1
|
Fajemisin JA, Gonzalez G, Rosenberg SA, Ullah G, Redler G, Latifi K, Moros EG, El Naqa I. Magnetic Resonance-Guided Cancer Therapy Radiomics and Machine Learning Models for Response Prediction. Tomography 2024; 10:1439-1454. [PMID: 39330753 PMCID: PMC11435563 DOI: 10.3390/tomography10090107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Magnetic resonance imaging (MRI) is known for its accurate soft tissue delineation of tumors and normal tissues. This development has significantly impacted the imaging and treatment of cancers. Radiomics is the process of extracting high-dimensional features from medical images. Several studies have shown that these extracted features may be used to build machine-learning models for the prediction of treatment outcomes of cancer patients. Various feature selection techniques and machine models interrogate the relevant radiomics features for predicting cancer treatment outcomes. This study aims to provide an overview of MRI radiomics features used in predicting clinical treatment outcomes with machine learning techniques. The review includes examples from different disease sites. It will also discuss the impact of magnetic field strength, sample size, and other characteristics on outcome prediction performance.
Collapse
Affiliation(s)
- Jesutofunmi Ayo Fajemisin
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Glebys Gonzalez
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Stephen A Rosenberg
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Gage Redler
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kujtim Latifi
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Eduardo G Moros
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Issam El Naqa
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Habibi MA, Shad N, Mirjnani MS, Fasihi S, Sadeghi S, Karami S, Ahmadvand MH, Delbari P, Zare AH, Zare AH, Alavi SAN. Is add-on Bevacizumab therapy to Temozolomide and radiotherapy associated with clinical utility for newly diagnosed Glioblastoma? A systematic review and meta-analysis. Neurosurg Rev 2024; 47:445. [PMID: 39162874 DOI: 10.1007/s10143-024-02667-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
Bevacizumab, temozolomide (TMZ), and radiotherapy are three therapeutic methods, but the combination of them as a new approach for the treatment of newly diagnosed high-grade gliomas (HGGs) is still under investigation. Therefore, this study aims to evaluate the safety, efficacy, and clinical utility of this treatment approach for patients with glioblastoma (GBM). PubMed/Medline, Scopus, Embase, and Web of Science were systematically reviewed from inception to 24 August 2023. Relevant studies evaluating the therapeutic effect of adding Bevacizumab to TMZ-based chemotherapy and radiation therapy were enrolled. All statistical analysis was performed using the "meta" package of R. A total of 21 studies were included in this study. Our meta-analysis found that adding bevacizumab to standard therapy improved progression-free survival (PFS) in patients with newly diagnosed GBM. The pooled 6-month PFS rate was significantly higher with bevacizumab (79% vs. 56%, odds ratio 3.17). Overall survival (OS) showed modest improvements, with 2-year OS rates of 39% vs. 20% favoring bevacizumab. Radiological response rates varied, with a pooled overall response rate of 44% for bevacizumab-treated patients. The complete response rate was 16%, partial response 32%, and progressive disease 25%. Adverse events occurred in 62% of bevacizumab-treated patients. Common complications included fatigue, thrombocytopenia, and thromboembolic events. When added to standard therapy, bevacizumab demonstrates modest improvements in PFS and OS for newly diagnosedGBM. While it shows promise in short-term outcomes and radiological responses, long-term survival benefits remain limited. The risk of adverse events, particularly CNS hemorrhage, necessitates careful patient selection. These findings suggest that bevacizumab may have a role in treating high-grade gliomas, but its use should be individualized based on patient characteristics and risk-benefit assessment.
Collapse
Affiliation(s)
- Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Niloufar Shad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sara Fasihi
- Department of Anesthesiology, Qom University of Medical Sciences, Qom, Iran
| | - Sara Sadeghi
- School of Medicine, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Shaghayegh Karami
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Pouria Delbari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Zare
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hessam Zare
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ahmad Naseri Alavi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
3
|
Grundberg O, Skribek M, Swerkersson S, Skorpil M, Kölbeck K, Grozman V, Nyren S, Tsakonas G. Diffusion weighted MRI and apparent diffusion coefficient as a prognostic biomarker in evaluating chemotherapy-antiangiogenic treated stage IV non-small cell lung cancer: A prospective, single-arm, open-label, clinical trial (BevMar). Eur J Radiol 2024; 177:111557. [PMID: 38954912 DOI: 10.1016/j.ejrad.2024.111557] [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: 01/29/2024] [Revised: 05/20/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE When treating Lung Cancer, it is necessary to identify early treatment failure to enable timely therapeutic adjustments. The Aim of this study was to investigate whether changes in tumor diffusion during treatment with chemotherapy and bevacizumab could serve as a predictor of treatment failure. MATERIAL AND METHODS A prospective single-arm, open-label, clinical trial was conducted between September 2014 and December 2020, enrolling patients with stage IV non-small cell lung cancer (NSCLC). The patients were treated with chemotherapy-antiangiogenic combination. Diffusion weighted magnetic resonance imaging (DW-MRI) was performed at baseline, two, four, and sixteen weeks after initiating treatment. The differences in apparent diffusion coefficient (ADC) values between pre- and post-treatment MRIs were recorded as Delta values (ΔADC). We assessed whether ΔADC could serve as a prognostic biomarker for overall survival (OS), with a five year follow up. RESULTS 18 patients were included in the final analysis. Patients with a ΔADC value ≥ -3 demonstrated a significantly longer OS with an HR of 0.12 (95 % CI; 0.03- 0.61; p = 0.003) The median OS in patients with a ΔADC value ≥ -3 was 18 months, (95 % C.I; 7-46) compared to 7 months (95 % C.I; 5-9) in those with a ΔADC value < -3. CONCLUSION Our findings suggest that early changes in tumor ADC values, may be indicative of a longer OS. Therefore, DW-MRI could serve as an early biomarker for assessing treatment response in patients receiving chemotherapy combined with antiangiogenic therapy.
Collapse
Affiliation(s)
- Oscar Grundberg
- Department of Thoracic Oncology, Karolinska University Hospital, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Marcus Skribek
- Department of Thoracic Oncology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Mikael Skorpil
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Karl Kölbeck
- Department of Thoracic Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Vitali Grozman
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Thoracic Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sven Nyren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Thoracic Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Georgios Tsakonas
- Department of Thoracic Oncology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Brown LJ, Khou V, Brown C, Alexander M, Jayamanne D, Wei J, Gray L, Chan WY, Smith S, Harden S, Mersiades A, Warburton L, Itchins M, Lee JH, Pavlakis N, Clarke SJ, Boyer M, Nagrial A, Hau E, Pires da Silva I, Kao S, Kong BY. First-line chemoimmunotherapy and immunotherapy in patients with non-small cell lung cancer and brain metastases: a registry study. Front Oncol 2024; 14:1305720. [PMID: 38406805 PMCID: PMC10885799 DOI: 10.3389/fonc.2024.1305720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/08/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Brain metastases commonly occur in patients with non-small cell lung cancer (NSCLC). Standard first-line treatment for NSCLC, without an EGFR, ALK or ROS1 mutation, is either chemoimmunotherapy or anti-PD-1 monotherapy. Traditionally, patients with symptomatic or untreated brain metastases were excluded from the pivotal clinical trials that established first-line treatment recommendations. The intracranial effectiveness of these treatment protocols has only recently been elucidated in small-scale prospective trials. Methods Patients with NSCLC and brain metastases, treated with first-line chemoimmunotherapy or anti-PD-1 monotherapy were selected from the Australian Registry and biObank of thoracic cancers (AURORA) clinical database covering seven institutions. The primary outcome was a composite time-to-event (TTE) outcome, including extracranial and intracranial progression, death, or need for local intracranial therapy, which served as a surrogate for disease progression. The secondary outcome included overall survival (OS), intracranial objective response rate (iORR) and objective response rate (ORR). Results 116 patients were included. 63% received combination chemoimmunotherapy and 37% received anti-PD-1 monotherapy. 69% of patients received upfront local therapy either with surgery, radiotherapy or both. The median TTE was 7.1 months (95% CI 5 - 9) with extracranial progression being the most common progression event. Neither type of systemic therapy or upfront local therapy were predictive of TTE in a multivariate analysis. The median OS was 17 months (95% CI 13-27). Treatment with chemoimmunotherapy was predictive of longer OS in multivariate analysis (HR 0.35; 95% CI 0.14 - 0.86; p=0.01). The iORR was 46.6%. The iORR was higher in patients treated with chemoimmunotherapy compared to immunotherapy (58% versus 31%, p=0.01). The use of chemoimmunotherapy being predictive of iORR in a multivariate analysis (OR 2.88; 95% CI 1.68 - 9.98; p=0.04). Conclusion The results of this study of real-world data demonstrate the promising intracranial efficacy of chemoimmunotherapy in the first-line setting, potentially surpassing that of immunotherapy alone. No demonstrable difference in survival or TTE was seen between receipt of upfront local therapy. Prospective studies are required to assist clinical decision making regarding optimal sequencing of local and systemic therapies.
Collapse
Affiliation(s)
- Lauren Julia Brown
- Translational Radiation Biology and Oncology Group, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, NSW, Australia
- Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Victor Khou
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Radiation Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
- Department of Radiation Oncology, North Coast Cancer Institute, Coffs Harbour, NSW, Australia
| | - Chris Brown
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
| | - Marliese Alexander
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Pharmacy Department, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Dasantha Jayamanne
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Radiation Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
- Genesis Care, St Leonards, NSW, Australia
| | - Joe Wei
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Lauren Gray
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Wei Yen Chan
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Samuel Smith
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Susan Harden
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Radiation Oncology, Sir Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Antony Mersiades
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Northern Beaches Hospital, Frenches Forest, NSW, Australia
| | - Lydia Warburton
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, WA, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| | - Malinda Itchins
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Genesis Care, St Leonards, NSW, Australia
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Jenny H. Lee
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Nick Pavlakis
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Genesis Care, St Leonards, NSW, Australia
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Stephen J. Clarke
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Genesis Care, St Leonards, NSW, Australia
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Michael Boyer
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Adnan Nagrial
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, NSW, Australia
- Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Group, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, NSW, Australia
- Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ines Pires da Silva
- Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Wollstonecraft, NSW, Australia
| | - Steven Kao
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Benjamin Y. Kong
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, Australia
- Sydney Partnership for Health, Education, Research and Enterprise (SPHERE) Cancer Clinical Academic Group, Faculty of Medicine, University of New South Wales (NSW), Sydney, NSW, Australia
| |
Collapse
|
5
|
Sener U, Islam M, Webb M, Kizilbash SH. Antiangiogenic exclusion rules in glioma trials: Historical perspectives and guidance for future trial design. Neurooncol Adv 2024; 6:vdae039. [PMID: 38596714 PMCID: PMC11003534 DOI: 10.1093/noajnl/vdae039] [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] [Indexed: 04/11/2024] Open
Abstract
Background Despite the lack of proven therapies for recurrent high-grade glioma (HGG), only 8%-11% of patients with glioblastoma participate in clinical trials, partly due to stringent eligibility criteria. Prior bevacizumab treatment is a frequent exclusion criterion, due to difficulty with response assessment and concerns for rebound edema following antiangiogenic discontinuation. There are no standardized trial eligibility rules related to prior antiangiogenic use. Methods We reviewed ClinicalTrials.gov listings for glioma studies starting between May 2009 and July 2022 for eligibility rules related to antiangiogenics. We also reviewed the literature pertaining to bevacizumab withdrawal. Results Two hundred and ninety-seven studies for patients with recurrent glioma were reviewed. Most were phase 1 (n = 145, 49%), non-randomized (n = 257, 87%), evaluated a drug-only intervention (n = 223, 75%), and had a safety and tolerability primary objective (n = 181, 61%). Fifty-one (17%) excluded participants who received any antiangiogenic, one (0.3%) excluded participants who received any non-temozolomide systemic therapy. Fifty-nine (20%) outlined washout rules for bevacizumab (range 2-24 weeks, 4-week washout n = 35, 12% most common). Seventy-eight required a systemic therapy washout (range 1-6 weeks, 4-week washout n = 34, 11% most common). Nine permitted prior bevacizumab use with limitations, 18 (6%) permitted any prior bevacizumab, 5 (2%) were for bevacizumab-refractory disease, and 76 (26%) had no rules regarding antiangiogenic use. A literature review is then presented to define standardized eligibility criteria with a 6-week washout period proposed for future trial design. Conclusions Interventional clinical trials for patients with HGG have substantial heterogeneity regarding eligibility criteria pertaining to bevacizumab use, demonstrating a need for standardizing clinical trial design.
