecular Physiology & Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK;
| | - Edward J. Delikatny
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Sungheon G. Kim
- Department of Radiology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Correspondence:
| |
Collapse
|
13
|
Hou W, Xue Y, Qian Y, Pan H, Xu M, Shen Y, Li X, Yu Y. Application of Intravoxel Incoherent Motion Diffusion-Weighted Imaging in Predicting and Monitoring Early Efficacy of Anti-Angiogenic Therapy in the C6 Glioma Rat Model. Front Oncol 2022; 11:842169. [PMID: 35155219 PMCID: PMC8831888 DOI: 10.3389/fonc.2021.842169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/31/2021] [Indexed: 12/30/2022] Open
Abstract
Objective To investigate the feasibility of intravoxel incoherent motion (IVIM) diffusion-weighted imaging (DWI) in evaluating early effects of anti-angiogenic therapy in the C6 glioma rat model. Methods Twenty-six rats of the C6 glioma model were randomly divided into a treatment group (received bevacizumab) and a control group (physiological saline). IVIM-DWI was performed on days 0, 1, 3, 5, and 7 after anti-angiogenic therapy and tumor growth and IVIM-DWI parameters were dynamically observed. Hematoxylin and eosin, CD34 microvessel density (MVD), proliferation of cell nuclear antigen (PCNA), and Hif-α staining were performed on day 7. One-way ANOVA was used to compare intra-group differences and an independent-samples t-test was used to compare inter-group differences of MRI parameters. Correlations between IVIM-DWI parameters, tumor size, and pathological results were analyzed. Results The relative change in tumor volume (ΔVolume) in the two groups differed significantly on days 5 and 7 (p = 0.038 and p < 0.001). The perfusion-related parameters D*- and f-values decreased in the treatment group and demonstrated significant differences compared with the control group on days 3, 5, and 7 (p = 0.033, p < 0.001, and p < 0.001, respectively). The diffusion-related parameters ADC and D-values increased in the treatment group and were found to be significantly differently different from the control group on days 5 and 7 (both p < 0.001). The initial D-value showed a negative correlation with ΔVolume (γ = −0.744, p < 0.001), whereas the initial D*-value and relative change of D-value had a positive correlation with ΔVolume (γ = 0.718, p < 0.001 and γ = 0.800, p < 0.001, respectively). MVD was strongly positively correlated with D*-value (r = 0.886, p = 0.019), PCNA was negatively correlated with ADC- and D-values (r = −0.848, p = 0.033; and r = −0.928 p = 0.008, respectively), and Hif-1α was strongly negatively correlated with D*-value (r = −0.879, p = 0.010). Conclusion IVIM-DWI was sensitive and accurate in predicting and monitoring the effects of early anti-angiogenesis therapy in a C6 glioma rat model.
Collapse
Affiliation(s)
- Weishu Hou
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyang Xue
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yinfeng Qian
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongli Pan
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Man Xu
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yujun Shen
- Department of Basic Medical Sciences, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiaohu Li
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Xiaohu Li, ; Yongqiang Yu,
| | - Yongqiang Yu
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Xiaohu Li, ; Yongqiang Yu,
| |
Collapse
|
14
|
Liu Z, Liu B, Bian L, Wang H, Jia Y, Wang Y, Zhang W, Wang Y, Han Z, Cheng X, Lian X, Ren Z, Gao Y. ITGB3BP is a potential biomarker associated with poor prognosis of glioma. J Cell Mol Med 2021; 26:813-827. [PMID: 34953037 PMCID: PMC8817129 DOI: 10.1111/jcmm.17127] [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: 08/12/2021] [Revised: 11/12/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022] Open
Abstract
Despite the growing recognition of ITGB3BP as an essential feature of various cancers, the relationship between ITGB3BP and glioma remains unclear. The main aim of this study was to determine the prognostic and diagnostic value of ITGB3BP in glioma. RNA-Seq and microarray data from 2222 glioma patients were included, and we found that the expression level of ITGB3BP in glioma tissues was significantly higher than that in normal brain tissues. Moreover, ITGB3BP can be considered an independent risk factor for poor prognosis and has great predictive value for the prognosis of glioma. Gene Set Enrichment Analysis results showed that ITGB3BP contributes to the poor prognosis of glioma by activating tumour-related signalling pathways. Some small-molecule drugs were identified, such as hexestrol, which may specifically inhibit ITGB3BP and be useful in the treatment of glioma. The TIMER database analysis results revealed a correlation between the expression of ITGB3BP and the infiltration of various immune cells in glioma. Our findings provide the first evidence that the up-regulation of ITGB3BP correlates with poor prognosis in human glioma. Thus, ITGB3BP is a potential new biomarker that can be used for the clinical diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| | - Binfeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, China
| | - Lu Bian
- Department of Dermatology, Henan University People's Hospital, Henan Provincial People's Hospital, Henan, China
| | - Hongbo Wang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| | - Yulong Jia
- Department of Neurosurgery of the Henan Provincial People's Hospital, Henan, China
| | - Yubo Wang
- College of Agriculture, Henan University of Science and Technology, Luoyang, China
| | - Wang Zhang
- Department of Neurosurgery of the First affiliate Hospital of Harbin Medical University, Harbin, China
| | - Yanbiao Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, China
| | - Zhibin Han
- Department of Neurosurgery of the First affiliate Hospital of Harbin Medical University, Harbin, China
| | - Xingbo Cheng
- Department of Neurosurgery of the First affiliate Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Lian
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, China
| | - Zhishuai Ren
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| |
Collapse
|
15
|
Lei J, Zhou Z. Efficacy and safety of bevacizumab combined with temozolomide in the treatment of recurrent malignant gliomas and its influence on serum tumor markers. Am J Transl Res 2021; 13:13886-13893. [PMID: 35035729 PMCID: PMC8748084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/14/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To determine the efficacy and safety of bevacizumab (Bz) combined with temozolomide (TMZ) in the treatment of recurrent malignant gliomas and its influence on serum tumor markers (STMs). METHODS The clinical data of 73 patients with recurrent malignant gliomas admitted to the First People's Hospital of Shuangliu District from April 2016 to June 2018 were analyzed retrospectively. Patients were divided into two groups according to different therapies: the control group (n=33) treated with TMZ, and the research group (n=40) treated with Bz combined with TMZ (Bz+TMZ). The overall response rate (ORR), disease control rate (DCR) and incidence of adverse reactions (ARs) were observed after 4 courses of treatment. The levels of STMs were detected. Additionally, the Karnofsky Performance Scale (KPS) score and quality of life (QoL) before and after treatment were compared between the two groups. The 1-year and 2-year survival rates as well as median survival time (MST) were also compared after 2-year follow-up. Treatment satisfaction was recorded and compared. RESULTS After treatment, the research group exhibited better ORR and DCR than the control group; The incidence of ARs differed insignificantly between the two arms; The serum levels of Vascular endothelial growth factor (VEGF), epidermal growth factor (EGF) and transforming growth factor β (TGF-β) in the research group were statistically lower than those in the control group; The KPS score and QoL score increased significantly in both arms, and were higher in the research group compared with the control group; the research group was also superior to the control group in treatment satisfaction. The research group showed higher 1-year and 2-year survival rates than the control group. CONCLUSIONS BZ+TMZ is effective in treating recurrent malignant gliomas, which can improve the QoL and survival of patients.
