Brain Tumor Outcome Studies

Published on 13/03/2015 by admin

Filed under Neurosurgery

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1344 times

CHAPTER 113 Brain Tumor Outcome Studies

Design and Interpretation

This chapter addresses issues in the design and interpretation of clinical studies in patients with brain tumors. Although the main focus is on surgical studies, most modern treatment of brain tumors is multidisciplinary, and some aspects of outcome studies for adjuvant treatments such as radiotherapy, chemotherapy, and immunotherapy are covered briefly. The goal of the chapter is to indicate the principles of sound clinical trial design for individuals who will be planning, conducting, reporting, or interpreting the results of brain tumor outcome studies. Because excellent texts on general and oncologic clinical trials already exist,17 this chapter concentrates on issues that are of special importance in brain tumor trials, especially those that involve neurosurgeons.8,9 Some elements common to all clinical studies are addressed first—defining the patient population, the treatment itself, and the end points used to measure benefit and harm from the treatment. Then, issues specific to different types of trial designs, tumor histologies, types of therapy, and delivery of therapy are covered in turn.

Defining the Patient Population

Age

Patients enrolled in most neurosurgical clinical studies are described by age, but this is particularly essential in brain tumor reports because age is such an important prognostic factor for most types of brain tumor. For malignant and low-grade glioma, for example, older patients have much shorter survival for reasons that are still not entirely clear.10 Older patients with malignant glioma are less likely than younger patients to have known low-grade tumors preceding their diagnosis of malignancy, more likely to have PTEN (phosphatase and tensin homologue from chromosome 10) abnormalities, and less likely to have p53 mutations.11 Pediatric malignant glioma patients have longer median survival than do young adults.12 Conversely, in medulloblastoma, patients younger than 2 years have shorter survival, partially because of reluctance to subject such young children to aggressive central nervous system radiotherapy, but also probably because of more biologically aggressive disease.13,14 For both acoustic neuromas and hemangioblastomas, younger patients with apparently isolated sporadic tumors are much more likely to have occult neurofibromatosis type 2 and von Hippel-Lindau disease, respectively, which potentially affects long-term outcome.15,16 Age at diagnosis or initiation of treatment and age limitations on eligibility for prospective studies should be carefully documented in all brain tumor reports.

Histology

In 1932, Harvey Cushing announced to a hushed crowd that he would never again attempt to report on the subject of brain tumor outcomes as a whole.17 Almost all subsequent brain tumor outcome studies have followed Cushing’s example in being limited to a single tumor histology and most to a single point in the course of the disease, such as at diagnosis or first recurrence. This seems straightforward enough, except that histologic definitions have changed over time, some diagnoses are highly inconsistent from pathologist to pathologist, and some are dependent on the amount of tissue sampled at surgery (or when diagnoses are based on imaging alone).

The present standard reference for describing the histology of brain tumors is the 2007 histologic classification of the World Health Organization (WHO).1820 In interpreting older studies, temporal changes in grading systems should be kept in mind. For glioblastoma, newer grading systems no longer mandate the presence of tissue necrosis, which was formerly required to make the diagnosis.18,21,22 This change should have little effect on most studies because the majority of glioblastomas still contain necrosis and prognoses are similar for patients whose glioblastomas do or do not contain necrosis.23 However, for studies that focus specifically on long-term glioblastoma survivors, the evolution of neuropathologic criteria constantly forces a re-evaluation of earlier research. This is so because some patients in whom glioblastoma was originally diagnosed are now recognized to have other tumors by current criteria, such as some anaplastic oligodendrogliomas that contain vascular proliferation or benign tumors whose histopathologic appearance formerly qualified for a diagnosis of glioblastoma. For example, increased awareness of the relatively new diagnosis of pleomorphic xanthoastrocytoma required removal of some patients from one long-term glioblastoma survivor series,24 and recognition of malignant oligodendroglioma elements caused another group to reclassify a third of their long-term glioblastoma survivors.25 Only 17% of long-term glioblastoma survivors in Sweden from 1958 to 1977 were not reclassified on later re-examination.26

At the other end of the glioma spectrum, the identification of pilomyxoid astrocytomas in 1999 caused some poor-prognosis patients to be reclassified from earlier pilocytic astrocytoma series,27,28 just as increasing recognition of atypical teratoma-rhabdoid tumors has removed some of the worst-prognosis patients from modern medulloblastoma series.29,30 Medulloblastoma series are also variably affected by categorization into subdiagnoses, such as desmoplastic, anaplastic, and nodular medulloblastoma.19