Collapse
Affiliation(s)
- Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mahnoor Islam
- Department of Neurology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mason Webb
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sani H Kizilbash
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Welch MR. Management of Complications in Neuro-oncology Patients. Continuum (Minneap Minn) 2023; 29:1844-1871. [PMID: 38085901 DOI: 10.1212/con.0000000000001359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
OBJECTIVE The purpose of this article is to familiarize the reader with the spectrum of neurologic and medical complications relevant to the care of patients with neurologic cancer while highlighting best practices to prevent morbidity and mortality. Topics include tumor-related epilepsy, vasogenic edema, complications of corticosteroid use, disruption of the hypothalamic-pituitary axis, venous thromboembolism, and opportunistic infection. LATEST DEVELOPMENTS In 2021, a joint guideline from the Society for Neuro-Oncology and the European Association of Neuro-Oncology reaffirmed recommendations first established in 2000 that patients with newly diagnosed brain tumors should not be prescribed an antiseizure medication prophylactically. For those with tumor-related epilepsy, monotherapy with a non-enzyme-inducing anticonvulsant is the preferred initial treatment, and levetiracetam remains the preferred first choice. Surveys of physician practice continue to demonstrate excessive use of glucocorticoids in the management of patients with both primary and metastatic central nervous system malignancy. This is particularly concerning among patients who require checkpoint inhibitors as the efficacy of these agents is blunted by concomitant glucocorticoid use, resulting in a reduction in overall survival. Finally, direct oral anticoagulants have been shown to be safe in patients with brain tumors and are now favored as first-line treatment among those who require treatment for venous thromboembolism. ESSENTIAL POINTS Medical care for patients impacted by primary and secondary central nervous system malignancy is complex and requires a committed team-based approach that routinely calls upon the expertise of physicians across multiple fields. Neurologists have an important role to play and should be familiar with the spectrum of complications impacting these patients as well as the latest recommendations for management.
Collapse
|
7
|
Khadka S, Lin YH, Ackroyd J, Chen YA, Sheng Y, Qian W, Guo S, Chen Y, Behr E, Barekatain Y, Uddin N, Arthur K, Yan V, Hsu WH, Chang Q, Poral A, Tran T, Chaurasia S, Georgiou DK, Asara JM, Barthel FP, Millward SW, DePinho RA, Muller FL. Anaplerotic nutrient stress drives synergy of angiogenesis inhibitors with therapeutics targeting tumor metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.07.539744. [PMID: 37214825 PMCID: PMC10197573 DOI: 10.1101/2023.05.07.539744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Tumor angiogenesis is a cancer hallmark, and its therapeutic inhibition has provided meaningful, albeit limited, clinical benefit. While anti-angiogenesis inhibitors deprive the tumor of oxygen and essential nutrients, cancer cells activate metabolic adaptations to diminish therapeutic response. Despite these adaptations, angiogenesis inhibition incurs extensive metabolic stress, prompting us to consider such metabolic stress as an induced vulnerability to therapies targeting cancer metabolism. Metabolomic profiling of angiogenesis-inhibited intracranial xenografts showed universal decrease in tricarboxylic acid cycle intermediates, corroborating a state of anaplerotic nutrient deficit or stress. Accordingly, we show strong synergy between angiogenesis inhibitors (Avastin, Tivozanib) and inhibitors of glycolysis or oxidative phosphorylation through exacerbation of anaplerotic nutrient stress in intracranial orthotopic xenografted gliomas. Our findings were recapitulated in GBM xenografts that do not have genetically predisposed metabolic vulnerabilities at baseline. Thus, our findings cement the central importance of the tricarboxylic acid cycle as the nexus of metabolic vulnerabilities and suggest clinical path hypothesis combining angiogenesis inhibitors with pharmacological cancer interventions targeting tumor metabolism for GBM tumors.
Collapse
Affiliation(s)
- Sunada Khadka
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yu-Hsi Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey Ackroyd
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yi-An Chen
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Yanghui Sheng
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Wubin Qian
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Sheng Guo
- Crown Bioscience Inc., Suzhou Industrial Park, 218 Xinghu Rd, Jiangsu, China
| | - Yining Chen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eliot Behr
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasaman Barekatain
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Nasir Uddin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenisha Arthur
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria Yan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anton Poral
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theresa Tran
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Surendra Chaurasia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Asara
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Floris P Barthel
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Steve W Millward
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florian L Muller
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
- Present address: Sporos Bioventures, Houston, TX, USA
| |
Collapse
|
8
|
Li M, Wang Y, Zhang L, Liu Q, Jiang F, Hou W, Wang Y, Fang H, Zhang Y. Cancer Cell Membrane-Enveloped Dexamethasone Normalizes the Tumor Microenvironment and Enhances Gynecologic Cancer Chemotherapy. ACS NANO 2023; 17:16703-16714. [PMID: 37603464 DOI: 10.1021/acsnano.3c03013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The aberrant tumor microenvironment (TME), especially immature and leaky vessels, prevents the penetration and accumulation of chemotherapeutics and results in the failure of chemotherapy to treat gynecologic cancer. Herein, dexamethasone (Dex), a glucocorticoid steroid used to moderate tumor extracellular matrix and normalize vessels, was enclosed within a biocompatible material known as poly(lactic-co-glycolic acid) (PLGA), and the obtained Dex@PLGA was further coated with a mouse cervical cancer cell membrane (CM). The formulated Dex@PLGA-CM nanoparticles showed efficient extravascular diffusion within the tumor owing to the homologous targeting abilities inherited from the source cancer cells. The Dex@PLGA-CM nanoparticles greatly reshaped the TME, enhancing the penetration of Doxil and thus markedly improving the therapeutic effect of this drug against cervical cancers. Excitingly, the Dex@PLGA-CM nanoparticles coated with mouse ovarian cancer cell membranes also promoted Doxil-mediated chemotherapy effects in metastatic ovarian cancer when administered intraperitoneally. This work presents an effective nanomedicine for the efficient modification of the TME to enhance the effects of gynecologic cancer chemotherapy.
Collapse
Affiliation(s)
- Mingzhuang Li
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Yingyao Wang
- Department of gynecology, Kunshan Maternity and Children's Health Care Hospital, Suzhou 215300, China
| | - Lin Zhang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qin Liu
- Department of gynecology, Kunshan Maternity and Children's Health Care Hospital, Suzhou 215300, China
| | - Feizhou Jiang
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Wenjie Hou
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huapan Fang
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yueming Zhang
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| |
Collapse
|
9
|
Kim S, Hoch MJ, Peng L, Somasundaram A, Chen Z, Weinberg BD. A brain tumor reporting and data system to optimize imaging surveillance and prognostication in high-grade gliomas. J Neuroimaging 2022; 32:1185-1192. [PMID: 36045502 DOI: 10.1111/jon.13044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE High-grade glioma (HGG), including glioblastoma, is the most common primary brain neoplasm and has a dismal prognosis. After initial treatment, follow-up decisions are guided by longitudinal MRI performed at routine intervals. The Brain Tumor Reporting and Data System (BT-RADS) is a proposed structured reporting system for posttreatment brain MRIs. The purpose of this study is to determine the relationship between BT-RADS scores and overall survival in HGG patients. METHODS Chart review of grade 4 glioma patients who had an MRI at a single institution from November 2018 to November 2019 was performed. BT-RADS scores, tumor characteristics, and overall survival were recorded. Likelihood of improvement, stability, or worsening on the subsequent study was calculated for each score. Survival analysis was performed using Kaplan-Meier method, log-rank test, and a time-dependent cox model. Significance level of .05 was used. RESULTS The study identified 91 HGG patients who underwent a total of 538 MRIs. Mean age of patients was 57 years old. Score with the highest likelihood for worsening on the next follow-up was 3b. The risk of death was 53% higher with each incremental increase in BT-RADS scores (hazard ratio, 1.53; 95% confidence interval [CI], 1.07-2.19; p = .019). The risk of death was 167% higher in O-6-methylguanine-DNA-methyltransferase unmethylated tumors (hazard ratio, 2.67; 95% CI, 1.34-5.33; p = .005). CONCLUSIONS BT-RADS scores can be used as a reference guide to anticipate whether patients' subsequent MRI will be improved, stable, or worsened. The scoring system can also be used to predict clinical outcomes and prognosis.
Collapse
Affiliation(s)
- Sera Kim
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | - Michael J Hoch
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lingyi Peng
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aravind Somasundaram
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| |
Collapse
|
10
|
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
|
11
|
Gahrmann R, Smits M, Vernhout RM, Taal W, Kapsas G, de Groot JC, Hanse M, Vos M, Beerepoot LV, Buter J, Flach ZH, van der Holt B, van den Bent M. The impact of different volumetric thresholds to determine progressive disease in patients with recurrent glioblastoma treated with bevacizumab. Neurooncol Adv 2022; 4:vdac032. [PMID: 35419519 PMCID: PMC9000300 DOI: 10.1093/noajnl/vdac032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background The optimal volumetric threshold for determining progressive disease (PD) in recurrent glioblastoma is yet to be determined. We investigated a range of thresholds in association with overall survival (OS). Methods First recurrent glioblastoma patients treated with bevacizumab and/or lomustine were included from the phase II BELOB and phase III EORTC26101 trials. Enhancing and nonenhancing tumor volumes were measured at baseline, first (6 weeks), and second (12 weeks) follow-up. Hazard ratios (HRs) for the appearance of new lesions and several thresholds for tumor volume increase were calculated using cox regression analysis. Results were corrected in a multivariate analysis for well-established prognostic factors. Results At first and second follow-up, 138 and 94 patients respectively, were deemed eligible for analysis of enhancing volumes, while 89 patients were included in the analysis of nonenhancing volumes at first follow-up. New lesions were associated with a significantly worse OS (3.2 versus 11.2 months, HR = 7.03, P < .001). At first follow-up a threshold of enhancing volume increase of ≥20% provided the highest HR (5.55, p = .001. At second follow-up, any increase in enhancing volume (≥0%) provided the highest HR (9.00, p < .001). When measuring nonenhancing volume at first follow-up, only 6 additional patients were scored as PD with the highest HR of ≥25% increase in volume (HR=3.25, p = .008). Conclusion Early appearing new lesions were associated with poor OS. Lowering the volumetric threshold for PD at both first and second follow-up improved survival prediction. However, the additional number of patients categorized as PD by lowering the threshold was very low. The per-RANO added change in nonenhancing volumes to the analyses was of limited value.
