clinical trials in brain

Upload: madhukar1978

Post on 07-Apr-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/4/2019 Clinical Trials in Brain

    1/16

    Cl i ni c a l T ri a l s i n Bra i nTumor Surgery

    Jason M. Hoover, MDa, Susan M. Chang, MDb,Ian F. Parney, MD, PhDa,*

    Central nervous system tumors represent

    a continued medical and surgical challenge.

    Primary intra-axial brain tumors, more specifically

    gliomas, have an annual incidence of approxi-mately 14 per 100,000.1,2 Glioblastoma is the

    most frequent and malignant histologic subtype

    (World Health Organization [WHO] grade IV).

    Surgery has an established role in the diagnosis

    and treatment of brain tumors and forms a key

    part of standard therapy along with radiation and

    chemotherapy. Outcomes for most brain tumor

    patients, however, remain humbling and continued

    efforts to improve and refine brain tumor treatment

    (including surgery) are needed. This article focuses

    on clinical trials in primary intra-axial brain tumorsurgery, including a review of outcome data with

    standard surgical maneuvers, techniques to maxi-

    mize safe surgical resection, and trials of surgically

    delivered therapeutics.

    STANDARD NEUROSURGICAL TECHNIQUES

    Standard neurosurgical techniques in brain tumor

    management can be divided into those primarily

    directed at diagnosis (ie, techniques for biopsy)

    and those directed at tumor debulking (ie, tech-

    niques for craniotomy and resection). This sectionreviews clinical trials of these standard tech-

    niques, including outcome studies.

    Frame-Based Stereotactic Biopsy

    Stereotactic biopsy can be defined as biopsy of

    intracranial lesions through a burr hole using

    stereotactic target coordinates determined on

    prebiopsy imaging. Stereotactic biopsy tech-

    niques were first developed using rigid frames

    attached to a patients skull.3 A rigid fiducial

    marker box is attached to the frame base beforeimaging (CT or MR imaging), thus defining the

    stereotactic space to determine the target, entry

    point, depth, and trajectory based on X, Y, and

    Z coordinates and a Cartesian system. The fidu-

    cial marker box is replaced by a stereotactic

    guide system attached to the frame that directs

    a biopsy needle from the entry point to the

    target along the selected trajectory to the depth

    calculated by the system (Fig. 1 ). Frame-based

    stereotactic needle biopsy can be performed

    with local or general anesthetic. The techniqueis generally safe and has a high chance of

    obtaining diagnostic material. Diagnostic yields

    are 91% to 98.4% with transient morbidity of

    2.9% to 9%, permanent morbidity of 1.5% to

    5%, and mortality of approximately 1% in

    modern series.46 Small, deep lesions in

    eloquent brain are associated with higher rates

    of morbidity and nondiagnosis. In addition, the

    number of separate trajectories needed to

    obtain diagnostic material has been associated

    with increased morbidity.5 Finally, there remains

    the potential for sampling error with stereotacticbiopsy. In a series of patients at the MD Ander-

    son Cancer Center who underwent craniotomy

    and tumor resection after previously undergoing

    stereotactic biopsy for glioma, the final patho-

    logic diagnosis changed in 38% once further

    tissue was available.7 This was largely ac-

    counted for by differences in tumor grade.

    a Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USAb

    Department of Neurologic Surgery, University of California, 505 Parnassus Avenue, San Francisco, CA 94143,USA* Corresponding author.E-mail address: [email protected]

    KEYWORDS

    Brain tumor Gliomas Central nervous system Cytoreductive surgery

    Neuroimag Clin N Am 20 (2010) 409424doi:10.1016/j.nic.2010.04.0061052-5149/10/$ see front matter 2010 Elsevier Inc. All rights reserved. n

    euroimaging.theclinics.c

    om

    mailto:[email protected]://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/http://neuroimaging.theclinics.com/mailto:[email protected]
  • 8/4/2019 Clinical Trials in Brain

    2/16

    Frameless Stereotactic Needle Biopsy

    Frameless stereotactic biopsy performed using an

    intraoperative neuronavigation system, such as

    Stealth, BrainLab, or ISG Wand, is increasingly

    common.8 Preoperative imaging (MR imaging or

    CT) is obtained after placing multiple adhesive

    fiducial markers on the patients scalp. This infor-

    mation is transferred to an intraoperative

    computer connected to a binocular sensing device

    (eg, infrared camera) that can register the preoper-

    ative imaging in stereotactic space using the fidu-

    cial markers (Fig. 2 ). Alternatively, some systems

    register preoperative imaging using bony land-

    marks. Once the imaging and patient are regis-tered, a viewing wand or other tool can be used

    to demonstrate the underlying anatomy in 3-D.

    An entry point, trajectory, depth, and target can

    then be determined for the biopsy. A guide system

    attached to the skull or head holder keeps the

    biopsy instrument steady, allowing careful depth

    measurement and strict adherence to theproposed trajectory. Frameless stereotactic nee-

    dle biopsy is often performed under general

    anesthetic.

    Frameless stereotactic biopsy has been exten-

    sively reported in recent years, with diagnostic

    yields of 85% to 91% and permanent morbidity

    rates of 3% to 6%.810 Studies with imaging

    phantoms have suggested that frameless and

    frame-based stereotactic biopsy accuracy are

    comparable.11 Fiducial markers or surface land-

    marks used to register images for frameless

    systems may shift, however, during positioning

    for surgery. This is not an issue for rigid frame-

    based systems. Thus, some investigators advo-

    cate continued reliance on frame-based biopsy

    Fig. 1. Postcontrast T1-weighted MR imaging and reconstructions of a 73-year-old right-handed man who pre-sented with a right hemiparesis secondary to an enhancing mass in his left insular cortex and basal ganglia.The hyperintense fiducial markers from the Compass stereotactic frame can be seen surrounding his head. Theentry point, trajectory, and target are shown in the upper and lower right images. The final pathology was GBM.

    Hoover et al410

  • 8/4/2019 Clinical Trials in Brain

    3/16

    for small or deep lesions. Furthermore, there are

    potential advantages to frame-based biopsies in

    terms of reduced use of anesthesia, operating

    room, and hospital resources,10 although this

    may vary with individual practice patterns. Incontrast, some investigators have concluded that

    frameless stereotactic biopsy is comparable with

    frame bases with regard to diagnostic yield and

    complications but has distinct advantages.

