1
|
A predictive microfluidic model of human glioblastoma to assess trafficking of blood-brain barrier-penetrant nanoparticles. Proc Natl Acad Sci U S A 2022; 119:e2118697119. [PMID: 35648828 PMCID: PMC9191661 DOI: 10.1073/pnas.2118697119] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The blood–brain barrier represents a significant challenge for the treatment of high-grade gliomas, and our understanding of drug transport across this critical biointerface remains limited. To advance preclinical therapeutic development for gliomas, there is an urgent need for predictive in vitro models with realistic blood–brain-barrier vasculature. Here, we report a vascularized human glioblastoma multiforme (GBM) model in a microfluidic device that accurately recapitulates brain tumor vasculature with self-assembled endothelial cells, astrocytes, and pericytes to investigate the transport of targeted nanotherapeutics across the blood–brain barrier and into GBM cells. Using modular layer-by-layer assembly, we functionalized the surface of nanoparticles with GBM-targeting motifs to improve trafficking to tumors. We directly compared nanoparticle transport in our in vitro platform with transport across mouse brain capillaries using intravital imaging, validating the ability of the platform to model in vivo blood–brain-barrier transport. We investigated the therapeutic potential of functionalized nanoparticles by encapsulating cisplatin and showed improved efficacy of these GBM-targeted nanoparticles both in vitro and in an in vivo orthotopic xenograft model. Our vascularized GBM model represents a significant biomaterials advance, enabling in-depth investigation of brain tumor vasculature and accelerating the development of targeted nanotherapeutics.
Collapse
|
2
|
Tseng YY, Chen TY, Liu SJ. Role of Polymeric Local Drug Delivery in Multimodal Treatment of Malignant Glioma: A Review. Int J Nanomedicine 2021; 16:4597-4614. [PMID: 34267515 PMCID: PMC8275179 DOI: 10.2147/ijn.s309937] [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: 03/11/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant gliomas (MGs) are the most common and devastating primary brain tumor. At present, surgical interventions, radiotherapy, and chemotherapy are only marginally effective in prolonging the life expectancy of patients with MGs. Inherent heterogeneity, aggressive invasion and infiltration, intact physical barriers, and the numerous mechanisms underlying chemotherapy and radiotherapy resistance contribute to the poor prognosis for patients with MGs. Various studies have investigated methods to overcome these obstacles in MG treatment. In this review, we address difficulties in MG treatment and focus on promising polymeric local drug delivery systems. In contrast to most local delivery systems, which are directly implanted into the residual cavity after intratumoral injection or the surgical removal of a tumor, some rapidly developing and promising nanotechnological methods—including surface-decorated nanoparticles, magnetic nanoparticles, and focused ultrasound assist transport—are administered through (systemic) intravascular injection. We also discuss further synergistic and multimodal strategies for heightening therapeutic efficacy. Finally, we outline the challenges and therapeutic potential of these polymeric drug delivery systems.
Collapse
Affiliation(s)
- Yuan-Yun Tseng
- Department of Neurosurgery, New Taipei Municipal Tu-Cheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City, Taiwan
| | - Tai-Yuan Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkuo, Tao-Yuan, Taiwan
| |
Collapse
|
3
|
Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis 2019; 23:55-74. [PMID: 31865479 DOI: 10.1007/s10456-019-09698-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Vessel co-option is a non-angiogenic mechanism of tumour vascularisation in which cancer cells utilise pre-existing blood vessels instead of inducing new blood vessel formation. Vessel co-option has been observed across a range of different tumour types, in both primary cancers and metastatic disease. Importantly, vessel co-option is now implicated as a major mechanism that mediates resistance to conventional anti-angiogenic drugs and this may help to explain the limited efficacy of this therapeutic approach in certain clinical settings. This includes the use of anti-angiogenic drugs to treat advanced-stage/metastatic disease, treatment in the adjuvant setting and the treatment of primary disease. In this article, we review the available evidence linking vessel co-option with resistance to anti-angiogenic therapy in numerous tumour types, including breast, colorectal, lung and pancreatic cancer, glioblastoma, melanoma, hepatocellular carcinoma, and renal cell carcinoma. The finding that vessel co-option is a significant mechanism of resistance to anti-angiogenic therapy may have important implications for the future of anti-cancer therapy, including (a) predicting response to anti-angiogenic drugs, (b) the need to develop therapies that target both angiogenesis and vessel co-option in tumours, and (c) predicting the response to other therapeutic modalities, including immunotherapy.
Collapse
|
4
|
Abstract
All solid tumours require a vascular supply in order to progress. Although the ability to induce angiogenesis (new blood vessel growth) has long been regarded as essential to this purpose, thus far, anti-angiogenic therapies have shown only modest efficacy in patients. Importantly, overshadowed by the literature on tumour angiogenesis is a long-standing, but continually emerging, body of research indicating that tumours can grow instead by hijacking pre-existing blood vessels of the surrounding nonmalignant tissue. This process, termed vessel co-option, is a frequently overlooked mechanism of tumour vascularization that can influence disease progression, metastasis and response to treatment. In this Review, we describe the evidence that tumours located at numerous anatomical sites can exploit vessel co-option. We also discuss the proposed molecular mechanisms involved and the multifaceted implications of vessel co-option for patient outcomes.
Collapse
Affiliation(s)
- Elizabeth A Kuczynski
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals St Augustinus, University of Antwerp, Wilrijk-Antwerp, Belgium
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Francesco Pezzella
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- Oncology Translational Medicine Unit, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
5
|
Eckert F, Schilbach K, Klumpp L, Bardoscia L, Sezgin EC, Schwab M, Zips D, Huber SM. Potential Role of CXCR4 Targeting in the Context of Radiotherapy and Immunotherapy of Cancer. Front Immunol 2018; 9:3018. [PMID: 30622535 PMCID: PMC6308162 DOI: 10.3389/fimmu.2018.03018] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/06/2018] [Indexed: 12/28/2022] Open
Abstract
Cancer immunotherapy has been established as standard of care in different tumor entities. After the first reports on synergistic effects with radiotherapy and the induction of abscopal effects-tumor shrinkage outside the irradiated volume attributed to immunological effects of radiotherapy-several treatment combinations have been evaluated. Different immunotherapy strategies (e.g., immune checkpoint inhibition, vaccination, cytokine based therapies) have been combined with local tumor irradiation in preclinical models. Clinical trials are ongoing in different cancer entities with a broad range of immunotherapeutics and radiation schedules. SDF-1 (CXCL12)/CXCR4 signaling has been described to play a major role in tumor biology, especially in hypoxia adaptation, metastasis and migration. Local tumor irradiation is a known inducer of SDF-1 expression and release. CXCR4 also plays a major role in immunological processes. CXCR4 antagonists have been approved for the use of hematopoietic stem cell mobilization from the bone marrow. In addition, several groups reported an influence of the SDF-1/CXCR4 axis on intratumoral immune cell subsets and anti-tumor immune response. The aim of this review is to merge the knowledge on the role of SDF-1/CXCR4 in tumor biology, radiotherapy and immunotherapy of cancer and in combinatorial approaches.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karin Schilbach
- Department of General Pediatrics/Pediatric Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Lilia Bardoscia
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Department of Radiation Oncology, University of Brescia, Brescia, Italy
| | - Efe Cumhur Sezgin
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University Hospital and University Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
6
|
Heinzel A, Müller D, Yekta-Michael SS, Ceccon G, Langen KJ, Mottaghy FM, Wiesmann M, Kocher M, Hattingen E, Galldiks N. O-(2-18F-fluoroethyl)-L-tyrosine PET for evaluation of brain metastasis recurrence after radiotherapy: an effectiveness and cost-effectiveness analysis. Neuro Oncol 2018; 19:1271-1278. [PMID: 28204572 DOI: 10.1093/neuonc/now310] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background Conventional MRI is the standard method to diagnose recurrence of brain metastases after radiation. However, following radiation therapy, reactive transient blood-brain barrier alterations with consecutive contrast enhancement can mimic brain metastasis recurrence. Recent studies have suggested that O-(2-18F-fluoroethyl)-L-tyrosine (FET) PET improves the correct differentiation of brain metastasis recurrence from radiation injury. Based on published evidence and clinical expert opinion, we analyzed effectiveness and cost-effectiveness of the use of FET PET in addition to MRI compared with MRI alone for the diagnosis of recurrent brain metastases. Methods A decision-tree model was designed to compare the 2 diagnostic strategies from the perspective of the German Statutory Health Insurance (SHI) system. Effectiveness was defined as correct diagnosis of recurrent brain metastasis and was compared between FET PET with MRI and MRI alone. Costs were calculated for a baseline scenario and for a more expensive scenario. Robustness of the results was tested using sensitivity analyses. Results Compared with MRI alone, FET PET in combination with MRI increases the rate of correct diagnoses by 42% (number needed to diagnose of 3) with an incremental cost-effectiveness ratio of €2821 (baseline scenario) and €4014 (more expensive scenario) per correct diagnosis. The sensitivity analyses confirmed the robustness of the results. Conclusions The model suggests that the additional use of FET PET with conventional MRI for the diagnosis of recurrent brain metastases may be cost-effective. Integration of FET PET has the potential to avoid overtreatment with corresponding costs as well as unnecessary side effects.