Collapse
Affiliation(s)
- Renske Gahrmann
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - René Michel Vernhout
- Clinical Trial Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Walter Taal
- The Brain Tumor Center at Erasmus MC Cancer Institute Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Giorgios Kapsas
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jan Cees de Groot
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Monique Hanse
- Department of Neurology, Catharina Hospital Eindhoven, The Netherlands
| | - Maaike Vos
- Department of Neurology, Medical Center Haaglanden, The Hague, The Netherlands
| | | | - Jan Buter
- Department of Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Bronno van der Holt
- Clinical Trial Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Siakallis L, Sudre CH, Mulholland P, Fersht N, Rees J, Topff L, Thust S, Jager R, Cardoso MJ, Panovska-Griffiths J, Bisdas S. Longitudinal structural and perfusion MRI enhanced by machine learning outperforms standalone modalities and radiological expertise in high-grade glioma surveillance. Neuroradiology 2021; 63:2047-2056. [PMID: 34047805 PMCID: PMC8589799 DOI: 10.1007/s00234-021-02719-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/12/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Surveillance of patients with high-grade glioma (HGG) and identification of disease progression remain a major challenge in neurooncology. This study aimed to develop a support vector machine (SVM) classifier, employing combined longitudinal structural and perfusion MRI studies, to classify between stable disease, pseudoprogression and progressive disease (3-class problem). METHODS Study participants were separated into two groups: group I (total cohort: 64 patients) with a single DSC time point and group II (19 patients) with longitudinal DSC time points (2-3). We retrospectively analysed 269 structural MRI and 92 dynamic susceptibility contrast perfusion (DSC) MRI scans. The SVM classifier was trained using all available MRI studies for each group. Classification accuracy was assessed for different feature dataset and time point combinations and compared to radiologists' classifications. RESULTS SVM classification based on combined perfusion and structural features outperformed radiologists' classification across all groups. For the identification of progressive disease, use of combined features and longitudinal DSC time points improved classification performance (lowest error rate 1.6%). Optimal performance was observed in group II (multiple time points) with SVM sensitivity/specificity/accuracy of 100/91.67/94.7% (first time point analysis) and 85.71/100/94.7% (longitudinal analysis), compared to 60/78/68% and 70/90/84.2% for the respective radiologist classifications. In group I (single time point), the SVM classifier also outperformed radiologists' classifications with sensitivity/specificity/accuracy of 86.49/75.00/81.53% (SVM) compared to 75.7/68.9/73.84% (radiologists). CONCLUSION Our results indicate that utilisation of a machine learning (SVM) classifier based on analysis of longitudinal perfusion time points and combined structural and perfusion features significantly enhances classification outcome (p value= 0.0001).
Collapse
Affiliation(s)
- Loizos Siakallis
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.
| | - Carole H Sudre
- Translational Imaging Group, Centre for Medical Image Computing, University College London , London, UK.,Department of Medical Physics, University College London, London, UK
| | - Paul Mulholland
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Naomi Fersht
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Jeremy Rees
- Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK.,Department of Neurooncology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Laurens Topff
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Steffi Thust
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Rolf Jager
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.,Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - M Jorge Cardoso
- Translational Imaging Group, Centre for Medical Image Computing, University College London , London, UK
| | - Jasmina Panovska-Griffiths
- Institute for Global Health, University College London, London, UK.,The Queen's College, University of Oxford, Oxford, UK
| | - Sotirios Bisdas
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.,Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| |
Collapse
|
13
|
Zoccarato M, Nardetto L, Basile AM, Giometto B, Zagonel V, Lombardi G. Seizures, Edema, Thrombosis, and Hemorrhages: An Update Review on the Medical Management of Gliomas. Front Oncol 2021; 11:617966. [PMID: 33828976 PMCID: PMC8019972 DOI: 10.3389/fonc.2021.617966] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/04/2021] [Indexed: 12/21/2022] Open
Abstract
Patients affected with gliomas develop a complex set of clinical manifestations that deeply impact on quality of life and overall survival. Brain tumor-related epilepsy is frequently the first manifestation of gliomas or may occur during the course of disease; the underlying mechanisms have not been fully explained and depend on both patient and tumor factors. Novel treatment options derive from the growing use of third-generation antiepileptic drugs. Vasogenic edema and elevated intracranial pressure cause a considerable burden of symptoms, especially in high-grade glioma, requiring an adequate use of corticosteroids. Patients with gliomas present with an elevated risk of tumor-associated venous thromboembolism whose prophylaxis and treatment are challenging, considering also the availability of new oral anticoagulant drugs. Moreover, intracerebral hemorrhages can complicate the course of the illness both due to tumor-specific characteristics, patient comorbidities, and side effects of antithrombotic and antitumoral therapies. This paper aims to review recent advances in these clinical issues, discussing the medical management of gliomas through an updated literature review.
Collapse
Affiliation(s)
- Marco Zoccarato
- Neurology Unit, O.S.A., Azienda Ospedale-Università, Padua, Italy
| | - Lucia Nardetto
- Neurology Unit, O.S.A., Azienda Ospedale-Università, Padua, Italy
| | | | - Bruno Giometto
- Neurology Unit, Trento Hospital, Azienda Provinciale per i Servizi Sanitari (APSS) di Trento, Trento, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| |
Collapse
|
14
|
Le Fèvre C, Lhermitte B, Ahle G, Chambrelant I, Cebula H, Antoni D, Keller A, Schott R, Thiery A, Constans JM, Noël G. Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review: Part 1 - Molecular, morphological and clinical features. Crit Rev Oncol Hematol 2020; 157:103188. [PMID: 33307200 DOI: 10.1016/j.critrevonc.2020.103188] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 01/04/2023] Open
Abstract
With new therapeutic protocols, more patients treated for glioblastoma have experienced a suspicious radiologic image of progression (pseudoprogression) during follow-up. Pseudoprogression should be differentiated from true progression because the disease management is completely different. In the case of pseudoprogression, the follow-up continues, and the patient is considered stable. In the case of true progression, a treatment adjustment is necessary. Presently, a pseudoprogression diagnosis certainly needs to be pathologically confirmed. Some important efforts in the radiological, histopathological, and genomic fields have been made to differentiate pseudoprogression from true progression, and the assessment of response criteria exists but remains limited. The aim of this paper is to highlight clinical and pathological markers to differentiate pseudoprogression from true progression through a literature review.
Collapse
Affiliation(s)
- Clara Le Fèvre
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Benoît Lhermitte
- Département of Pathology, Hautepierre University Hospital, 1, Avenue Molière, 67200, Strasbourg, France
| | - Guido Ahle
- Departement of Neurology, Hôpitaux Civils de Colmar, 39 Avenue de la Liberté, 68024, Colmar, France
| | - Isabelle Chambrelant
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Hélène Cebula
- Departement of Neurosurgery, Hautepierre University Hospital, 1, Avenue Molière, 67200, Strasbourg, France
| | - Delphine Antoni
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Audrey Keller
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Roland Schott
- Departement of Medical Oncology, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Alicia Thiery
- Department of Public Health, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Jean-Marc Constans
- Department of Radiology, Amiens-Pïcardie University Hospital, 1 rond point du Professeur Christian Cabrol, 80054 Amiens Cedex 1, France
| | - Georges Noël
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France.
| |
Collapse
|
15
|
Cho SJ, Kim HS, Suh CH, Park JE. Radiological Recurrence Patterns after Bevacizumab Treatment of Recurrent High-Grade Glioma: A Systematic Review and Meta-Analysis. Korean J Radiol 2020; 21:908-918. [PMID: 32524791 PMCID: PMC7289701 DOI: 10.3348/kjr.2019.0898] [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: 12/02/2019] [Revised: 02/10/2020] [Accepted: 03/05/2020] [Indexed: 12/28/2022] Open
Abstract
Objective To categorize the radiological patterns of recurrence after bevacizumab treatment and to derive the pooled proportions of patients with recurrent malignant glioma showing the different radiological patterns. Materials and Methods A systematic literature search in the Ovid-MEDLINE and EMBASE databases was performed to identify studies reporting radiological recurrence patterns in patients with recurrent malignant glioma after bevacizumab treatment failure until April 10, 2019. The pooled proportions according to radiological recurrence patterns (geographically local versus non-local recurrence) and predominant tumor portions (enhancing tumor versus non-enhancing tumor) after bevacizumab treatment were calculated. Subgroup and meta-regression analyses were also performed. Results The systematic review and meta-analysis included 17 articles. The pooled proportions were 38.3% (95% confidence interval [CI], 30.6–46.1%) for a geographical radiologic pattern of non-local recurrence and 34.2% (95% CI, 27.3–41.5%) for a non-enhancing tumor-predominant recurrence pattern. In the subgroup analysis, the pooled proportion of non-local recurrence in the patients treated with bevacizumab only was slightly higher than that in patients treated with the combination with cytotoxic chemotherapy (34.9% [95% CI, 22.8–49.4%] versus 22.5% [95% CI, 9.5–44.6%]). Conclusion A substantial proportion of high-grade glioma patients show non-local or non-enhancing radiologic patterns of recurrence after bevacizumab treatment, which may provide insight into surrogate endpoints for treatment failure in clinical trials of recurrent high-grade glioma.
Collapse
Affiliation(s)
- Se Jin Cho
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
16
|
Aptamer-Based In Vivo Therapeutic Targeting of Glioblastoma. Molecules 2020; 25:molecules25184267. [PMID: 32957732 PMCID: PMC7570863 DOI: 10.3390/molecules25184267] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive, infiltrative, and lethal brain tumor in humans. Despite the extensive advancement in the knowledge about tumor progression and treatment over the last few years, the prognosis of GBM is still very poor due to the difficulty of targeting drugs or anticancer molecules to GBM cells. The major challenge in improving GBM treatment implicates the development of a targeted drug delivery system, capable of crossing the blood–brain barrier (BBB) and specifically targeting GBM cells. Aptamers possess many characteristics that make them ideal novel therapeutic agents for the treatment of GBM. They are short single-stranded nucleic acids (RNA or ssDNA) able to bind to a molecular target with high affinity and specificity. Several GBM-targeting aptamers have been developed for imaging, tumor cell isolation from biopsies, and drug/anticancer molecule delivery to the tumor cells. Due to their properties (low immunogenicity, long stability, and toxicity), a large number of aptamers have been selected against GBM biomarkers and tested in GBM cell lines, while only a few of them have also been tested in in vivo models of GBM. Herein, we specifically focus on aptamers tested in GBM in vivo models that can be considered as new diagnostic and/or therapeutic tools for GBM patients’ treatment.
Collapse
|
17
|
Hou W, Li X, Pan H, Xu M, Bi S, Shen Y, Yu Y. Dynamic contrast-enhanced magnetic resonance imaging for monitoring the anti-angiogenesis efficacy in a C6 glioma rat model. Acta Radiol 2020; 61:973-982. [PMID: 31739674 DOI: 10.1177/0284185119887598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) is useful in predicting responses to angiogenic therapy of malignant tumors. PURPOSE To observe the dynamics of DCE-MRI parameters in evaluating early effects of antiangiogenic therapy in a C6 glioma rat model. MATERIAL AND METHODS The Bevacizumab or vehicle treatment was started from the 14th day after glioma model was established. The treated and control groups (n = 13 per group) underwent DCE-MRI scans on days 0, 1, 3, 5, and 7 after treatment. Tumor volume was calculated according to T2-weighted images. Hematoxylin and eosin, microvessel density (MVD), and proliferating cell nuclear antigen (PCNA) examination were performed on day 7. The MRI parameters between the two groups were compared and correlations with immunohistochemical scores were analyzed. RESULTS The average tumor volume of treated group was significantly lower than that of control group on day 7 (81.764 ± 1.043 vs. 103.634 ± 3.868 mm3, P = 0.002). Ktrans and Kep decreased in the treated group while they increased in the control group. The differences were observed on day 5 (Ktrans: 0.045 ± 0.018 vs. 0.093 ± 0.014 min-1, P < 0.001; Kep: 0.062 ± 0.018 vs. 0.134 ± 0.047 min-1, P = 0.005) and day 7 (Ktrans: 0.032 ± 0.010 vs. 0.115 ± 0.025 min-1, P < 0.001; Kep: 0.045 ± 0.016 vs. 0.144 ± 0.042 min-1, P < 0.001). The difference of Ve was observed on day 5 (0.847 ± 0.248 vs. 0.397 ± 0.151, P = 0.009) and 7 (0.920 ± 0.154 vs. 0.364 ± 0.105, P = 0.006). Ktrans and Kep showed positive correlations with MVD and Ve showed negative correlation with PCNA. CONCLUSION DCE-MRI can assess the changes of early effects of anti-angiogenic therapy in preclinical practice.