    Although a recent series from the Johns Hopkins

    Hospital found no significant differences in diag-

    nostic yield or complications, more than one nee-

    dle trajectory was required more frequently to

    obtain diagnostic tissue from cortical lesions withframe-based compared with frameless stereo-

    taxy.8 In other studies, increased number of nee-

    dle trajectories has been strongly associated with

    increased morbidity.5 The Johns Hopkins

    Fig. 2. (A) Axial T1-weighted and T2-weighted postcontrast images and (B) frameless stereotaxic biopsy plan(using the Stealth system) for an 18-year-old right-handed woman presenting with diplopia secondary toa partially enhancing mass in the left medial thalamus and midbrain tectum. The final pathology was primitiveneuroectodermal tumor.

    Clinical Trials in Brain Tumor Surgery 411

  • 8/4/2019 Clinical Trials in Brain

    4/16

  • 8/4/2019 Clinical Trials in Brain

    5/16

    response to adjuvant therapy with more extensive

    resection. The definition of maximal safe resection,

    however, differs widely among neurosurgeons.

    The authors reviewed the literature from the past

    20 years using a similar strategy to that described

    previously for malignant gliomas. A PubMed

    search was performed using combinations of thekey words, low-grade, grade I, diffuse,

    glioma, astrocytoma, oligodendroglioma,

    oligoastrocytoma, surgery, and outcome. In

    addition, further key publications were identified

    by cross-references in the resulting articles.

    Studies without information on outcome with

    varying degrees of resection, where extent of

    resection was not determined on the basis of post-

    operative imaging or where formal statistical eval-

    uation was lacking, were excluded. Similarly,

    studies including children or patients with grade

    I, III, or IV tumors were excluded unless these pop-

    ulations were analyzed separately. In addition,

    studies with short median follow-up (

  • 8/4/2019 Clinical Trials in Brain

    6/16

    (magnetic resonance spectroscopy [MRS], cere-

    bral perfusion, and positron emission tomography

    [PET]), which can be used to provide information

    about the nature and extent of the tumor itself;

    and (2) functional imaging (functional MR imaging,

    diffusion tensor imaging, PET, and magnetoence-

    phalography), which can provide informationabout adjacent eloquent brain. This section

    reviews recent clinical studies about physiologic

    and functional imaging in brain tumor surgery.

    Physiologic imagingSeveral imaging modalities have been used to

    elucidate information about tumor physiology

    that can be beneficial in surgical planning by high-

    lighting more aggressive areas or further delin-

    eating the extent of tumor beyond enhancing

    margins.57 For example, proton-based MRS can

    distinguish tumor (increased choline to N-acety-

    laspartate [NAA] ratio) from normal tissue (choline:

    NAA ratio

  • 8/4/2019 Clinical Trials in Brain

    7/16

    imaging scanners. Furthermore, they can be used

    to reregister image-guidance systems. A recent

    study from the University of California in Los An-

    geles reported a series documenting extent of

    resection with varying image guidance and intrao-

    perative MR imaging systems.80 As expected,

    extent of resection was generally larger with image

    guidance than without. Low-field intraoperative or

    high-field intraoperative MR imaging alone did not

    improve on this but high-field intraoperative MR

    imaging with reregistration of a neuronavigation

    system resulted in the highest degree of resection

    of any combination of image guidance and intrao-

    perative imaging systems. (See Fig. 5 for an

    example of imaging obtained with high-field intra-

    operative MR imaging.)

    Electrophysiologic Mapping

    Although preoperative functional imaging can be

    incorporated into many neuronavigation systems,

    intraoperative electrophysiologic mapping remains

    the gold standard for identifying functional cortex

    and subcortical fiber tracts. This is not a new tech-

    nique, having been originally developed by Pen-

    field and Boldrey in the 1930s to facilitate

    epilepsy surgery.81 It has since been adapted to

    a variety of neurosurgical indications, including

    intra-axial tumor resection.82,83 It has been sug-

    gested that, as long as the resection remains

    exclusively within the tumor itself, the risk of

    neurologic injury is low. This is not always

    possible, however, and data from large prospec-

    tive series suggest that functional neurons occur

    within tumors adjacent to functional cortex in up

    to 8% of cases.84 Mapping motor cortex and

    subcortical fibers can be done while patients are

    awake or asleep (with appropriate anesthetic

    management)85 but speech mapping requires

    awake craniotomy and patient cooperation. As

    experience has grown with speech mapping in

    tumor surgery, it has become evident that thiscan allow aggressive tumor resection in brain

    areas traditionally felt to be inoperable due to their

    proximity to critical areas for speech. In a recent

    prospective series of 250 patients with gliomas,

    Fig. 3. (A) Standard axial T1-weighted postcontrast MR imaging of the head demonstrating a right frontal glio-blastoma. (B) Cerebral blood volume/perfusion MR imaging demonstrating increased blood flow (red) inenhancing areas of same glioblastoma. (C) Multivoxel 3-D MRS of the same patient demonstrating increased chol-ine:NAA ratio in tumor and surrounding T2 hyperintensity.

    Clinical Trials in Brain Tumor Surgery 415

  • 8/4/2019 Clinical Trials in Brain

    8/16

    Berger and colleagues86 reported that critical

    speech areas were identified in 58%. One weekafter surgery, speech function was worsened in

    22%; it resolved in most patients, however, and

    only 1.6% of surviving patients at 6 months

    continued to have increased speech deficits.86

    Surface cortical electrophysiologic mapping is

    useful for identifying appropriate safe entry pointsfor surgical resection. It does not identify subcor-

    tical fibers, however, that may deviate from their

    surface origin as they pass. For this, subcortical

    electrophysiologic mapping is critical, particularly

    Fig. 4. (A) Standard axial T1-weighted postcontrast MR imaging of the head demonstrating a left parietal glio-blastoma. (B) Functional MR imaging demonstrating areas of activation (red and yellow) with right ankle andhand motor tasks anterior and superior to the tumor. (C) DTI demonstrating displacement of white matter tractsby the tumor. (D) Frameless stereotactic images of the same patient with motor areas for right hand (green) andankle (pink) superimposed. The viewing wand (blue) has been placed at a critical motor area for right hand func-tion identified by intraoperative electrical stimulation mapping with a bipolar electrode. Note that this corre-sponds to only a portion of the larger area identified by functional MR imaging, suggesting that functionalMR imaging was sensitive but not specific for motor function in this case.