Collapse
|
16
|
Gerwing M, Krähling T, Schliemann C, Harrach S, Schwöppe C, Berdel AF, Klein S, Hartmann W, Wardelmann E, Heindel WL, Lenz G, Berdel WE, Wildgruber M. Multiparametric Magnetic Resonance Imaging for Immediate Target Hit Assessment of CD13-Targeted Tissue Factor tTF-NGR in Advanced Malignant Disease. Cancers (Basel) 2021; 13:cancers13235880. [PMID: 34884988 PMCID: PMC8657298 DOI: 10.3390/cancers13235880] [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: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Since the knowledge of tumor biology has advanced, a variety of targeted therapies has been developed. These do not immediately affect the tumor size, so optimized oncological imaging is needed. In this phase I study of patients with advanced malignant disease, a multiparametric imaging approach was used to assess changes in tumor perfusion after vessel-occluding therapy with the CD13 targeted truncated tissue factor with a C-terminal NGR-peptide. It comprises different sequences and the use of two different contrast media, ferucarbotran and gadobutrol. This multiparametric MRI protocol enables assessing the therapy effectiveness as early as five hours after therapy initiation. Abstract Early assessment of target hit in anti-cancer therapies is a major task in oncologic imaging. In this study, immediate target hit and effectiveness of CD13-targeted tissue factor tTF-NGR in patients with advanced malignant disease enrolled in a phase I trial was assessed using a multiparametric MRI protocol. Seventeen patients with advanced solid malignancies were enrolled in the trial and received tTF-NGR for at least one cycle of five daily infusions. Tumor target lesions were imaged with multiparametric MRI before therapy initiation, five hours after the first infusion and after five days. The imaging protocol comprised ADC, calculated from DWI, and DCE imaging and vascular volume fraction (VVF) assessment. DCE and VVF values decreased within 5 h after therapy initiation, indicating early target hit with a subsequent decrease in tumor perfusion due to selective tumor vessel occlusion and thrombosis induced by tTF-NGR. Simultaneously, ADC values increased at five hours after tTF-NGR administration. In four patients, treatment had to be stopped due to an increase in troponin T hs, with subsequent anticoagulation. In these patients, a reversed effect, with DCE and VVF values increasing and ADC values decreasing, was observed after anticoagulation. Changes in imaging parameters were independent of the mean vessel density determined by immunohistochemistry. By using a multiparametric imaging approach, changes in tumor perfusion after initiation of a tumor vessel occluding therapy can be evaluated as early as five hours after therapy initiation, enabling early assessment of target hit.
Collapse
Affiliation(s)
- Mirjam Gerwing
- Clinic of Radiology, University Hospital Muenster, D-48149 Muenster, Germany; (T.K.); (W.L.H.); (M.W.)
- Correspondence:
| | - Tobias Krähling
- Clinic of Radiology, University Hospital Muenster, D-48149 Muenster, Germany; (T.K.); (W.L.H.); (M.W.)
| | - Christoph Schliemann
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Saliha Harrach
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Christian Schwöppe
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Andrew F. Berdel
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Sebastian Klein
- Gerhard-Domagk-Institute for Pathology, University of Muenster, D-48149 Muenster, Germany; (S.K.); (W.H.); (E.W.)
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute for Pathology, University of Muenster, D-48149 Muenster, Germany; (S.K.); (W.H.); (E.W.)
| | - Eva Wardelmann
- Gerhard-Domagk-Institute for Pathology, University of Muenster, D-48149 Muenster, Germany; (S.K.); (W.H.); (E.W.)
| | - Walter L. Heindel
- Clinic of Radiology, University Hospital Muenster, D-48149 Muenster, Germany; (T.K.); (W.L.H.); (M.W.)
| | - Georg Lenz
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pulmonology, University Hospital Muenster, D-48149 Muenster, Germany; (C.S.); (S.H.); (C.S.); (A.F.B.); (G.L.); (W.E.B.)
| | - Moritz Wildgruber
- Clinic of Radiology, University Hospital Muenster, D-48149 Muenster, Germany; (T.K.); (W.L.H.); (M.W.)