For lower grade gliomas, changes in formal grading systems rather than the development of new diagnoses have been more significant. Although the classic histologic criteria for defining oligodendroglial portions of a tumor have generally been established, the subjective nature of their application (in the absence of an immunohistochemical marker), in the context of intense clinical interest in oligodendroglial sensitivity to chemotherapy,31 has resulted in important epidemiologic shifts in the frequency of diagnosing oligodendroglioma in the past 2 decades, with a remarkable increase in the proportion of low-grade tumors now thought to contain oligodendroglial elements.32 The most recent WHO revision has introduced a new grade IV diagnosis of “glioblastoma with an oligodendroglioma component” for many tumors that were previously diagnosed as grade III anaplastic oligodendrogliomas.18,19 The impact of these changes on outcome studies is complex and difficult to predict. Revisions in the WHO definition of atypical meningioma have also been shown to have a concrete impact on the recommended treatment of many patients at certain centers.3335

Interpathologist disagreement on brain tumor diagnoses is sufficiently common to require central review for many studies. Discordant results on central review are more common with diagnoses originating at community hospitals, especially those without a neuropathologist, and affect some diagnoses more than others.3638 For example, in the San Francisco Bay area, Aldape and coauthors reported more than 50% discordant results on central review of tumors diagnosed as containing an oligodendroglial component, thus indicating that the distinction between oligodendroglial and astrocytic histology is an especially subjective one.38 The diagnosis of glioblastoma in this study was much more reliable. For studies using data sources that include many community hospital diagnoses, such as population-based case registries, it has been recommended that all diagnoses except glioblastoma and meningioma be subjected to central pathology review whenever possible.36 Most multicenter randomized studies of brain tumor treatment now require central pathology review before registration.

An important consideration in studies on glioma surgery is that the pathologic diagnosis may vary with the extent of tumor resection because of the combination of sampling bias and the high degree of spatial pathologic heterogeneity in many gliomas. Glioma grading systems are based on counting individual malignant features that may be seen in successive microscopic fields examined, so the idea that more tissue examined leads to a more malignant diagnosis makes intrinsic sense. This has been demonstrated in clinical studies comparing rates of high-grade diagnoses in patient cohorts that differ in the extent of resection,39 as well as in studies comparing needle biopsy diagnoses with later resections, especially in larger tumors.4042 Similarly, within glioblastoma, detection of necrosis is more common with more extensive resection.23 Less widely recognized is the tendency for more extensive resections to produce more histologic diagnoses that include an oligodendroglial component. Perry and colleagues described a cohort of grade 3 gliomas in which the percentage containing an oligodendroglial component increased from 3% in biopsied tumors to 29% in subtotal resection specimens and 43% in gross total resections.43 Similar results have been described for grade 2 gliomas, both in a single-institution study44 (46% oligodendroglial-containing tumors in biopsy specimens, 89% in gross total resections) and in a population-based study45 (32% oligodendroglial-containing tumors in biopsy specimens, 62% in resection specimens). Because of sampling bias, when one group in a treatment comparison selectively contains patients with less extensive surgical resection, the pathologic diagnoses in that group will be biased toward lower grade histology and less common oligodendroglial diagnoses than will the true pathology.

Studies in which some or all patients are enrolled on the basis of imaging diagnoses alone, without histologic confirmation, face special problems in reliably defining diagnostic categories. Radiosurgery series and series of tumors undergoing observation without other treatment are particularly dependent on accurate imaging diagnoses. Randomized clinical trials (RCTs) in which patients are registered before surgery provide some insight into the error rates of imaging diagnoses. For example, in three RCTs of patients undergoing resection for single metastases, all of whom had single brain lesions and established diagnoses of systemic cancer, 7 of 127 presumed metastases (6%) proved to be other lesions (gliomas, abscesses, or inflammatory changes)4648; in a phase II trial involving implantation of carmustine (BCNU)-impregnated wafers for single metastases with presurgical enrollment that did not require a previous history of cancer, 6 of 35 patients (17%) had pathology other than metastasis.49 These results are relevant to radiosurgical series for single brain metastases. Nonenhancing cerebral masses are usually thought to represent low-grade gliomas, but up to a third will be malignant gliomas, especially in older patients,50 and many low-grade gliomas enhance on magnetic resonance imaging (MRI), a feature that characteristically portends a poorer prognosis.51,52 An observational study of “low-grade gliomas” that is limited to nonenhancing tumors will therefore include some malignant gliomas and exclude some poor-prognosis low-grade tumors, thus making the combined effect of the two biases difficult to predict. Treatment of malignant gliomas without a histologic diagnosis is less common, but knowing the rate of misdiagnosis based on imaging can assist in power calculations for trials. Of 30 patients enrolled in an RCT of glioblastoma resection based on characteristic images,53 7 (23%) had nonglioma lesions and another 4 had grade 3 gliomas. In a larger study that required an MRI diagnosis of contrast-enhancing glioma amenable to complete resection for enrollment, 34 of 322 patients (11%) had nonglioma pathology; 97% of the remaining tumors were grade 4 astrocytomas.54