    Hoover et al416

  • 8/4/2019 Clinical Trials in Brain

    9/16

    for motor mapping,85 and uses techniques similar

    to surface mapping and can be used intermittently

    as resection proceeds.

    Fluorescence-Guided Resection

    Fluorescence-guided resection relies on adminis-

    tration of a pharmacologic agent that localizes totumor but not surrounding brain and fluoresces

    when exposed to light of the appropriate wave-

    length. Tumor resection can be guided by the

    fluorescence seen at surgery, potentially allowing

    a surgeon to identify tumor tissue that otherwise

    might not be obvious at craniotomy. Fluores-

    cence-guided surgery for brain tumors has been

    used most commonly with 5-ALA. 5-ALA is orally

    available and accumulates in glioma tissue where

    it is metabolized to protoporphyrin IX and fluo-

    resces pink when exposed to light at 375 to

    440 nm.87 Stummer and colleagues22 have pub-lished results of a multicenter randomized

    controlled trial comparing 5-ALA fluorescence-

    guided resection with standard white light resec-

    tion in 322 newly diagnosed GBM patients.

    Patients undergoing fluorescence-guided

    resection were more likely to have a gross total

    resection (65% vs 36%, P

  • 8/4/2019 Clinical Trials in Brain

    10/16

    Brachytherapy

    Brachytherapy can be defined as implanting inter-

    stitial radioactive sources directly into the tumor at

    surgery to allow delivery of local high-dose irradia-

    tion while leaving surrounding tissues relatively

    spared. Iodine 125 and iridium 132 are commonlyused sources. At least two forms have been investi-

    gated in malignant glioma treatment: temporary/

    high-dose brachytherapy and permanent/low-

    dose brachytherapy.89,90 Although initial studies

    with high-dose temporary brachytherapy were

    promising, they failed to show benefit in a random-

    ized controlled trial.88 Single-arm studies of radical

    resection of recurrent malignant gliomas followed

    by permanent low-dose brachytherapy have shown

    modest efficacy.91,92 GliaSite, another method for

    delivering temporary brachytherapy, has also

    been investigated. In this method, an inflatableballoon catheter is placed in the resection cavity

    at craniotomy for a recurrent malignant glioma.93

    An aqueous solution of organically bound 125I is

    delivered to the balloon via a subcutaneous port 1

    to 2 weeks after craniotomy and removed after

    3 to 6 days. Gliasite has been safe and rela-

    tively promising in single-arm studies.9395

    These results remain to be evaluated, however, in

    randomized studies.

    Chemotherapy with Biodegradable Wafers

    Several strategies have been used to deliver

    chemotherapeutic agents surgically to patients

    with malignant gliomas. Initial efforts focused on

    direct injection into tumor or surrounding paren-

    chyma had limited efficacy but relatively high

    toxicity.96 As an alternative to bolus injection,

    Brem and colleagues97 developed methods to

    line malignant glioma resection cavities with

    biodegradable wafers impregnated with chemo-

    therapeutic agents. In randomized controlled

    trials, recurrent malignant glioma patientsreceiving carmustine (BCNU)-impregnated poly-

    mer wafers had modest but significantly prolonged

    median survival compared with patients who

    underwent simple resection (31 weeks vs 23

    weeks).9698 In the largest study, a statistical

    survival benefit persisted for up to 3 years.99 In

    a recent institutional review, 1013 patients under-

    going craniotomy for malignant astrocytomas

    over a 10-year period at Johns Hopkins Hospital

    were reviewed. Perioperative morbidity (within 3

    months) and overall survival were assessed. Of

    1013 patients who had craniotomies for malignantastrocytoma, 288 (28%) received Gliadel wafers

    (250 patients had GBM and 38 patients had AA/

    anaplastic oligodendroglioma [AA/AO]). Patients

    receiving Gliadel were older and more frequently

    underwent gross total resection (75% vs 36%).

    The patients in Gliadel versus non-Gliadel cohorts

    had similar perioperative morbidity. For patients

    receiving Gliadel for GBM, median survival was

    13.5 months after primary resection (20% alive at

    2 years) and 11.3 months after revision resection

    (13% alive at 2 years). For patients receiving Glia-del for AA/AO, median survival was 57 months

    after primary resection (66% alive at 2 years) and

    23.6 months after revision resection (47% alive at

    2 years).100

    Convection-Enhanced Delivery

    There remains concern as to whether or not

    chemotherapy-impregnated wafers are the most

    effective way to deliver therapeutic agents locally

    to patients with malignant gliomas at surgery.

    Just like bolus injection, chemotherapy wafers

    rely on diffusion to carry their agents into the

    surrounding brain parenchyma. Concerns have

    been raised that delivery via chemotherapy wafers

    may be too limited. As an alternative, Oldfield and

    colleagues101,102 at the National Institutes of

    Health pioneered an approach, termed, convec-

    tion-enhanced delivery (CED). Between 1 and 3

    small-diameter sialastic tubes are placed stereo-

    tactically in the tumor itself or in brain parenchyma

    surrounding a resection cavity and tunneled

    subcutaneously through the scalp. Therapeuticagents are then delivered via slow constant infu-

    sion (eg, 0.2 mL/h) through these catheters over

    3 to 5 days postoperatively. The pressure from

    the slow infusion sets up a local convection current

    within the brain from the catheter tip (convection-

    enhanced) that results in a large volume of

    distribution for the agent in question. In ideal

    circumstances, an entire lobe can be perfused

    from a single catheter.