- Department of Radiology, University Hospital, LMU Munich, D-81377 Munich, Germany
| |
Collapse
|
17
|
Shi W, Qu C, Wang X, Liang X, Tan Y, Zhang H. Diffusion kurtosis imaging combined with dynamic susceptibility contrast-enhanced MRI in differentiating high-grade glioma recurrence from pseudoprogression. Eur J Radiol 2021; 144:109941. [PMID: 34735828 DOI: 10.1016/j.ejrad.2021.109941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To compare the added value of diffusion kurtosis imaging (DKI) with the combination of dynamic susceptibility contrast-enhanced (DSC) MRI in differentiating glioma recurrence from pseudoprogression. METHODS Thirty-four patients with high-grade gliomas developing new and/or increasing enhanced lesions within six months after surgery and chemoradiotherapy were retrospectively analyzed. All patients were pathologically confirmed to have recurrent glioma (n = 22) or pseudoprogression (n = 12). The DKI and DSC MRI parameters were calculated based on the enhanced lesions on contrast-enhanced T1WI. ROC analysis was performed on significant variables to determine their diagnostic performance. Multivariate logistic regression was used to determine the best prediction model for discrimination. RESULTS The relative mean kurtosis (rMK), relative axial kurtosis (rKa), relative cerebral blood volume (rCBV), and relative mean transit time (rMTT) of glioma recurrence were higher than those of pseudoprogression (all, P < 0.05). The AUCs and diagnostic accuracy were 0.879 and 82.35% for rMK, 0.723 and 70.59% for rKa, 0.890 and 82.35% for rCBV, 0.765 and 73.53% for rMTT, respectively. A multivariate logistic regression model showed a significant contribution of rMK (P = 0.006) and rCBV (P = 0.009) as independent imaging classifiers for discrimination. The combined use of rMK and rCBV improved the AUC to 0.924 (P < 0.001) and the diagnostic accuracy to 88.24%. CONCLUSION DKI may be a valuable non-invasive tool in differentiating glioma recurrence from pseudoprogression, and its use in combination with DSC MRI can improve diagnostic performance in assessing treatment response compared with either technique alone.
Collapse
Affiliation(s)
- Wenwei Shi
- Department of Radiology, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, Jiangsu Province, PR China
| | - Chongxiao Qu
- Department of Pathology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, No. 29 of Twin Towers Temple Street, Taiyuan 030012, Shanxi Province, PR China
| | - Xiaochun Wang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China
| | - Xiao Liang
- Department of Radiology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, No. 29 of Twin Towers Temple Street, Taiyuan 030012, Shanxi Province, PR China
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China.
| | - Hui Zhang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China.
| |
Collapse
|
18
|
Zhang B, Yao J, Lian X, Liu B, Wang Y, Wang H, Wang J, Zhang M, Zhao Y, Zhu Y, Liu R, Gao Y. Role of RHOC in evaluating an adverse prognosis in patients with glioma and its potential prognostic value. Mol Clin Oncol 2021; 15:171. [PMID: 34276990 PMCID: PMC8278397 DOI: 10.3892/mco.2021.2333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/07/2021] [Indexed: 12/09/2022] Open
Abstract
In recent years, major discoveries have indicated that Ras homology family member C (RHOC) is involved in the occurrence and pathological progression of a number of malignant tumours; nevertheless, the role served by RHOC in glioma remains unclear. The present study aimed to gain further insight into the biological function and expression of RHOC in human glioma based on the Chinese Glioma Genome Atlas (CGGA). The current study analysed ~1,000 glioma samples from the CGGA. First, RHOC expression was analysed according to the clinical features associated with the prognosis of glioma, such as clinical stage, histological type and age. Second, the Kaplan-Meier method was used, revealing that the survival rate of patients with glioma with high RHOC expression was significantly lower than that of patients with low RHOC expression. Receiver operating characteristic curve analysis indicated that RHOC had moderate diagnostic value for patients with glioma. Gene set enrichment analysis indirectly indicated that RHOC mainly participated in the pathological mechanism of glioma through p53, extracellular matrix receptor interaction and focal adhesion. Finally, the aforementioned results were further verified using The Cancer Genome Atlas data and reverse transcription-quantitative PCR technology. To the best of our knowledge, the present study was the first comprehensive in-depth analysis of RHOC, revealing the potential value of RHOC as a novel oncogene in glioma. The current study provided a novel potential biomarker for the diagnosis and prognosis of glioma, and re-examined the pathological mechanism of glioma from a new perspective.
Collapse
Affiliation(s)
- Bo Zhang
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jiawei Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaoyu Lian
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Binfeng Liu
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yanbiao Wang
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Hongbo Wang
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jialin Wang
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Mengjun Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150086, P.R. China
| | - Yaoye Zhao
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yongjie Zhu
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Runze Liu
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
19
|
Mweempwa A, Rosenthal MA, Dimou J, Drummond KJ, Whittle JR. Perioperative clinical trials for glioma: Raising the bar. J Clin Neurosci 2021; 89:144-150. [PMID: 34119258 DOI: 10.1016/j.jocn.2021.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/25/2021] [Indexed: 02/07/2023]
Abstract
Gliomas are a heterogeneous group of primary brain cancers with poor survival despite multimodality therapy that includes surgery, radiation and chemotherapy. Numerous clinical trials have investigated systemic therapies in glioma, but have largely been negative. Multiple factors have contributed to the lack of progress including tumour heterogeneity, the tumour micro-environment and presence of the blood-brain barrier, as well as extrinsic factors relating to trial design, such as the lack of a contemporaneous biopsy at the time of treatment. A number of strategies have been proposed to progress new agents into the clinic. Here, we review the progress of perioperative, including phase 0 and 'window of opportunity', studies and provide recommendations for trial design in the development of new agents for glioma. The incorporation of pre- and post-treatment biopsies in glioma early phase trials will provide valuable pharmacokinetic and pharmacodynamic data and also determine the target or biomarker effect, which will guide further development of new agents. Perioperative 'window of opportunity' studies must use drugs with a recommended-phase-2-dose, known safety profile and adequate blood-brain barrier penetration. Drugs shown to have on-target effects in perioperative trials can then be evaluated further in a larger cohort of patients in an adaptive trial to increase the efficiency of drug development.