Collapse
Affiliation(s)
- Alexander Heinzel
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Dirk Müller
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Sareh Said Yekta-Michael
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Garry Ceccon
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Karl-Josef Langen
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Martin Wiesmann
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Martin Kocher
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Elke Hattingen
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| | - Norbert Galldiks
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany; Institute for Health Economics and Clinical Epidemiology, University of Cologne, Cologne, Germany; Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University of Aachen, Aachen, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Institute for Neuroscience and Medicine, Research Center Juelich, Juelich, Germany; Department of Neuroradiology University of Aachen, Aachen, Germany; Department of Radiation Oncology, University of Cologne, Cologne, Germany; Department of Radiology, University of Bonn, Bonn, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Germany
| |
Collapse
|
7
|
Liu SJ, Yang TC, Yang ST, Chen YC, Tseng YY. Biodegradable hybrid-structured nanofibrous membrane supported chemoprotective gene therapy enhances chemotherapy tolerance and efficacy in malignant glioma rats. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:515-526. [PMID: 29658349 DOI: 10.1080/21691401.2018.1460374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotherapy is ineffective for treating malignant glioma (MG) because of the low therapeutic levels of pharmaceuticals in tumour tissues and the well-known tumour resistance. The resistance to alkylators is modulated by the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT). O6-benzylguanine (O6-BG) can irreversibly inactivate AGT by competing with O6-methylguanine and has been confirmed to increase the therapeutic activity of alkylators. We developed hybrid-structured poly[(d,l)-lactide-co-glycolide] nanofibrous membranes (HSNMs) that enable the sequential and sustained release of O6-BG and two alkylators (carmustine and temozolomide [TMZ]). HSNMs were surgically instilled into the cerebral cavity of pathogen-free rats and F98 glioma-bearing rats. The release behaviours of loaded drugs were quantified by using high-performance liquid chromatography. The treatment results were compared with the rats treated with intraperitoneal injection of O6-BG combined with surgical implantation of carmustine wafer and oral TMZ. The HSNMs revealed a sequential drug release behaviour with the elution of high drug concentrations of O6-BG in the early phase, followed by high levels of two alkylators. All drug concentrations remained high for over 14 weeks. Tumour growth was slower and the mean survival time was significantly prolonged in the HSNM-treated group. Biodegradable HSNMs can enhance therapeutic efficacy and prevent toxic systemic effects.
Collapse
Affiliation(s)
- Shih-Jung Liu
- a Department of Mechanical Engineering , Chang Gung University , Tao-Yuan , Taiwan, ROC.,b Department of Orthopedic Surgery , Chang Gung Memorial Hospital , Tao-Yuan , Taiwan, ROC
| | - Tao-Chieh Yang
- c Department of Neurosurgery , Asia University Hospital , Taichung , Taiwan, ROC
| | - Shun-Tai Yang
- d Division of Neurosurgery, Department of Surgery , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan, ROC.,e Department of Surgery, School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ying-Chun Chen
- a Department of Mechanical Engineering , Chang Gung University , Tao-Yuan , Taiwan, ROC
| | - Yuan-Yun Tseng
- d Division of Neurosurgery, Department of Surgery , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan, ROC.,e Department of Surgery, School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan, ROC
| |
Collapse
|
8
|
Blumenthal DT, Kanner AA, Aizenstein O, Cagnano E, Greenberg A, Hershkovitz D, Ram Z, Bokstein F. Surgery for Recurrent High-Grade Glioma After Treatment with Bevacizumab. World Neurosurg 2018; 110:e727-e737. [DOI: 10.1016/j.wneu.2017.11.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 01/04/2023]
|
9
|
Yamamoto Y, Tamura R, Tanaka T, Ohara K, Tokuda Y, Miyake K, Takei J, Akasaki Y, Yoshida K, Murayama Y, Sasaki H. "Paradoxical" findings of tumor vascularity and oxygenation in recurrent glioblastomas refractory to bevacizumab. Oncotarget 2017; 8:103890-103899. [PMID: 29262608 PMCID: PMC5732774 DOI: 10.18632/oncotarget.21978] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/17/2017] [Indexed: 11/25/2022] Open
Abstract
Anti-angiogenic therapy induces the apparent normalization of vascular structure, decreases microvessel density (MVD), and improves tumor oxygenation in glioblastomas (GBMs). Six initial and recurrent tumor pairs after bevacizumab (Bev) treatment were compared with GBMs from nine patients resected under neoadjuvant Bev treatment with regard to histological characteristics; MVD; MIB-1 index; and expression of vascular endothelial growth factor (VEGF) and its receptors, hypoxia markers (hypoxia-inducible factor 1 alpha, carbonic anhydrase 9), and nestin as a marker of glioma stem-like cells. In recurrent tumors post-Bev treatment, while the MVD remained low compared with the paired initial tumors (pre-Bev tumors), the expression of hypoxic markers were increased and were even higher in expression compared with the paired pre-Bev tumors in three of the six cases. MIB-1 indices were similar among the initial GBMs, neoadjuvant group, and recurrent tumors post-Bev treatment. The nestin-positive cell ratio of the post-Bev recurrent tumors was as high as that of the pre-Bev tumors. The expression of VEGF and VEGFR1 was increased in the post-Bev recurrent tumors in three and four cases, respectively, compared with the paired pre-Bev tumors. In the majority of Bev-refractory GBMs, tumor hypoxia was present with a paradoxical decrease in MVD. These findings suggest that re-activation of tumor angiogenesis is not initially involved in the acquisition of resistance to Bev.