    Most CED clinical trials to date have focused on

    small fusion proteins that have linked an intracel-lular toxin (eg, pseudomonas exotoxin or diph-

    theria toxin) to a ligand for surface receptors that

    are overexpressed on malignant glioma cells (eg,

    interleukin-13 or transferrin). Promising results

    from several phase I and II studies have been re-

    ported in newly diagnosed and recurrent malig-

    nant gliomas.103,104 These therapies have

    potential for local inflammatory reactions that can

    be followed on post-treatment MR imaging scans

    and occasionally require treatment.105 Two large

    multicenter randomized controlled trials have

    been completed in patients with recurrent glio-blastomas (PRECISE and TransMID). These trials

    demonstrated that CED of these agents was safe

    and well tolerated. Unfortunately, they did not

    show any survival benefit. To date, these results

    Hoover et al418

  • 8/4/2019 Clinical Trials in Brain

    11/16

    have only been reported as press releases. Exper-

    imental efforts are ongoing to track and improve

    delivery by CED106 and to expand the types of

    agents that can be delivered using this

    approach.107

    Oncolytic Viral Vectors and SuicideGene Therapy

    Oncolytic virus therapy is a novel treatment option

    for GBM patients. These viruses are naturally

    occurring or genetically engineered viruses with

    a predilection to infect and kill tumor cells while

    leaving normal cells unharmed. They are derived

    from a variety of viruses, including herpes simplex

    virus (HSV-1), adenovirus, reovirus, measles, and

    Newcastle disease virus. The mechanisms under-

    lying their relative tumor specificity are not always

    clear. Although work is ongoing to develop onco-

    lytic viruses that can be systemically delivered,

    clinical trials so far in malignant gliomas have

    been limited to viruses that are delivered locally

    at surgery.108110 Oncolytic measles virus strains

    derived from the Edmonston vaccine lineage serve

    as a good example of the types viruses used in

    this manner. They have been effective in preclin-

    ical studies when delivered to intracranial

    tumors.111,112 This strain was engineered to

    express the marker peptide measles viruscarci-

    noembryonic antigen (MV-CEA) and MV-CEAlevels can serve as a correlate of viral gene expres-

    sion. In support of a phase I clinical trial of intratu-

    moral and resection cavity administration of

    MV-CEA to patients with recurrent gliomas, safety

    was demonstrated in rhesus macaques. The

    investigators found that more than 36 months

    from study initiation there had been no clinical or

    biochemical evidence of toxicity, including lack

    of neurologic symptoms, fever, or other systemic

    symptoms and lack of immunosuppression.113

    This approach is currently in phase I clinical trials.Finally, Ram and colleagues114 have evaluated

    the use of sitimagene ceradenovec (Cerepro) and

    ganciclovir gene therapy in the treatment of

    primary high-grade gliomas. This differs from

    a true oncolytic virus approach in that the virus

    has no inherent anticancer properties. Instead,

    a genetically engineered adenovirus delivers

    a suicide gene (HSV thymidine kinase) that kills

    tumor cells only when they are exposed to ganci-

    clovir. The primary outcome measures for this

    randomized controlled trial are time from surgery

    to reintervention or death. Although data analysesare not complete, preliminary results suggest that

    patients in the Cerepro arm have a statistically

    significant improvement in outcome. Further data

    will be reported as they become available.114

    SUMMARY

    Brain tumor surgery, specifically glioma surgery,

    spans a broad range of pathologies and tech-

    niques. There is a continued need to improve the

    therapies available for treating these devastating

    tumors. As reviewed in this article, many clinicalstudies have examined new surgical techniques

    to perform and guide brain tumor biopsy and

    resection, the value of cytoreductive surgery, and

    the potential to introduce novel therapeutic agents

    locally at surgery. These studies have expanded

    the breadth of knowledge regarding optimal

    surgical approaches and have opened the door

    to new technologies that will hopefully have an

    impact on survival for patients with gliomas in the

    future.

    REFERENCES

    1. Davis FG, Kupelian V, Freels S, et al. Prevalence

    estimates for primary brain tumors in the United

    States by behavior and major histology groups.

    Neuro Oncol 2001;3(3):1528.

    2. Wrensch M, Minn Y, Chew T, et al. Epidemiology of

    primary brain tumors: current concepts and review

    of the literature. Neuro Oncol 2002;4(4):27899.

    3. Bernstein M, Parrent AG. Complications of CT-

    guided stereotactic biopsy of intra-axial brain

    lesions. J Neurosurg 1994;81(2):1658.4. Kongkham PN, Knifed E, Tamber MS, et al. Compli-

    cations in 622 cases of frame-based stereotactic

    biopsy, a decreasing procedure. Can J Neurol

    Sci 2008;35(1):7984.

    5. McGirt MJ, Woodworth GF, Coon AL, et al.

    Independent predictors of morbidity after image-

    guided stereotactic brain biopsy: a risk assess-

    ment of 270 cases. J Neurosurg 2005;102(5):

    897901.

    6. Owen CM, Linskey ME. Frame-based stereotaxy in

    a frameless era: current capabilities, relative role,

    and the positive- and negative predictive values

    of blood through the needle. J Neurooncol 2009;

    93(1):13949.

    7. Jackson RJ, Fuller GN, Abi-Said D, et al. Limita-

    tions of stereotactic biopsy in the initial manage-

    ment of gliomas. Neuro Oncol 2001;3(3):193200.

    8. Woodworth GF, McGirt MJ, Samdani A, et al.

    Frameless image-guided stereotactic brain biopsy

    procedure: diagnostic yield, surgical morbidity,

    and comparison with the frame-based technique.

    J Neurosurg 2006;104(2):2337.

    9. Air EL, Leach JL, Warnick RE, et al. Comparing therisks of frameless stereotactic biopsy in eloquent

    and noneloquent regions of the brain: a retrospec-

    tive review of 284 cases. J Neurosurg 2009;111(4):

    8204.

    Clinical Trials in Brain Tumor Surgery 419

  • 8/4/2019 Clinical Trials in Brain

    12/16

    10. Smith JS, Quinones-Hinojosa A, Barbaro NM, et al.

    Frame-based stereotactic biopsy remains an

    important diagnostic tool with distinct advantages

    over frameless stereotactic biopsy. J Neurooncol

    2005;73(2):1739.

    11. Quinones-Hinojosa A, Ware ML, Sanai N, et al.

    Assessment of image guided accuracy in a skullmodel: comparison of frameless stereotaxy tech-

    niques vs. frame-based localization. J Neurooncol

    2006;76(1):6570.

    12. Kreth FW, Warnke PC, Ostertag CB. Progress in

    malignant glioma. J Neurosurg 2004;100:11323.