Collapse
Affiliation(s)
- Angela Mweempwa
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Mark A Rosenthal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - James Dimou
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC 3050, Australia; Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katharine J Drummond
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC 3050, Australia; Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - James R Whittle
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
20
|
Álvarez-Torres MDM, Fuster-García E, Reynés G, Juan-Albarracín J, Chelebian E, Oleaga L, Pineda J, Auger C, Rovira A, Emblem KE, Filice S, Mollà-Olmos E, García-Gómez JM. Differential effect of vascularity between long- and short-term survivors with IDH1/2 wild-type glioblastoma. NMR IN BIOMEDICINE 2021; 34:e4462. [PMID: 33470039 DOI: 10.1002/nbm.4462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/28/2020] [Indexed: 06/12/2023]
Abstract
INTRODUCTION IDH1/2 wt glioblastoma (GB) represents the most lethal tumour of the central nervous system. Tumour vascularity is associated with overall survival (OS), and the clinical relevance of vascular markers, such as rCBV, has already been validated. Nevertheless, molecular and clinical factors may have different influences on the beneficial effect of a favourable vascular signature. PURPOSE To evaluate the association between the rCBV and OS of IDH1/2 wt GB patients for long-term survivors (LTSs) and short-term survivors (STSs). Given that initial high rCBV may affect the patient's OS in follow-up stages, we will assess whether a moderate vascularity is beneficial for OS in both groups of patients. MATERIALS AND METHODS Ninety-nine IDH1/2 wt GB patients were divided into LTSs (OS ≥ 400 days) and STSs (OS < 400 days). Mann-Whitney and Fisher, uni- and multiparametric Cox, Aalen's additive regression and Kaplan-Meier tests were carried out. Tumour vascularity was represented by the mean rCBV of the high angiogenic tumour (HAT) habitat computed through the haemodynamic tissue signature methodology (available on the ONCOhabitats platform). RESULTS For LTSs, we found a significant association between a moderate value of rCBVmean and higher OS (uni- and multiparametric Cox and Aalen's regression) (p = 0.0140, HR = 1.19; p = 0.0085, HR = 1.22) and significant stratification capability (p = 0.0343). For the STS group, no association between rCBVmean and survival was observed. Moreover, no significant differences (p > 0.05) in gender, age, resection status, chemoradiation, or MGMT methylation were observed between LTSs and STSs. CONCLUSION We have found different prognostic and stratification effects of the vascular marker for the LTS and STS groups. We propose the use of rCBVmean at HAT as a vascular marker clinically relevant for LTSs with IDH1/2 wt GB and maybe as a potential target for randomized clinical trials focused on this group of patients.
Collapse
Affiliation(s)
| | | | - Gaspar Reynés
- Cancer Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | | | | | | | - Jose Pineda
- Hospital Clinic de Barcelona, Barcelona, Spain
| | - Cristina Auger
- Magnetic Resonance Unit, Department of Radiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Rovira
- Magnetic Resonance Unit, Department of Radiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kyrre E Emblem
- Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway
| | - Silvano Filice
- Medical Physics, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Enrique Mollà-Olmos
- Departamento de Radiodiagnóstico, Hospital Universitario de la Ribera, Alzira, Spain
| | | |
Collapse
|
21
|
Vogelbaum MA, Krivosheya D, Borghei-Razavi H, Sanai N, Weller M, Wick W, Soffietti R, Reardon DA, Aghi MK, Galanis E, Wen PY, van den Bent M, Chang S. Phase 0 and window of opportunity clinical trial design in neuro-oncology: a RANO review. Neuro Oncol 2021; 22:1568-1579. [PMID: 32598442 DOI: 10.1093/neuonc/noaa149] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is a devastating disease with poor prognosis. Few effective chemotherapeutics are currently available, and much effort has been expended to identify new drugs capable of slowing tumor progression. The phase 0 trial design was developed to facilitate early identification of promising agents for cancer that should undergo accelerated approval. This design features an early in-human study that enrolls a small number of patients who receive subtherapeutic doses of medication with the goals of describing pharmacokinetics through drug blood level measurements and determining intratumoral concentrations of the investigational compound as well as pharmacodynamics by studying the biochemical and physiological effects of drugs. In neuro-oncology, however, the presence of the blood-brain barrier and difficulty in obtaining brain tumor tissue warrant a separate set of considerations. In this paper, we critically reviewed the protocols used in all brain tumor related in-human phase 0 and phase 0-like ("window of opportunity") studies between 1993 and 2018, as well as ongoing clinical trials, and identified major challenges in trial design as applied to central nervous system tumors that include surgical specimen collection and storage, brain tumor drug level analysis, and confirmation of drug action. We therefore propose that phase 0 trials in neuro-oncology should include (i) only patients in whom a resection of the tumor is planned, (ii) use of clinical doses of an investigational agent, (iii) tissue sampling from enhancing and non-enhancing portions of the tumor, and (iv) assessment of drug-specific target effects. Standardization of clinical protocols for phase 0/window of opportunity studies can help accelerate the development of effective treatments for glioblastoma.