Defining the stage of disease (i.e., at diagnosis versus recurrent) is generally straightforward. However, care should be exercised in defining the date of diagnosis in glioblastoma studies that include (or exclude55) patients with previous known low-grade tumors (“secondary glioblastomas”). Defining the date of recurrence in treated malignant gliomas is discussed later in the chapter.

Composite Baseline Risk Scores—Recursive Partitioning Analysis and Others

Several specially developed composite baseline risk classifications have found widespread use in reporting treatment of glioma, metastasis, and medulloblastoma. Baseline prognostic classifications or scores are useful in constructing historical control groups for case series without internal controls and for stratifying randomization in RCTs so that both treatment groups will contain patients with comparable prognoses given standard treatment.

For malignant glioma patients, the 1993 Radiation Therapy and Oncology Group (RTOG) recursive partitioning analysis (RPA)56 is the most commonly used prognostic classification. This scheme uses patient age, functional status (Karnofsky performance scale [KPS] and work ability), mental status and neurological function, length of symptoms, type of surgery, radiation dose delivered, and tumor histology to define six patient groups with distinctly different survival prognoses. The U.K. Medical Research Council developed and validated a similar six-group classification of malignant glioma patients that has been less used widely,57,58 and a glioblastoma-specific RPA has also been published.59 For patients with low-grade glioma, Pignatti and coworkers identified five baseline prognostic factors (age >40, astrocytoma histology, tumor diameter >6 cm, tumor crossing the midline, and presence of a neurological deficit before surgery) and split patients into two prognostic groups, low risk (two or fewer factors) and high risk (three or more factors).60 Brain metastasis patients are classified with another RTOG RPA scheme that uses patient age, presence of metastases outside the brain, and state of control of the primary tumor to define three prognostic groups.61,62 A 2008 revision of RPA, called graded prognostic assessment (GPA), was developed by the RTOG to be easier to use and less subjective than RPA.63 GPA uses patient age, KPS, number of brain metastases, and the presence of known extracranial metastases to define four prognostic classes, as opposed to three for RPA. The authors found that GPA was as prognostic as RPA and superior to other published prognostic indexes for brain metastasis patients, but GPA has not yet been validated on other data sets. For medulloblastoma, various means of combining baseline risk factors, including age, dissemination, and residual disease after surgery, have been used to construct “average-risk” and “poor-risk” groups.14

Prognostic indexes are only valid to construct historical control patient sets that share diagnostic, prognostic, and treatment characteristics with the original cohort from which the index was developed, and they should not be used as comparisons for patient cohorts that differ substantially from the original cohort. For example, the RTOG conducted a phase II trial of interferon beta-1a after adjuvant radiation therapy for malignant glioma patients in which eligible patients were those who had stable disease at the completion of radiation treatment.64 Initial analyses showed superiority over expected survival with RPA, but the comparison was invalid because patients whose tumors progressed during radiation therapy (a marker for poor prognosis)65 were included in the RPA but excluded from the trial (about a third of the original cohort). A matched-pair analysis that accounted for disease stability during radiation therapy showed no difference between survival of interferon-treated patients in the trial and appropriate historical controls. Similarly, malignant glioma trials that stipulate gross total resection for entry (such as many immunotherapy trials) require a special prognostic index constructed from similar patients.66 When the standard of care shifts for a disease, as it has recently for malignant glioma because of the introduction of adjuvant temozolomide,67 new indexes must be developed.

