301
|
Cho JM, Colen CB, Li G, Wang VY, Dahdaleh N, Choy W, Yang I, Smith ZA. Neurosurgery Concepts: Key perspectives on Traumatic Brain Injury, New Treatments for Glioblastoma, Hemicraniectomy for Extensive Middle-Cerebral-Artery Stroke, Minimally Invasive Spine Surgery and Lumbar Epidural Injections for Radiculopathy. Surg Neurol Int 2015; 6:98. [PMID: 26110080 PMCID: PMC4466788 DOI: 10.4103/2152-7806.158374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 11/15/2022] Open
Affiliation(s)
- Jin Mo Cho
- Department of Neurological Surgery, Ajou University School of Medicine, Suwon, South Korea
| | - Chaim B Colen
- Department of Neurological Surgery, Beaumont Hospital, Grosse Pointe, MI, USA
| | - Gordon Li
- Department of Neurological Surgery, Stanford School of Medicine, San Francisco, CA, USA
| | - Vincent Yat Wang
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Nader Dahdaleh
- Department of Neurological Surgery, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Winward Choy
- Department of Neurological Surgery, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Isaac Yang
- Department of Neurological Surgery, David Geffen School of Medicine at University of California Los Angeles, CA, USA
| | - Zachary A Smith
- Department of Neurological Surgery, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| |
Collapse
|
302
|
Movement of magnetic nanoparticles in brain tissue: mechanisms and impact on normal neuronal function. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1821-9. [PMID: 26115639 DOI: 10.1016/j.nano.2015.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 06/01/2015] [Accepted: 06/09/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Magnetic nanoparticles (MNPs) have been used as effective vehicles for targeted delivery of theranostic agents in the brain. The advantage of magnetic targeting lies in the ability to control the concentration and distribution of therapy to a desired target region using external driving magnets. In this study, we investigated the behavior and safety of MNP motion in brain tissue. We found that MNPs move and form nanoparticle chains in the presence of a uniform magnetic field, and that this chaining is influenced by the applied magnetic field intensity and the concentration of MNPs in the tissue. Using electrophysiology recordings, immunohistochemistry and fluorescent imaging we assessed the functional health of neurons and neural circuits and found no adverse effects associated with MNP motion through brain tissue. FROM THE CLINICAL EDITOR Much research has been done to test the use of nanocarriers for gaining access across the blood brain barrier (BBB). In this respect, magnetic nanoparticles (MNPs) are one of the most studied candidates. Nonetheless, the behavior and safety of MNP once inside brain tissue remains unknown. In this article, the authors thus studied this very important subject.
Collapse
|
303
|
Galle JO, McDonald MW, Simoneaux V, Buchsbaum JC. Reirradiation with Proton Therapy for Recurrent Gliomas. Int J Part Ther 2015. [DOI: 10.14338/theijpt-14-00029.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
304
|
Hasan S, Chen E, Lanciano R, Yang J, Hanlon A, Lamond J, Arrigo S, Ding W, Mikhail M, Ghaneie A, Brady L. Salvage Fractionated Stereotactic Radiotherapy with or without Chemotherapy and Immunotherapy for Recurrent Glioblastoma Multiforme: A Single Institution Experience. Front Oncol 2015; 5:106. [PMID: 26029663 PMCID: PMC4432688 DOI: 10.3389/fonc.2015.00106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/21/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The current standard of care for salvage treatment of glioblastoma multiforme (GBM) is gross total resection and adjuvant chemoradiation for operable patients. Limited evidence exists to suggest that any particular treatment modality improves survival for recurrent GBM, especially if inoperable. We report our experience with fractionated stereotactic radiotherapy (fSRT) with and without chemo/immunotherapy, identifying prognostic factors associated with prolonged survival. METHODS From 2007 to 2014, 19 patients between 29 and 78 years old (median 55) with recurrent GBM following resection and chemoradiation for their initial tumor, received 18-35 Gy (median 25) in three to five fractions via CyberKnife fSRT. Clinical target volume (CTV) ranged from 0.9 to 152 cc. Sixteen patients received adjuvant systemic therapy with bevacizumab (BEV), temozolomide (TMZ), anti-epidermal growth factor receptor (125)I-mAb 425, or some combination thereof. RESULTS The median overall survival (OS) from date of recurrence was 8 months (2.5-61) and 5.3 months (0.6-58) from the end of fSRT. The OS at 6 and 12 months was 47 and 32%, respectively. Three of 19 patients were alive at the time of this review at 20, 49, and 58 months from completion of fSRT. Hazard ratios for survival indicated that patients with a frontal lobe tumor, adjuvant treatment with either BEV or TMZ, time to first recurrence >16 months, CTV <36 cc, recursive partitioning analysis <5, and Eastern Cooperative Oncology Group performance status <2 were all associated with improved survival (P < 0.05). There was no evidence of radionecrosis for any patient. CONCLUSION Radiation Therapy Oncology Group (RTOG) 1205 will establish the role of re-irradiation for recurrent GBM, however our study suggests that CyberKnife with chemotherapy can be safely delivered, and is most effective in patients with smaller frontal lobe tumors, good performance status, or long interval from diagnosis.
Collapse
Affiliation(s)
- Shaakir Hasan
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Eda Chen
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Rachelle Lanciano
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Jun Yang
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Alex Hanlon
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Nursing, University of Pennsylvania , Philadelphia, PA , USA
| | - John Lamond
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Stephen Arrigo
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - William Ding
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Michael Mikhail
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Arezoo Ghaneie
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Luther Brady
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| |
Collapse
|
305
|
Quirk BJ, Brandal G, Donlon S, Vera JC, Mang TS, Foy AB, Lew SM, Girotti AW, Jogal S, LaViolette PS, Connelly JM, Whelan HT. Photodynamic therapy (PDT) for malignant brain tumors--where do we stand? Photodiagnosis Photodyn Ther 2015; 12:530-44. [PMID: 25960361 DOI: 10.1016/j.pdpdt.2015.04.009] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 11/16/2022]
Abstract
INTRODUCTION What is the current status of photodynamic therapy (PDT) with regard to treating malignant brain tumors? Despite several decades of effort, PDT has yet to achieve standard of care. PURPOSE The questions we wish to answer are: where are we clinically with PDT, why is it not standard of care, and what is being done in clinical trials to get us there. METHOD Rather than a meta-analysis or comprehensive review, our review focuses on who the major research groups are, what their approaches to the problem are, and how their results compare to standard of care. Secondary questions include what the effective depth of light penetration is, and how deep can we expect to kill tumor cells. CURRENT RESULTS A measurable degree of necrosis is seen to a depth of about 5mm. Cavitary PDT with hematoporphyrin derivative (HpD) results are encouraging, but need an adequate Phase III trial. Talaporfin with cavitary light application appears promising, although only a small case series has been reported. Foscan for fluorescence guided resection (FGR) plus intraoperative cavitary PDT results were improved over controls, but are poor compared to other groups. 5-Aminolevulinic acid-FGR plus postop cavitary HpD PDT show improvement over controls, but the comparison to standard of care is still poor. CONCLUSION Continued research in PDT will determine whether the advances shown will mitigate morbidity and mortality, but certainly the potential for this modality to revolutionize the treatment of brain tumors remains. The various uses for PDT in clinical practice should be pursued.
Collapse
Affiliation(s)
- Brendan J Quirk
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Garth Brandal
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Steven Donlon
- Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Thomas S Mang
- Department of Oral and Maxillofacial Surgery, University at Buffalo, Buffalo, NY, United States
| | - Andrew B Foy
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sean M Lew
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sachin Jogal
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Peter S LaViolette
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M Connelly
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Harry T Whelan
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
306
|
Le Rhun E, Taillibert S, Chamberlain MC. Anaplastic glioma: current treatment and management. Expert Rev Neurother 2015; 15:601-20. [PMID: 25936680 DOI: 10.1586/14737175.2015.1042455] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anaplastic glioma (AG) is divided into three morphology-based groups (anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic oligoastrocytoma) as well as three molecular groups (glioma-CpG island methylation phenotype [G-CIMP] negative, G-CIMP positive non-1p19q codeleted tumors and G-CIMP positive codeleted tumors). The RTOG 9402 and EORTC 26951 trials established radiotherapy plus (procarbazine, lomustine, vincristine) chemotherapy as the standard of care in 1p/19q codeleted AG. Uni- or non-codeleted AG are currently best treated with radiotherapy only or alkylator-based chemotherapy only as determined by the NOA-04 trial. Maturation of NOA-04 and results of the currently accruing studies, CODEL (for codeleted AG) and CATNON (for uni or non-codeleted AG), will likely refine current up-front treatment recommendations for AG.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, France
| | | | | |
Collapse
|
307
|
Sun M, Wang M, Chen M, Dagnaes-Hansen F, Le DQS, Baatrup A, Horsman MR, Kjems J, Bünger CE. A tissue-engineered therapeutic device inhibits tumor growth in vitro and in vivo. Acta Biomater 2015; 18:21-9. [PMID: 25686557 DOI: 10.1016/j.actbio.2015.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/20/2015] [Accepted: 02/03/2015] [Indexed: 01/13/2023]
Abstract
Bone metastasis is one of the leading causes of death in breast cancer patients. The current treatment is performed as a palliative therapy and the adverse side effects can compromise the patients' quality of life. In order to both effectively treat bone metastasis and avoid the limitation of current strategies, we have invented a drug eluting scaffold with clay matrix release doxorubicin (DESCLAYMR_DOX) to mechanically support the structure after resecting the metastatic tissue while also releasing the anticancer drug doxorubicin which supplements growth inhibition and elimination of the remaining tumor cells. We have previously demonstrated that this device has the capacity to regenerate the bone and provide sustained release of the anticancer drug in vitro. In this study, we focus on the ability of the device to inhibit cancer cell growth in vitro as well as in vivo. Drug-release kinetics was investigated and the cell viability test showed that the tumor inhibitory effect is sustained for up to 4weeks in vitro. Subcutaneous implantation of DESCLAYMR_DOX in athymic mice resulted in significant growth inhibition of human tumor xenografts of breast origin and decelerated multi-organ metastasis formation. Fluorescence images, visualizing doxorubicin, showed a sustained drug release from the DESCLAYMR device in vivo. Furthermore, local use of DESCLAYMR_DOX implantation reduced the incidence of doxorubicin's cardio-toxicity. These results suggest that DESCLAYMR_DOX can be used in reconstructive surgery to support the structure after bone tumor resection and facilitate a sustained release of anticancer drugs in order to prevent tumor recurrence.