Collapse
Affiliation(s)
- Weishu Hou
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Xiaohu Li
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Hongli Pan
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Man Xu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Sixing Bi
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Yujun Shen
- Biopharmaceutical Research Institute, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, PR China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| |
Collapse
|
18
|
Park JE, Kim HS, Park SY, Jung SC, Kim JH, Heo HY. Identification of Early Response to Anti-Angiogenic Therapy in Recurrent Glioblastoma: Amide Proton Transfer–weighted and Perfusion-weighted MRI compared with Diffusion-weighted MRI. Radiology 2020; 295:397-406. [DOI: 10.1148/radiol.2020191376] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ji Eun Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Ho Sung Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Seo Young Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Seung Chai Jung
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Jeong Hoon Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Hye-Young Heo
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| |
Collapse
|
19
|
Ahir BK, Engelhard HH, Lakka SS. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol Neurobiol 2020; 57:2461-2478. [PMID: 32152825 PMCID: PMC7170819 DOI: 10.1007/s12035-020-01892-8] [Citation(s) in RCA: 251] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is the growth of new capillaries from the preexisting blood vessels. Glioblastoma (GBM) tumors are highly vascularized tumors, and glioma growth depends on the formation of new blood vessels. Angiogenesis is a complex process involving proliferation, migration, and differentiation of vascular endothelial cells (ECs) under the stimulation of specific signals. It is controlled by the balance between its promoting and inhibiting factors. Various angiogenic factors and genes have been identified that stimulate glioma angiogenesis. Therefore, attention has been directed to anti-angiogenesis therapy in which glioma proliferation is inhibited by inhibiting the formation of new tumor vessels using angiogenesis inhibitory factors and drugs. Here, in this review, we highlight and summarize the various molecular mediators that regulate GBM angiogenesis with focus on recent clinical research on the potential of exploiting angiogenic pathways as a strategy in the treatment of GBM patients.
Collapse
Affiliation(s)
- Bhavesh K Ahir
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Herbert H Engelhard
- Department of Neurosurgery, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Sajani S Lakka
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
20
|
Super selective intra-arterial cerebral infusion of modern chemotherapeutics after blood–brain barrier disruption: where are we now, and where we are going. J Neurooncol 2020; 147:261-278. [DOI: 10.1007/s11060-020-03435-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
|
21
|
Ramezani S, Vousooghi N, Joghataei MT, Chabok SY. The Role of Kinase Signaling in Resistance to Bevacizumab Therapy for Glioblastoma Multiforme. Cancer Biother Radiopharm 2020; 34:345-354. [PMID: 31411929 DOI: 10.1089/cbr.2018.2651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant primary brain tumor and is characterized by vascular hyperplasia, necrosis, and high cell proliferation. Despite current standard therapies, including surgical resection and chemoradiotherapy, GBM patients survive for only about 15 months after diagnosis. Recently, the U.S. Food and Drug Administration (FDA) has approved an antiangiogenesis medication for recurrent GBM-bevacizumab-which has improved progression-free survival in GBM patients. Although bevacizumab has resulted in significant early clinical benefit, it inescapably predisposes tumor to relapse that can be represented as an infiltrative phenotype. Fundamentally, bevacizumab antagonizes the vascular endothelial growth factor A (VEGFA), which is consistently released on both endothelial cells (ECs) and GBM cells. Actually, VEGFA inhibition on the ECs leads to the suppression of vascular progression, permeability, and the vasogenic edema. However, the consequence of the VEGFA pathway blockage on the GBM cells remains controversial. Nevertheless, a piece of evidence supports the relationship between bevacizumab application and compensatory activation of kinase signaling within GBM cells, leading to a tumor cell invasion known as the main mechanism of bevacizumab-induced tumor resistance. A complete understanding of kinase responses associated with tumor invasion in bevacizumab-resistant GBMs offers new therapeutic opportunities. Thus, this study aimed at presenting a brief overview of preclinical and clinical data of the tumor invasion and resistance induced by bevacizumab administration in GBMs, with a focus on the kinase responses during treatment. The novel therapeutic strategies to overcome this resistance by targeting protein kinases have also been summarized.
Collapse
Affiliation(s)
- Sara Ramezani
- 1Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,2Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasim Vousooghi
- 3Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,4Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran.,5Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- 6Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,7Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Yousefzadeh Chabok
- 1Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,2Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
22
|
Progression-Free but No Overall Survival Benefit for Adult Patients with Bevacizumab Therapy for the Treatment of Newly Diagnosed Glioblastoma: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:cancers11111723. [PMID: 31689995 PMCID: PMC6895972 DOI: 10.3390/cancers11111723] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is the most common high-grade primary brain tumor in adults. Standard multi-modality treatment of glioblastoma with surgery, temozolomide chemotherapy, and radiation results in transient tumor control but inevitably gives way to disease progression. The need for additional therapeutic avenues for patients with GBM led to interest in anti-angiogenic therapies, and in particular, bevacizumab. We sought to determine the efficacy of bevacizumab as a treatment for newly diagnosed GBM. We conducted a literature search using the PubMed database and Google Scholar to identify randomized controlled trials (RCTs) since 2014 investigating the safety and efficacy of bevacizumab in the treatment of adult patients (18 years and older) with newly diagnosed GBM. Only Level Ι data that reported progression-free survival (PFS) and overall survival (OS) were included for analysis. Random effects meta-analyses on studies with newly diagnosed glioblastoma were conducted in R to estimate the pooled hazard ratio (HR) for PFS and OS. Six RCTs met requirements for meta-analysis, revealing a pooled estimate of PFS HR suggesting a 33% decreased risk of disease progression (HR 0.67, 95% CI, 0.58–0.78; p < 0.001) with bevacizumab therapy, but no effect on OS (HR = 1, 95% CI, 0.85–1.18; p = 0.97). A pooled estimate of the mean difference in OS months of −0.13 predicts little difference in time of survival between treatment groups (95% CI, −1.87–1.61). The pooled estimate for the mean difference in PFS months was 2.70 (95% CI, 1.89–3.50; p < 0.001). Meta-analysis shows that bevacizumab therapy is associated with a longer PFS in adult patients with newly diagnosed glioblastoma, but had an inconsistent effect on OS in this patient population.
Collapse
|
23
|
Ramezani S, Vousooghi N, Ramezani Kapourchali F, Yousefzadeh-Chabok S, Reihanian Z, Alizadeh AM, Khodayari S, Khodayari H. Rolipram optimizes therapeutic effect of bevacizumab by enhancing proapoptotic, antiproliferative signals in a glioblastoma heterotopic model. Life Sci 2019; 239:116880. [PMID: 31678282 DOI: 10.1016/j.lfs.2019.116880] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/18/2019] [Accepted: 09/15/2019] [Indexed: 01/24/2023]
Abstract
The unstable response to bevacizumab is a big dilemma in the antiangiogenic therapy of high-grade glioma that appears to be linked to an increase in the post-treatment intratumor levels of hypoxia-inducible factor 1 α (HIF1α) and active AKT. Particularly, a selective phosphodiesterase IV (PDE4) inhibitor, rolipram is capable of inhibiting HIF1α and AKT in cancer cells. Here, the effect of bevacizumab alone and in presence of rolipram on therapeutic efficacy, intratumor hypoxia levels, angiogenesis, apoptosis and proliferation mechanisms were evaluated. BALB/c mice bearing C6 glioma were received bevacizumab and rolipram either alone or combined for 30 days (n = 11/group). At the last day of treatments, apoptosis, proliferation and microvessel density, in xenografts (3/group) were detected by TUNEL staining, Ki67 and CD31 markers, respectively. Relative expression of target proteins was measured using western blotting. Bevacizumab initially hindered the tumor progression but its antitumor effect was weakened later despite the vascular regression and apoptosis induction. Unpredictably, bevacizumab-treated tumors exhibited the highest cell proliferation coupled with PDE4A, HIF1α and AKT upregulation and p53 downregulation and reversed by co-treatment with rolipram. Unlike a similar antivascular pattern to bevacizumab, rolipram consistently led to a more tumor growth suppression and proapoptotic effect versus bevacizumab. Co-treatment maximally hampered the tumor progression and elongated survival along with the major vascular regression, hypoxia, apoptosis induction, p53 and caspase activities. In conclusion, superior and persistent therapeutic efficacy of co-treatment provides a new insight into antiangiogenic therapy of malignant gliomas, suggesting to be a potential substitute in selected patients.
Collapse
Affiliation(s)
- Sara Ramezani
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nasim Vousooghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran; Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Shahrokh Yousefzadeh-Chabok
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Zoheir Reihanian
- Neurosurgery Department, Guilan University of Medical Sciences, Guilan, Iran
| | | | - Saeed Khodayari
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Khodayari
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Ellingson BM, Aftab DT, Schwab GM, Hessel C, Harris RJ, Woodworth DC, Leu K, Chakhoyan A, Raymond C, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Wen PY, Cloughesy TF. Volumetric response quantified using T1 subtraction predicts long-term survival benefit from cabozantinib monotherapy in recurrent glioblastoma. Neuro Oncol 2019; 20:1411-1418. [PMID: 29660005 DOI: 10.1093/neuonc/noy054] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background To overcome challenges with traditional response assessment in anti-angiogenic agents, the current study uses T1 subtraction maps to quantify volumetric radiographic response in monotherapy with cabozantinib, an orally bioavailable tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2), hepatocyte growth factor receptor (MET), and AXL, in an open-label, phase II trial in patients with recurrent glioblastoma (GBM) (NCT00704288). Methods A total of 108 patients with adequate imaging data and confirmed recurrent GBM were included in this retrospective study from a phase II multicenter trial of cabozantinib monotherapy (XL184-201) at either 100 mg (N = 87) or 140 mg (N = 21) per day. Contrast enhanced T1-weighted digital subtraction maps were used to define volume of contrast-enhancing tumor at baseline and subsequent follow-up time points. Volumetric radiographic response (>65% reduction in contrast-enhancing tumor volume from pretreatment baseline tumor volume sustained for more than 4 wk) was tested as an independent predictor of overall survival (OS). Results Volumetric response rate for all therapeutic doses was 38.9% (41.4% and 28.6% for 100 mg and 140 mg doses, respectively). A log-linear association between baseline tumor volume and OS (P = 0.0006) and a linear correlation between initial change in tumor volume and OS (P = 0.0256) were observed. A significant difference in OS was observed between responders (median OS = 20.6 mo) and nonresponders (median OS = 8.0 mo) (hazard ratio [HR] = 0.3050, P < 0.0001). Multivariable analyses showed that continuous measures of baseline tumor volume (HR = 1.0233, P < 0.0001) and volumetric response (HR = 0.2240, P < 0.0001) were independent predictors of OS. Conclusions T1 subtraction maps provide value in determining response in recurrent GBM treated with cabozantinib and correlated with survival benefit.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | | | | | - Robert J Harris
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Davis C Woodworth
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jan Drappatz
- Department of Neurology and Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, California
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Schiff
- Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia
| | - Marc Chamberlain
- Department of Neurology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | | | - Jerry Ping
- Exelixis, South San Francisco, California
| | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
25
|
Anthony C, Mladkova-Suchy N, Adamson DC. The evolving role of antiangiogenic therapies in glioblastoma multiforme: current clinical significance and future potential. Expert Opin Investig Drugs 2019; 28:787-797. [PMID: 31356114 DOI: 10.1080/13543784.2019.1650019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults, but its prognosis remains poor despite significant advances in our understanding of its molecular biology and investigation of numerous treatment modalities. Despite conventional treatment consisting of surgical resection, radiotherapy, and temozolomide marginally prolonging survival, most GBM patients die within 2 years of initial diagnosis. Bevacizumab (Bev) is the best-studied antiangiogenic agent for GBM and currently the only FDA-approved second-line treatment. Areas covered: Areas covered in this review include the molecular pathways of angiogenesis in glioblastoma, specifically the overexpression of vascular endothelial growth factor (VEGF) and robust formation of tumor neovasculature. In addition, this review covers pharmacological targeting of this process as a longstanding attractive clinical strategy, specifically by Bev. Expert opinion: This review attempts to discuss the history of early studies of antiangiogenic treatment for GBM that eventually failed in subsequent studies and the evolving modern role of Bev in the course of treatment for a variety of indications, including symptom control, reduced glucocorticoid use, and improved quality of life.