Collapse
Affiliation(s)
- Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa-shi, Chiba 277-8567, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa-shi, Chiba 277-8567, Japan
| | - Kentaro Ohara
- Division of Diagnostic Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yukina Tokuda
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Keisuke Miyake
- Department of Neurosurgery, Kagawa University Hospital, Kita-gun, Kagawa 761-0793, Japan
| | - Jun Takei
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa-shi, Chiba 277-8567, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
10
|
Han S, Wang C, Qin X, Xia J, Wu A. LPS alters the immuno-phenotype of glioma and glioma stem-like cells and induces in vivo antitumor immunity via TLR4. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017. [PMID: 28641579 PMCID: PMC5480420 DOI: 10.1186/s13046-017-0552-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND This study examined the ability of lipopolysaccharide (LPS) to affect glioma and glioma stem-like cells (GSCs) in vitro and to induce antitumor immunity in vivo and the role of TLR4 in these processes. METHODS Using RT-PCR and immunohistochemistry, we examined the expression of TLR4 in 34 glioblastoma clinical samples. Using real time-PCR, western blot and ELISA analyses, the effect of LPS stimulation on the expression of immune related molecules was evaluated in RG2 and U87 GSCs. Control or LPS-pretreated RG2 GSCs were intracranially or subcutaneously implanted into wild-type or nude Fisher 344 rats. Histopathological examinations were used to assess tumor progression and immune infiltration and Kaplan-Meier analyses to compare survival times of the animal models. RESULTS TLR4 was highly expressed in glioblastoma clinical samples. In vitro LPS stimulation for 6 h significantly altered expression of immune related molecules in RG2 and U87 GSCs. However, prolonged LPS stimulation diminished this effect. Rats inoculated intracranially with LPS-pretreated RG2 GSCs survived significantly longer than rats inoculated with control RG2 GSCs. In vivo, LPS-pretreated RG2 GSCs expressed higher levels of MHC molecules, CXCL10 and TNF-α and recruited more CD8+ lymphocytes. However, intratumoral LPS treatment was not equally beneficial. Furthermore, the in vitro and in vivo effects of LPS stimulation appeared to be largely TLR4-dependent. CONCLUSION LPS pretreatment promotes the recognition and eradication of tumor GSCs in vivo when the immune function of the tumor-bearing host is intact. In addition, our data indicate a complex relationship between bacterial infection and glioma prognosis.
Collapse
Affiliation(s)
- Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China
| | - Chao Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China
| | - Xiaofei Qin
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China
| | - Junzhe Xia
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China.
| |
Collapse
|
11
|
Uhm JH, Porter AB. Treatment of Glioma in the 21st Century: An Exciting Decade of Postsurgical Treatment Advances in the Molecular Era. Mayo Clin Proc 2017; 92:995-1004. [PMID: 28578786 DOI: 10.1016/j.mayocp.2017.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/21/2016] [Accepted: 01/25/2017] [Indexed: 11/18/2022]
Abstract
The past decade has brought about major changes in the way we classify and have begun to approach patients with high-grade glioma. As we trend toward personalized medicine, we are now able to utilize the molecular characteristics of each individual's tumor in order to tailor their treatment, particularly if the patient is elderly or has a poor performance status at baseline. We address the state of the practice as of 2016 in regard to chemotherapy, immunotherapy, and tumor-treating fields. The goal of this review is to enhance readers' understanding of the nuances that are allowing clinicians to tailor the treatment of high-grade glioma more specifically.
Collapse
Affiliation(s)
- Joon H Uhm
- Department of Neurology and Division of Neuro-Oncology, Mayo Clinic, Rochester, MN.
| | - Alyx B Porter
- Department of Neurology, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
12
|
Incidence of Tumour Progression and Pseudoprogression in High-Grade Gliomas: a Systematic Review and Meta-Analysis. Clin Neuroradiol 2017; 28:401-411. [PMID: 28466127 PMCID: PMC6105173 DOI: 10.1007/s00062-017-0584-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/04/2017] [Indexed: 12/29/2022]
Abstract
Background High-grade gliomas are the most common primary brain tumours. Pseudoprogression describes the false appearance of radiation-induced progression on MRI. A distinction should be made from true tumour progression to correctly plan treatment. However, there is wide variation of reported pseudoprogression. We thus aimed to establish the incidence of pseudoprogression and tumour progression in high-grade glioma patients with a systematic review and meta-analysis. Methods We searched PubMed, Embase and Web of Science on the incidence of pseudoprogression and tumour progression in adult high-grade glioma patients from 2005, the latest on 8 October 2014. Histology or imaging follow-up was used as reference standard. Extracted data included number of patients with worsening of imaging findings on T1 postcontrast or T2/FLAIR, pseudoprogression and tumour progression. Study quality was assessed. Heterogeneity was tested with I2. Pooling of the results was done with random models using Metaprop in STATA (StataCorp. Stata Statistical Software. College Station, TX: StataCorp LP). Results We identified 73 studies. MRI progression occurred in 2603 patients. Of these, 36% (95% confidence interval [CI] 33–40%) demonstrated pseudoprogression, 60% (95%CI 56–64%) tumour progression and unknown outcome was present in the remaining 4% of the patients (range 1–37%). Conclusion This meta-analysis demonstrated for the first time a notably high pooled incidence of pseudoprogression in patients with a form of progression across the available literature. This highlighted the full extent of the problem of the currently conventional MRI-based Response Assessment in Neuro-Oncology (RANO) criteria for treatment evaluation in high-grade gliomas. This underscores the need for more accurate treatment evaluation using advanced imaging to improve diagnostic accuracy and therapeutic approach. Electronic supplementary material The online version of this article (doi: 10.1007/s00062-017-0584-x) contains supplementary material, which is available to authorized users. It contains the characteristics of the included studies (supplementary table 1) and a full search strategy (see supplementary search strategy).
Collapse
|
13
|
Kuczynski EA, Yin M, Bar-Zion A, Lee CR, Butz H, Man S, Daley F, Vermeulen PB, Yousef GM, Foster FS, Reynolds AR, Kerbel RS. Co-option of Liver Vessels and Not Sprouting Angiogenesis Drives Acquired Sorafenib Resistance in Hepatocellular Carcinoma. J Natl Cancer Inst 2016; 108:djw030. [PMID: 27059374 PMCID: PMC5017954 DOI: 10.1093/jnci/djw030] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/24/2015] [Accepted: 02/08/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The anti-angiogenic Sorafenib is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC). However, acquired resistance limits its efficacy. An emerging theory to explain intrinsic resistance to other anti-angiogenic drugs is 'vessel co-option,' ie, the ability of tumors to hijack the existing vasculature in organs such as the lungs or liver, thus limiting the need for sprouting angiogenesis. Vessel co-option has not been evaluated as a potential mechanism for acquired resistance to anti-angiogenic agents. METHODS To study sorafenib resistance mechanisms, we used an orthotopic human HCC model (n = 4-11 per group), where tumor cells are tagged with a secreted protein biomarker to monitor disease burden and response to therapy. Histopathology, vessel perfusion assessed by contrast-enhanced ultrasound, and miRNA sequencing and quantitative real-time polymerase chain reaction were used to monitor changes in tumor biology. RESULTS While sorafenib initially inhibited angiogenesis and stabilized tumor growth, no angiogenic 'rebound' effect was observed during development of resistance unless therapy was stopped. Instead, resistant tumors became more locally infiltrative, which facilitated extensive incorporation of liver parenchyma and the co-option of liver-associated vessels. Up to 75% (±10.9%) of total vessels were provided by vessel co-option in resistant tumors relative to 23.3% (±10.3%) in untreated controls. miRNA sequencing implicated pro-invasive signaling and epithelial-to-mesenchymal-like transition during resistance development while functional imaging further supported a shift from angiogenesis to vessel co-option. CONCLUSIONS This is the first documentation of vessel co-option as a mechanism of acquired resistance to anti-angiogenic therapy and could have important implications including the potential therapeutic benefits of targeting vessel co-option in conjunction with vascular endothelial growth factor receptor signaling.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Antigens, CD34/metabolism
- Antineoplastic Agents/therapeutic use
- Blood Vessels/diagnostic imaging
- Blood Vessels/pathology
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Contrast Media