Collapse
|
308
|
Chaichana KL, Kone L, Bettegowda C, Weingart JD, Olivi A, Lim M, Quinones-Hinojosa A, Gallia GL, Brem H. Risk of surgical site infection in 401 consecutive patients with glioblastoma with and without carmustine wafer implantation. Neurol Res 2015; 37:717-26. [PMID: 25916669 DOI: 10.1179/1743132815y.0000000042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Patients with glioblastoma (GBM) have an inherently shortened survival because of their disease. It has been recently shown that carmustine wafers in addition to other therapies (surgery, temozolomide, and radiation) can further extend survival. There is concern, however, that these therapies may increase infection risk. The goals of this study were to calculate the incidence of postoperative infection, evaluate if carmustine wafers changes the risk of infection and identify factors independently associated with an infection following GBM surgery. METHODS All patients who underwent non-biopsy, surgical resection of an intracranial GBM from 2007 to 2011 at a single institution were retrospectively reviewed. Stepwise multivariate proportional hazards regression analysis was used to identify factors associated with infection, including the use of carmustine wafers. Variables with P < 0.05 were considered statistically significant. RESULTS Four hundred and one patients underwent resection of an intracranial GBM during the reviewed period, and 21 (5%) patients developed an infection at a median time of 40 [28-286] days following surgery. The incidence of infection was not higher in patients who had carmustine wafers, and this remained true in multivariate analyses to account for differences in treatment cohorts. The factors that remained significantly associated with an increased risk of infection were prior surgery [RR (95% CI); 2.026 (1.473-4.428), P = 0.01], diabetes mellitus [RR (95% CI); 6.090 (1.380-9.354)], P = 0.02], and increasing duration of hospital stay [RR (95% CI); 1.048 (1.006-1.078); P = 0.02], where the greatest risk occurred with hospital stays > 5 days [RR (95% CI); 3.904 (1.003-11.620), P = 0.05]. DISCUSSION These findings may help guide treatment regimens aimed at minimizing infection for patients with GBM.
Collapse
|
309
|
Abstract
The treatment of glial brain tumors begins with surgery, and standard adjuvant treatment at the end of the past millennium for high-grade glioma and high-risk low-grade glioma was radiotherapy and chemotherapy was given at recurrence. However, over the past 10 years much has changed regarding the role of chemotherapy in gliomas and it is now clear that chemotherapy has a role in the treatment of almost all newly diagnosed diffuse gliomas (WHO grade II-IV). This is the result of several prospective studies that showed survival benefit after combined chemoradiotherapy with temozolomide in glioblastoma (WHO grade IV) or after procarbazine, CCNU (lomustine) and vincristine chemotherapy in diffuse low-grade (WHO grade II) and anaplastic (WHO grade III) glioma. The current standard of treatment for diffuse gliomas is described in this overview and in addition some attention is given to targeted therapies.
Collapse
Affiliation(s)
- Walter Taal
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Jacoline EC Bromberg
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Martin J van den Bent
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| |
Collapse
|
310
|
Chowdhary SA, Ryken T, Newton HB. Survival outcomes and safety of carmustine wafers in the treatment of high-grade gliomas: a meta-analysis. J Neurooncol 2015; 122:367-82. [PMID: 25630625 PMCID: PMC4368843 DOI: 10.1007/s11060-015-1724-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 01/19/2015] [Indexed: 11/24/2022]
Abstract
Carmustine wafers (CW; Gliadel(®) wafers) are approved to treat newly-diagnosed high-grade glioma (HGG) and recurrent glioblastoma. Widespread use has been limited for several reasons, including concern that their use may preclude enrollment in subsequent clinical trials due to uncertainty about confounding of results and potential toxicities. This meta-analysis estimated survival following treatment with CW for HGG. A literature search identified relevant studies. Overall survival (OS), median survival, and adverse events (AEs) were summarized. Analysis of variance evaluated effects of treatment (CW vs non-CW) and diagnosis (new vs recurrent) on median survival. The analysis included 62 publications, which reported data for 60 studies (CW: n = 3,162; non-CW: n = 1,736). For newly-diagnosed HGG, 1-year OS was 67 % with CW and 48 % without; 2-year OS was 26 and 15 %, respectively; median survival was 16.4 ± 21.6 months and 13.1 ± 29.9 months, respectively. For recurrent HGG, 1-year OS was 37 % with CW and 34 % without; 2-year OS was 15 and 12 %, respectively; median survival was 9.7 ± 20.9 months and 8.6 ± 22.6 months, respectively. Effects of treatment (longer median survival with CW than without; P = 0.043) and diagnosis (longer median survival for newly-diagnosed HGG than recurrent; P < 0.001) on median survival were significant, with no significant treatment-by-diagnosis interaction (P = 0.620). The most common AE associated with wafer removal was surgical site infection (SSI); the most common AEs for repeat surgery were mass effect, SSI, hydrocephalus, cysts in resection cavity, acute hematoma, wound healing complications, and brain necrosis. These data may be useful in the context of utilizing CW in HGG management, and in designing future clinical trials to allow CW-treated patients to participate in experimental protocols.
Collapse
Affiliation(s)
- Sajeel A. Chowdhary
- Department of Neuro-Oncology, Florida Hospital Cancer Institute, 2501 N. Orange Avenue, Suite 286, Orlando, FL 32804 USA
| | - Timothy Ryken
- Department of Neurosurgery, Iowa Spine and Brain Institute, 2710 St. Francis Drive, Waterloo, IA 50702 USA
| | - Herbert B. Newton
- Departments of Neurology, Neurosurgery, and Oncology, Wexner Medical Center at the Ohio State University and James Cancer Hospital, M410-B Starling-Loving Hall, 320 West 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
311
|
Abstract
Neurosurgical oncology for intrinsic glioma is evolving rapidly. It must be patient-centered, consultant-led and research-orientated. The value of specialist neurosurgical engagement is becoming more widely recognized. Detailed evaluation tailored to each patient is essential before the surgical admission, in conjunction with clinical oncology input. Medical optimization, collation of magnetic resonance datasets for preoperative planning and providing an informed explanation of the proposed management and its alternatives are all part of the neurosurgeon's remit. Meticulous microsurgical technique during surgery utilizing modern neuronavigation and physiological monitoring are integral components of the specialist armamentarium. A clear understanding of the rationale for surgical intervention, including its place alongside radiotherapy and chemotherapy, informs surgical decision-making. Recognition and understanding of these issues are driving the evolution of neurosurgical management of high-grade glioma. New challenges are emerging and need to be critically evaluated in robustly designed clinical trials.
Collapse
Affiliation(s)
- Colin Watts
- University of Cambridge Department of Clinical Neurosciences, Division of Neurosurgery, Box 167 Addenbrookes Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
312
|
Warren KE. Novel therapeutic delivery approaches in development for pediatric gliomas. CNS Oncol 2015; 2:427-35. [PMID: 24511389 DOI: 10.2217/cns.13.37] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pediatric gliomas are a heterogeneous group of diseases, ranging from relatively benign pilocytic astrocytomas with >90% 5-year survival, to glioblastomas and diffuse intrinsic pontine gliomas with <20% 5-year survival. Chemotherapy plays an important role in the management of these tumors, particularly in low-grade gliomas, but many high-grade tumors are resistant to chemotherapy. A major obstacle and contributor to this resistance is the blood–brain barrier, which protects the CNS by limiting entry of potential toxins, including chemotherapeutic agents. Several novel delivery approaches that circumvent the blood–brain barrier have been developed, including some currently in clinical trials. This review describes several of these novel approaches to improve delivery of chemotherapeutic agents to their site of action at the tumor, in attempts to improve their efficacy and the prognosis of children with this disease.