Collapse
Affiliation(s)
- Casey Anthony
- Department of Neurosurgery, Emory University , Atlanta , GA , USA
| | - Nikol Mladkova-Suchy
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London , UK
| | - David Cory Adamson
- Department of Neurosurgery, Emory University , Atlanta , GA , USA.,Neurosurgery section, Atlanta VA Medical Center , Decatur , GA , USA
| |
Collapse
|
26
|
Miyoshi F, Shinohara Y, Kambe A, Kuya K, Murakami A, Kurosaki M, Ogawa T. Utility of intravoxel incoherent motion magnetic resonance imaging and arterial spin labeling for recurrent glioma after bevacizumab treatment. Acta Radiol 2018; 59:1372-1379. [PMID: 29471670 DOI: 10.1177/0284185118759707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Detecting recurrence of glioma on magnetic resonance imaging (MRI) is getting more and more important, especially after administration of new anti-tumor agent. However, it is still hard to identify. Purpose To examine the utility of intravoxel incoherent motion (IVIM) MRI and arterial spin labeling-cerebral blood flow (ASL-CBF) for recurrent glioma after initiation of bevacizumab (BEV) treatment. Material and Methods Thirteen patients (7 men, 6 women; age range = 41-82 years) with glioma (high grade, n = 11; low grade, n = 2) were enrolled in the study. IVIM parameters including apparent diffusion coefficient (ADC), true diffusion coefficient (D), and perfusion fraction (f) were obtained with 14 different b-values. We identified tumor progression during BEV therapy by MRI monitoring consisting of diffusion-weighted imaging (DWI), fluid-attenuated inversion recovery (FLAIR) imaging, and contrast-enhanced T1-weighted (CE-T1W) imaging by measuring tumor area. We also measured each parameter of IVIM and ASL-CBF, and calculated relative ADC (rADC), relative D (rD), relative f (rf), and relative CBF (rCBF) by obtaining the ratio between each area and the contralateral cerebral white matter. We calculated the rate of change (Δ) by subtracting values from those from the preceding MRI study, and obtained Spearman's rank correlation coefficient (rs). Results Tumor progression was identified in nine patients (high grade, n = 7; low grade, n = 2). Negative correlations were identified between ΔrD and ΔDWI area (rs = -0.583), and between ΔrD and ΔCE-T1W imaging area (rs = -0.605). Conclusion Tumor progression after BEV treatment can be identified by decreasing rD.
Collapse
Affiliation(s)
- Fuminori Miyoshi
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yuki Shinohara
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Kambe
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Keita Kuya
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Murakami
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Masamichi Kurosaki
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshihide Ogawa
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
27
|
Bahrami N, Piccioni D, Karunamuni R, Chang YH, White N, Delfanti R, Seibert TM, Hattangadi-Gluth JA, Dale A, Farid N, McDonald CR. Edge Contrast of the FLAIR Hyperintense Region Predicts Survival in Patients with High-Grade Gliomas following Treatment with Bevacizumab. AJNR Am J Neuroradiol 2018; 39:1017-1024. [PMID: 29622553 PMCID: PMC6002890 DOI: 10.3174/ajnr.a5620] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/07/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Treatment with bevacizumab is standard of care for recurrent high-grade gliomas; however, monitoring response to treatment following bevacizumab remains a challenge. The purpose of this study was to determine whether quantifying the sharpness of the fluid-attenuated inversion recovery hyperintense border using a measure derived from texture analysis-edge contrast-improves the evaluation of response to bevacizumab in patients with high-grade gliomas. MATERIALS AND METHODS MRIs were evaluated in 33 patients with high-grade gliomas before and after the initiation of bevacizumab. Volumes of interest within the FLAIR hyperintense region were segmented. Edge contrast magnitude for each VOI was extracted using gradients of the 3D FLAIR images. Cox proportional hazards models were generated to determine the relationship between edge contrast and progression-free survival/overall survival using age and the extent of surgical resection as covariates. RESULTS After bevacizumab, lower edge contrast of the FLAIR hyperintense region was associated with poorer progression-free survival (P = .009) and overall survival (P = .022) among patients with high-grade gliomas. Kaplan-Meier curves revealed that edge contrast cutoff significantly stratified patients for both progression-free survival (log-rank χ2 = 8.3, P = .003) and overall survival (log-rank χ2 = 5.5, P = .019). CONCLUSIONS Texture analysis using edge contrast of the FLAIR hyperintense region may be an important predictive indicator in patients with high-grade gliomas following treatment with bevacizumab. Specifically, low FLAIR edge contrast may partially reflect areas of early tumor infiltration. This study adds to a growing body of literature proposing that quantifying features may be important for determining outcomes in patients with high-grade gliomas.
Collapse
Affiliation(s)
- N Bahrami
- From the Center for Multimodal Imaging and Genetics (N.B., N.W., C.R.M.)
- Department of Psychiatry (N.B., Y.-H.C., C.R.M.)
- Department of Radiology (N.B., N.W., R.D., A.D., N.F., C.R.M.)
- Multimodal Imaging Laboratory (N.B., N.W., A.D., C.R.M.)
| | - D Piccioni
- Department of Neurosciences (D.P., A.D., N.F.)
| | - R Karunamuni
- Department of Radiation Medicine (R.K., T.M.S., J.A.H.-G.), University of California, San Diego, La Jolla, California
| | - Y-H Chang
- Department of Psychiatry (N.B., Y.-H.C., C.R.M.)
| | - N White
- From the Center for Multimodal Imaging and Genetics (N.B., N.W., C.R.M.)
- Department of Radiology (N.B., N.W., R.D., A.D., N.F., C.R.M.)
- Multimodal Imaging Laboratory (N.B., N.W., A.D., C.R.M.)
| | - R Delfanti
- Department of Radiology (N.B., N.W., R.D., A.D., N.F., C.R.M.)
| | - T M Seibert
- Department of Radiation Medicine (R.K., T.M.S., J.A.H.-G.), University of California, San Diego, La Jolla, California
| | - J A Hattangadi-Gluth
- Department of Radiation Medicine (R.K., T.M.S., J.A.H.-G.), University of California, San Diego, La Jolla, California
| | - A Dale
- Multimodal Imaging Laboratory (N.B., N.W., A.D., C.R.M.)
- Department of Neurosciences (D.P., A.D., N.F.)
| | - N Farid
- Department of Radiology (N.B., N.W., R.D., A.D., N.F., C.R.M.)
- Department of Neurosciences (D.P., A.D., N.F.)
| | - C R McDonald
- From the Center for Multimodal Imaging and Genetics (N.B., N.W., C.R.M.)
- Department of Psychiatry (N.B., Y.-H.C., C.R.M.)
- Department of Radiology (N.B., N.W., R.D., A.D., N.F., C.R.M.)
- Multimodal Imaging Laboratory (N.B., N.W., A.D., C.R.M.)
| |
Collapse
|
28
|
Mantica M, Pritchard A, Lieberman F, Drappatz J. Retrospective study of nivolumab for patients with recurrent high grade gliomas. J Neurooncol 2018; 139:625-631. [PMID: 29779086 DOI: 10.1007/s11060-018-2907-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/16/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Patients with recurrent high-grade gliomas (HGG) have limited treatment options. HGG utilize the PD-1 pathway to evade immune responses. Checkpoint inhibitors have demonstrated safety and clinical activity in patients with recurrent glioblastoma. We explored the efficacy of nivolumab in recurrent HGG with a primary objective of progression free survival (PFS) and overall survival (OS). METHODS We retrospectively analyzed HGG patients treated with nivolumab in our institution. We included patients with advanced HGG who received nivolumab at their oncologist's decision. Patients received nivolumab 3 mg/kg every 2 weeks until confirmed progression, intolerable toxicity, death, or physician decision. Radiographic assessments were performed every 8 weeks. RESULTS Between April 2015 and October 2017, 50 HGG patients received nivolumab. 43 patients received nivolumab with bevacizumab. 44 patients were bevacizumab refractory and 7 patients received nivolumab monotherapy. All had received prior radiation and chemotherapy. 39 adverse events (AEs) were noted [most commonly fatigue (16%) and constipation (10%)]. 4 (8%) patients experienced grade 3-4 AEs. 36 (72%) patients experienced stable disease (SD) at the 2-month assessment. Median duration of SD was 4.3 months (5.1 months in the bevacizumab naïve, 3.8 months in the bevacizumab refractory). Median PFS was 4.3 months (95% CI 3.5-5.3); median OS was 6.5 months (95% CI 6.0-8.8). CONCLUSION Treatment with nivolumab therapy was associated with a manageable safety profile. In a subset of patients, there was disease stabilization in heavily pre-treated recurrent HGG.
Collapse
Affiliation(s)
- Megan Mantica
- Division of Hematology-Oncology, Departments of Neurology and Medicine, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | - Ashley Pritchard
- Division of Hematology-Oncology, Departments of Neurology and Medicine, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Frank Lieberman
- Division of Hematology-Oncology, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Jan Drappatz
- Division of Hematology-Oncology, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA
| |
Collapse
|
29
|
Ellingson BM, Wen PY, Cloughesy TF. Evidence and context of use for contrast enhancement as a surrogate of disease burden and treatment response in malignant glioma. Neuro Oncol 2018; 20:457-471. [PMID: 29040703 PMCID: PMC5909663 DOI: 10.1093/neuonc/nox193] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The use of contrast enhancement within the brain on CT or MRI has been the gold standard for diagnosis and therapeutic response assessment in malignant gliomas for decades. The use of contrast enhancing tumor size, however, remains controversial as a tool for accurately diagnosing and assessing treatment efficacy in malignant gliomas, particularly in the current, quickly evolving therapeutic landscape. The current article consolidates overwhelming evidence from hundreds of studies in the field of neuro-oncology, providing the necessary evidence base and specific contexts of use for consideration of contrast enhancing tumor size as an appropriate surrogate biomarker for disease burden and as a tool for measuring treatment response in malignant glioma, including glioblastoma.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Brain Research Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Physics in Medicine and Biology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA, University of California Los Angeles, Los Angeles, California
| | - Patrick Y Wen
- Department of Neurooncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
30
|
Kaloshi G, Brace G, Rroji A, Bushati T, Roci E, Hoxha M, Fejzo G, Petrela M. Bevacizumab Alone at 5 mg/kg in an Every-3-Week Schedule for Patients with Recurrent Glioblastomas: A Single Center Experience. TUMORI JOURNAL 2018; 99:601-3. [DOI: 10.1177/030089161309900507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The use of bevacizumab is increasingly reported in neuro-oncology. The most common schedule is 10 mg/kg every 2 weeks. We retrospectively investigated the efficacy of a 3-week schedule of 5 mg/kg bevacizumab in patients with recurrent glioblastomas. Fourteen patients (median age, 46 years) were included in the study. The median number of bevacizumab cycles was 4 (range, 2–8). Five patients (36%) had a partial response, 7 (50%) had stable disease, and 2 (14%) had progressive disease. No grade III-IV toxicities were observed. The median progression-free and overall survival were 3.6 months and 6.4 months, respectively. Every-3-week low-dose single-agent bevacizumab showed substantial activity and a safe profile in patients with recurrent glioblastoma.