    13. BrownPD, MaurerMJ, RummansTA,et al. A prospec-

    tive study of quality of life in adults with newly diag-

    nosed high-grade gliomas: the impact of the extent

    of resection on quality of life and survival. Neurosur-

    gery 2005;57(3):495504 [discussion: 495504].

    14. Buckner JC, Schomberg PJ, McGinnis WL, et al. A

    phase III study of radiation therapy plus carmustine

    with or without recombinant interferon-alpha in the

    treatment of patients with newly diagnosed high-

    grade glioma. Cancer 2001;92(2):42033.

    15. Keles GE, Anderson B, Berger MS. The effect of

    extent of resection on time to tumor progression

    and survival in patients with glioblastoma multi-

    forme of the cerebral hemisphere. Surg Neurol

    1999;52(4):3719.

    16. Kiwit JC, Floeth FW, Bock WJ. Survival in malignant

    glioma: analysis of prognostic factors with special

    regard to cytoreductive surgery. Zentralbl Neuro-chir 1996;57(2):7688.

    17. Lacroix M, Abi-Said D, Fourney DR, et al. A multi-

    variate analysis of 416 patients with glioblastoma

    multiforme: prognosis, extent of resection, and

    survival. J Neurosurg 2001;95(2):1908.

    18. Lamborn K, Chang S, Prados M. Prognostic

    factors for survival of patients with glioblastoma:

    recursive partitioning analysis. Neuro Oncol

    2004;6:22735.

    19. Laws ER, Parney IF, Huang W, et al. Survival

    following surgery and prognostic factors for

    recently diagnosed malignant glioma: data from

    the Glioma Outcomes Project. J Neurosurg 2003;

    99(3):46773.

    20. Mohan DS, Suh JH, Phan JL, et al. Outcome in

    elderly patients undergoing definitive surgery and

    radiation therapy for supratentorial glioblastoma

    multiforme at a tertiary care institution. Int J Radiat

    Oncol Biol Phys 1998;42(5):9817.

    21. Nitta T, Sato K. Prognostic implications of the extent

    of surgical resection in patients with intracranial

    malignant gliomas. Cancer 1995;75:272731.

    22. Stummer W, Pichlmeier U, Meinel T, et al. Fluores-cence-guided surgery with 5-aminolevulinic acid

    for resection of malignant glioma: a randomised

    controlled multicentre phase III trial. Lancet Oncol

    2006;7(5):392401.

    23. Nomiya T, Nemoto K, Kumabe T, et al. Prognostic

    significance of surgery and radiation therapy in

    cases of anaplastic astrocytoma: retrospective

    analysis of 170 cases. J Neurosurg 2007;106(4):

    57581.

    24. Quigley MR, Flores N, Maroon JC, et al. Value of

    surgical intervention in the treatment of glioma.Stereotact Funct Neurosurg 1995;65(14):1715.

    25. Slotman BJ, Kralendonk JH, van Alphen HA, et al.

    Hypofractionated radiation therapy in patients

    with glioblastoma multiforme: results of treatment

    and impact of prognostic factors. Int J Radiat On-

    col Biol Phys 1996;34(4):8958.

    26. Stark AM, Nabavi A, Mehdorn HM, et al. Glioblas-

    toma multiforme-report of 267 cases treated at

    a single institution. Surg Neurol 2005;63(2):1629

    [discussion: 169].

    27. Albert FK, Forsting M, Sartor K, et al. Early postop-

    erative magnetic resonance imaging after resection

    of malignant glioma: objective evaluation of residual

    tumor and its influence on regrowth and prognosis.

    Neurosurgery 1994;34(1):4560 [discussion: 1].

    28. Ammirati M, Vick N, Liao YL, et al. Effect of the

    extent of surgical resection on survival and quality

    of life in patients with supratentorial glioblastomas

    and anaplastic astrocytomas. Neurosurgery 1987;

    21(2):2016.

    29. Devaux BC, OFallon JR, Kelly PJ. Resection,

    biopsy, and survival in malignant glial neoplasms.

    A retrospective study of clinical parameters,therapy, and outcome. J Neurosurg 1993;78(5):

    76775.

    30. Jeremic B, Grujicic D, Antunovic V, et al. Hyper-

    fractionated radiation therapy followed by multi-

    agent chemotherapy in patients with malignant

    glioma: a phase II study. Int J Radiat Oncol Biol

    Phys 1994;30:117985.

    31. Kelly PJ, Hunt C. The limited value of cytoreductive

    surgery in elderly patients with malignant gliomas.

    Neurosurgery 1994;34(1):626 [discussion: 667].

    32. Ushio Y, Kochi M, Hamada J, et al. Effect of

    surgical removal on survival and quality of life in

    patients with supratentorial glioblastoma. Neurol

    Med Chir (Tokyo) 2005;45(9):45460 [discussion:

    4601].

    33. Vuorinen V, Hinkka S, Farkkila M, et al. Debulking or

    biopsy of malignant glioma in elderly people -

    a randomised study. Acta Neurochir (Wien) 2003;

    145(1):510.

    34. Shibamoto Y, Yamashita J, Takahashi M, et al.

    Supratentorial malignant glioma: an analysis of

    radiation therapy in 178 cases. Radiother Oncol

    1990;18(1):917.35. Simpson JR, Horton J, Scott C, et al. Influence of

    location and extent of surgical resection on survival

    of patients with glioblastoma multiforme: results of

    three consecutive Radiation Therapy Oncology

    Hoover et al420

  • 8/4/2019 Clinical Trials in Brain

    13/16

    Group (RTOG) clinical trials. Int J Radiat Oncol Biol

    Phys 1993;26(2):23944.

    36. Vecht CJ, Avezaat CJ, van Putten WL, et al. The

    influence of the extent of surgery on the neurolog-

    ical function and survival in malignant glioma. A

    retrospective analysis in 243 patients. J Neurol

    Neurosurg Psychiatry 1990;53(6):46671.37. Winger MJ, Macdonald DR, Cairncross JG. Supra-

    tentorial anaplastic gliomas in adults. The prog-

    nostic importance of extent of resection and prior

    low-grade glioma. J Neurosurg 1989;71(4):48793.