Collapse
|
313
|
Nanomedicine to overcome radioresistance in glioblastoma stem-like cells and surviving clones. Trends Pharmacol Sci 2015; 36:236-52. [PMID: 25799457 DOI: 10.1016/j.tips.2015.02.002] [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] [Received: 12/10/2014] [Revised: 01/28/2015] [Accepted: 02/03/2015] [Indexed: 12/14/2022]
Abstract
Radiotherapy is one of the standard treatments for glioblastoma, but its effectiveness often encounters the phenomenon of radioresistance. This resistance was recently attributed to distinct cell contingents known as glioblastoma stem-like cells (GSCs) and dominant clones. It is characterized in particular by the activation of signaling pathways and DNA repair mechanisms. Recent advances in the field of nanomedicine offer new possibilities for radiosensitizing these cell populations. Several strategies have been developed in this direction, the first consisting of encapsulating a contrast agent or synthesizing metal-based nanocarriers to concentrate the dose gradient at the level of the target tissue. In the second strategy the physicochemical properties of the vectors are used to encapsulate a wide range of pharmacological agents which act in synergy with the ionizing radiation to destroy the cancerous cells. This review reports on the various molecular anomalies present in GSCs and the predominant role of nanomedicines in the development of radiosensitization strategies.
Collapse
|
314
|
Walker AJ, Ruzevick J, Malayeri AA, Rigamonti D, Lim M, Redmond KJ, Kleinberg L. Postradiation imaging changes in the CNS: how can we differentiate between treatment effect and disease progression? Future Oncol 2015; 10:1277-97. [PMID: 24947265 DOI: 10.2217/fon.13.271] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A familiar challenge for neuroradiologists and neuro-oncologists is differentiating between radiation treatment effect and disease progression in the CNS. Both entities are characterized by an increase in contrast enhancement on MRI and present with similar clinical signs and symptoms that may occur either in close temporal proximity to the treatment or later in the disease course. When radiation-related imaging changes or clinical deterioration are mistaken for disease progression, patients may be subject to unnecessary surgery and/or a change from otherwise effective therapy. Similarly, when disease progression is mistaken for treatment effect, a potentially ineffective therapy may be continued in the face of progressive disease. Here we describe the three types of radiation injury to the brain based on the time to development of signs and symptoms--acute, subacute and late--and then review specific imaging changes after intensity-modulated radiation therapy, stereotactic radiosurgery and brachytherapy. We provide an overview of these phenomena in the treatment of a wide range of malignant and benign CNS illnesses. Finally, we review the published data regarding imaging techniques under investigation to address this well-known problem.
Collapse
Affiliation(s)
- Amanda J Walker
- Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
315
|
Neagu MR, Huang RY, Reardon DA, Wen PY. How treatment monitoring is influencing treatment decisions in glioblastomas. Curr Treat Options Neurol 2015; 17:343. [PMID: 25749847 DOI: 10.1007/s11940-015-0343-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Glioblastoma (GBM), the most common malignant primary tumor in adults, carries a dismal prognosis with an average median survival of 14-16 months. The current standard of care for newly diagnosed GBM consists of maximal safe resection followed by fractionated radiotherapy combined with concurrent temozolomide and 6 to 12 cycles of adjuvant temozolomide. The determination of treatment response and clinical decision-making in the treatment of GBM depends on accurate radiographic assessment. Differentiating treatment response from tumor progression is challenging and combines long-term follow-up using standard MRI, with assessing clinical status and corticosteroid dependency. At progression, bevacizumab is the mainstay of treatment. Incorporation of antiangiogenic therapies leads to rapid blood-brain barrier normalization with remarkable radiographic response often not accompanied by the expected survival benefit, further complicating imaging assessment. Improved radiographic interpretation criteria, such as the Response Assessment in Neuro-Oncology (RANO) criteria, incorporate non-enhancing disease but still fall short of definitely distinguishing tumor progression, pseudoresponse, and pseudoprogression. With new evolving treatment modalities for this devastating disease, advanced imaging modalities are increasingly becoming part of routine clinical care in a field where neuroimaging has such essential role in guiding treatment decisions and defining clinical trial eligibility and efficacy.
Collapse
Affiliation(s)
- Martha R Neagu
- Dana Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA
| | | | | | | |
Collapse
|
316
|
Zembko I, Ahmed I, Farooq A, Dail J, Tawari P, Wang W, Mcconville C. Development of Disulfiram-Loaded Poly(Lactic-co-Glycolic Acid) Wafers for the Localised Treatment of Glioblastoma Multiforme: A Comparison of Manufacturing Techniques. J Pharm Sci 2015; 104:1076-86. [DOI: 10.1002/jps.24304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/19/2014] [Accepted: 11/19/2014] [Indexed: 11/06/2022]
|
317
|
Tumor resection with carmustine wafer placement as salvage therapy after local failure of radiosurgery for brain metastasis. J Clin Neurosci 2015; 22:561-5. [PMID: 25560387 PMCID: PMC10373436 DOI: 10.1016/j.jocn.2014.08.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 08/14/2014] [Accepted: 08/16/2014] [Indexed: 11/17/2022]
Abstract
Prolonged survival in brain metastasis patients increases recurrence rates and places added importance on salvage therapies. Research examining carmustine polymer wafers as an adjuvant therapy for brain metastasis is limited. We present a single institution retrospective series documenting the use of BCNU wafers placed in the cavity of resected recurrent brain metastases that had failed prior stereotactic radiosurgery (SRS). Between February 2002 and April 2013, a total of 31 patients with brain metastases failed SRS and underwent resection with intracavitary placement of carmustine wafers. Clinical outcomes including local control, survival, cause of death, and toxicity were determined from electronic medical records. Kaplan-Meier analysis was performed to assess local control and survival. Imaging features were reviewed and described for patients with serial post-operative follow-up imaging examinations over time. Overall survival at 6 months and 12 months was 63% and 36%, respectively. Fourteen of 31 patients (45%) died from neurologic causes. Local control within the resection cavity was 87% and 70% at 6 and 12 months, respectively. Five patients (16%) underwent further salvage therapy following carmustine wafer placement after local failure. Resection cavities of all six patients with follow-up imaging showed linear peripheral enhancement. Pericavity and wafer enhancement was present as early as the same day as surgery and persisted in all cases to 6 months or longer. Carmustine polymer wafers are an effective salvage treatment following resection of a brain metastasis that has failed prior SRS. For patients with successful local control after wafer implantation, linear enhancement at the cavity is common.
Collapse
|
318
|
van Tellingen O, Yetkin-Arik B, de Gooijer M, Wesseling P, Wurdinger T, de Vries H. Overcoming the blood–brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat 2015; 19:1-12. [DOI: 10.1016/j.drup.2015.02.002] [Citation(s) in RCA: 662] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 12/23/2022]
|
319
|
Pavlov V, Page P, Abi-Lahoud G, Nataf F, Dezamis E, Robin A, Varlet P, Turak B, Dhermain F, Domont J, Louvel G, Souillard-Scemama R, Parraga E, Meder JF, Chrétien F, Devaux B, Pallud J. Combining intraoperative carmustine wafers and Stupp regimen in multimodal first-line treatment of primary glioblastomas. Br J Neurosurg 2015; 29:524-31. [PMID: 25724425 DOI: 10.3109/02688697.2015.1012051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The study investigated if intraoperative use of carmustine wafers, particularly in combination with Stupp regimen, is a viable and safe first-line treatment option of glioblastomas. METHODS Eighty-three consecutive adult patients (50 men; mean age 60 years) with newly diagnosed supratentorial primary glioblastomas that underwent surgical resection with intraoperative carmustine wafers implantation (n = 7.1 ± 1.7) were retrospectively studied. RESULTS The median overall survival (OS) was 15.8 months with 56 patients dying over the course of the study. There was no significant association between the number of implanted carmustine wafers and complication rates (four surgical site infections, one death). The OS was significantly longer in Stupp regimen patients (19.5 months) as compared with patients with other postoperative treatments (13 months; p = 0.002). In addition patients with eight or more implanted carmustine wafers survived longer (24.5 months) than patients with seven or less implanted wafers (13 months; p = 0.021). Finally, regardless of the number of carmustine wafers, median OS was significantly longer in patients with a subtotal or total resection (21.5 months) than in patients with a partial resection (13 months; p = 0.011). CONCLUSIONS The intraoperative use of carmustine wafers in combination with Stupp regimen is a viable first-line treatment option of glioblastomas. The prognostic value of this treatment association should be evaluated in a multicenter trial, ideally in a randomized and placebo-controlled one.
Collapse
Affiliation(s)
- Vladislav Pavlov
- a Department of Neurosurgery , Sainte-Anne Hospital , Paris , France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
320
|
Yang G, Wang J, Wang Y, Li L, Guo X, Zhou S. An implantable active-targeting micelle-in-nanofiber device for efficient and safe cancer therapy. ACS NANO 2015; 9:1161-74. [PMID: 25602381 DOI: 10.1021/nn504573u] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanocarriers have attracted broad attention in cancer therapy because of their ability to carry drugs preferentially into cancer tissue, but their application is still limited due to the systemic toxicity and low delivery efficacy of intravenously delivered chemotherapeutics. In this study, we develop a localized drug delivery device with combination of an active-targeting micellar system and implantable polymeric nanofibers. This device is achieved first by the formation of hydrophobic doxorubicin (Dox)-encapsulated active-targeting micelles assembled from a folate-conjugated PCL-PEG copolymer. Then, fabrication of the core-shell polymeric nanofibers is achieved with coaxial electrospinning in which the core region consists of a mixture of poly(vinyl alcohol) and the micelles and the outer shell layer consists of cross-linked gelatin. In contrast to the systematic administration of therapeutics via repeatedly intravenous injections of micelles, this implantable device has these capacities of greatly reducing the drug dose, the frequency of administration and side effect of chemotherapeutic agents while maintaining highly therapeutic efficacy against artificial solid tumors. This micelle-based nanofiber device can be developed toward the next generation of nanomedicine for efficient and safe cancer therapy.