Collapse
Affiliation(s)
- Gentian Kaloshi
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Gramoz Brace
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Arben Rroji
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Teona Bushati
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Ermir Roci
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Mehmet Hoxha
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Geldon Fejzo
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| | - Mentor Petrela
- Department of Neurosurgery, University Hospital Center “Mother Theresa”, Faculty of Medicine, University of Tirana, Tirana, Albania
| |
Collapse
|
31
|
|
32
|
Abstract
BACKGROUND Corticosteroids are commonly used in the management of primary central nervous system (CNS) tumors and CNS metastases to treat cancer- and treatment-related cerebral edema and improve neurologic function. However, they are also associated with significant morbidity and mortality, given their wide range of adverse effects. PURPOSE OF REVIEW To review the mechanism of action, pharmacology, and toxicity profile of corticosteroids and to critically appraise the evidence that supports their use in neuro-oncologic practice based on the latest scientific and clinical data. RECENT FINDINGS Recent data suggest that corticosteroids may negatively impact survival in glioma patients. In addition, corticosteroids should be incorporated as a standard criterion to assess a patient's clinical and radiographic response to treatment. Corticosteroids should be used judiciously in neuro-oncologic patients, given the potential deleterious effects on clinical outcome and patient survival. Anti-angiogenic agents, which lack these adverse effects, may be a reasonable alternative to corticosteroids.
Collapse
Affiliation(s)
- K Ina Ly
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
33
|
Solinas C, Porcu M, Hlavata Z, De Silva P, Puzzoni M, Willard-Gallo K, Scartozzi M, Saba L. Critical features and challenges associated with imaging in patients undergoing cancer immunotherapy. Crit Rev Oncol Hematol 2017; 120:13-21. [PMID: 29198327 DOI: 10.1016/j.critrevonc.2017.09.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/13/2017] [Accepted: 09/30/2017] [Indexed: 01/03/2023] Open
Abstract
Manipulating an individual's immune system through immune checkpoint blockade is revolutionizing the paradigms of cancer treatment. Peculiar patterns and kinetics of response have been observed with these new drugs, rendering the assessment of tumor burden particularly challenging in cancer immunotherapy. The mechanisms of action for immune checkpoint blockade, based upon engagement of the adaptive immune system, can generate unusual response patterns, including pseudoprogression, hyperprogression, atypical and delayed responses. In patients treated with immune checkpoint blockade and radiotherapy, a reduction in tumor burden at metastatic sites distant from the irradiation field (abscopal effect) has been observed, with synergistic systemic immune effects provoked by this combination. New toxicities have also been observed, due to excessive immune activity in several organs, including lung, colon, liver and endocrine glands. Efforts to standardize assessment of cancer immunotherapy responses include novel consensus guidelines derived by modifying World Health Organization (WHO) and Response Evaluation Criteria In Solid Tumors (RECIST) criteria. The aim of this review is to evaluate imaging techniques currently used routinely in the clinic and those being used as investigational tools in immunotherapy clinical trials.
Collapse
Affiliation(s)
- Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet and Université Libre de Bruxelles, Boulevard de Waterloo, n. 127, Brussels, Belgium
| | - Michele Porcu
- Department of Radiology, Azienda Ospedaliero Universitaria of Cagliari, SS 554 Monserrato, CA, Italy.
| | - Zuzana Hlavata
- Department of Medical Oncology, CHR Mons - Hainaut, Avenue Baudouin de Constantinople, n. 5, Mons, Hainaut, Belgium
| | - Pushpamali De Silva
- Molecular Immunology Unit, Institut Jules Bordet and Université Libre de Bruxelles, Boulevard de Waterloo, n. 127, Brussels, Belgium
| | - Marco Puzzoni
- Department of Medical Oncology, Azienda Ospedaliero Universitaria of Cagliari, SS 554 Monserrato, CA, Italy
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet and Université Libre de Bruxelles, Boulevard de Waterloo, n. 127, Brussels, Belgium
| | - Mario Scartozzi
- Department of Medical Oncology, Azienda Ospedaliero Universitaria of Cagliari, SS 554 Monserrato, CA, Italy
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria of Cagliari, SS 554 Monserrato, CA, Italy
| |
Collapse
|
34
|
Ellingson BM, Chung C, Pope WB, Boxerman JL, Kaufmann TJ. Pseudoprogression, radionecrosis, inflammation or true tumor progression? challenges associated with glioblastoma response assessment in an evolving therapeutic landscape. J Neurooncol 2017; 134:495-504. [PMID: 28382534 PMCID: PMC7893814 DOI: 10.1007/s11060-017-2375-2] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/13/2017] [Indexed: 01/24/2023]
Abstract
The wide variety of treatment options that exist for glioblastoma, including surgery, ionizing radiation, anti-neoplastic chemotherapies, anti-angiogenic therapies, and active or passive immunotherapies, all may alter aspects of vascular permeability within the tumor and/or normal parenchyma. These alterations manifest as changes in the degree of contrast enhancement or T2-weighted signal hyperintensity on standard anatomic MRI scans, posing a potential challenge for accurate radiographic response assessment for identifying anti-tumor effects. The current review highlights the challenges that remain in differentiating true disease progression from changes due to radiation therapy, including pseudoprogression and radionecrosis, as well as immune or inflammatory changes that may occur as either an undesired result of cytotoxic therapy or as a desired consequence of immunotherapies.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), David Geffen School of Medicine, Center for Computer Vision and Imaging, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Caroline Chung
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | | |
Collapse
|
35
|
Stadlbauer A, Zimmermann M, Oberndorfer S, Doerfler A, Buchfelder M, Heinz G, Roessler K. Vascular Hysteresis Loops and Vascular Architecture Mapping in Patients with Glioblastoma treated with Antiangiogenic Therapy. Sci Rep 2017; 7:8508. [PMID: 28819189 PMCID: PMC5561153 DOI: 10.1038/s41598-017-09048-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/19/2017] [Indexed: 01/06/2023] Open
Abstract
In this study, we investigated the variability of vascular hysteresis loop (VHL) shapes and the spatial heterogeneity of neovascularization and microvascular alterations using vascular architecture mapping (VAM) in patients with recurrent glioblastoma during bevacizumab mono-therapy. VAM data were acquired in 13 patients suffering from recurrent glioblastoma prior to and 3 months after bevacizumab treatment onset using a dual contrast agent injections approach as part of routine MRI. Two patients were additionally examined after the first cycle of bevacizumab to check for early treatment response. VHLs were evaluated as biomarker maps of neovascularization activity: microvessel type indicator (MTI) and curvature (Curv) of the VHL-long-axis. Early response to bevacizumab was dominated by reduction of smaller microvasculature (around 10 µm). In the 3-month follow-up, responding tumors additionally showed a reduction in larger microvasculature (>20 µm). VAM biomarker images revealed spatially heterogeneous microvascular alterations during bevacizumab treatment. Responding, non-responding, progressive, and remote-progressive tumor areas were observed. MTI may be useful to predict responding and non-responding tumor regions, and Curv to assess severity of vasogenic edema. Analysis of VHLs in combination with VAM biomarkers may lead to a new perspective on investigating the spatial heterogeneity of neovascularization and microvascular alterations in glioblastoma during antiangiogenic therapy.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany. .,Institute of Medical Radiology, University Clinic of St. Pölten, St. Pölten, Austria.
| | - Max Zimmermann
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Arnd Doerfler
- Department of Neuroradiology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gertraud Heinz
- Institute of Medical Radiology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Karl Roessler
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
36
|
Kim JH, Jung TY, Hwang EC, Jung SH, Jung S, Kim IY, Jang WY, Moon KS, Lee KH, Kim SK. Disease progression patterns of bevacizumab responders with recurrent malignant gliomas. Oncol Lett 2017; 14:3529-3535. [PMID: 28927109 PMCID: PMC5588053 DOI: 10.3892/ol.2017.6630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 03/03/2017] [Indexed: 01/21/2023] Open
Abstract
Tumor progression in patients with recurrent malignant glioma who respond to bevacizumab (BEV) is difficult to assess. The current study reviewed the clinical and radiological results of patients following a BEV-based chemotherapy regimen, and evaluated disease progression patterns in patients who responded to BEV therapy. From August 2011 to November 2015, 24 patients (18 glioblastoma cases and 6 anaplastic astrocytoma cases) were treated with BEV-based chemotherapy. In total, 6 patients were treated with BEV alone and 18 patients were treated with BEV combined with irinotecan. The male-female ratio was 10:14, and the median age was 47.5 years (range, 29-69). Patient performance status (PS) was classified using the Eastern Cooperative Oncology Group PS scores as follows: PS 1 (n=3), PS 2 (n=9), PS 3 (n=9) and PS 4 (n=3). Treatment-associated complications were also analyzed according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Treatment responses were estimated using the Response Assessment in Neuro-Oncology Criteria. Progression-free survival (PFS) following treatment, patterns of disease progression and overall survival following treatment failure were also analyzed. The median PFS was 2.8 months (range, 0.6-10.1). In total, 2 patients did not continue treatment due to rectal bleeding and severe hematologic toxicity. Amongst the BEV responders (n=16, 72.7%), there was clinical deterioration without significant radiological progression in 2 patients (n=2, 12.5%). Radiological progression of non-enhancing lesions without enhancement flare-ups was observed in 6 patients (42.9%). A total of 3 of those lesions were diffuse and 3 were focal. Increased lesion enhancement was observed in 8 patients (57.1%). Of the non-responders (n=6, 27.3%), diffuse enlargement of non-enhancing lesions was detected in 2 patients and an increase in lesion enhancement occurred in 4 patients. BEV complete responders (n=3) radiologically progressed with enlarged T2/fluid attenuation inversion recovery lesions without enhancement, followed by enhancement flare-ups. Following BEV treatment failure, 8 patients received a number of adjuvant treatments and the overall survival was 4.5 months (range, 0.4-34.0). Clinical symptoms and radiological alterations of non-enhancing lesions must be evaluated in order to assess tumor progression in the BEV responders, particularly in patients who have achieved complete remission.
Collapse
Affiliation(s)
- Ju-Hwi Kim
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Eu Chang Hwang
- Department of Urology, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Sung-Hoon Jung
- Department of Hemato-oncology, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Shin Jung
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - In-Young Kim
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Woo-Youl Jang
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| | - Seul-Kee Kim
- Department of Radiology, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Chonnam 519-763, Republic of Korea
| |
Collapse
|
37
|
Li Y, Ali S, Clarke J, Cha S. Bevacizumab in Recurrent Glioma: Patterns of Treatment Failure and Implications. Brain Tumor Res Treat 2017; 5:1-9. [PMID: 28516072 PMCID: PMC5433944 DOI: 10.14791/btrt.2017.5.1.1] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/03/2016] [Accepted: 12/14/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma, the most common primary malignant brain tumor in adults, is highly aggressive and associated with a poor prognosis. Bevacizumab, a monoclonal antibody against the vascular endothelial growth factor receptor, has increasingly been used in the treatment of recurrent glioblastoma. It has achieved excellent rates of radiographic response, but most patients will progress after only a few months. Upon recurrence, tumors may not enhance, secondary to vascular normalization. We describe four patterns of radiographic progression commonly associated with Bevacizumab failure: 1) Distant enhancing tumor, 2) Local tumor progression without enhancement, 3) Diffuse gliomatosis-like infiltration, and 4) Local or multifocal progression, with enhancement. Some have noted an increased incidence of distant or diffuse disease upon recurrence, suggestive of a transition to a more aggressive phenotype, but a review of the literature suggests there is no conclusive evidence that Bevacizumab treatment is associated with an increased rate of distant or diffuse recurrence.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Saad Ali
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | - Jennifer Clarke
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
38
|
Ellingson BM, Wen PY, Cloughesy TF. Modified Criteria for Radiographic Response Assessment in Glioblastoma Clinical Trials. Neurotherapeutics 2017; 14:307-320. [PMID: 28108885 PMCID: PMC5398984 DOI: 10.1007/s13311-016-0507-6] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Radiographic endpoints including response and progression are important for the evaluation of new glioblastoma therapies. The current RANO criteria was developed to overcome many of the challenges identified with previous guidelines for response assessment, however, significant challenges and limitations remain. The current recommendations build on the strengths of the current RANO criteria, while addressing many of these limitations. Modifications to the current RANO criteria include suggestions for volumetric response evaluation, use contrast enhanced T1 subtraction maps to increase lesion conspicuity, removal of qualitative non-enhancing tumor assessment requirements, use of the post-radiation time point as the baseline for newly diagnosed glioblastoma response assessment, and "treatment-agnostic" response assessment rubrics for identifying pseudoprogression, pseudoresponse, and a confirmed durable response in newly diagnosed and recurrent glioblastoma trials.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- UCLA Neuro-Oncology Program, University of California Los Angeles, Los Angeles, CA, USA.