    38. Shinoda J, Sakai N, Murase S, et al. Selection of

    eligible patients with supratentorial glioblastoma

    multiforme for gross total resection. J Neurooncol

    2001;52(2):16171.

    39. Keles GE, Chang EF, Lamborn KR, et al. Volumetric

    extent of resection and residual contrast enhance-

    ment on initial surgery as predictors of outcome in

    adult patients with hemispheric anaplastic astrocy-

    toma. J Neurosurg 2006;105(1):3440.

    40. Pope WB, Sayre J, Perlina A, et al. MR imaging

    correlates of survival in patients with high-grade

    gliomas. AJNR Am J Neuroradiol 2005;26(10):

    246674.

    41. Schmidt M, Berger M, Lamborn K, et al. Repeated

    operations for infiltrative low-grade gliomas

    without intervening therapy. J Neurosurg 2003;

    98:11659.

    42. Frappaz D, Chinot O, Bataillard A, et al. Summary

    version of the standards, options and recommen-dations for the management of adult patients with

    intracranial glioma (2002). Br J Cancer 2003;

    89(Suppl 1):S7383.

    43. Team L-GGG. Practice parameters in adults with

    suspected or known supratentorial nonoptic

    pathway low-grade glioma. Neurosurg Focus

    1998;4:e10.

    44. Keles G, Lamborn K, Berger M. Low-grade hemi-

    spheric gliomas in adults: a critical review of extent

    of resection as a factor influencing outcome. J Neu-

    rosurg 2001;95:73545.

    45. Claus E, Horlacher A, Hsu L, et al. Survival rates in

    patients with low-grade glioma after intraoperative

    magnetic resonance image guidance. Cancer

    2005;103:122733.

    46. Johannesen T, Langmark F, Lote K. Progress in

    long-term survival in adult patients with supratento-

    rial low-grade gliomas: a population-based study

    of 993 patients in whom tumors were diagnosed

    between 1970 and 1993. J Neurosurg 2003;99:

    85462.

    47. Leighton C, Fisher B, Bauman G, et al. Supratento-

    rial low-grade glioma in adults: an analysis of prog-nostic factors and timing of radiation. J Clin Oncol

    1997;15(4):1294301.

    48. Nakamura M, Konishi N, Tsunoda S, et al. Analysis

    of prognostic and survival factors related to

    treatment of low-grade astrocytomas in adults.

    Oncology 2000;58(2):10816.

    49. Philippon JH, Clemenceau SH, Fauchon FH, et al.

    Supratentorial low-grade astrocytomas in adults.

    Neurosurgery 1993;32(4):5549.

    50. Rajan B, Pickuth D, Ashley S, et al. The manage-

    ment of histologically unverified presumed cerebralgliomas with radiotherapy. Int J Radiat Oncol Biol

    Phys 1994;28(2):40513.

    51. Shaw E, Arusell R, Scheithauer B, et al .

    Prospective randomized trial of low- versus

    high-dose radiation therapy in adults with supra-

    tentorial low-grade glioma: initial report of a North

    Central Cancer Treatment Group/Radiation

    Therapy Oncology Group/Eastern Cooperative

    Oncology Group study. J Clin Oncol 2002;

    20(9):226776.

    52. van Veelen M, Avezaat C, Kros J, et al. Supratento-

    rial low grade astrocytoma: prognostic factors,

    dedifferentiation, and the issue of early versus

    late surgery. J Neurol Neurosurg Psychiatry 1998;

    64:5817.

    53. Yeh SA, Ho JT, Lui CC, et al. Treatment outcomes

    and prognostic factors in patients with supratento-

    rial low-grade gliomas. Br J Radiol 2005;78(927):

    2305.

    54. Smith JS, Chang EF, Lamborn KR, et al. Role of

    extent of resection in the long-term outcome of

    low-grade hemispheric gliomas. J Clin Oncol

    2008;26(8):133845.55. Shaw EG, Berkey B, Coons SW, et al. Recurrence

    following neurosurgeon-determined gross-total

    resection of adult supratentorial low-grade-glioma:

    results of a prospective clinical trial. J Neurosurg

    2008;109(5):83541.

    56. Yeh DD, Warnick RE, Ernst RJ. Management

    strategy for adult patients with dorsal midbrain

    gliomas. Neurosurgery 2002;50(4):7358 [discus-

    sion: 73840].

    57. Chang SM, Nelson S, Vandenberg S, et al. Integra-

    tion of preoperative anatomic and metabolic phys-

    iologic imaging of newly diagnosed glioma.

    J Neurooncol 2009;92(3):40115.

    58. Dowling C, Bollen AW, Noworolski SM, et al. Preop-

    erative proton MR spectroscopic imaging of brain

    tumors: correlation with histopathologic analysis

    of resection specimens. AJNR Am J Neuroradiol

    2001;22(4):60412.

    59. Lupo JM, Cha S, Chang SM, et al. Dynamic

    susceptibility-weighted perfusion imaging of high-

    grade gliomas: characterization of spatial hetero-

    geneity. AJNR Am J Neuroradiol 2005;26(6):

    144654.60. Kwong K, Belliveau J, Chesler D, et al. Dynamic

    magnetic resonance imaging of human brain

    activity during primary sensory stimulation. Proc

    Natl Acad Sci U S A 1992;89:56759.

    Clinical Trials in Brain Tumor Surgery 421

  • 8/4/2019 Clinical Trials in Brain

    14/16

    61. Basser P, Jones D. Diffusion-tensor MRI: theory,

    experimental design and data analysis - a technical

    review. NMR Biomed 2002;15:45667.

    62. Binder J, Rao S, Hammeke T, et al. Lateralized

    human brain language systems demonstrated by

    task subtraction functional magnetic resonance

    imaging. Arch Neurol 1995;52:593601.63. Bittar R, Olivier A, Sadikot A, et al. Presurgical

    motor and somatosensory cortex mapping with

    functional magnetic resonance imaging and posi-

    tron emission tomography. J Neurosurg 1999;91:

    91521.

    64. Nimsky C, Ganslandt O, Fahlbusch R. Comparing

    0.2 tesla with 1.5 tesla intraoperative magnetic

    resonance imaging analysis of setup, workflow,

    and efficiency. Acad Radiol 2005;12(9):106579.