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Epithelial-Mesenchymal Transition/genetics
- Homeodomain Proteins/genetics
- Humans
- Liver/blood supply
- Liver Neoplasms/blood supply
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, SCID
- MicroRNAs/analysis
- Neoplasm Invasiveness
- Neoplasm Transplantation
- Neovascularization, Pathologic/diagnostic imaging
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Osteopontin/blood
- Phenylurea Compounds/therapeutic use
- Repressor Proteins/genetics
- Sequence Analysis, RNA
- Signal Transduction/genetics
- Sorafenib
- Ultrasonography
- Up-Regulation
- Vascular Endothelial Growth Factor A/blood
- Vimentin/genetics
- Zinc Finger E-box Binding Homeobox 2
- Zinc Finger E-box-Binding Homeobox 1/genetics
Collapse
Affiliation(s)
- Elizabeth A Kuczynski
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Melissa Yin
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Avinoam Bar-Zion
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Christina R Lee
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Henriett Butz
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Shan Man
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Frances Daley
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Peter B Vermeulen
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - George M Yousef
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - F Stuart Foster
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Andrew R Reynolds
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| | - Robert S Kerbel
- Affiliations of authors: Department of Medical Biophysics, University of Toronto, Toronto, Canada (EAK, FSF, RSK); Physical Sciences Platform (MY, FSF) and Biological Sciences Platform (CRL, SM, RSK), Sunnybrook Research Institute, Toronto, Canada; Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel (ABZ); Keenan Research Centre, St. Michael's Hospital, Toronto, Canada (HB, GMY); The Breast Cancer Now Toby Robins Research Centre, Mary-Jean Mitchell Green Building, The Institute of Cancer Research, London, UK (FD, PBV, ARR); Translational Cancer Research Unit, GZA Hospitals St. Augustinus, Antwerp, Belgium (PBV)
| |
Collapse
|
14
|
Tseng YY, Kau YC, Liu SJ. Advanced interstitial chemotherapy for treating malignant glioma. Expert Opin Drug Deliv 2016; 13:1533-1544. [DOI: 10.1080/17425247.2016.1193153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yuan-Yun Tseng
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chuan Kau
- Department of Anesthesiology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| |
Collapse
|
15
|
Kang JH, Adamson C. Novel chemotherapeutics and other therapies for treating high-grade glioma. Expert Opin Investig Drugs 2015; 24:1361-79. [PMID: 26289791 DOI: 10.1517/13543784.2015.1048332] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Despite extensive research, high-grade glioma (HGG) remains a dire diagnosis with no change in the standard of care in almost a decade. However, recent advancements uncovering molecular biomarkers of brain tumors and tumor-specific antigens targeted by immunotherapies provide opportunities for novel personalized treatment regimens to improve survival. AREAS COVERED In this review, the authors provide a comprehensive overview of recent therapeutic advancements in HGG. Furthermore, they describe new molecular biomarkers and molecular classifications, in addition to updated research on bevacizumab, targeted molecular therapies, immunotherapy and alternative delivery methods that overcome the blood-brain barrier to reach the target tumor tissue. Challenges regarding each therapy are also outlined. The authors also provide some insight into a novel non-chemotherapeutic treatment for malignant glioma, NovoTTFA, as well as a summary of current treatment options for recurrence. EXPERT OPINION Current research for treating malignant gliomas are paving the path to personalized therapy, including immunotherapy, that involve integrated genomic and histolopathologic data, as well as a multi-modal treatment regimen. Immunotherapy will potentially be the next addition to the current standard of care, specialized to the antigens presented on the tumors. The results of the current trials of multi-antigen vaccines are eagerly anticipated.
Collapse
Affiliation(s)
- Jennifer H Kang
- a 1 Duke University School of Medicine , Box 3807, Durham, NC, USA
| | - Cory Adamson
- b 2 Director, Molecular Neuro-oncology Lab, Duke Medical Center , DUMC Box 3807, Durham, NC, USA.,c 3 Chief of Neurosurgery, Durham VA Medical Center , 508 Fulton Street, Durham, NC, USA +1 919 698 3152 ; .,d 4 Duke Medical Center , DUMC Box 3807, Durham, NC, USA
| |
Collapse
|
16
|
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
|
17
|
Concurrent delivery of carmustine, irinotecan, and cisplatin to the cerebral cavity using biodegradable nanofibers: In vitro and in vivo studies. Colloids Surf B Biointerfaces 2015. [PMID: 26209775 DOI: 10.1016/j.colsurfb.2015.06.055] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor, and the prognosis of patients afflicted with GBM has been dismal, exhibiting progressive neurologic impairment and imminent death. Even with the most active regimens currently available, chemotherapy achieves only modest improvement in the overall survival. New chemotherapeutic agents and novel approaches to therapy are required for improving clinical outcomes. In this study, we used an electrospinning technique and developed biodegradable poly[(d,l)-lactide-co-glycolide] nanofibrous membranes that facilitated a sustained release of carmustine (or bis-chloroethylnitrosourea, BCNU), irinotecan, and cisplatin. An elution method and a high-performance liquid chromatography assay were employed to characterize the in vitro and in vivo release behaviors of pharmaceuticals from the nanofibrous membranes. The experimental results showed that the biodegradable, nanofibrous membranes released high concentrations of BCNU, irinotecan, and cisplatin for more than 8 weeks in the cerebral cavity of rats. A histological examination revealed progressive atrophy of the brain tissues without inflammatory reactions. Biodegradable drug-eluting nanofibrous membranes may facilitate sustained delivery of various and concurrent chemotherapeutic agents in the cerebral cavity, enhancing the therapeutic efficacy of GBM treatment and preventing toxic effects resulting from the systemic administration of chemotherapeutic agents.
Collapse
|
18
|
Prognostic implication of progression pattern after anti-VEGF bevacizumab treatment for recurrent malignant gliomas. J Neurooncol 2015; 124:101-10. [DOI: 10.1007/s11060-015-1808-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 05/09/2015] [Indexed: 12/22/2022]
|
19
|
Lu KV, Bergers G. Mechanisms of evasive resistance to anti-VEGF therapy in glioblastoma. CNS Oncol 2015; 2:49-65. [PMID: 23750318 DOI: 10.2217/cns.12.36] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Angiogenesis inhibitors targeting the VEGF signaling pathway have been US FDA approved for various cancers including glioblastoma (GBM), one of the most lethal and angiogenic tumors. This has led to the routine use of the anti-VEGF antibody bevacizumab in recurrent GBM, conveying substantial improvements in radiographic response, progression-free survival and quality of life. Despite these encouraging beneficial effects, patients inevitably develop resistance and frequently fail to demonstrate significantly better overall survival. Unlike chemotherapies, to which tumors exhibit resistance due to genetic mutation of drug targets, emerging evidence suggests that tumors bypass antiangiogenic therapy while VEGF signaling remains inhibited through a variety of mechanisms that are just beginning to be recognized. Because of the indirect nature of resistance to VEGF inhibitors there is promise that strategies combining angiogenesis inhibitors with drugs targeting such evasive resistance pathways will lead to more durable antiangiogenic efficacy and improved patient outcomes. Further identifying and understanding of evasive resistance mechanisms and their clinical importance in GBM relapse is therefore a timely and critical issue.
Collapse
|
20
|
Wang W, Sivakumar W, Torres S, Jhaveri N, Vaikari VP, Gong A, Howard A, Golden EB, Louie SG, Schönthal AH, Hofman FM, Chen TC. Effects of convection-enhanced delivery of bevacizumab on survival of glioma-bearing animals. Neurosurg Focus 2015; 38:E8. [DOI: 10.3171/2015.1.focus14743] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECT
Bevacizumab (Avastin), an antibody to vascular endothelial growth factor (VEGF), alone or in combination with irinotecan (Camptosar [CPT-11]), is a promising treatment for recurrent glioblastoma. However, the intravenous (IV) administration of bevacizumab produces a number of systemic side effects, and the increase in survival it provides for patients with recurrent glioblastoma is still only a few months. Because bevacizumab is an antibody against VEGF, which is secreted into the extracellular milieu by glioma cells, the authors hypothesized that direct chronic intratumoral delivery techniques (i.e., convection-enhanced delivery [CED]) can be more effective than IV administration. To test this hypothesis, the authors compared outcomes for these routes of bevacizumab application with respect to animal survival, microvessel density (MVD), and inflammatory cell distribution.