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
39
|
Apparent diffusion coefficient changes predict survival after intra-arterial bevacizumab treatment in recurrent glioblastoma. Neuroradiology 2017; 59:499-505. [PMID: 28343250 DOI: 10.1007/s00234-017-1820-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/14/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Superselective intra-arterial cerebral infusion (SIACI) of bevacizumab (BV) has emerged as a novel therapy in the treatment of recurrent glioblastoma (GB). This study assessed the use of apparent diffusion coefficient (ADC) in predicting length of survival after SIACI BV and overall survival in patients with recurrent GB. METHODS Sixty-five patients from a cohort enrolled in a phase I/II trial of SIACI BV for treatment of recurrent GB were retrospectively included in this analysis. MR imaging with a diffusion-weighted (DWI) sequence was performed before and after treatment. ROIs were manually delineated on ADC maps corresponding to the enhancing and non-enhancing portions of the tumor. Cox and logistic regression analyses were performed to determine which ADC values best predicted survival. RESULTS The change in minimum ADC in the enhancing portion of the tumor after SIACI BV therapy was associated with an increased risk of death (hazard ratio = 2.0, 95% confidence interval(CI) [1.04-3.79], p = 0.038), adjusting for age, tumor size, BV dose, and prior IV BV treatments. Similarly, the change in ADC after SIACI BV therapy was associated with greater likelihood of surviving less than 1 year after therapy (odds ratio = 7.0, 95% CI [1.08-45.7], p = 0.04). Having previously received IV BV was associated with increased risk of death (OR 18, 95% CI [1.8-180.0], p = 0.014). CONCLUSION In patients with recurrent GB treated with SIACI BV, the change in ADC value after treatment is predictive of overall survival.
Collapse
|
40
|
Nelson SJ, Li Y, Lupo JM, Olson M, Crane JC, Molinaro A, Roy R, Clarke J, Butowski N, Prados M, Cha S, Chang SM. Serial analysis of 3D H-1 MRSI for patients with newly diagnosed GBM treated with combination therapy that includes bevacizumab. J Neurooncol 2016; 130:171-179. [PMID: 27535746 PMCID: PMC5069332 DOI: 10.1007/s11060-016-2229-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/31/2016] [Indexed: 10/26/2022]
Abstract
Interpretation of changes in the T1- and T2-weighted MR images from patients with newly diagnosed glioblastoma (GBM) treated with standard of care in conjunction with anti-angiogenic agents is complicated by pseudoprogression and pseudoresponse. The hypothesis being tested in this study was that 3D H-1 magnetic resonance spectroscopic imaging (MRSI) provides estimates of levels of choline, creatine, N-acetylaspartate (NAA), lactate and lipid that change in response to treatment and that metrics describing these characteristics are associated with survival. Thirty-one patients with newly diagnosed GBM and being treated with radiation therapy (RT), temozolomide, erlotinib and bevacizumab were recruited to receive serial MR scans that included 3-D lactate edited MRSI at baseline, mid-RT, post-RT and at specific follow-up time points. The data were processed to provide estimates of metrics representing changes in metabolite levels relative to normal appearing brain. Cox proportional hazards analysis was applied to examine the relationship of these parameters with progression free survival (PFS) and overall survival (OS). There were significant reductions in parameters that describe relative levels of choline to NAA and creatine, indicating that the treatment caused a decrease in tumor cellularity. Changes in the levels of lactate and lipid relative to the NAA from contralateral brain were consistent with vascular normalization. Metabolic parameters from the first serial follow-up scan were associated with PFS and OS, when accounting for age and extent of resection. Integrating metabolic parameters into the assessment of patients with newly diagnosed GBM receiving therapies that include anti-angiogenic agents may be helpful for tracking changes in tumor burden, resolving ambiguities in anatomic images caused by non-specific treatment effects and for predicting outcome.
Collapse
Affiliation(s)
- Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Marram Olson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jason C Crane
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Annette Molinaro
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
41
|
Abstract
Neurologic complications of cancer are common and are frequently life-threatening events. Certain neurologic emergencies occur more frequently in the cancer population, specifically elevated intracranial pressure, epidural cord compression, status epilepticus, ischemic and hemorrhagic stroke, central nervous system infection, and treatment-associated neurologic dysfunction. These emergencies require early diagnosis and prompt treatment to ensure the best possible outcome and are best managed in the intensive care unit. This article reviews the presentation, pathophysiology, and management of the most common causes of acute neurologic decompensation in the patient with cancer.
Collapse
Affiliation(s)
- Andrew L Lin
- 1 Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edward K Avila
- 1 Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
42
|
Abstract
Magnetic resonance imaging (MRI) is the most useful imaging tool in the evaluation of patients with brain tumors. Most information is supplied by standard anatomic images that were developed in the 1980s and 1990s. More recently, functional imaging including diffusion and perfusion MRI has been investigated as a way to generate predictive and prognostic biomarkers for high-grade glioma evaluation, but additional research is needed to establish the added benefits of these indices to standard MRI. Response critieria for high-grade gliomas have recently been updated by the Response Assessment in Neuro-Oncology (RANO) working group. The new criteria account for nonenhancing tumor in addition to the contrast-enhancing abnormalities on which older criteria relied. This issue has recently come to the fore with the introduction of the antiangiogenic agent bevacizumab into standard treatment for recurrent glioblastoma. Because of its potent antipermeability effect, contrast enhancement is markedly reduced in patients who receive bevacizumab. The RANO criteria also address the phenomenon of pseudoprogression, in which there may be transient MRI worsening of a glioblastoma following concurrent radiotherapy and temozolomide.
Collapse
Affiliation(s)
- Andrew D Norden
- From Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; University of California, San Francisco, Department of Neurological Surgery, San Francisco, CA
| | - Whitney B Pope
- From Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; University of California, San Francisco, Department of Neurological Surgery, San Francisco, CA
| | - Susan M Chang
- From Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; University of California, San Francisco, Department of Neurological Surgery, San Francisco, CA
| |
Collapse
|
43
|
Hilario A, Sepulveda JM, Hernandez-Lain A, Salvador E, Koren L, Manneh R, Ruano Y, Perez-Nuñez A, Lagares A, Ramos A. Leakage decrease detected by dynamic susceptibility-weighted contrast-enhanced perfusion MRI predicts survival in recurrent glioblastoma treated with bevacizumab. Clin Transl Oncol 2016; 19:51-57. [PMID: 27026567 DOI: 10.1007/s12094-016-1502-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/09/2016] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND PURPOSE In glioblastoma, tumor progression appears to be triggered by expression of VEGF, a regulator of blood vessel permeability. Bevacizumab is a monoclonal antibody that inhibits angiogenesis by clearing circulating VEGF, resulting in a decline in the contrast-enhancing tumor, which does not always correlate with treatment response. Our objectives were: (1) to evaluate whether changes in DSC perfusion MRI-derived leakage could predict survival in recurrent glioblastoma, and (2) to estimate whether leakage at baseline was related to treatment outcome. MATERIALS AND METHODS We retrospectively analyzed DSC perfusion MRI in 24 recurrent glioblastomas treated with bevacizumab as second line chemotherapy. Leakage at baseline and changes in maximum leakage between baseline and the first follow-up after treatment were selected for quantitative analysis. Survival univariate analysis was made constructing survival curves using Kaplan-Meier method and comparing subgroups by log rank probability test. RESULTS Leakage reduction at 8 weeks after initiation of bevacizumab treatment had a significant influence on overall survival (OS) and progression-free survival (PFS). Median OS and PFS were 2.4 and 2.8 months longer for patients with leakage reduction at the first follow-up. Higher leakage at baseline was associated with leakage reduction after treatment. Odds ratio of treatment response was 9 for patients with maximum leakage at baseline >5. CONCLUSIONS Leakage decrease may predict OS and PFS in recurrent glioblastomas treated with bevacizumab. Leakage reduction postulates as a potential biomarker for treatment response evaluation. Leakage at baseline seems to predict response to treatment, but was not independently associated with survival.
Collapse
Affiliation(s)
- A Hilario
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain.
| | - J M Sepulveda
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - A Hernandez-Lain
- Department of Neuropathology, Hospital 12 de Octubre, Madrid, Spain
| | - E Salvador
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - L Koren
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - R Manneh
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Y Ruano
- Department of Neuropathology, Hospital 12 de Octubre, Madrid, Spain
| | - A Perez-Nuñez
- Department of Neurosurgery, Hospital 12 de Octubre, Madrid, Spain
| | - A Lagares
- Department of Neurosurgery, Hospital 12 de Octubre, Madrid, Spain
| | - A Ramos
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| |
Collapse
|
44
|
Mechanistic interrogation of combination bevacizumab/dual PI3K/mTOR inhibitor response in glioblastoma implementing novel MR and PET imaging biomarkers. Eur J Nucl Med Mol Imaging 2016; 43:1673-83. [PMID: 26975402 DOI: 10.1007/s00259-016-3343-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/16/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE Resistance to bevacizumab (BEV) in glioblastoma is believed to occur via activation of molecular networks including the mTOR/PI3K pathway. Using an MR/PET molecular imaging biomarker approach, we investigated the response to combining BEV with the mTOR/PI3K inhibitor BEZ235. METHODS Tumours were established by orthotopically implanting U87MG-luc2 cells in mice. Animals were treated with BEZ235 and/or BEV, and imaged using diffusion-weighted-MRI, T2-weighted and T2*-weighted before and after administration of superparamagnetic iron oxide contrast agent. Maps for changes in relaxation rates (ΔR2, ΔR2* and apparent diffusion coefficient) were calculated. Vessel size index and microvessel density index were derived. 3'-Deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) PET and O-(2-[(18)F]fluoroethyl)-L-tyrosine ([(18)F]FET) PET were further performed and tumour endothelium/proliferation markers assessed by immunohistochemistry. RESULTS Treatment with BEV resulted in a pronounced decrease in tumour volume (T2-weighted MRI). No additive effect on tumour volume was observed with the BEV/BEZ235 combination compared with BEV monotherapy. The Ki67 proliferation index and [(18)F]FLT uptake studies were used to support the observations. Using ΔR2* and ΔR2 values, respectively, the BEV/BEZ235 combination significantly reduced tumour microvessel volume in comparison to BEV alone. Decreased microvessel density index was further observed in animals treated with the combination, supported by von Willebrand factor (vWF) immunohistochemistry. [(18)F]FET uptake was decreased following treatment with BEV alone, but was not further reduced following treatment with the combination. vWF immunohistochemistry analysis showed that the mean tumour vessel size was increased in all cohorts. CONCLUSION Assessing MR imaging biomarker parameters together with [(18)F]FET and [(18)F]FLT PET provided information on mechanism of action of the drug combination and clues as to potential clinical responses. Following translation to clinical use, treatment with a BEV/BEZ235 combination could reduce peritumoral oedema obviating the requirement for steroids. The use of hypothesis-driven molecular imaging studies facilitates the preclinical evaluation of drug response. Studies of this kind may more accurately predict the clinical potential of the BEV/BEZ235 combination regimen as a novel therapeutic approach in oncology.
Collapse
|
45
|
Response Assessment and Magnetic Resonance Imaging Issues for Clinical Trials Involving High-Grade Gliomas. Top Magn Reson Imaging 2016; 24:127-36. [PMID: 26049816 DOI: 10.1097/rmr.0000000000000054] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There exist multiple challenges associated with the current response assessment criteria for high-grade gliomas, including the uncertain role of changes in nonenhancing T2 hyperintensity, and the phenomena of pseudoresponse and pseudoprogression in the setting of antiangiogenic and chemoradiation therapies, respectively. Advanced physiological magnetic resonance imaging (MRI), including diffusion and perfusion (dynamic susceptibility contrast MRI and dynamic contrast-enhanced MRI) sensitive techniques for overcoming response assessment challenges, has been proposed, with their own potential advantages and inherent shortcomings. Measurement variability exists for conventional and advanced MRI techniques, necessitating the standardization of image acquisition parameters in order to establish the utility of these imaging methods in multicenter trials for high-grade gliomas. This review chapter highlights the important features of MRI in clinical brain tumor trials, focusing on the current state of response assessment in brain tumors, advanced imaging techniques that may provide additional value for determining response, and imaging issues to be considered for multicenter trials.