A basic characteristic of all such prognostic indexes in current use is that they were developed from patients enrolled in prospective clinical trials. In the United States, only 2% to 3% of patients with malignant glioma enter clinical trials (Barker FG, Curry WT, unpublished data). It is well known that trial patients have decidedly favorable prognostic factors in comparison to population-based cohorts because of trial eligibility requirements, self-selection, and social factors.6870 Valid historical controls for population-based patient cohorts cannot be generated with the standard trial-based prognostic indexes. Conversely, single-center trial patients usually have even better baseline prognostic factors than do patients in multicenter trials, which partially accounts for the tendency of many therapies that appear promising in early single-center trials to fail in RCTs7072; this renders the use of multicenter trial–based prognostic indexes suspect as historical controls for single-center trial reports that use survival end points.

Other Factors

All brain tumor studies should include functional status in baseline descriptions because it is a very strong predictor of both outcome and survival.10 Commonly used scales include the KPS73 and European Cooperative Oncology Group (ECOG) scale.74 For special tumor types, other baseline neurological status needs to be specified. An example is preoperative hearing status for acoustic neuromas, which is usually described in terms of the Gardner-Robertson classification,75 a strong predictor of hearing preservation results. This same scale is used for studies of radiosurgical treatment. Many specific neurological measures are more often used to describe complications of surgery (such as facial nerve weakness)76 and are described later. Tumor size, location, focality and multiplicity, and proximity to or involvement of eloquent structures all contribute to the concept of “resectability,” an important prognostic factor described subsequently.

In metastasis studies, additional important baseline variables include histology and time since diagnosis of the primary tumor (longer intervals suggest a better outcome),77 “expected survival” at enrollment (most surgical trials require a minimum expected survival, although physicians are notoriously bad at such predictions7880), and whether the systemic cancer is stable or progressive (a very subjective determination with no known guidelines). Despite their subjective and imprecise nature, all these factors are strong predictors of actual patient survival.

Describing Treatment—Extent of Resection

Descriptions of chemotherapy or radiation treatment of brain tumors follow established practice in terms of doses, schedules, and treatment fields, but how to describe surgical treatment is less well established. The main variable of interest is typically the extent of resection. Older studies relied on the surgeon’s operative notes to grade the extent of resection, but with more routine use of early postoperative imaging it became clear that surgeons’ assessments are much too optimistic.81,82 Many studies base grading the extent of resection on preoperative and postoperative images by using scales such as “biopsy, subtotal resection, gross total resection” and give percentages of resection to define the cutoff points between the grades (such as >90% resection defining gross total resection),65,66 but the method of deriving the percentage of resection is not usually specified, the number of grades in the system can vary (such as “partial, subtotal, total” excision), and the cutoff points between grades vary substantially between studies.83,84 The extent of resection has also been graded by using the greatest linear dimension of residual disease on cross-sectional images85 or commonly by subjective visual impression of overall volumetric change based on imaging studies without formal cutoff points between grades.

In modern studies the extent of resection is typically defined and reported in two ways with the use of volumetric MRI: by the volume of residual tumor and as a percentage of resection based on preoperative and postoperative volumes.8688 In high-grade tumors, the enhancing volume on T1-weighted images is defined as the tumor volume, and for nonenhancing tumors, the T2-bright volume is used. Early postoperative imaging (i.e., within 48 or 72 hours) is important because of the rapid development of contrast enhancement in the margin of cerebral resections, as confirmed by imaging studies after resection for non-neoplastic indications and probably representing infarcted tissue.8991 Although “complete” or “gross total” resection is often defined as the absence of all enhancing postoperative disease on images, in practice some reasonable threshold for minimal residual disease should be defined.

Even though removal of all tumor volume according to the aforementioned definitions is a reasonable surgical goal, it is well established that tissue surrounding a glioma that appears normal or has T2-bright signal without enhancement (“swelling”) contains pathologically detectable tumor cells,92 and pattern-of-failure studies demonstrate that most glioma recurrences develop within 2 cm of the tumor margin.85,93 This offers the opportunity for potentially greater treatment effect by performing a resection that includes a volume of apparently normal tissue surrounding the imaging-defined tumor. Although some early RTOG studies included an extent-of-resection group described as “lobectomy,” this was not necessarily synonymous with resection of all tumor plus a defined margin.94 There are currently no accepted standards for defining this type of surgical resection, nor is pathologic examination of surgical margins for tumor cells—a frequent method in general oncologic surgery—in common clinical use by glioma surgeons at present.