Collapse
Affiliation(s)
| | - Daria Krivosheya
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Nader Sanai
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Wolfgang Wick
- Department of Neurology Heidelberg University Hospital and German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science, Turin, Italy
| | - David A Reardon
- Center For Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | | | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam
| | - Susan Chang
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
22
|
Alsaab HO, Al-Hibs AS, Alzhrani R, Alrabighi KK, Alqathama A, Alwithenani A, Almalki AH, Althobaiti YS. Nanomaterials for Antiangiogenic Therapies for Cancer: A Promising Tool for Personalized Medicine. Int J Mol Sci 2021; 22:1631. [PMID: 33562829 PMCID: PMC7915670 DOI: 10.3390/ijms22041631] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the hallmarks of cancer. Several studies have shown that vascular endothelium growth factor (VEGF) plays a leading role in angiogenesis progression. Antiangiogenic medication has gained substantial recognition and is commonly administered in many forms of human cancer, leading to a rising interest in cancer therapy. However, this treatment method can lead to a deteriorating outcome of resistance, invasion, distant metastasis, and overall survival relative to its cytotoxicity. Furthermore, there are significant obstacles in tracking the efficacy of antiangiogenic treatments by incorporating positive biomarkers into clinical settings. These shortcomings underline the essential need to identify additional angiogenic inhibitors that target numerous angiogenic factors or to develop a new method for drug delivery of current inhibitors. The great benefits of nanoparticles are their potential, based on their specific properties, to be effective mechanisms that concentrate on the biological system and control various important functions. Among various therapeutic approaches, nanotechnology has emerged as a new strategy for treating different cancer types. This article attempts to demonstrate the huge potential for targeted nanoparticles and their molecular imaging applications. Notably, several nanoparticles have been developed and engineered to demonstrate antiangiogenic features. This nanomedicine could effectively treat a number of cancers using antiangiogenic therapies as an alternative approach. We also discuss the latest antiangiogenic and nanotherapeutic strategies and highlight tumor vessels and their microenvironments.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
| | - Alanoud S. Al-Hibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia;
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Khawlah K. Alrabighi
- Batterjee Medical College for Sciences and Technology, Jeddah 21577, Saudi Arabia;
| | - Aljawharah Alqathama
- Department of Pharmacognosy, Pharmacy College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Akram Alwithenani
- Department of Laboratory Medicine, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Atiah H. Almalki
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
- Department of Pharmaceutical Chemistry, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Yusuf S. Althobaiti
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (A.H.A.); (Y.S.A.)
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
23
|
Advanced magnetic resonance imaging to support clinical drug development for malignant glioma. Drug Discov Today 2020; 26:429-441. [PMID: 33249294 DOI: 10.1016/j.drudis.2020.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/23/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022]
Abstract
Even though the treatment options and survival of patients with glioblastoma multiforme (GBM), the most common type of malignant glioma, have improved over the past decade, there is still a high unmet medical need to develop novel therapies. Complexity in pathology and therapy require biomarkers to characterize tumors, to define malignant and active areas, to assess disease prognosis, and to quantify and monitor therapy response. While conventional magnetic resonance imaging (MRI) techniques have improved these assessments, limitations remain. In this review, we evaluate the role of various non-invasive biomarkers based on advanced structural and functional MRI techniques in the context of GBM drug development over the past 5 years.
Collapse
|
24
|
Soliman MA, Guccione J, Reiter AM, Moawad AW, Etchison A, Kamel S, Khatchikian AD, Elsayes KM. Current Concepts in Multi-Modality Imaging of Solid Tumor Angiogenesis. Cancers (Basel) 2020; 12:cancers12113239. [PMID: 33153067 PMCID: PMC7692820 DOI: 10.3390/cancers12113239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The recent increase in the use of targeted molecular therapy including anti-angiogenetic agents in cancer treatment necessitate the use of robust tools to assess and guide treatment. Angiogenesis, the formation of new disorganized blood vessels, is used by tumor cells to grow and spread using different mechanisms that could be targeted by anti-angiogenetic agents. In this review, we discuss the biological principles of tumor angiogenesis and the imaging modalities that could provide information beyond gross tumor size and morphology to capture the efficacy of anti-angiogenetic therapeutic response. Abstract There have been rapid advancements in cancer treatment in recent years, including targeted molecular therapy and the emergence of anti-angiogenic agents, which necessitate the need to quickly and accurately assess treatment response. The ideal tool is robust and non-invasive so that the treatment can be rapidly adjusted or discontinued based on efficacy. Since targeted therapies primarily affect tumor angiogenesis, morphological assessment based on tumor size alone may be insufficient, and other imaging modalities and features may be more helpful in assessing response. This review aims to discuss the biological principles of tumor angiogenesis and the multi-modality imaging evaluation of anti-angiogenic therapeutic responses.