    65. Berman J, Berger M, Mukherjee P, et al. Diffusion-

    tensor imaging-guided tracking of fibers of the

    pyramidal tract combined with intraoperative

    cortical stimulation mapping in patients with

    gliomas. J Neurosurg 2004;101:6672.

    66. Roberts T, Disbrow E, Roberts H, et al. Quantifica-

    tion and reproducibility of tracking cortical extent of

    activation by use of functional MR imaging and

    magnetoencephalography. AJNR Am J Neurora-

    diol 2000;21:137787.

    67. Nimsky C, Ganslandt O, Hastreiter P, et al. Preop-

    erative and intraoperative diffusion tensor

    imaging-based fiber tracking in glioma surgery.

    Neurosurgery 2007;61(Suppl 1):17885 [discus-sion: 86].

    68. Nagarajan S, Kirsch H, Lin P, et al. Preoperative

    localization of hand motor cortex by adaptive

    spatial filtering of magnetoencephalography data.

    J Neurosurg 2008;109(2):22837.

    69. Gaetz W, Cheyne D, Rutka JT, et al. Presurgical

    localization of primary motor cortex in pediatric

    patients with brain lesions by the use of spatially

    filtered magnetoencephalography. Neurosurgery

    2009;64(Suppl 3):17785 [discussion: 186].

    70. Berger MS, Hadjipanayis CG. Surgery of intrinsic

    cerebral tumors. Neurosurgery 2007;61(Suppl 1):

    279304 [discussion: 305].

    71. Litofsky NS, Bauer AM, Kasper RS, et al. Image-

    guided resection of high-grade glioma: patient

    selection factors and outcome. Neurosurg Focus

    2006;20(4):E16.

    72. Keles G, Lamborn K, Berger M. Coregistration

    accuracy and detection of brain shift using intrao-

    perative sononavigation during resection of hemi-

    spheric tumors. Neurosurgery 2003;53:55662.

    73. Ram Z, Hadani M. Intraoperative imagingMRI.

    Acta Neurochir Suppl 2003;88:14.74. Benveniste R, Germano IM. Evaluation of factors

    predicting accurate resection of high-grade

    gliomas by using frameless image-guided stereo-

    tactic guidance. Neurosurg Focus 2003;14(2):e5.

    75. Nimsky C, Ganslandt O, Tomandl B, et al. Low-field

    magnetic resonance imaging for intraoperative use

    in neurosurgery: a 5-year experience. Eur Radiol

    2002;12(11):2690703.

    76. Senft C, Seifert V, Hermann E, et al. Usefulness of

    intraoperative ultra low-field magnetic resonance

    imaging in glioma surgery. Neurosurgery 2008;63(4 Suppl 2):25766 [discussion: 2667].

    77. Schneider JP, Trantakis C, Rubach M, et al. Intrao-

    perative MRI to guide the resection of primary

    supratentorial glioblastoma multiformea quantita-

    tive radiological analysis. Neuroradiology 2005;

    47(7):489500.

    78. Kaibara T, Saunders JK, Sutherland GR. Advances

    in mobile intraoperative magnetic resonance

    imaging. Neurosurgery 2000;47(1):1317 [discus-

    sion: 1378].

    79. Hatiboglu MA, Weinberg JS, Suki D, et al. Impact

    of intraoperative high-field magnetic resonance

    imaging guidance on glioma surgery: a prospective

    volumetric analysis. Neurosurgery 2009;64(6):

    107381 [discussion: 1081].

    80. Bergsneider M, Sehati N, Villablanca P, et al. Maha-

    ley Clinical Research Award: extent of glioma

    resection using low-field (0.2 T) versus high-field

    (1.5 T) intraoperative MRI and image-guided

    frameless neuronavigation. Clin Neurosurg 2005;

    52:38999.

    81. Penfield W, Boldrey E. Somatic motor and sensory

    representation in the cerebral cortex of man asstudied by electrical stimulation. Brain 1937;60:

    389443.

    82. Berger M, Cohen W, Ojemann G. Correlation of

    motor cortex brain mapping data with magnetic

    resonance imaging. J Neurosurg 1990;72:3837.

    83. Ojemann G, Ojemann J, Lettich E, et al. Cortical

    language localization in left, dominant hemisphere.

    An electrical stimulation mapping investigation in

    117 patients. J Neurosurg 1989;71(3):31626.

    84. Skirboll S, Ojemann G, Berger M, et al. Functional

    cortex and subcortical white matter located within

    gliomas. Neurosurgery 1996;38:67884.

    85. Keles G, Lundin D, Lamborn K, et al. Intraopera-

    tive subcortical stimulation mapping for hemi-

    spherical perirolandic gliomas located within or

    adjacent to the descending motor pathways:

    evaluation of morbidity and assessment of func-

    tional outcome in 294 patients. J Neurosurg

    2004;100:36975.

    86. Sanai N, Mirzadeh Z, Berger MS. Functional

    outcome after language mapping for glioma resec-

    tion. N Engl J Med 2008;358(1):1827.

    87. Stummer W, Novotny A, Stepp H, et al. Fluores-cence-guided resection of glioblastoma multiforme

    by using 5-aminolevulinic acid-induced porphy-

    rins: a prospective study in 52 consecutive

    patients. J Neurosurg 2000;93(6):100313.

    Hoover et al422

  • 8/4/2019 Clinical Trials in Brain

    15/16

    88. Stummer W, Reulen HJ, Meinel T, et al. Extent of

    resection and survival in glioblastoma multiforme:

    identification of and adjustment for bias. Neurosur-

    gery 2008;62(3):56476 [discussion: 576].

    89. McDermott M, Berger M, Kunwar S, et al. Stereo-

    tactic radiosurgery and interstitial brachytherapy

    for glial neoplasms. J Neurooncol 2004;69:83100.90. Laperriere NJ, Leung PM, McKenzie S, et al.

    Randomized study of brachytherapy in the initial

    management of patients with malignant astrocy-

    toma. Int J Radiat Oncol Biol Phys 1998;41(5):

    100511.

    91. Larson D, Suplica J, Chang S, et al. Permanent

    iodine 125 brachytherapy in patients with progres-

    sive or recurrent glioblastoma multiforme. Neuro

    Oncol 2004;6:11926.