Tumors other than gliomas have different schemes for grading the extent of resection. Medulloblastomas typically enhance, and the volumetric extent of resection can be measured as for gliomas. Current standard practice is to use a threshold of 1.5 cm2 of postoperative residual disease (by bidimensional area) to define risk groups.14 For meningiomas, the Simpson class (Table 113-1),95 which uses a five-grade system, is usually reported, with one group defining an additional resection class (“grade 0 resection”) as including gross total resection of convexity meningiomas with a 2-cm normal dural margin.96 Acoustic neuromas are reported as gross total or subtotal resection based on the surgeon’s impression, with some investigators distinguishing between more and less aggressive subtotal resection according to various ad hoc criteria.9799 The limitations of pituitary tumor imaging are such that for endocrine-active tumors, follow-up endocrine testing is usually more sensitive to minimal residual disease; volumetric tumor measures may have a larger role in reports of nonfunctioning macroadenoma.

TABLE 113-1 Simpson’s Classification of Extent of Resection in Meningioma Surgery,95 with Later Addition of “Grade 0” Resection by Kinjo et al96

GRADE DEFINITION
0 Gross total resection of a convexity meningioma plus a 2-cm normal dural margin
1 Gross total resection, including the dural attachment and abnormal bone; resection of the venous sinus if the wall is involved
2 Gross total resection not including the dural attachment, which is “charred” with electrocautery
3 Gross total resection of the mass, dural attachment not resected or “charred”
4 Partial resection of the intradural mass lesion
5 Decompression only, with or without biopsy

Describing Outcomes of Brain Tumor Therapy: End Point Choices and Definitions

End Points for Cancer Studies

A bewildering variety of end points have been used in cancer outcome studies, many of which are useful in brain tumor reports.5,100 Careful selection of end points for both efficacy and toxicity is one of the most important aspects of planning a brain tumor trial. Appropriate end points for reports of disease treatment typically shift over time as those treatments improve. For example, in the treatment of acoustic neuroma, operative mortality has been supplemented in turn by complete resection, anatomic facial nerve preservation, good facial function, and hearing preservation as additional important end points, and with the introduction of radiosurgery, tumor stability has been added.101 Because many cancer innovations eventually involve approval of a drug or product by regulatory authorities, the perspective of the U.S. Food and Drug Administration (FDA) on appropriate end points for cancer product approval will be touched on briefly in this chapter as an example.102104

Survival, Operative Mortality, and Disease-Specific Mortality

The original criterion for success of brain tumor surgery was operative mortality, and overall patient survival is still the “gold standard” for success of therapy for malignant brain tumors, including low-grade gliomas. A survey of neurosurgeons showed that mortality was considered the most important end point far more often than morbidity or quality of life.105 Survival (or overall survival) is usually defined as the time from initial surgery, date of diagnosis, or date of trial registration until death. Events (such as loss to follow-up) that will be considered censoring events in survival analysis should be carefully defined.106 The obvious advantages of overall survival as an end point are its undeniable importance to the patient (within reasonable limits imposed by quality of life) and lack of ambiguity in definition. Governments typically maintain death indices that can be used to obtain follow-up even for patients who have gone missing during the study.107,108 Survival is an ideal end point for diseases with high acute mortality, such as glioblastoma. In diseases with longer expected survival, trials with survival end points must plan for lengthy follow-up (with an increasing amount of patient dropout over time), and when effective salvage therapies are available, the survival benefit of the initial treatment becomes difficult to distinguish. In these situations, progression-free survival (see later) may be a more attractive end point.

In surgical studies, operative mortality is a short-term end point of importance to many investigators. Cushing, in an era when many patients had prolonged hospitalization after brain tumor surgery, used mortality before hospital discharge as his definition of operative mortality.17 Later investigators substituted 30-day mortality as the standard measure to capture events occurring soon after hospital discharge (such as death from pulmonary embolism). With the increasing use of administrative databases for brain tumor outcome studies,109111 mortality before hospital discharge is again frequently reported by investigators. Use of this end point can introduce bias into analyses. For example, studies showed a substantial decrease in in-hospital mortality for patients undergoing brain tumor craniotomy in the United States from 1988 to 2000.109111 However, a Medicare analysis showed that although in-hospital mortality after craniotomy (all types) fell 8.5% between 1998 and 2003, 30-day mortality remained almost unchanged. This strongly suggests that the observed decrease in in-hospital mortality is largely an artifact of decreasing length of hospital stay over the study period.112 In general, when patients with acute conditions remain in the hospital longer (even for administrative reasons), their observed in-hospital mortality will be higher.