Collapse
Affiliation(s)
- Moataz A. Soliman
- Department of Diagnostic Radiology, Northwestern University, Evanston, IL 60201, USA;
| | - Jeffrey Guccione
- Department of Diagnostic and Interventional Imaging, The University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA;
| | - Anna M. Reiter
- School of Medicine, University of Texas Southwestern, Dallas, TX 75390, USA;
| | - Ahmed W. Moawad
- Department of Diagnostic Radiology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Ashley Etchison
- Department of Diagnostic Radiology, Baylor College of Medicine, Houston, TX 76798, USA;
| | - Serageldin Kamel
- Department of Lymphoma and Myeloma, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Aline D. Khatchikian
- Department of Diagnostic Radiology, McGill University, Montreal, QC H3G 1A4, Canada;
| | - Khaled M. Elsayes
- Department of Diagnostic Radiology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence:
| |
Collapse
|
25
|
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
|
26
|
Jiang YW, Wang R, Zhuang YD, Chen CM. Identification and validation of potential novel prognostic biomarkers for patients with glioma based on a gene co-expression network. Transl Cancer Res 2020; 9:6444-6454. [PMID: 35117252 PMCID: PMC8798165 DOI: 10.21037/tcr-20-492] [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: 01/12/2020] [Accepted: 08/28/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Glioma is widely regarded as one of most lethal and challenging diseases of the nervous system. The aim of this study was to identify novel biomarkers that offer better prognosis prediction for Chinese patients with glioma. METHODS By using systematic approaches, the co-expression modules were identified from the Chinese Glioma Genome Atlas (CGGA) database through weighted gene co-expression network analysis and functional enrichment of essential modules of Kyoto Encyclopedia of Genes and Genomes terms. The co-expression modules were validated using The Cancer Genome Atlas database and the protein-protein interaction (PPI) network. RESULTS For network construction, 5,374 among 21,494 genes were selected, and an increasing genetic variance was associated with the prognosis of glioma. By using functional enrichment analysis, the involvement of multiple vital processes, including metabolism of fatty acids, was correlated with the patient prognosis. Notably, five hub genes (KCNB1, UST, SOX8, KLHL42, and HDAC4) were identified for these processes. Accordingly, using the Kaplan-Meier method, there was enhanced expression of these genes in patients with significantly lower overall survival rates, especially those from the CGGA database. CONCLUSIONS This study not only revealed the essential co-expression gene modules in patients with glioma, but it also unraveled the potential signaling pathways underlying these functional processes.
Collapse
Affiliation(s)
- Yan-Wei Jiang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Wang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan-Dong Zhuang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chun-Mei Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
27
|
Zhang J, Xue W, Xu K, Yi L, Guo Y, Xie T, Tong H, Zhou B, Wang S, Li Q, Liu H, Chen X, Fang J, Zhang W. Dual inhibition of PFKFB3 and VEGF normalizes tumor vasculature, reduces lactate production, and improves chemotherapy in glioblastoma: insights from protein expression profiling and MRI. Theranostics 2020; 10:7245-7259. [PMID: 32641990 PMCID: PMC7330843 DOI: 10.7150/thno.44427] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Tumor vascular normalization (TVN) is emerging to enhance the efficacy of anticancer treatment in many cancers including glioblastoma (GBM). However, a common and severe challenge being currently faced is the transient TVN effect, hampering the sustained administration of anticancer therapy during TVN window. Additionally, the lack of non-contrast agent-based imaging biomarkers to monitor TVN process postpones the clinical translation of TVN strategy. In this study, we investigated whether dual inhibition of VEGF and the glycolytic activator PFKFB3 could reinforce the TVN effect in GBM. Dynamic contrast-enhanced-magnetic resonance imaging (DCE-MRI) and intravoxel incoherent motion (IVIM)-MRI were performed to monitor TVN process and to identify whether IVIM-MRI is a candidate or complementary imaging biomarker for monitoring TVN window without exogenous contrast agent administration. Methods: Patient-derived orthotopic GBM xenografts in mice were established and treated with bevacizumab (BEV), 3PO (PFKFB3 inhibitor), BEV+3PO dual therapy, or saline. The vascular morphology, tumor hypoxia, and lactate level were evaluated before and at different time points after treatments. Doxorubicin was used to evaluate chemotherapeutic efficacy and drug delivery. Microarray of angiogenesis cytokines and western blotting were conducted to characterize post-treatment molecular profiling. TVN process was monitored by DCE- and IVIM-MRI. Correlation analysis of pathological indicators and MRI parameters was further analyzed. Results: Dual therapy extended survival and delayed tumor growth over each therapy alone, concomitant with a decrease of cell proliferation and an increase of cell apoptosis. The dual therapy reinforces TVN effect, thereby alleviating tumor hypoxia, reducing lactate production, and improving the efficacy and delivery of doxorubicin. Mechanistically, several angiogenic cytokines and pathways were downregulated after dual therapy. Notably, dual therapy inhibited Tie1 expression, the key regulator of TVN, in both endothelial cells and tumor cells. DCE- and IVIM-MRI data showed that dual therapy induced a more homogenous and prominent TVN effect characterized by improved vascular function in tumor core and tumor rim. Correlation analysis revealed that IVIM-MRI parameter D* had better correlations with TVN pathological indicators compared with the DCE-MRI parameter Ktrans. Conclusions: Our results propose a rationale to overcome the current limitation of BEV monotherapy by integrating the synergistic effects of VEGF and PFKFB3 blockade to enhance chemotherapy efficacy through a sustained TVN effect. Moreover, we unveil IVIM-MRI parameter D* has much potential as a complementary imaging biomarker to monitor TVN window more precisely without exogenous contrast agent injection.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Kai Xu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qing Li
- Department of Oncology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, 400042, China
| |
Collapse
|
28
|