METHODS
Two human glioma cell lines, U87 and U251, were used as sources of intracranial tumor cells. The glioma cell lines were implanted into the brains of mice in an orthotopic xenograft mouse tumor model. After 7 days, the mice were treated with one of the following: 1) vehicle, 2) CED bevacizumab, 3) IV bevacizumab, 4) intraperitoneal (IP) irinotecan, 5) CED bevacizumab plus IP irinotecan, or 6) IV bevacizumab plus IP irinotecan. Alzet micro-osmotic pumps were used to introduce bevacizumab directly into the tumor. Survival was monitored. Excised tumor tissue samples were immunostained to measure MVD and inflammatory cell and growth factor levels.
RESULTS
The results demonstrate that mice treated with CED of bevacizumab alone or in combination with irinotecan survived longer than those treated systemically; CED-treated animals survived 30% longer than IV-treated animals. In combination studies, CED bevacizumab plus CPT-11 increased survival by more than 90%, whereas IV bevacizumab plus CPT-11 increased survival by 40%. Furthermore, CED bevacizumab-treated tissues exhibited decreased MVD compared with that of IV-treated tissues. In additional studies, the infiltration of macrophages and dendritic cells into CED-treated animals were increased compared with those in IV-treated animals, suggesting a highly active inflammatory response taking place in CED-treated mice.
CONCLUSIONS
The administration of bevacizumab via CED increases survival over that of treatment with IV bevacizumab. Thus, CED of bevacizumab alone or in combination with chemotherapy can be an effective protocol for treating gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Stan G. Louie
- 4Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Axel H. Schönthal
- 3Molecular Microbiology and Immunology, Keck School of Medicine; and
| | | | | |
Collapse
|
21
|
Bota DA, Alexandru-Abrams D, Pretto C, Hofman FM, Chen TC, Fu B, Carrillo JA, Schijns VE, Stathopoulos A. Use of ERC-1671 Vaccine in a Patient with Recurrent Glioblastoma Multiforme after Progression during Bevacizumab Therapy: First Published Report. Perm J 2015; 19:41-6. [PMID: 25785641 DOI: 10.7812/tpp/14-042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Glioblastoma multiforme is a highy aggressive tumor that recurs despite resection, focal beam radiation, and temozolamide chemotherapy. ERC-1671 is an experimental treatment strategy that uses the patient's own immune system to attack the tumor cells. The authors report preliminary data on the first human administration of ERC-1671 vaccination under a single-patient, compassionate-use protocol. The patient survived for ten months after the vaccine administration without any other adjuvant therapy and died of complications related to his previous chemotherapies.
Collapse
Affiliation(s)
- Daniela A Bota
- Associate Professor of Neurology and Neurosurgery at the University of California, Irvine in the City of Orange.
| | - Daniela Alexandru-Abrams
- Neurosurgery Resident at the University of California, Irvine Medical Center in the City of Orange.
| | - Chrystel Pretto
- Laboratory Manager for Epitopoietic Research Corporation at the Scientific Park Crealys in Gemblous, Les Ines, Belgium.
| | - Florence M Hofman
- Professor of Pathology at the Keck School of Medicine, University of Southern California, Los Angeles.
| | - Thomas C Chen
- Chief Medical Officer of Epitopoietic Research Corporation and a Professor of Neurosurgery at the Keck School of Medicine, University of Southern California, Los Angeles.
| | - Beverly Fu
- Neurological Oncology Nurse Practitioner in the Neurology Department of the University of California, Irvine Medical Center in the City of Orange.
| | - Jose A Carrillo
- Assistant Professor of Neurology at the University of California, Irvine Medical Center in the City of Orange.
| | - Virgil Ejc Schijns
- Chief Security Officer of Epitopoietic Research Corporation and a Professor of Immune Intervention in the Department of Cell Biology and Immunology at the Wageningen University in Netherlands.
| | - Apostolos Stathopoulos
- Chief Executive Officer for Epitopoietic Research Corporation at the Scientific Park Crealys in Gemblous, Les Ines, Belgium and Chief Neurosurgeon, Department of Neurosurgery, Arlon Hospital, Belgium.
| |
Collapse
|
22
|
Macas J, Ku MC, Nern C, Xu Y, Bühler H, Remke M, Synowitz M, Franz K, Seifert V, Plate KH, Kettenmann H, Glass R, Momma S. Generation of neuronal progenitor cells in response to tumors in the human brain. Stem Cells 2014; 32:244-57. [PMID: 24170295 DOI: 10.1002/stem.1581] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 09/26/2013] [Indexed: 01/18/2023]
Abstract
Data from transgenic mouse models show that neuronal progenitor cells (NPCs) migrate toward experimental brain tumors and modulate the course of pathology. However, the pathways whereby NPCs are attracted to CNS neoplasms are not fully understood and it is unexplored if NPCs migrate toward brain tumors (high-grade astrocytomas) in humans. We analyzed the tumor-parenchyma interface of neurosurgical resections for the presence of (NPCs) and distinguished these physiological cells from the tumor mass. We observed that polysialic acid neural cell adhesion molecule-positive NPCs accumulate at the border of high-grade astrocytomas and display a marker profile consistent with immature migratory NPCs. Importantly, these high-grade astrocytoma-associated NPCs did not carry genetic aberrations that are indicative of the tumor. Additionally, we observed NPCs accumulating in CNS metastases. These metastatic tumors are distinguished from neural cells by defined sets of markers. Transplanting murine glioma cells embedded in a cell-impermeable hollow fiber capsule into the brains of nestin-gfp reporter mice showed that diffusible factors are sufficient to induce a neurogenic reaction. In vitro, vascular endothelial growth factor (VEGF) secreted from glioma cells increases the migratory and proliferative behavior of adult human brain-derived neural stem and progenitor cells via stimulation of VEGF receptor-2 (VEGFR-2). In vivo, inhibiting VEGFR-2 signaling with a function-blocking antibody led to a reduction in NPC migration toward tumors. Overall, our data reveal a mechanism by which NPCs are attracted to CNS tumors and suggest that NPCs accumulate in human high-grade astrocytomas.
Collapse
Affiliation(s)
- Jadranka Macas
- Institute of Neurology (Edinger Institute), Department of Neurosurgery, Frankfurt University Medical School, Frankfurt, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mansour J, Fields B, Macomson S, Rixe O. Significant anti-tumor effect of bevacizumab in treatment of pineal gland glioblastoma multiforme. Target Oncol 2014; 9:395-8. [DOI: 10.1007/s11523-014-0327-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/20/2014] [Indexed: 10/25/2022]
|
24
|
|
25
|
Ogura K, Mizowaki T, Arakawa Y, Sakanaka K, Miyamoto S, Hiraoka M. Efficacy of salvage stereotactic radiotherapy for recurrent glioma: impact of tumor morphology and method of target delineation on local control. Cancer Med 2013; 2:942-9. [PMID: 24403268 PMCID: PMC3892399 DOI: 10.1002/cam4.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 01/17/2023] Open
Abstract
In this study, we assessed the efficacy of salvage stereotactic radiotherapy (SRT) for recurrent glioma. From August 2008 to December 2012, 30 patients with recurrent glioma underwent salvage SRT. The initial histological diagnoses were World Health Organization (WHO) grades II, III, and IV in 6, 9, and 15 patients, respectively. Morphologically, the type of recurrence was classified as diffuse or other. Two methods of clinical target delineation were used: A, a contrast-enhancing tumor; or B, a contrast-enhancing tumor with a 3–10-mm margin and/or surrounding fluid attenuation inversion recovery (FLAIR) high-intensity areas. The prescribed dose was 22.5–35 Gy delivered in five fractions at an isocenter using a dynamic conformal arc technique. The overall survival (OS) and local control probability (LCP) after SRT were calculated using the Kaplan–Meier method. A univariate analysis was used to test the effect of clinical variables on OS/LCP. The median follow-up period was 272 days after SRT. The OS and LCP were 83% and 56% at 6 months after SRT, respectively. Morphologically, the tumor type correlated significantly with both OS and LCP (P = 0.006 and <0.001, respectively). The method of target delineation also had a significant influence on LCP (P = 0.016). Grade 3 radiation necrosis was observed in two patients according to Common Terminology Criteria for Adverse Events, version 3. Salvage SRT was safe and effective for recurrent glioma, especially non-diffuse recurrences. Improved local control might be obtained by adding a margin to contrast-enhancing tumors or including increased FLAIR high-intensity areas.