Collapse
|
46
|
Siegal T, Charbit H, Paldor I, Zelikovitch B, Canello T, Benis A, Wong ML, Morokoff AP, Kaye AH, Lavon I. Dynamics of circulating hypoxia-mediated miRNAs and tumor response in patients with high-grade glioma treated with bevacizumab. J Neurosurg 2016; 125:1008-1015. [PMID: 26799295 DOI: 10.3171/2015.8.jns15437] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Bevacizumab is an antiangiogenic agent under investigation for use in patients with high-grade glioma. It produces a high rate of radiological response; however, this response should be interpreted with caution because it may reflect normalization of the tumor vasculature and not necessarily a true antitumor effect. The authors previously demonstrated that 4 hypoxia-mediated microRNAs (miRNA)-miR-210, miR-21, miR-10b, and miR-196b-are upregulated in glioma as compared with normal brain tissue. The authors hypothesized that the regulation and expression of these miRNAs would be altered in response to bevacizumab treatment. The object of this study was to perform longitudinal monitoring of circulating miRNA levels in patients undergoing bevacizumab treatment and to correlate it with tumor response. METHODS A total of 120 serum samples from 28 patients with high-grade glioma were prospectively collected prior to bevacizumab (n = 15) or temozolomide (TMZ; n = 13) treatment and then longitudinally during treatment. Quantification of the 4 miRNAs was evaluated by real-time polymerase chain reaction using total RNA extracted from the serum. At each time point, tumor response was assessed by Response Assessment in Neuro-Oncology criteria and by performing MRI using fluid attenuated inversion recovery (FLAIR) and contrast-enhanced images. RESULTS As compared with pretreatment levels, high levels of miR-10b and miR-21 were observed in the majority of patients throughout the bevacizumab treatment period. miR-10b and miR-21 levels correlated negatively and significantly with changes in enhancing tumor diameters (r = -0.648, p < 0.0001) in the bevacizumab group but not in the TMZ group. FLAIR images and the RANO assessment did not correlate with the sum quantification of these miRNAs in either group. CONCLUSIONS Circulating levels of miR-10b and miR-21 probably reflect the antiangiogenic effect of therapy, but their role as biomarkers for tumor response remains uncertain and requires further investigation.
Collapse
Affiliation(s)
- Tali Siegal
- Center for Neuro-Oncology, Davidoff Institute of Oncology, Rabin Medical Center, Campus Beilinson, Petach Tikva.,Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| | - Hanna Charbit
- Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| | - Iddo Paldor
- Department of Neurosurgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel; and
| | - Bracha Zelikovitch
- Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| | - Tamar Canello
- Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| | - Arriel Benis
- Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| | | | - Andrew P Morokoff
- Departments of 4 Neurosurgery and.,Surgery, The Royal Melbourne Hospital and The University of Melbourne, Australia
| | - Andrew H Kaye
- Departments of 4 Neurosurgery and.,Surgery, The Royal Melbourne Hospital and The University of Melbourne, Australia
| | - Iris Lavon
- Leslie and Michael Gaffin Center for Neuro-Oncology and Department of Neurology, The Agnes Ginges Center for Human Neurogenetics
| |
Collapse
|
47
|
Wen Q, Jalilian L, Lupo JM, Li Y, Roy R, Molinaro AM, Chang SM, Prados M, Butowski N, Clarke J, Nelson SJ. Association of Diffusion and Anatomic Imaging Parameters with Survival for Patients with Newly Diagnosed Glioblastoma Participating in Two Different Clinical Trials. Transl Oncol 2015; 8:446-55. [PMID: 26692525 PMCID: PMC4700297 DOI: 10.1016/j.tranon.2015.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/20/2023] Open
Abstract
PURPOSE: To evaluate the time course and association with survival of anatomic lesion volumes and diffusion imaging parameters for patients with newly diagnosed glioblastoma who were treated with radiation and concurrently with either temozolomide and enzastaurin (TMZ+enza cohort) or temozolomide, erlotonib, and bevaciumab (TMZ+erl+bev cohort). MATERIALS AND METHODS: Regions of interest corresponding to the contrast-enhancing and hyperintense lesions on T2-weighted images were generated. Diffusion-weighted images were processed to provide maps of apparent diffusion coefficient, fractional anisotropy, and longitudinal and radial eigenvalues. Histograms of diffusion values were generated and summary statistics calculated. Cox proportional hazards models were employed to assess the association of representative imaging parameters with survival with adjustments for age, Karnofsky performance status, and extent of resection. RESULTS: Although progression-free survival was significantly longer for the TMZ+erl+bev cohort (12.8 vs 7.3 months), there was no significant difference in overall survival between the two populations (17.0 vs 17.8 months). The median contrast-enhancing lesion volumes decreased from 6.3 to 1.9 cm3 from baseline to the postradiotherapy scan for patients in the TMZ+enza cohort and from 2.8 to 0.9cm3 for the TMZ+erl+bev cohort. Changes in the T2 lesion volumes were only significant for the latter cohort (26.5 to 11.9 cm3). The median apparent diffusion coefficient and related diffusion parameters were significantly increased for the TMZ+enza cohort (1054 to 1225 μm2/s). More of the anatomic parameters were associated with survival for the TMZ+enza cohort, whereas more diffusion parameters were associated with survival for the TMZ+erl+bev cohort. CONCLUSION: The early changes in anatomic and diffusion imaging parameters and their association with survival reflected differences in the mechanisms of action of the treatments that were being given. This suggests that integrating diffusion metrics and anatomic lesion volumes into the Response Assessment in Neuro-Oncology criteria would assist in interpreting treatment-induced changes and predicting outcome in patients with newly diagnosed glioblastoma who are receiving such combination treatments.
Collapse
Affiliation(s)
- Qiuting Wen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States
| | - Laleh Jalilian
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Ritu Roy
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA United States.
| |
Collapse
|
48
|
Yu Z, Zhao G, Zhang Z, Li Y, Chen Y, Wang N, Zhao Z, Xie G. Efficacy and safety of bevacizumab for the treatment of glioblastoma. Exp Ther Med 2015; 11:371-380. [PMID: 26893618 DOI: 10.3892/etm.2015.2947] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/13/2015] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary malignant intracranial tumor in adults. The current first-line treatment for patients with newly diagnosed GBM is surgical resection followed by radiotherapy plus concomitant and adjuvant temozolomide. This treatment protocol may prolong the survival period of the patient, however it is not curative and more effective therapeutic strategies are required. GBM is a type of highly vascularized tumor with increased expression levels of vascular endothelial growth factor (VEGF), which is a significant mediator of angiogenesis. Since angiogenesis is essential for tumor growth, anti-angiogenic therapies hold potential for the treatment of GBM, and targeting VEGF has demonstrated promising results in previous studies. Bevacizumab (BEV) is a recombinant humanized monoclonal antibody that inhibits VEGF and is approved by the US Food and Drug Administration as a monotherapy treatment for patients with recurrent GBM and is associated with manageable toxicity. Previous studies have demonstrated that BEV may be an effective treatment for recurrent GBM, with prolonged progression-free survival and overall survival, and maintained patient quality of life and functional status. The present review article briefly outlines the mechanism of action of BEV and summarizes the current literature and clinical trial research on the role of BEV for the treatment of patients with recurrent and newly diagnosed GBM.
Collapse
Affiliation(s)
- Zhiyun Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Gang Zhao
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhonghua Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yunqian Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yong Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Nan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhongying Zhao
- Department of Neurosurgery, Siping Central People's Hospital, Siping, Jilin 136000, P.R. China
| | - Guifang Xie
- Department of Obstetrics and Gynecology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
49
|
Wang K, Wang YY, Wang JF, Ma J, Jiang T, Dai JP. Radiologic Features and Expression of Vascular Endothelial Growth Factor Stratify Survival Outcomes in Patients with Glioblastoma. AJNR Am J Neuroradiol 2015; 37:629-35. [PMID: 26585263 DOI: 10.3174/ajnr.a4567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/19/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Vascular endothelial growth factor is a well-known tumor-specific biomarker that mediates angiogenesis in glioblastoma via hypoxia-dependent mechanisms. Our aim was to investigate the correlation of clinical characteristics, radiologic features, and vascular endothelial growth factor expression with survival outcomes in patients with glioblastoma. MATERIALS AND METHODS Clinical and radiologic data of 185 patients with glioblastoma were retrospectively reviewed. Vascular endothelial growth factor expression was examined in all cases via immunohistochemical analysis. Univariate and multivariate analyses were performed to identify the prognostic factors of progression-free survival and overall survival. RESULTS Vascular endothelial growth factor expression levels were associated with the presence of ringlike tumor contrast enhancement. Age, preoperative Karnofsky Performance Scale score, gross total resection, and adjuvant therapy were identified as prognostic factors. Among patients undergoing gross total resection, high vascular endothelial growth factor expression was associated with longer progression-free survival (P = .011) and overall survival (P = .039). For tumors with high vascular endothelial growth factor expression, both the non-contrast-enhancing tumor component and peritumoral edema could stratify overall survival (P = .039 and .018, respectively), while only the presence of the non-contrast-enhancing tumor component predicted a longer progression-free survival (P = .024). CONCLUSIONS Vascular endothelial growth factor expression level was not an independent prognostic factor in glioblastoma. However, high vascular endothelial growth factor expression might predict longer survival in patients in whom gross total resection was achieved. Furthermore, peritumoral edema and the non-contrast-enhancing tumor component could stratify survival outcomes in patients with high vascular endothelial growth factor tumors.
Collapse
Affiliation(s)
- K Wang
- From the Departments of Neuroradiology (K.W., J.M., J.P.D.)
| | - Y Y Wang
- Neurosurgery (Y.Y.W., J.F.W., T.J.), Beijing Tiantan Hospital Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China
| | - J F Wang
- Neurosurgery (Y.Y.W., J.F.W., T.J.), Beijing Tiantan Hospital Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China
| | - J Ma
- From the Departments of Neuroradiology (K.W., J.M., J.P.D.)
| | - T Jiang
- Neurosurgery (Y.Y.W., J.F.W., T.J.), Beijing Tiantan Hospital Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China Beijing Institute for Brain Disorders (T.J.), Brain Tumor Center, Beijing, China.
| | - J P Dai
- From the Departments of Neuroradiology (K.W., J.M., J.P.D.) Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Ellingson BM, Wen PY, van den Bent MJ, Cloughesy TF. Pros and cons of current brain tumor imaging. Neuro Oncol 2015; 16 Suppl 7:vii2-11. [PMID: 25313235 DOI: 10.1093/neuonc/nou224] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Over the past 20 years, very few agents have been approved for the treatment of brain tumors. Recent studies have highlighted some of the challenges in assessing activity in novel agents for the treatment of brain tumors. This paper reviews some of the key challenges related to assessment of tumor response to therapy in adult high-grade gliomas and discusses the strengths and limitations of imaging-based endpoints. Although overall survival is considered the "gold standard" endpoint in the field of oncology, progression-free survival and response rate are endpoints that hold great value in neuro-oncology. Particular focus is given to advancements made since the January 2006 Brain Tumor Endpoints Workshop, including the development of Response Assessment in Neuro-Oncology criteria, the value of T2/fluid-attenuated inversion recovery, use of objective response rates and progression-free survival in clinical trials, and the evaluation of pseudoprogression, pseudoresponse, and inflammatory response in radiographic images.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Patrick Y Wen
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Martin J van den Bent
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Timothy F Cloughesy
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| |
Collapse
|