Liu Z, Shen F, Wang H, Li A, Wang J, Du L, Liu B, Zhang B, Lian X, Pang B, Liu L, Gao Y. Abnormally high expression of HOXA2 as an independent factor for poor prognosis in glioma patients. Cell Cycle 2020; 19:1632-1640. [PMID: 32436804 DOI: 10.1080/15384101.2020.1762038] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, studies have revealed HOXA2 as a new oncogene, but its function is unknown in gliomas. We aimed to reveal the relationship between HOXA2 and glioma based on the Chinese Glioma Genome Atlas(CGGA) and the cancer genome atlas (TCGA). HOXA2 expression data and clinically relevant information of glioma patients were obtained from the CGGA and TCGA containing 1447 glioma tissues and five non-tumor brain tissues. The Wilcox or Kruskal tests were used to detect the correlation between the HOXA2 expression level and clinical data of glioma patients. the Kaplan-Meier method were used to examine the relationship between HOXA2 and overall patient survival. Gene set enrichment analysis (GSEA) was conducted to indirectly reveal the signaling pathways involved in HOXA2, and RT-PCR was used to detect HOXA2 expression in gliomas and non-tumor brain tissues. High HOXA2 expression was found to be positively correlated with clinical grade, histological type, age, and tumor recurrence, but negatively correlated with 1p19 codeletion and isocitrate dehydrogenase mutation status.RT-PCR results showed that HOXA2 expression levels were significantly higher in tumor tissues than in non-tumor brain tissues. GSEA showed that HOXA2 promoted the activation of the activation of the JAK-STAT-signaling pathway, focal adhesion, cell-adhesion-molecules-CAMS pathway, cytosolic DNA sensing pathway, and natural killer cell-mediated cytotoxicity. This study revealed for the first time that the novel oncogene,HOXA2, leads to poor prognosis in gliomas, and can be used as a biomarker for the diagnosis and treatment of gliomas.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University , Zhengzhou, Henan, China
| | - Fei Shen
- Department of Ophthalmology, Kaifeng Central Hospital , Kaifeng, Henan, China
| | - Hongbo Wang
- Henan Provincial People's Hospital, Henan University People's Hospital , Zhengzhou, Henan, China
| | - Ang Li
- Department of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University , Zhengzhou, Henan, China
| | - Jialin Wang
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital , Zhengzhou, Henan, China
| | - Lin Du
- Department of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University , Zhengzhou, Henan, China
| | - Binfeng Liu
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital , Zhengzhou, Henan, China
| | - Bo Zhang
- Henan Provincial People's Hospital, Henan University People's Hospital , Zhengzhou, Henan, China
| | - Xiaoyu Lian
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital , Zhengzhou, Henan, China
| | - Bo Pang
- Department of Neurosurgery, The Fourth Medical Center of Chinese PLA General Hospital , Beijing, China
| | - Liyun Liu
- Department of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University , Zhengzhou, Henan, China
| | - Yanzheng Gao
- Department of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University , Zhengzhou, Henan, China
| |
Collapse
|
29
|
Xue W, Zhang J, Tong H, Xie T, Chen X, Zhou B, Wu P, Zhong P, Du X, Guo Y, Yang Y, Liu H, Fang J, Wang S, Wu H, Xu K, Zhang W. Effects of BMPER, CXCL10, and HOXA9 on Neovascularization During Early-Growth Stage of Primary High-Grade Glioma and Their Corresponding MRI Biomarkers. Front Oncol 2020; 10:711. [PMID: 32432046 PMCID: PMC7214627 DOI: 10.3389/fonc.2020.00711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/15/2020] [Indexed: 02/02/2023] Open
Abstract
Neovascularization is required in high-grade glioma (HGG). The objective of this study was to explore neovascularization-related genes and their corresponding MRI biomarkers during the early-growth stage of HGG. Tumor tissues from 30 HGG patients underwent perfusion MRI scanning prior to surgery were used to establish orthotopic xenograft models, pathologically analyze the tumor vasculature and perform transcriptome sequencing. The cases were divided into two groups based on whether the xenograft was successfully established. Microvascular density and BMPER, CXCL10, and HOXA9 expression of surgical specimens in the xenograft-forming group was significantly elevated and the microvascular diameter was significantly reduced, in vitro inhibition of BMPER, CXCL10, or HOXA9 in the glioma stem cell significantly suppressed its tube formation abilities. The in vivo experiment showed that BMPER was highly expressed in the early tumor growth phase (20 days), CXCL10 and HOXA9 expression was elevated with tumor progress, and spatially associated with tumor vasculature. Perfusion weighted MRI (PWI-MRI) derived parameters, rCBV, rCBF, Ktrans, and Vp, were also increased in the xenograft-forming group. In conclusion BMPER, CXCL10, and HOXA9 promote early tumor growth and progression by stimulating neovascularization of primary HGG. The rCBV, rCBF, Ktrans, and Vp can be used as imaging biomarkers to predict the expression statuses of these genes.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Pengfei Wu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xuesong Du
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Youyuan Yang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Heng Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Wu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Kai Xu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Clinical Research Centre of Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
30
|
Comparison of Model-Free and Model-Based Dynamic Contrast-Enhanced Magnetic Resonance Imaging Pharmacokinetic Parameters for Predicting Breast Cancers' Response to Neoadjuvant Chemotherapy. J Comput Assist Tomogr 2020; 44:269-274. [PMID: 32195807 DOI: 10.1097/rct.0000000000001001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To prospectively compare the performance of model-based and model-free dynamic contrast-enhanced (DCE) pharmacokinetic parameters in monitoring breast cancers' early response to neoadjuvant chemotherapy (NACT). METHODS Sixty patients, with 61 pathology-proven breast cancers, were examined using DCE magnetic resonance imaging before, after the first cycle, and after full cycles of NACT. Both model-based (Ktrans and others) and model-free parameters, mainly time-intensity curve (TIC), were measured. According to Miller-Payne grading, patients were divided into response and nonresponse group. Mann-Whitney U test, Fisher exact test, multivariate logistic regression, and receiver operating characteristic curve were used in analysis. RESULTS After the first cycle, among all the parameters, Ktrans and TIC were strongly associated with tumors' early response. There was no significant difference between the areas under receiver operating characteristic curve of Ktrans and TIC (0.768, 0.852, respectively). CONCLUSIONS Model-based and model-free DCE parameters, especially Ktrans and TIC, have similar performance in predicting the efficacy of NACT for breast cancers.