Collapse
Affiliation(s)
- Guang Yang
- Key Laboratory of Advanced Technologies of Material, Minister of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, Sichuan, PR China
| | | | | | | | | | | |
Collapse
|
321
|
Huang RY, Neagu MR, Reardon DA, Wen PY. Pitfalls in the neuroimaging of glioblastoma in the era of antiangiogenic and immuno/targeted therapy - detecting illusive disease, defining response. Front Neurol 2015; 6:33. [PMID: 25755649 PMCID: PMC4337341 DOI: 10.3389/fneur.2015.00033] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/09/2015] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma, the most common malignant primary brain tumor in adults is a devastating diagnosis with an average survival of 14–16 months using the current standard of care treatment. The determination of treatment response and clinical decision making is based on the accuracy of radiographic assessment. Notwithstanding, challenges exist in the neuroimaging evaluation of patients undergoing treatment for malignant glioma. Differentiating treatment response from tumor progression is problematic and currently combines long-term follow-up using standard magnetic resonance imaging (MRI), with clinical status and corticosteroid-dependency assessments. In the clinical trial setting, treatment with gene therapy, vaccines, immunotherapy, and targeted biologicals similarly produces MRI changes mimicking disease progression. A neuroimaging method to clearly distinguish between pseudoprogression and tumor progression has unfortunately not been found to date. With the incorporation of antiangiogenic therapies, a further pitfall in imaging interpretation is pseudoresponse. The Macdonald criteria that correlate tumor burden with contrast-enhanced imaging proved insufficient and misleading in the context of rapid blood–brain barrier normalization following antiangiogenic treatment that is not accompanied by expected survival benefit. Even improved criteria, such as the RANO criteria, which incorporate non-enhancing disease, clinical status, and need for corticosteroid use, fall short of definitively distinguishing tumor progression, pseudoresponse, and pseudoprogression. This review focuses on advanced imaging techniques including perfusion MRI, diffusion MRI, MR spectroscopy, and new positron emission tomography imaging tracers. The relevant image analysis algorithms and interpretation methods of these promising techniques are discussed in the context of determining response and progression during treatment of glioblastoma both in the standard of care and in clinical trial context.
Collapse
Affiliation(s)
- Raymond Y Huang
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center , Boston, MA , USA
| | - Martha R Neagu
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center , Boston, MA , USA
| | - David A Reardon
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center , Boston, MA , USA
| | - Patrick Y Wen
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center , Boston, MA , USA
| |
Collapse
|
322
|
Parrish KE, Sarkaria JN, Elmquist WF. Improving drug delivery to primary and metastatic brain tumors: strategies to overcome the blood-brain barrier. Clin Pharmacol Ther 2015; 97:336-46. [PMID: 25669487 DOI: 10.1002/cpt.71] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022]
Abstract
Brain tumor diagnosis has an extremely poor prognosis, due in part to the blood-brain barrier (BBB) that prevents both early diagnosis and effective drug delivery. The infiltrative nature of primary brain tumors and the presence of micro-metastases lead to tumor cells that reside behind an intact BBB. Recent genomic technologies have identified many genetic mutations present in glioma and other central nervous system (CNS) tumors, and this information has been instrumental in guiding the development of molecularly targeted therapies. However, the majority of these agents are unable to penetrate an intact BBB, leading to one mechanism by which the invasive brain tumor cells effectively escape treatment. The diagnosis and treatment of a brain tumor remains a serious challenge and new therapeutic agents that either penetrate the BBB or disrupt mechanisms that limit brain penetration, such as endothelial efflux transporters or tight junctions, are required in order to improve patient outcomes in this devastating disease.
Collapse
Affiliation(s)
- K E Parrish
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
323
|
Mangraviti A, Tyler B, Brem H. Interstitial chemotherapy for malignant glioma: Future prospects in the era of multimodal therapy. Surg Neurol Int 2015; 6:S78-84. [PMID: 25722936 PMCID: PMC4338488 DOI: 10.4103/2152-7806.151345] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/15/2014] [Indexed: 11/05/2022] Open
Abstract
The advent of interstitial chemotherapy has significantly increased therapeutic options for patients with malignant glioma. Interstitial chemotherapy can deliver high concentrations of chemotherapeutic agents, directly at the site of the brain tumor while bypassing systemic toxicities. Gliadel, a locally implanted polymer that releases the alkylating agent carmustine, given alone and in combination with various other antitumor and resistance modifying therapies, has significantly increased the median survival for patients with malignant glioma. Convection enhanced delivery, a technique used to directly infuse drugs into brain tissue, has shown promise for the delivery of immunotoxins, monoclonal antibodies, and chemotherapeutic agents. Preclinical studies include delivery of chemotherapeutic and immunomodulating agents by polymer and microchips. Interstitial chemotherapy was shown to maximize local efficacy and is an important strategy for the efficacy of any multimodal approach.
Collapse
Affiliation(s)
- Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA ; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA ; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
324
|
Healy AT, Vogelbaum MA. Convection-enhanced drug delivery for gliomas. Surg Neurol Int 2015; 6:S59-67. [PMID: 25722934 PMCID: PMC4338487 DOI: 10.4103/2152-7806.151337] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 10/15/2014] [Indexed: 11/09/2022] Open
Abstract
In spite of aggressive multi-modality treatments, patients diagnosed with anaplastic astrocytoma and glioblastoma continue to display poor median survival. The success of our current conventional and targeted chemotherapies are largely hindered by systemic- and neurotoxicity, as well as poor central nervous system (CNS) penetration. Interstitial drug administration via convection-enhanced delivery (CED) is an alternative that potentially overcomes systemic toxicities and CNS delivery issues by directly bypassing the blood–brain barrier (BBB). This novel approach not only allows for directed administration, but also allows for newer, tumor-selective agents, which would normally be excluded from the CNS due to molecular size alone. To date, randomized trials of CED therapy have yet to definitely show survival advantage as compared with today's standard of care, however, early studies appear to have been limited by “first generation” delivery techniques. Taking into consideration lessons learned from early trials along with decades of research, newer CED technologies and therapeutic agents are emerging, which are reviewed herein.
Collapse
Affiliation(s)
- Andrew T Healy
- Neurosurgical Resident, Department of Neurological Surgery, Director, Center for Translational Therapeutics, Associate Director, Brain Tumor and Neuro-Oncology Center, ND40, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Michael A Vogelbaum
- Department of Neurological Surgery, Director, Center for Translational Therapeutics, Associate Director, Brain Tumor and Neuro-Oncology Center, ND40, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
325
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
326
|
Chamberlain MC. Salvage therapy with lomustine for temozolomide refractory recurrent anaplastic astrocytoma: a retrospective study. J Neurooncol 2015; 122:329-38. [PMID: 25563816 DOI: 10.1007/s11060-014-1714-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/24/2014] [Indexed: 01/06/2023]
Abstract
There is no standard therapy for recurrent anaplastic astrocytoma (AA). Assess response and toxicity of lomustine (CCNU) in recurrent AA following prior surgery, radiotherapy and TMZ in a retrospective case series. Thirty-five adults (18 males; 17 females: median age 42.5 years) with TMZ refractory recurrent AA were treated with lomustine. Seven patients were treated at 1st recurrence and 28 patients were treated at 2nd recurrence. Prior salvage therapy included re-resection in 19, TMZ in 20 and radiotherapy in 7. A cycle of lomustine was defined as 110 mg/m(2) on day 1 only administered once every 6-8 weeks. Success of treatment was defined as progression free survival at 6 months of 40 % or better. Grade 3 or 4 toxicities included anemia (14 patients), constipation (1), fatigue (4), lymphopenia (5), nausea/vomiting (2), neutropenia (8) and thrombocytopenia (10). No grade five toxicities were seen. The median number of cycles of therapy was 3 (range 1-6). Best radiographic response was progressive disease in 14 (40 %), stable disease in 19 (54 %) and partial response in 2 (5.7 %). Median progression free survival (PFS) was 4.5 months (range 1.5-12 months), 6-month PFS was 40 % and 12 month PFS was 11.4 %. Median survival after onset of CCNU was 9.5 months (range 2.5-15 months). Median overall survival was 2.7 years (range 1.7-4.3). In this small retrospective series of patients with recurrent AA refractory to TMZ, lomustine appears to have modest single agent with manageable toxicity. Confirmation in a larger series of similar patients is required.