    92. Patel S, Breneman J, Warnick R, et al. Permanent

    iodine-125 interstitial implants for the treatment of

    recurrent glioblastoma multiforme. Neurosurgery

    2000;46:11238.

    93. Tatter S, Shaw E, Rosenblum M, et al. An inflatable

    balloon catheter and liquid 125I radiation source

    (GliaSite Radiation Therapy System) for treatment

    of recurrent malignant glioma: multicenter safety

    and feasibility trial. J Neurosurg 2003;99:297303.

    94. Chan TA, Weingart JD, Parisi M, et al. Treatment of

    recurrent glioblastoma multiforme with GliaSite bra-

    chytherapy. Int J Radiat Oncol Biol Phys 2005;

    62(4):11339.

    95. Gabayan AJ, Green SB, Sanan A, et al. GliaSitebrachytherapy for treatment of recurrent malignant

    gliomas: a retrospective multi-institutional analysis.

    Neurosurgery 2006;58(4):7019 [discussion: 9].

    96. Walter KA, Tamargo RJ, Olivi A, et al. Intratumoral

    chemotherapy. Neurosurgery 1995;37(6):112845.

    97. Brem H, Mahaley MS Jr, Vick NA, et al. Interstitial

    chemotherapy with drug polymer implants for the

    treatment of recurrent gliomas. J Neurosurg 1991;

    74(3):4416.

    98. Westphal M, Hilt D, Bortey E, et al. A phase 3 trial of

    local chemotherapy with biodegradable carmus-

    tine (BCNU) wafers (Gliadel wafers) in patients

    with primary malignant glioma. Neuro Oncol

    2003;5:7988.

    99. Westphal M, Ram Z, Riddle V, et al. Gliadel wafer in

    initial surgery for malignant glioma: long-term

    follow-up of a multicenter controlled trial. Acta Neu-

    rochir (Wien) 2006;148:26975.

    100. Attenello FJ, Mukherjee D, Datoo G, et al. Use of

    Gliadel (BCNU) wafer in the surgical treatment of

    malignant glioma: a 10-year institutional experi-

    ence. Ann Surg Oncol 2008;15(10):288793.

    101. Bobo RH, Laske DW, Akbasak A, et al. Convection-enhanced delivery of macromolecules in the brain.

    Proc Natl Acad Sci U S A 1994;91(6):207680.

    102. Lieberman DM, Laske DW, Morrison PF, et al.

    Convection-enhanced distribution of large

    molecules in gray matter during interstitial drug

    infusion. J Neurosurg 1995;82(6):10219.

    103. Vogelbaum MA, Sampson JH, Kunwar S, et al.

    Convection-enhanced delivery of cintredekin besu-

    dotox (Interleukin-13-Pe38qqr) followed by radia-

    tion therapy with and without temozolomide in

    newly diagnosed malignant gliomas: phase 1 studyof final safety results. Neurosurgery 2007;61(5):

    10317.

    104. Kunwar S, Prados MD, Chang SM, et al. Direct intra-

    cerebral delivery of cintredekin besudotox (IL13-

    PE38QQR) in recurrent malignant glioma: a report

    by the Cintredekin Besudotox Intraparenchymal

    Study Group. J Clin Oncol 2007;25(7):83744.

    105. Parney IF, Kunwar S, McDermott M, et al. Neurora-

    diographic changes following convection-

    enhanced delivery of the recombinant cytotoxin

    interleukin 13-PE38QQR for recurrent malignant

    glioma. J Neurosurg 2005;102(2):26775.

    106. Sampson J, Brady M, Petry N, et al. Intracerebral

    infusate distribution by convection-enhanced

    delivery in humans with malignant gliomas:

    descriptive effects of target anatomy and catheter

    positioning. Neurosurgery 2007;60(2 Suppl 1):

    ONS8998.

    107. Kaiser MG, Parsa AT, Fine RL, et al. Tissue distribu-

    tion and antitumor activity of topotecan delivered

    by intracerebral clysis in a rat glioma model. Neuro-

    surgery 2000;47(6):13918 [discussion: 13989].

    108. Forsyth P, Roldan G, George D, et al. A phase I trialof intratumoral administration of reovirus in patients

    with histologically confirmed recurrent malignant

    gliomas. Mol Ther 2008;16(3):62732.

    109. Chiocca EA, Abbed KM, Tatter S, et al. A phase I

    open-label, dose-escalation, multi-institutional trial

    of injection with an E1B-Attenuated adenovirus,

    ONYX-015, into the peritumoral region of recurrent

    malignant gliomas, in the adjuvant setting. Mol

    Ther 2004;10(5):95866.

    110. Liu TC, Galanis E, Kirn D. Clinical trial results with

    oncolytic virotherapy: a century of promise,

    a decade of progress. Nat Clin Pract 2007;4(2):

    10117.

    111. Liu C, Sarkaria JN, Petell CA, et al. Combination of

    measles virus virotherapy and radiation therapy

    has synergistic activity in the treatment of glioblas-

    toma multiforme. Clin Cancer Res 2007;13(23):

    715565.

    112. Phuong LK, Allen C, Peng KW, et al. Use of

    a vaccine strain of measles virus genetically engi-

    neered to produce carcinoembryonic antigen as

    a novel therapeutic agent against glioblastoma

    multiforme. Cancer Res 2003;63(10):24629.113. Myers R, Harvey M, Kaufmann TJ, et al. Toxi-

    cology study of repeat intracerebral administra-

    tion of a measles virus derivative producing

    carcinoembryonic antigen in rhesus macaques

    Clinical Trials in Brain Tumor Surgery 423

  • 8/4/2019 Clinical Trials in Brain

    16/16

    in support of a phase I/II clinical trial for patients

    with recurrent gliomas. Hum Gene Ther 2008;

    19(7):6908.

    114. Ram Z, Westphal M, Warnke P, et al. Treatment of

    primary high-grade glioma with sitimagene

    ceradenovec (Cerepro) and ganciclovir gene

    medicine: update of phase III results. Abstract

    presentation at the 2009 Joint Meeting of the

    Society for Neuro-Oncology and the AANS/CNS

    Section on Tumors.

    Hoover et al424