Collapse
|
31
|
Yang Y, Yang Y, Wu X, Pan Y, Zhou D, Zhang H, Chen Y, Zhao J, Mo Z, Huang B. Adding DSC PWI and DWI to BT-RADS can help identify postoperative recurrence in patients with high-grade gliomas. J Neurooncol 2020; 146:363-371. [PMID: 31902040 DOI: 10.1007/s11060-019-03387-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/27/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The Brain Tumor Reporting and Data System (BT-RADS) category 3 is suitable for identifying cases with intermediate probability of tumor recurrence that do not meet the Response Assessment in Neuro-Oncology (RANO) criteria for progression. The aim of this study was to evaluate the added value of dynamic susceptibility contrast-enhanced perfusion-weighted imaging (DSC PWI) and diffusion-weighted imaging (DWI) to BT-RADS for differentiating tumor recurrence from non-recurrence in postoperative high-grade glioma (HGG) patients with category 3 lesions. METHODS Patients with BT-RADS category 3 lesions were included. The maximal relative cerebral blood volume (rCBVmax) and the mean apparent diffusion coefficient (ADCmean) values were measured. The added value of DSC PWI and DWI to BT-RADS was evaluated by receiver operating characteristic (ROC) curve analysis. RESULTS Fifty-one of 91 patients had tumor recurrence, and 40 patients did not. There were significant differences in rCBVmax and ADCmean between the tumor recurrence group and non-recurrence group. Compared to BT-RADS alone, the addition of DSC PWI to BT-RADS increased the area under curve (AUC) from 0.76 (95% confidence interval [CI] 0.66-0.84) to 0.90 (95% CI 0.81-0.95) for differentiating tumor recurrence from non-recurrence. The addition of DWI to BT-RADS increased the AUC from 0.76 (95% CI 0.66-0.84) to 0.88 (95% CI 0.80-0.94). The combination of BT-RADS, DSC PWI, and DWI exhibited the best diagnostic performance (AUC = 0.95; 95% CI 0.88-0.98) for differentiating tumor recurrence from non-recurrence. CONCLUSION Adding DSC PWI and DWI to BT-RADS can significantly improve the diagnostic performance for differentiating tumor recurrence from non-recurrence in BT-RADS category 3 lesions.
Collapse
Affiliation(s)
- Yuelong Yang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yunjun Yang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xiaoling Wu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yi Pan
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Dong Zhou
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hongdan Zhang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yonglu Chen
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Jiayun Zhao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zihua Mo
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Biao Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
32
|
Silencing of Mig-7 expression inhibits in-vitro invasiveness and vasculogenic mimicry of human glioma U87 Cells. Neuroreport 2019; 30:1135-1142. [DOI: 10.1097/wnr.0000000000001317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Whole brain apparent diffusion coefficient measurements correlate with survival in glioblastoma patients. J Neurooncol 2019; 146:157-162. [PMID: 31797235 PMCID: PMC6938471 DOI: 10.1007/s11060-019-03357-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant primary brain tumor, and methods to improve the early detection of disease progression and evaluate treatment response are highly desirable. We therefore explored changes in whole-brain apparent diffusion coefficient (ADC) values with respect to survival (progression-free [PFS], overall [OS]) in a cohort of GBM patients followed at regular intervals until disease progression. METHODS A total of 43 subjects met inclusion criteria and were analyzed retrospectively. Histogram data were extracted from standardized whole-brain ADC maps including skewness, kurtosis, entropy, median, mode, 15th percentile (p15) and 85th percentile (p85) values, and linear regression slopes (metrics versus time) were fitted. Regression slope directionality (positive/negative) was subjected to univariate Cox regression. The final model was determined by aLASSO on metrics above threshold. RESULTS Skewness, kurtosis, median, p15 and p85 were all below threshold for both PFS and OS and were analyzed further. Median regression slope directionality best modeled PFS (p = 0.001; HR 3.3; 95% CI 1.6-6.7), while p85 was selected for OS (p = 0.002; HR 0.29; 95% CI 0.13-0.64). CONCLUSIONS Our data show tantalizing potential in the use of whole-brain ADC measurements in the follow up of GBM patients, specifically serial median ADC values which correlated with PFS, and serial p85 values which correlated with OS. Whole-brain ADC measurements are fast and easy to perform, and free of ROI-placement bias.
Collapse
|
34
|
Li X, Xue Y, Liu X, Zheng J, Shen S, Yang C, Chen J, Li Z, Liu L, Ma J, Ma T, Liu Y. ZRANB2/SNHG20/FOXK1 Axis regulates Vasculogenic mimicry formation in glioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:68. [PMID: 30744670 PMCID: PMC6371528 DOI: 10.1186/s13046-019-1073-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Background Glioma is the most common intracranial neoplasm with vasculogenic mimicry formation as one form of blood supply. Many RNA-binding proteins and long non-coding RNAs are involved in tumorigenesis of glioma. Methods The expression of ZRANB2, SNHG20 and FOXK1 in glioma were detected by real-time PCR or western blot. The function of ZRANB2/SNHG20/FOXK1 axis in glioma associated with vasculogenic mimicry formation was analyzed. Results ZRANB2 is up-regulated in glioma tissues and glioma cells. ZRANB2 knockdown inhibits the proliferation, migration, invasion and vasculogenic mimicry formation of glioma cells. ZRANB2 binds to SNHG20 and increases its stability. Knockdown of SNHG20 reduces the degradation of FOXK1 mRNA by SMD pathway. FOXK1 inhibits transcription by binding to the promoters of MMP1, MMP9 and VE-Cadherin and inhibits vasculogenic mimicry formation of glioma cells. Conclusions ZRANB2/SNHG20/FOXK1 axis plays an important role in regulating vasculogenic mimicry formation of glioma, which might provide new targets of glioma therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1073-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaozhi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Shuyuan Shen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Jiajia Chen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China. .,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China. .,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|