Collapse
Affiliation(s)
- Marc C Chamberlain
- Division of Neuro-Oncology, Department of Neurology and Neurological Surgery, University of Washington/Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, 825 Eastlake Ave E, MS: G4-940, Seattle, WA, 98109, USA,
| |
Collapse
|
327
|
Abstract
Glioblastoma is the most common adult malignant primary brain tumor. Despite the advances in therapeutic options, survival of patients with glioblastoma remains dismal at 15-18 months. Current standard of care for newly diagnosed glioblastoma is maximal possible safe resection consistent with the preservation of neurologic function followed by concurrent temozolomide with radiation and adjuvant. Treatment options at recurrence include surgical resection with or without the placement of carmustine wafers, re-irradiation and chemotherapeutics such as nitrosoureas (lomustine, carmustine) or bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor (VEGF).
Collapse
Affiliation(s)
- Vyshak Alva Venur
- Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, S73, Cleveland, OH, 44195, USA
| | | | | |
Collapse
|
328
|
Schneider CS, Perez JG, Cheng E, Zhang C, Mastorakos P, Hanes J, Winkles JA, Woodworth GF, Kim AJ. Minimizing the non-specific binding of nanoparticles to the brain enables active targeting of Fn14-positive glioblastoma cells. Biomaterials 2014; 42:42-51. [PMID: 25542792 DOI: 10.1016/j.biomaterials.2014.11.054] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/11/2014] [Accepted: 11/25/2014] [Indexed: 12/31/2022]
Abstract
A major limitation in the treatment of glioblastoma (GBM), the most common and deadly primary brain cancer, is delivery of therapeutics to invading tumor cells outside of the area that is safe for surgical removal. A promising way to target invading GBM cells is via drug-loaded nanoparticles that bind to fibroblast growth factor-inducible 14 (Fn14), thereby potentially improving efficacy and reducing toxicity. However, achieving broad particle distribution and nanoparticle targeting within the brain remains a significant challenge due to the adhesive extracellular matrix (ECM) and clearance mechanisms in the brain. In this work, we developed Fn14 monoclonal antibody-decorated nanoparticles that can efficiently penetrate brain tissue. We show these Fn14-targeted brain tissue penetrating nanoparticles are able to (i) selectively bind to recombinant Fn14 but not brain ECM proteins, (ii) associate with and be internalized by Fn14-positive GBM cells, and (iii) diffuse within brain tissue in a manner similar to non-targeted brain penetrating nanoparticles. In addition, when administered intracranially, Fn14-targeted nanoparticles showed improved tumor cell co-localization in mice bearing human GBM xenografts compared to non-targeted nanoparticles. Minimizing non-specific binding of targeted nanoparticles in the brain may greatly improve the access of particulate delivery systems to remote brain tumor cells and other brain targets.
Collapse
Affiliation(s)
- Craig S Schneider
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jimena G Perez
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Emily Cheng
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Clark Zhang
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 North Broadway Street, Baltimore, MD 21231, USA
| | - Panagiotis Mastorakos
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 North Broadway Street, Baltimore, MD 21231, USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 North Broadway Street, Baltimore, MD 21231, USA
| | - Jeffrey A Winkles
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| |
Collapse
|
329
|
Kim HR, Kim SH, Lee JI, Seol HJ, Nam DH, Kim ST, Park K, Kim JH, Kong DS. Outcome of radiosurgery for recurrent malignant gliomas: assessment of treatment response using relative cerebral blood volume. J Neurooncol 2014; 121:311-8. [PMID: 25488072 DOI: 10.1007/s11060-014-1634-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 10/17/2014] [Indexed: 11/28/2022]
Abstract
Gamma knife radiosurgery (GKS) is efficacious for treating recurrent malignant gliomas as a salvage treatment. However, contrast enhancement alone on MR imaging remains difficult to determine the treatment response following GKS. The purpose of this study was to evaluate the radiosurgical effect for recurrent malignant gliomas and to clarify if relative cerebral blood volume (rCBV) derived from dynamic susceptibility-weighted contrast-enhanced (DSC) perfusion MR imaging could represent the treatment response. Between March 2006 and December 2008, 38 patients underwent GKS for recurrent malignant gliomas. Before and after GKS, DSC perfusion MR imaging datasets were retrospectively reprocessed and regions of interest were drawn around the contrast-enhancing region targeted with GKS. DSC-perfusion MR scans were assessed at a regular interval of two months. Following GKS for the recurrent lesions, MR images showed response (stable disease or partial response) in 26 of 38 patients (68.4 %) at post-GKS 2 months and 18 of 38 patients (47.3 %) at post-GKS 4 months. Initial mean rCBV value was 2.552 (0.586-6.178) at the pre-GKS MRI. In the response group, mean rCBV value was significantly decreased (P < 0.05) at the follow up of 2 and 4 months. However, in the treatment-failure group, mean rCBV value had no significant change. We suggest that GKS is an alternative treatment choice for the recurrent glioma. DSC-perfusion MR images are helpful to predict the treatment response after GKS.
Collapse
Affiliation(s)
- Hong Rye Kim
- Department of Neurosurgery, Konyang University Hospital, Konyang University School of Medicine, Daejeon, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Messali A, Villacorta R, Hay JW. A review of the economic burden of glioblastoma and the cost effectiveness of pharmacologic treatments. PHARMACOECONOMICS 2014; 32:1201-1212. [PMID: 25085219 DOI: 10.1007/s40273-014-0198-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Grade IV glioma (glioblastoma) is one of the most common brain/central nervous system cancers. In 2005, the standard of care for adjuvant treatment was significantly changed with the approval of temozolomide. Carmustine wafers have also gained some popularity. Phase III trials are currently evaluating bevacizumab in conjunction with the standard temozolomide regimen. Despite these recent advances in pharmacotherapy, roughly two-thirds of patients do not survive longer than 2 years after diagnosis. Meanwhile, the costs of treatment are substantial. The goal of this study is to review the clinical, cost-of-illness, and cost-effectiveness literature relevant to treating glioblastoma. Estimates of the economic burden of glioblastoma within different healthcare systems were converted to 2013 US dollars. Temozolomide has demonstrated a 2.5-month increase in overall survival and a 1.9-month increase in progression-free survival, relative to radiotherapy alone. Carmustine wafers have also been shown to increase overall survival by 2.3 months, compared with placebo wafers. Cost-effectiveness studies of temozolomide have produced incremental cost-effectiveness ratios, adjusted to 2013 US dollars, with a range from US$73,586 per quality-adjusted life-year (QALY) (UK National Health Service perspective) to US$105,234 per QALY (US societal perspective). More research is needed to quantify the full societal burden of illness.
Collapse
Affiliation(s)
- Andrew Messali
- Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, Leonard D. Schaeffer Center for Health Policy and Economics, University of Southern California, 3335 S. Figueroa St., Unit A, University Park Campus, UGW-Unit A, Los Angeles, CA, 90089-7273, USA,
| | | | | |
Collapse
|
331
|
23Na-MRI of recurrent glioblastoma multiforme after intraoperative radiotherapy: technical note. Neuroradiology 2014; 57:321-6. [DOI: 10.1007/s00234-014-1468-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/17/2014] [Indexed: 11/25/2022]
|
332
|
Smith SJ, Rahman CV, Clarke PA, Ritchie AA, Gould TW, Ward JH, Shakesheff KM, Grundy RG, Rahman R. Surgical delivery of drug releasing poly(lactic-co-glycolic acid)/poly(ethylene glycol) paste with in vivo effects against glioblastoma. Ann R Coll Surg Engl 2014; 96:495-501. [PMID: 25245726 DOI: 10.1308/003588414x13946184903568] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION The median survival of patients with glioblastoma multiforme (astrocytoma grade 4) remains less than 18 months despite radical surgery, radiotherapy and systemic chemotherapy. Surgical implantation of chemotherapy eluting wafers into the resection cavity has been shown to improve length of survival but the current licensed therapy has several drawbacks. This paper investigates in vivo efficacy of a novel drug eluting paste in glioblastoma. METHODS Poly(lactic-co-glycolic acid)/poly(ethylene glycol) (PLGA/PEG) self-sintering paste was loaded with the chemotherapeutic agent etoposide and delivered surgically into partially resected tumours in a flank murine glioblastoma xenograft model. RESULTS Surgical delivery of the paste was successful and practical, with no toxicity or surgical morbidity to the animals. The paste was retained in the tumour cavity, and preliminary results suggest a useful antitumour and antiangiogenic effect, particularly at higher doses. Bioluminescent imaging was not affected significantly by the presence of the paste in the tumour. CONCLUSIONS Chemotherapy loaded PLGA/PEG paste seems to be a promising technology capable of delivering active drugs into partially resected tumours. The preliminary results of this study suggest efficacy with no toxicity and will lead to larger scale efficacy studies in orthotopic glioblastoma models.