Collapse
Affiliation(s)
- Kengo Ogura
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Johnson DR, Leeper HE, Uhm JH. Glioblastoma survival in the United States improved after Food and Drug Administration approval of bevacizumab: A population-based analysis. Cancer 2013; 119:3489-95. [DOI: 10.1002/cncr.28259] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/16/2013] [Accepted: 06/18/2013] [Indexed: 11/11/2022]
Affiliation(s)
| | | | - Joon H. Uhm
- Department of Neurology; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
27
|
Galldiks N, Rapp M, Stoffels G, Dunkl V, Sabel M, Langen KJ. Earlier Diagnosis of Progressive Disease during Bevacizumab Treatment Using O-(2-18F-Fluorethyl)-L-Tyrosine Positron Emission Tomography in Comparison with Magnetic Resonance Imaging. Mol Imaging 2013. [DOI: 10.2310/7290.2013.00051] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Norbert Galldiks
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Marion Rapp
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Gabriele Stoffels
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Veronika Dunkl
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Michael Sabel
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Karl-Josef Langen
- From the Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany; and Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| |
Collapse
|
28
|
Heinzel A, Müller D, Langen KJ, Blaum M, Verburg FA, Mottaghy FM, Galldiks N. The Use of O-(2-18F-Fluoroethyl)-l-Tyrosine PET for Treatment Management of Bevacizumab and Irinotecan in Patients with Recurrent High-Grade Glioma: A Cost-Effectiveness Analysis. J Nucl Med 2013; 54:1217-22. [DOI: 10.2967/jnumed.113.120089] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Dasgupta T, Haas-Kogan DA. The combination of novel targeted molecular agents and radiation in the treatment of pediatric gliomas. Front Oncol 2013; 3:110. [PMID: 23717811 PMCID: PMC3650671 DOI: 10.3389/fonc.2013.00110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/22/2013] [Indexed: 11/13/2022] Open
Abstract
Brain tumors are the most common solid pediatric malignancy. For high-grade, recurrent, or refractory pediatric brain tumors, radiation therapy (XRT) is an integral treatment modality. In the era of personalized cancer therapy, molecularly targeted agents have been designed to inhibit pathways critical to tumorigenesis. Our evolving knowledge of genetic aberrations in pediatric gliomas is being exploited with the use of specific targeted inhibitors. These agents are additionally being combined with XRT to increase the efficacy and duration of local control. In this review, we discuss novel agents targeting three different pathways in gliomas, and their potential combination with XRT. BRAF is a serine/threonine kinase in the RAS/RAF/MAPK kinase pathway, which is integral to cellular division, survival, and metabolism. Two-thirds of pilocytic astrocytomas, a low-grade pediatric glioma, contain a translocation within the BRAF gene called KIAA1549:BRAF that causes an overactivation of the MEK/MAPK signaling cascade. In vitro and in vivo data support the use of MEK or mammalian target of rapamycin (mTOR) inhibitors in low-grade gliomas expressing this translocation. Additionally, 15-20% of high-grade pediatric gliomas express BRAF V600E, an activating mutation of the BRAF gene. Pre-clinical in vivo and in vitro data in BRAF V600E gliomas demonstrate dramatic cooperation between XRT and small molecule inhibitors of BRAF V600E. Another major signaling cascade that plays a role in pediatric glioma pathogenesis is the PI3-kinase (PI3K)/mTOR pathway, known to be upregulated in the majority of high- and low-grade pediatric gliomas. Dual PI3K/mTOR inhibitors are in clinical trials for adult high-grade gliomas and are poised to enter studies of pediatric tumors. Finally, many brain tumors express potent stimulators of angiogenesis that render them refractory to treatment. An analog of thalidomide, CC-5103 increases the secretion of critical cytokines of the tumor microenvironment, including IL-2, IFN-γ, TNF-α, and IL-10, and is currently being evaluated in clinical trials for the treatment of recurrent or refractory pediatric central nervous system tumors. In summary, several targeted inhibitors with radiation are currently under investigation in both translational bench research and early clinical trials. This review article summarizes the molecular rationale for, and the pre-clinical data supporting the combinations of these targeted agents with other anti-cancer agents and XRT in pediatric gliomas. In many cases, parallels are drawn to molecular mechanisms and targeted inhibitors of adult gliomas. We additionally discuss the potential mechanisms underlying the efficacy of these agents.
Collapse
Affiliation(s)
- Tina Dasgupta
- Department of Radiation Oncology, University of California San FranciscoSan Francisco, CA, USA
| | - Daphne A. Haas-Kogan
- Department of Radiation Oncology, University of California San FranciscoSan Francisco, CA, USA
| |
Collapse
|
30
|
Javerzat S, Godard V, Bikfalvi A. Balancing risks and benefits of anti-angiogenic drugs for malignant glioma. FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.12.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Angiogenesis is a delicate process that has been programmed over the time of evolution of vertebrates to provide optimized quantities of oxygen and nutrients to the developing embryo and the growing newborn. Similarly, angiogenesis induction pathways are used during tumor development. Angiogenesis and tumor cell invasion are closely linked. Anti-angiogenesis treatment strategies have entered the clinic and show some promising results. However, recent research using preclinical models have pointed to possible harmful effects, including evasive resistance and increase in tumor cell invasion when VEGF activity is inhibited. This has been corroborated by observations in treated glioblastoma patients. However, the meaning of these observations is still in question. The results of Phase III clinical trials that are ongoing will certainly provide more definitive answers with regard to evasive resistance in glioblastoma treated with anti-angiogenic drugs.