Collapse
|
333
|
Intracranial microcapsule chemotherapy delivery for the localized treatment of rodent metastatic breast adenocarcinoma in the brain. Proc Natl Acad Sci U S A 2014; 111:16071-6. [PMID: 25349381 DOI: 10.1073/pnas.1313420110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Metastases represent the most common brain tumors in adults. Surgical resection alone results in 45% recurrence and is usually accompanied by radiation and chemotherapy. Adequate chemotherapy delivery to the CNS is hindered by the blood-brain barrier. Efforts at delivering chemotherapy locally to gliomas have shown modest increases in survival, likely limited by the infiltrative nature of the tumor. Temozolomide (TMZ) is first-line treatment for gliomas and recurrent brain metastases. Doxorubicin (DOX) is used in treating many types of breast cancer, although its use is limited by severe cardiac toxicity. Intracranially implanted DOX and TMZ microcapsules are compared with systemic administration of the same treatments in a rodent model of breast adenocarcinoma brain metastases. Outcomes were animal survival, quantified drug exposure, and distribution of cleaved caspase 3. Intracranial delivery of TMZ and systemic DOX administration prolong survival more than intracranial DOX or systemic TMZ. Intracranial TMZ generates the more robust induction of apoptotic pathways. We postulate that these differences may be explained by distribution profiles of each drug when administered intracranially: TMZ displays a broader distribution profile than DOX. These microcapsule devices provide a safe, reliable vehicle for intracranial chemotherapy delivery and have the capacity to be efficacious and superior to systemic delivery of chemotherapy. Future work should include strategies to improve the distribution profile. These findings also have broader implications in localized drug delivery to all tissue, because the efficacy of a drug will always be limited by its ability to diffuse into surrounding tissue past its delivery source.
Collapse
|
334
|
Kuramitsu S, Motomura K, Natsume A, Wakabayashi T. Double-edged Sword in the Placement of Carmustine (BCNU) Wafers along the Eloquent Area: A Case Report. NMC Case Rep J 2014; 2:40-45. [PMID: 28663961 PMCID: PMC5364933 DOI: 10.2176/nmccrj.2014-0025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/22/2014] [Indexed: 11/28/2022] Open
Abstract
Although direct Gliadel® wafer implantation into the resection cavity has been shown to significantly improve survival in patients with high-grade gliomas, several complications have been associated with the implantations of Gliadel wafers, including brain edema, healing delay, cerebral spinal fluid leak, intracranial infections, and cyst formation. The brain edema that is associated with Gliadel wafer implantation might result in neurological deficits and significant morbidities and mortalities. In particular, it is not clear if they should be placed in the eloquent areas, such as language areas, motor areas, and areas related to cognitive function, even if these areas contain a remnant tumor. Here, we present a case of profound brain edema along the pyramidal tract due to Gliadel wafer implantation, which resulted in severe neurological deficits. This treatment represents a double-edged sword due to the possibility of severe symptomatic brain edema along the eloquent area, even though Gliadel wafers might be effective in controlling local tumor growth. We should keep in mind that Gliadel wafer placement in eloquent areas may result in severe disadvantages to patients and a loss of their quality of life.
Collapse
Affiliation(s)
- Shunichiro Kuramitsu
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Toshihiko Wakabayashi
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| |
Collapse
|
335
|
Wang C, Tan Z, Louis TA. An Exponential Tilt Mixture Model for Time-to-Event Data to Evaluate Treatment Effect Heterogeneity in Randomized Clinical Trials. BIOMETRICS & BIOSTATISTICS INTERNATIONAL JOURNAL 2014; 1:00006. [PMID: 29546253 PMCID: PMC5849265 DOI: 10.15406/bbij.2014.01.00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Evaluating the effect of a treatment on a time-to-event outcome is the focus of many randomized clinical trials. It is often observed that the treatment effect is heterogeneous, where only a subgroup of the patients may respond to the treatment due to some unknown mechanism such as genetic polymorphism. In this paper, we propose a semiparametric exponential tilt mixture model to estimate the proportion of patients who respond to the treatment and to assess the treatment effect. Our model is a natural extension of parametric mixture models to a semiparametric setting with a time-to-event outcome. We propose a nonparametric maximum likelihood estimation approach for inference and establish related asymptotic properties. Our method is illustrated by a randomized clinical trial on biodegradable polymer-delivered chemotherapy for malignant gliomas patients.
Collapse
Affiliation(s)
- Chi Wang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY 40536, USA
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Zhiqiang Tan
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Thomas A. Louis
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
336
|
Batich KA, Sampson JH. Standard of care and future pharmacological treatment options for malignant glioma: an urgent need for screening and identification of novel tumor-specific antigens. Expert Opin Pharmacother 2014; 15:2047-61. [PMID: 25139628 DOI: 10.1517/14656566.2014.947266] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Malignant gliomas (MGs) represent the most common primary brain tumors in adults, the most deadly of which is grade IV glioblastoma. Patients with glioblastoma undergoing current standard-of-care therapy have a median survival of 12 - 15 months. AREAS COVERED Over the past 25 years, there have been modest advancements in the treatment of MGs. Assessment of therapeutic responses has continued to evolve to account for the increasing number of agents being tested in the clinic. Currently approved therapies for primary tumors have been extended for use in the setting of recurrent disease with modest efficacy. Agents initially approved for recurrent gliomas have begun to demonstrate efficacy against de novo tumors but will ultimately need to be evaluated in future studies for scheduling, timing and dosing relative to chemotherapy. EXPERT OPINION Screening and identification of tumor-specific mutations is critical for the advancement of effective therapy that is both safe and precise for the patient. Two unique antigens found in glioblastoma are currently being employed as targets for immunotherapeutic vaccines, one of which has advanced to Phase III testing. Whole genome sequencing of MGs has yielded two other novel mutations that offer great promise for the development of molecular inhibitors.
Collapse
Affiliation(s)
- Kristen A Batich
- Duke University Medical Center, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery , DUMC Box 3050, 303 Research Drive, 220 Sands Building, Durham, NC 27710 , USA +1 919 684 9041 ; +1 919 684 9045 ;
| | | |
Collapse
|
337
|
Sai K, Zhong MG, Wang J, Chen YS, Mou YG, Ke C, Zhang XH, Yang QY, Lin FH, Guo CC, Chen ZH, Zeng J, Lv YC, Li X, Gao WC, Chen ZP. Safety evaluation of high-dose BCNU-loaded biodegradable implants in Chinese patients with recurrent malignant gliomas. J Neurol Sci 2014; 343:60-5. [PMID: 24874252 DOI: 10.1016/j.jns.2014.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 04/10/2014] [Accepted: 05/12/2014] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Malignant gliomas are common primary brain tumors with dismal prognosis. The blood-brain barrier and unacceptable systemic toxicity limit the employment of chemotherapeutic agents. BCNU-impregnated biodegradable polymers (Gliadel®) have been demonstrated to prolong the survival of patients with malignant gliomas. Until now, no biodegradable drug delivery system has been commercially available in China. In the present study, we evaluated the safety of implants with high-dose BCNU in Chinese patients with recurrent malignant gliomas. PATIENTS AND METHODS Adults with supratentorial recurrent malignant glioma were eligible. High-dose BCNU-loaded PLGA implants (20mg of BCNU in each implant) were placed in the debulking cavity. The implants were investigated by a classical 3+3 design. Four levels of BCNU, up to 12 implants, were evaluated. Pharmacokinetic sampling was performed. The toxicity of the implants and the survival of patients were recorded. RESULTS Fifteen recurrent patients were enrolled with 12 glioblastomas and 3 anaplastic gliomas. Among 15 patients, 3 were treated with 3 implants (60 mg of BCNU), 3 with 6 implants (120 mg), 3 with 9 implants (180 mg) and 6 with 12 implants (240 mg). No dose-limiting toxicity was observed in the cohort of patients. Subgaleal effusion was the most common adverse event, presenting in 7 patients (46.7%). The median overall survival (OS) was 322 days (95% CI, 173-471 days). The 6-month, 1-year and 2-year survival rates were 66.7%, 40% and 13.3%, respectively. CONCLUSIONS The high-dose BCNU-loaded PLGA implants were safe for Chinese patients with recurrent malignant gliomas and further investigation for efficacy is warranted.
Collapse
Affiliation(s)
- Ke Sai
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Ming-Gu Zhong
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Jian Wang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Yin-Sheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Yong-Gao Mou
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Chao Ke
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Xiang-Heng Zhang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Qun-Ying Yang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Fu-Hua Lin
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Cheng-Cheng Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Zheng-He Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Jing Zeng
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Yan-Chun Lv
- Department of Imaging and Minimally Invasive Interventional Center, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| | - Xiang Li
- Department of Neurosurgery, Xuzhou Medical College Hospital, 99 West Huaihai Road, Xuzhou 221000, China.
| | - Wen-Chang Gao
- Department of Neurosurgery, Xuzhou Medical College Hospital, 99 West Huaihai Road, Xuzhou 221000, China.
| | - Zhong-Ping Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, 651 Dongfeng Road East, Guangzhou 510060, China.
| |
Collapse
|
338
|
Chiu B, Coburn J, Pilichowska M, Holcroft C, Seib FP, Charest A, Kaplan DL. Surgery combined with controlled-release doxorubicin silk films as a treatment strategy in an orthotopic neuroblastoma mouse model. Br J Cancer 2014; 111:708-15. [PMID: 24921912 PMCID: PMC4134491 DOI: 10.1038/bjc.2014.324] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/07/2014] [Accepted: 05/13/2014] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Neuroblastoma tumour resection goal is maximal tumour removal. We hypothesise that combining surgery with sustained, local doxorubicin application can control tumour growth. METHODS We injected human neuroblastoma cells into immunocompromised mouse adrenal gland. When KELLY cell-induced tumour volume was >300 mm(3), 80-90% of tumour was resected and treated as follows: instantaneous-release silk film with 100 μg doxorubicin (100IR), controlled-release film with 200 μg (200CR) over residual tumour bed; and 100 and 200 μg intravenous doxorubicin (100IV and 200IV). Tumour volume was measured and histology analysed. RESULTS Orthotopic tumours formed with KELLY, SK-N-AS, IMR-32, SH-SY5Y cells. Tumours reached 1800±180 mm(3) after 28 days, 2200±290 mm(3) after 35 days, 1280±260 mm(3) after 63 days, and 1700±360 mm(3) after 84 days, respectively. At 3 days post KELLY tumour resection, tumour volumes were similar across all groups (P=0.6210). Tumour growth rate was similar in untreated vs control film, 100IV vs 100IR, and 100IV vs 200IV. There was significant difference in 100IR vs 200CR (P=0.0004) and 200IV vs 200CR (P=0.0003). Tumour growth with all doxorubicin groups was slower than that of control (P: <0.0001-0.0069). At the interface of the 200CR film and tumour, there was cellular necrosis, surrounded by apoptotic cells before reaching viable tumour cells. CONCLUSIONS Combining surgical resection and sustained local doxorubicin treatment is effective in tumour control. Administering doxorubicin in a local, controlled manner is superior to giving an equivalent intravenous dose in tumour control.