Collapse
Affiliation(s)
- Sophie Javerzat
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
| | - Virginie Godard
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
| | - Andreas Bikfalvi
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France.
| |
Collapse
|
31
|
Kumar K, Wigfield S, Gee HE, Devlin CM, Singleton D, Li JL, Buffa F, Huffman M, Sinn AL, Silver J, Turley H, Leek R, Harris AL, Ivan M. Dichloroacetate reverses the hypoxic adaptation to bevacizumab and enhances its antitumor effects in mouse xenografts. J Mol Med (Berl) 2013; 91:749-58. [PMID: 23361368 DOI: 10.1007/s00109-013-0996-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 12/20/2012] [Accepted: 01/02/2013] [Indexed: 11/30/2022]
Abstract
Inhibition of vascular endothelial growth factor increases response rates to chemotherapy and progression-free survival in glioblastoma. However, resistance invariably occurs, prompting the urgent need for identification of synergizing agents. One possible strategy is to understand tumor adaptation to microenvironmental changes induced by antiangiogenic drugs and test agents that exploit this process. We used an in vivo glioblastoma-derived xenograft model of tumor escape in presence of continuous treatment with bevacizumab. U87-MG or U118-MG cells were subcutaneously implanted into either BALB/c SCID or athymic nude mice. Bevacizumab was given by intraperitoneal injection every 3 days (2.5 mg/kg/dose) and/or dichloroacetate (DCA) was administered by oral gavage twice daily (50 mg/kg/dose) when tumor volumes reached 0.3 cm(3) and continued until tumors reached approximately 1.5-2.0 cm(3). Microarray analysis of resistant U87 tumors revealed coordinated changes at the level of metabolic genes, in particular, a widening gap between glycolysis and mitochondrial respiration. There was a highly significant difference between U87-MG-implanted athymic nude mice 1 week after drug treatment. By 2 weeks of treatment, bevacizumab and DCA together dramatically blocked tumor growth compared to either drug alone. Similar results were seen in athymic nude mice implanted with U118-MG cells. We demonstrate for the first time that reversal of the bevacizumab-induced shift in metabolism using DCA is detrimental to neoplastic growth in vivo. As DCA is viewed as a promising agent targeting tumor metabolism, our data establish the timely proof of concept that combining it with antiangiogenic therapy represents a potent antineoplastic strategy.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Re-irradiation with and without bevacizumab as salvage therapy for recurrent or progressive high-grade gliomas. J Neurooncol 2013; 112:133-9. [DOI: 10.1007/s11060-013-1044-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/01/2013] [Indexed: 10/27/2022]
|
33
|
Tseng YY, Liao JY, Chen WA, Kao YC, Liu SJ. Sustainable release of carmustine from biodegradable poly[((D,L))-lactide-co-glycolide] nanofibrous membranes in the cerebral cavity: in vitro and in vivo studies. Expert Opin Drug Deliv 2013; 10:879-88. [PMID: 23289446 DOI: 10.1517/17425247.2013.758102] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) is the most common and most aggressive malignant primary brain tumor in humans. The only interstitial chemotherapy pharmaceutical approved to date for GBM treatment is the Gliadel® wafer. Despite the safety and efficacy of this approach that have been demonstrated in patients undergoing resection of both newly diagnosed and recurrent malignant gliomas, the wafer provides an effective release of the anticancer 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) for only 5 days. METHODS In this study, the authors developed biodegradable poly[(d,l)-lactide-co-glycolide] nanofibrous membranes via electrospinning that provided a sustained release of BCNU. An elution method and a HPLC assay were employed to characterize the in vitro and in vivo release behaviors of pharmaceuticals from the electrospun membranes. RESULTS The experimental results show that the biodegradable, nanofibrous membranes released high concentrations of BCNU for more than 6 weeks in the cerebral cavity of rats. Furthermore, the membranes can better conform to the geometry of the brain tissue and can cover more completely the tissue after the removal of tumors, achieving better drug transport without interfering with the normal function of the brain. Histological examination showed no obvious inflammation reactions of the brain tissues. CONCLUSION Adopting the electrospinning technique will help in manufacturing biodegradable, nanofibrous membranes for the long-term deliveries of various anticancer drugs in the cerebral cavity, which will further enhance the therapeutic efficacy of GBM treatment.
Collapse
Affiliation(s)
- Yuan-Yun Tseng
- Taipei Medical University, Shuang Ho Hospital, Department of Neurosurgery, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
34
|
Increased invasion of malignant gliomas after 15-LO-1 and HSV-tk/ganciclovir combination gene therapy. Cancer Gene Ther 2012; 19:870-4. [DOI: 10.1038/cgt.2012.76] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Response assessment of bevacizumab in patients with recurrent malignant glioma using [18F]Fluoroethyl-L-tyrosine PET in comparison to MRI. Eur J Nucl Med Mol Imaging 2012; 40:22-33. [PMID: 23053325 DOI: 10.1007/s00259-012-2251-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/09/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE To investigate prospectively the potential of O-(2-[(18)F]fluoroethyl)-L-tyrosine ((18)F-FET) PET in comparison to MRI for the assessment of the response of patients with recurrent high-grade glioma (rHGG) to antiangiogenic treatment. METHODS Ten patients with rHGG were treated biweekly with bevacizumab/irinotecan (BEV/IR). MR images and dynamic (18)F-FET PET scans were obtained at baseline and at follow-up after the start of treatment (median 4.9 weeks). Using MRI treatment response was evaluated according to RANO (Response Assessment in Neuro-Oncology) criteria. For (18)F-FET PET evaluation, a reduction >45 % of the metabolically active tumour volume was considered as a treatment response, with the metabolically active tumour being defined as a tumour-to-brain ratio (TBR) of ≥1.6. The results of the treatment assessments were related to progression-free survival (PFS) and overall survival (OS). For further evaluation of PET data, maximum and mean TBR were calculated using region-of-interest analysis at baseline and at follow-up. Additionally, (18)F-FET uptake kinetic studies were performed at baseline and at follow-up in all patients. Time-activity curves were generated and the times to peak (TTP) uptake (in minutes from the beginning of the dynamic acquisition to the maximum uptake) were calculated. RESULTS At follow-up, MRI showed a complete response according to RANO criteria in one of the ten patients (10 %), a partial response in five patients (50 %), and stable disease in four patients (40 %). Thus, MRI did not detect tumour progression. In contrast, (18)F-FET PET revealed six metabolic responders (60 %) and four nonresponders (40 %). In the univariate survival analyses, a response detected by (18)F-FET PET predicted a significantly longer PFS (median PFS, 9 vs. 3 months; P = 0.001) and OS (median OS 23.0 months vs. 3.5 months; P = 0.001). Furthermore, in four patients (40 %), diagnosis according to RANO criteria and by (18)F-FET PET was discordant. In these patients, PET was able to detect tumour progression earlier than MRI (median time benefit 10.5 weeks; range 6-12 weeks). At baseline and at follow-up, in nonresponders TTP was significantly shorter than in responders (baseline TTP 10 ± 8 min vs. 35 ± 9 min; P = 0.002; follow-up TTP 23 ± 9 min vs. 39 ± 8 min; P = 0.02). Additionally, at baseline a kinetic pattern characterized by an early peak of (18)F-FET uptake followed by a constant descent was more frequently observed in the nonresponders (P = 0.018). CONCLUSION Both standard and kinetic imaging parameters derived from(18)F-FET PET seem to predict BEV/IR treatment failure and thus contribute important additional information for clinical management over and above the information obtained by MRI response assessment based on RANO criteria.
Collapse
|
36
|
Galldiks N, Filss CP, Goldbrunner R, Langen KJ. Discrepant MR and [(18)F]Fluoroethyl-L-Tyrosine PET Imaging Findings in a Patient with Bevacizumab Failure. Case Rep Oncol 2012; 5:490-4. [PMID: 23271997 PMCID: PMC3529567 DOI: 10.1159/000342480] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antiangiogenic treatment using bevacizumab may cause difficulties in distinguishing between antivascular and true antitumor effects when using MRI response criteria based on changes of contrast enhancement (i.e., Macdonald criteria). Furthermore, more precise tumor response assessment criteria (i.e., RANO criteria), which incorporate nonenhancing T2/FLAIR sequences into Macdonald criteria, may be influenced by other causes of T2/FLAIR hyperintensity (e.g., radiation-induced gliosis). The authors present discrepant MR and [18F]fluoroethyl-L-tyrosine PET imaging findings in a patient with bevacizumab treatment failure.
Collapse
Affiliation(s)
- Norbert Galldiks
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich, Juelich ; Departments of Neurology, University Hospital Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
37
|
Lu KV, Chang JP, Parachoniak CA, Pandika MM, Aghi MK, Meyronet D, Isachenko N, Fouse SD, Phillips JJ, Cheresh DA, Park M, Bergers G. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 2012; 22:21-35. [PMID: 22789536 PMCID: PMC4068350 DOI: 10.1016/j.ccr.2012.05.037] [Citation(s) in RCA: 440] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 04/08/2012] [Accepted: 05/31/2012] [Indexed: 01/09/2023]
Abstract
Inhibition of VEGF signaling leads to a proinvasive phenotype in mouse models of glioblastoma multiforme (GBM) and in a subset of GBM patients treated with bevacizumab. Here, we demonstrate that vascular endothelial growth factor (VEGF) directly and negatively regulates tumor cell invasion through enhanced recruitment of the protein tyrosine phosphatase 1B (PTP1B) to a MET/VEGFR2 heterocomplex, thereby suppressing HGF-dependent MET phosphorylation and tumor cell migration. Consequently, VEGF blockade restores and increases MET activity in GBM cells in a hypoxia-independent manner, while inducing a program reminiscent of epithelial-to-mesenchymal transition highlighted by a T-cadherin to N-cadherin switch and enhanced mesenchymal features. Inhibition of MET in GBM mouse models blocks mesenchymal transition and invasion provoked by VEGF ablation, resulting in substantial survival benefit.
Collapse
Affiliation(s)
- Kan V. Lu
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Jeffrey P. Chang
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Christine A. Parachoniak
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Melissa M. Pandika
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Manish K. Aghi
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- UCSF Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - David Meyronet
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Nadezda Isachenko
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Shaun D. Fouse
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - Joanna J. Phillips
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- UCSF Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
| | - David A. Cheresh
- Department of Pathology and Moore’s UCSD Cancer Center, University of California, San Diego, California 92093, USA
| | - Morag Park
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Gabriele Bergers
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Anatomy, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Brain Tumor Research Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- UCSF Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, California 94143, USA
- Correspondence should be addressed to: University of California, San Francisco (UCSF) Helen Diller Family Cancer Research Center Department of Neurological Surgery 1450 3rd Street San Francisco, California 94143, USA Telephone: 415-476-6786 Fax: 415-476-0388
| |
Collapse
|
38
|
Salmaggi A, Gaviani P, Botturi A, Lamperti E, Simonetti G, Ferrari D, Silvani A. Bevacizumab at recurrence in high-grade glioma. Neurol Sci 2012; 32 Suppl 2:S251-3. [PMID: 21987287 DOI: 10.1007/s10072-011-0799-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bevacizumab has been introduced in the management of high-grade gliomas after preliminary studies that showed an acceptable safety and a marked increase in clinico-radiological responses in comparison with second-line chemotherapy. The objective is to synthetically review the present use of bevacizumab--alone or in combination--in the context of recurrent high-grade glioma and highlight the future developments. The methodology of this study is to analyse and discuss relevant literature studies using bevacizumab in recurrent high-grade glioma. Bevacizumab may be used as single-agent therapy in recurrent high-grade glioma, with good clinico-radiological responses having little effect on survival. The open questions and developments include new MRI criteria for evaluation of response to anti-angiogenic agents, the identification of putative factors predicting response/failure of bevacizumab and the introduction of bevacizumab in first-line management of high-grade glioma.
Collapse
Affiliation(s)
- Andrea Salmaggi
- Neuro-Oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Radbruch A, Lutz K, Wiestler B, Bäumer P, Heiland S, Wick W, Bendszus M. Relevance of T2 signal changes in the assessment of progression of glioblastoma according to the Response Assessment in Neurooncology criteria. Neuro Oncol 2011; 14:222-9. [PMID: 22146386 DOI: 10.1093/neuonc/nor200] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND According to the Response Assessment in Neurooncology (RANO) criteria, significant nonenhancing signal increase in T2-weighted images qualifies for progression in high-grade glioma (T2-progress), even if there is no change in the contrast-enhancing tumor portion. The purpose of this retrospective study was to assess the frequency of isolated T2-progress and its predictive value on subsequent T1-progress, as determined by a T2 signal increase of 15% or 25%, respectively. The frequency of T2-progress was correlated with antiangiogenic therapy. PATIENTS AND METHODS MRI follow-up examinations (n = 777) of 144 patients with histologically proven glioblastoma were assessed for contrast-enhanced T1 and T2-weighted images. Examinations were classified as T1-progress, T2-progress with 15% or 25% T2-signal increase, stable disease, or partial or complete response. RESULTS Thirty-five examinations revealed exclusive T2-progress using the 15% criterion, and only 2 examinations qualified for the 25% criterion; 61.8% of the scans presenting T2-progress and 31.5% of the scans presenting stable disease revealed T1-progress in the next follow-up examination. The χ(2) test showed a highly significant correlation (P < .001) between T2-progress, with the 15% criterion and subsequent T1-progress. No correlation between antiangiogenic therapy and T2-progress was shown. CONCLUSION Tumor progression, as determined by both contrast-enhanced T1 and T2 sequences is more frequently diagnosed than when considering only contrast-enhanced T1 sequences. Definition of T2-progress by a 15% T2-signal increase criterion is superior to a 25% criterion. The missing correlation of T2-progress and antiangiogenic therapy supports the hypothesis of T2-progress as part of the natural course of the tumor disease.
Collapse
Affiliation(s)
- Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, Heidelberg 69120, Germany.
| | | | | | | | | | | | | |
Collapse
|
41
|
Uncommon Presentations of Uncommon Conditions. J Neuroophthalmol 2011; 31:297-8. [DOI: 10.1097/wno.0b013e318235f553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Hattingen E, Jurcoane A, Bähr O, Rieger J, Magerkurth J, Anti S, Steinbach JP, Pilatus U. Bevacizumab impairs oxidative energy metabolism and shows antitumoral effects in recurrent glioblastomas: a 31P/1H MRSI and quantitative magnetic resonance imaging study. Neuro Oncol 2011; 13:1349-63. [PMID: 21890539 DOI: 10.1093/neuonc/nor132] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bevacizumab shows unprecedented rates of response in recurrent glioblastomas (GBM), but the detailed mechanisms are still unclear. We employed in vivo magnetic resonance spectroscopic imaging (MRSI) and quantitative magnetic resonance imaging to investigate whether bevacizumab alters oxygen and energy metabolism and whether this effect has antitumoral activity in recurrent GBM. (31)P and (1)H MRSI, apparent diffusion coefficient (ADC), and high-resolution T2 and T2' mapping (indirect marker of oxygen extraction) were investigated in 16 patients with recurrent GBM at 3 Tesla before and 1.5-2 months after initiation of therapy with bevacizumab. Changes of metabolite concentrations and of the quantitative values in the tumor and normal appearing brain tissue were calculated. The Wilcoxon signed-ranks test was used to evaluate differences for tumor/edema versus control as well as changes before versus after commencement of therapy. Survival analyses were performed for significant parameters. Tumor T2', pH, ADC, and T2 decreased significantly in patients responding to bevacizumab therapy (n = 10). Patients with at least 25% T2' decrease during treatment showed longer progression-free and overall survival durations. Levels of high-energy metabolites were lower at baseline; these persisted under therapy. Glycerophosphoethanolamine as catabolic phospholipid metabolite increased in responders. The MRSI data support the hypothesis that bevacizumab induces relative tumor hypoxia (T2' decrease) and affects energy homeostasis in recurrent GBM, suggesting that bevacizumab impairs vascular function. The antiangiogenic effect of bevacizumab is predictive of better outcome and seems to induce antitumoral activity in the responding GBMs.
Collapse
Affiliation(s)
- Elke Hattingen
- Goethe University Frankfurt, Institute of Neuroradiology, Schleusenweg 2-16 (Haus 95), 60528 Frankfurt/M, Germany.
| | | | | | | | | | | | | | | |
Collapse
|