Collapse
Affiliation(s)
- B Chiu
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood Street, Suite 416, Chicago, IL 60612, USA
| | - J Coburn
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - M Pilichowska
- Department of Pathology, Tufts Medical Center, 800 Washington Street, Box 115, Boston, MA 02111, USA
| | - C Holcroft
- Tufts Clinical and Translational Science Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - F P Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - A Charest
- Department of Neurosurgery, Tufts Medical Center, 800 Washington Street, Tufts Medical Center, Box 5609, Boston, MA 02111, USA
| | - D L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
339
|
Murphy M, Parney IF. Clinical trials in neurosurgical oncology. J Neurooncol 2014; 119:569-76. [PMID: 25106866 DOI: 10.1007/s11060-014-1569-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 07/23/2014] [Indexed: 10/24/2022]
Abstract
Brain tumors such as diffuse infiltrating gliomas continue to represent a major clinical challenge. Overall survival for patients diagnosed with glioblastoma, the most common primary brain tumor, remains less than 2 years despite intensive multimodal therapy with surgery, radiation, and chemotherapy. However, advances have been made in standard therapies and novel treatments that are showing great potential. These advances reflect careful study performed in the context of clinical trials. Neurosurgeons have played and will continue to play key parts in these studies. In this manuscript, we review clinical trials in neuro-oncology from a neurosurgical point of view and discuss potential roles for neurosurgeons in advancing glioma therapy in the future.
Collapse
Affiliation(s)
- Meghan Murphy
- Department of Neurological Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | |
Collapse
|
340
|
Wicks RT, Azadi J, Mangraviti A, Zhang I, Hwang L, Joshi A, Bow H, Hutt-Cabezas M, Martin KL, Rudek MA, Zhao M, Brem H, Tyler BM. Local delivery of cancer-cell glycolytic inhibitors in high-grade glioma. Neuro Oncol 2014; 17:70-80. [PMID: 25053853 DOI: 10.1093/neuonc/nou143] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND 3-bromopyruvate (3-BrPA) and dichloroacetate (DCA) are inhibitors of cancer-cell specific aerobic glycolysis. Their application in glioma is limited by 3-BrPA's inability to cross the blood-brain-barrier and DCA's dose-limiting toxicity. The safety and efficacy of intracranial delivery of these compounds were assessed. METHODS Cytotoxicity of 3-BrPA and DCA were analyzed in U87, 9L, and F98 glioma cell lines. 3-BrPA and DCA were incorporated into biodegradable pCPP:SA wafers, and the maximally tolerated dose was determined in F344 rats. Efficacies of the intracranial 3-BrPA wafer and DCA wafer were assessed in a rodent allograft model of high-grade glioma, both as a monotherapy and in combination with temozolomide (TMZ) and radiation therapy (XRT). RESULTS 3-BrPA and DCA were found to have similar IC50 values across the 3 glioma cell lines. 5% 3-BrPA wafer-treated animals had significantly increased survival compared with controls (P = .0027). The median survival of rats with the 50% DCA wafer increased significantly compared with both the oral DCA group (P = .050) and the controls (P = .02). Rats implanted on day 0 with a 5% 3-BrPA wafer in combination with TMZ had significantly increased survival over either therapy alone. No statistical difference in survival was noted when the wafers were added to the combination therapy of TMZ and XRT, but the 5% 3-BrPA wafer given on day 0 in combination with TMZ and XRT resulted in long-term survivorship of 30%. CONCLUSION Intracranial delivery of 3-BrPA and DCA polymer was safe and significantly increased survival in an animal model of glioma, a potential novel therapeutic approach. The combination of intracranial 3-BrPA and TMZ provided a synergistic effect.
Collapse
Affiliation(s)
- Robert T Wicks
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Javad Azadi
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Antonella Mangraviti
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Irma Zhang
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Lee Hwang
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Avadhut Joshi
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Hansen Bow
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Marianne Hutt-Cabezas
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Kristin L Martin
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Michelle A Rudek
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Ming Zhao
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Henry Brem
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| | - Betty M Tyler
- Department of Neurosurgery (R.T.W., J.A., A.M., I.Z., L.H., A.J., H.B., M.H.-C., K.L.M., H.B., B.M.T.); Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland (H.B.); Division of Chemical Therapeutics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland (M.A.R., M.Z.)
| |
Collapse
|
341
|
Woodworth GF, Dunn GP, Nance EA, Hanes J, Brem H. Emerging insights into barriers to effective brain tumor therapeutics. Front Oncol 2014; 4:126. [PMID: 25101239 PMCID: PMC4104487 DOI: 10.3389/fonc.2014.00126] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/13/2014] [Indexed: 12/27/2022] Open
Abstract
There is great promise that ongoing advances in the delivery of therapeutics to the central nervous system (CNS) combined with rapidly expanding knowledge of brain tumor patho-biology will provide new, more effective therapies. Brain tumors that form from brain cells, as opposed to those that come from other parts of the body, rarely metastasize outside of the CNS. Instead, the tumor cells invade deep into the brain itself, causing disruption in brain circuits, blood vessel and blood flow changes, and tissue swelling. Patients with the most common and deadly form, glioblastoma (GBM) rarely live more than 2 years even with the most aggressive treatments and often with devastating neurological consequences. Current treatments include maximal safe surgical removal or biopsy followed by radiation and chemotherapy to address the residual tumor mass and invading tumor cells. However, delivering effective and sustained treatments to these invading cells without damaging healthy brain tissue is a major challenge and focus of the emerging fields of nanomedicine and viral and cell-based therapies. New treatment strategies, particularly those directed against the invasive component of this devastating CNS disease, are sorely needed. In this review, we (1) discuss the history and evolution of treatments for GBM, (2) define and explore three critical barriers to improving therapeutic delivery to invasive brain tumors, specifically, the neuro-vascular unit as it relates to the blood brain barrier, the extra-cellular space in regard to the brain penetration barrier, and the tumor genetic heterogeneity and instability in association with the treatment efficacy barrier, and (3) identify promising new therapeutic delivery approaches that have the potential to address these barriers and create sustained, meaningful efficacy against GBM.
Collapse
Affiliation(s)
- Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine , Baltimore, MD , USA ; Department of Anatomy and Neurobiology, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Gavin P Dunn
- Department of Neurosurgery, Pathology and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine , St. Louis, MO , USA
| | - Elizabeth A Nance
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
342
|
Watts C, Price SJ, Santarius T. Current concepts in the surgical management of glioma patients. Clin Oncol (R Coll Radiol) 2014; 26:385-94. [PMID: 24882149 DOI: 10.1016/j.clon.2014.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 04/01/2014] [Indexed: 12/16/2022]
Abstract
The scientific basis for the surgical management of patients with glioma is rapidly evolving. The infiltrative nature of these cancers precludes a surgical cure, but despite this, cytoreductive surgery remains central to high-quality patient care. In addition to tissue sampling for accurate histopathological diagnosis and molecular genetic characterisation, clinical benefit from decompression of space-occupying lesions and microsurgical cytoreduction has been reported in patients with different grades of glioma. By integrating advanced surgical techniques with molecular genetic characterisation of the disease and targeted radiotherapy and chemotherapy, it is possible to construct a programme of personalised surgical therapy throughout the patient journey. The goal of therapeutic packages tailored to each patient is to optimise patient safety and clinical outcome and must be delivered in a multidisciplinary setting. Here we review the current concepts that underlie surgical subspecialisation in the management of patients with glioma.
Collapse
Affiliation(s)
- C Watts
- University of Cambridge, Department of Clinical Neurosciences, Division of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK; Department of Clinical Neurosciences, Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, UK.
| | - S J Price
- University of Cambridge, Department of Clinical Neurosciences, Division of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK
| | - T Santarius
- University of Cambridge, Department of Clinical Neurosciences, Division of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
343
|
Abstract
Malignant astrocytomas constitute the most aggressive and common primary tumors of the central nervous system. The standard treatment protocol for these tumors involves maximum safe surgical resection with adjuvant chemoradiotherapy. Despite numerous advances in surgical techniques and adjuncts, as well as the ongoing renaissance in the genetic and molecular characterization of these tumors, malignant astrocytomas continue to be associated with poor prognosis, with median overall survival averaging 15 months for grade IV astrocytomas after standard-of-care treatment. In this article, the goals, principles, techniques, prognostic factors, and modern outcomes of malignant astrocytoma surgery are reviewed. Particular attention is paid to contemporary methods of neuronavigation and functional mapping, the prognostic significance of the extent of resection, surgically delivered adjunctive therapies, and future avenues of research.
Collapse
Affiliation(s)
- Eli T Sayegh
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Taemin Oh
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shayan Fakurnejad
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Daniel E Oyon
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Orin Bloch
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Andrew T Parsa
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|
344
|
D'Amico RS, Kennedy BC, Bruce JN. Neurosurgical oncology: advances in operative technologies and adjuncts. J Neurooncol 2014; 119:451-63. [PMID: 24969924 DOI: 10.1007/s11060-014-1493-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/22/2014] [Indexed: 12/31/2022]
Abstract
Modern glioma surgery has evolved around the central tenet of safely maximizing resection. Recent surgical adjuncts have focused on increasing the maximum extent of resection while minimizing risk to functional brain. Technologies such as cortical and subcortical stimulation mapping, intraoperative magnetic resonance imaging, functional neuronavigation, navigable intraoperative ultrasound, neuroendoscopy, and fluorescence-guided resection have been developed to augment the identification of tumor while preserving brain anatomy and function. However, whether these technologies offer additional long-term benefits to glioma patients remains to be determined. Here we review advances over the past decade in operative technologies that have offered the most promising benefits for glioblastoma patients.
Collapse
Affiliation(s)
- Randy S D'Amico
- Department of Neurological Surgery, Neurological Institute, Columbia University Medical Center, 4th Floor, 710 West 168th Street, New York, NY, 10032, USA,
| | | | | |
Collapse
|
345
|
Archavlis E, Tselis N, Birn G, Ulrich P, Zamboglou N. Combined salvage therapies for recurrent glioblastoma multiforme: evaluation of an interdisciplinary treatment algorithm. J Neurooncol 2014; 119:387-95. [DOI: 10.1007/s11060-014-1500-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/04/2014] [Indexed: 11/24/2022]
|
346
|
Suzuki A, Leland P, Kobayashi H, Choyke PL, Jagoda EM, Inoue T, Joshi BH, Puri RK. Analysis of biodistribution of intracranially infused radiolabeled interleukin-13 receptor-targeted immunotoxin IL-13PE by SPECT/CT in an orthotopic mouse model of human glioma. J Nucl Med 2014; 55:1323-9. [PMID: 24947060 DOI: 10.2967/jnumed.114.138404] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/01/2014] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Interleukin-13 Pseudomonas exotoxin (IL-13PE), a targeted agent for interleukin-13 receptor α2 (IL-13Rα2)-expressing tumors, has been administered intracranially by convection-enhanced delivery (CED) for glioma therapy in several clinical trials including a randomized phase 3 clinical trial. However, its intracranial distribution was not optimally evaluated. We investigated the intracranial distribution of radiolabeled IL-13PE after CED in a murine model of glioblastoma multiforme. METHODS IL-13PE was radiolabeled with Na(125)I and evaluated for its activity in vitro in receptor-positive U251 or -negative T98G human glioma cell lines. Gliomas were grown in nude mice after intracranial implantation with U251 cells, and (125)I-IL-13PE was stereotactically administered by bolus or CED for 3 d, followed by micro-SPECT/CT imaging. SPECT images were evaluated quantitatively and compared with histology and autoradiography results. RESULTS The radioiodination technique resulted in a specific and biologically active (125)I-IL-13PE, which bound and was cytotoxic to IL-13Rα2-positive but not to IL-13Rα2-negative tumor cells. Both the binding and the cytotoxic activities were blocked by a 100-fold excess of IL-13, which indicated the specificity of binding and cytotoxicity. SPECT/CT imaging revealed retention of (125)I-IL-13PE administered by CED in U251 tumors and showed significantly higher volumes of distribution and maintained detectable drug levels for a longer period of time than the bolus route. These results were confirmed by autoradiography. CONCLUSION IL-13PE can be radioiodinated without the loss of specificity, binding, or cytotoxic activity. Intracranial CED administration produces a higher volume of distribution for a longer period of time than the bolus route. Thus, CED of IL-13PE is superior to bolus injection in delivering the drug to the entire tumor.
Collapse
Affiliation(s)
- Akiko Suzuki
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| | - Pamela Leland
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| | - Hisataka Kobayashi
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Peter L Choyke
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Elaine M Jagoda
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Tomio Inoue
- Department of Radiology, Yokohama City University, Yokohama, Japan
| | - Bharat H Joshi
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| | - Raj K Puri
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| |
Collapse
|
347
|
Lacouture ME, Elizabeth Davis M, Elzinga G, Butowski N, Tran D, Villano JL, DiMeglio L, Davies AM, Wong ET. Characterization and Management of Dermatologic Adverse Events With the NovoTTF-100A System, a Novel Anti-mitotic Electric Field Device for the Treatment of Recurrent Glioblastoma. Semin Oncol 2014; 41 Suppl 4:S1-14. [DOI: 10.1053/j.seminoncol.2014.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
348
|
Mohammadi AM, Hawasli AH, Rodriguez A, Schroeder JL, Laxton AW, Elson P, Tatter SB, Barnett GH, Leuthardt EC. The role of laser interstitial thermal therapy in enhancing progression-free survival of difficult-to-access high-grade gliomas: a multicenter study. Cancer Med 2014; 3:971-9. [PMID: 24810945 PMCID: PMC4303165 DOI: 10.1002/cam4.266] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 12/21/2022] Open
Abstract
Surgical extent-of-resection has been shown to have an impact on high-grade glioma (HGG) outcomes; however, complete resection is rarely achievable in difficult-to-access (DTA) tumors. Controlled thermal damage to the tumor may have the same impact in DTA-HGGs. We report our multicenter results of laser interstitial thermal therapy (LITT) in DTA-HGGs. We retrospectively reviewed 34 consecutive DTA-HGG patients (24 glioblastoma, 10 anaplastic) who underwent LITT at Cleveland Clinic, Washington University, and Wake Forest University (May 2011-December 2012) using the NeuroBlate(®) System. The extent of thermal damage was determined using thermal damage threshold (TDT) lines: yellow TDT line (43 °C for 2 min) and blue TDT line (43°C for 10 min). Volumetric analysis was performed to determine the extent-of-coverage of tumor volume by TDT lines. Patient outcomes were evaluated statistically. LITT was delivered as upfront in 19 and delivered as salvage in 16 cases. After 7.2 months of follow-up, 71% of cases demonstrated progression and 34% died. The median overall survival (OS) for the cohort was not reached; however, the 1-year estimate of OS was 68 ± 9%. Median progression-free survival (PFS) was 5.1 months. Thirteen cases who met the following two criteria-(1) <0.05 cm(3) tumor volume not covered by the yellow TDT line and (2) <1.5 cm(3) additional tumor volume not covered by the blue TDT line-had better PFS than the other 21 cases (9.7 vs. 4.6 months; P = 0.02). LITT can be used effectively for treatment of DTA-HGGs. More complete coverage of tumor by TDT lines improves PFS which can be translated as the extent of resection concept for surgery.
Collapse
Affiliation(s)
- Alireza M Mohammadi
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, 9500 Euclid Avenue, S70, Cleveland Clinic, Cleveland, Ohio, 44195
| | | | | | | | | | | | | | | | | |
Collapse
|
349
|
Stupp R, Brada M, van den Bent MJ, Tonn JC, Pentheroudakis G. High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014; 25 Suppl 3:iii93-101. [PMID: 24782454 DOI: 10.1093/annonc/mdu050] [Citation(s) in RCA: 511] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- R Stupp
- Department of Oncology and Cancer Centre, University Hospital Zurich, Zurich, Switzerland
| | - M Brada
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Clatterbridge Cancer Centre, Wirral, UK
| | - M J van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Center, Rotterdam, Netherlands
| | - J-C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
| | - G Pentheroudakis
- Department of Medical Oncology, Medical School, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
350
|
Ryken TC, Kalkanis SN, Buatti JM, Olson JJ. The role of cytoreductive surgery in the management of progressive glioblastoma : a systematic review and evidence-based clinical practice guideline. J Neurooncol 2014; 118:479-88. [PMID: 24756348 DOI: 10.1007/s11060-013-1336-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/28/2013] [Indexed: 02/04/2023]
Abstract
QUESTION Should patients with previously diagnosed malignant glioma who are suspected of experiencing progression of the neoplasm process undergo repeat open surgical resection? TARGET POPULATION These recommendations apply to adults with previously diagnosed malignant glioma who are suspected of experiencing progression of the neoplastic process and are amenable to surgical resection. RECOMMENDATIONS LEVEL II Repeat cytoreductive surgery is recommended in symptomatic patients with locally recurrent or progressive malignant glioma. The median survival in these patient diagnosed with glioblastoma is expected to range from 6 to 17 months following a second procedure. It is recommended that the following preoperative factors be considered when evaluating a patient for repeat operation: location of recurrence in eloquent/critical brain regions, Karnofsky Performance Status and tumor volume.
Collapse
|