In malignant glioma patients, studies show that virtually all deaths result from the primary tumor, so the difference between overall mortality (death from any cause) and disease-specific mortality (death from the brain tumor) is negligible. Although the same holds true for death in patients with neurofibromatosis type 2,113 for patients who undergo operations for sporadic benign tumors such as acoustic neuromas, meningiomas, or pituitary adenomas, most eventual deaths are not directly related to the brain tumor or its treatment. In fact, there is a bias toward better survival in such patients than in the general population because of preferential selection of healthy patients for surgery, as seen in patients with other benign neurosurgical conditions.114116 This bias is related to the “healthy worker” effect in epidemiology—when cases are selected only from a working population, a spurious increase in survival over general population controls is seen.117 In these situations, analysis of overall survival can produce confusing or misleading results. Not surprisingly, patient age at diagnosis is the strongest prognostic factor for survival in studies where few deaths result from the disease under study, such as meningioma.118 Disease-specific survival is the appropriate end point for determining relevant prognostic survival factors in the treatment of benign tumors: in one study of radiation therapy after subtotal resection of benign skull base meningiomas, the 15-year cause-specific survival rate was 92%, but the overall survival rate was just 62%.119

In detailing results, all time-dependent end points, such as survival, completeness of follow-up, and extent of patient dropout, should be reported. Most reports simply state the number of patients lost to follow-up, but when the loss occurred is also important. Better available methods include calculating the maximal theoretically possible follow-up (i.e., all patients monitored from enrollment to the time of analysis) and reporting the actual follow-up achieved as a percentage of the former.120 Another method is the “reverse Kaplan-Meier” method in which the indicator for events and censoring is reversed.120 Loss to follow-up in survival studies is usually nonrandom. Younger patients are more likely to be lost, as are those who have experienced a complication of therapy or other poor outcome,114,121,122 so including only patients with complete follow-up can bias a study toward better results.

Progression-Free Survival, Time to Progression, and Time to Treatment Failure

In many brain tumor treatment studies, progression-free survival is an important end point. It typically reflects a period of relative freedom from symptoms for the patient, in addition to being an excellent surrogate for overall survival.123,124 However, defining tumor progression in brain tumors (especially malignant gliomas) is complex and subjective; definitions of tumor progression and response are discussed later in this chapter. Progression-free survival starts at diagnosis, surgery, or trial enrollment and ends at either tumor progression or death. In contrast, time to progression ends only when tumor progression is actually observed (typically with an imaging study). The difference is important because many patients fail to keep clinical or imaging appointments when their tumors recur, so tumor progression is never formally demonstrated before death. Such patients are censored observations in time-to-progression analyses, which limits the usefulness of this end point. Similarly, “time to treatment failure” ends either at death, at tumor progression, or when treatment is stopped for any other reason, such as toxicity or patient choice. This is only rarely a useful end point in brain tumor trials because efficacy and toxicity are generally better assessed separately.

Quality of Life, Functional Status, and Measures of Symptoms

In recent years there has been increasing emphasis on patient-reported outcomes (PROs) in cancer therapy in addition to length of survival.105,126133 PROs include, in order of increasing specificity, health-related quality of life (HRQoL), functional status, and measures of specific neurological deficits. In the WHO framework for discussing health-related problems, decreases in overall HRQoL correspond roughly to “handicap,” deficits in functional status to “disability,” and specific symptoms to “impairment.” In the United States, the increasing value placed on PROs is reflected by their acceptance by the FDA as supporting evidence in the approval process for a growing number of anticancer drugs.128

Methods for measuring HRQoL are still in the course of development, and specialized texts are available.5,100,134 HRQoL is a measure of an individual’s satisfaction with life insofar as it is affected by health. It excludes many domains of “quality of life” defined generally, such as financial security, political and religious freedom, quality of interpersonal relationships, and so on, except as they are affected by the person’s health status. Generic HRQoL measures are applicable to all individuals regardless of health state; such measures include familiar instruments such as the short form health survey SF-36 (www.sf-36.org). Other measures are designed to capture HRQoL in cancer patients generally, such as the Functional Assessment of Cancer Therapy—General version (FACT-G; www.facit.org) and the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) C30 (www.eortc.be/home/qol/ExplQLQ-C30_1.htm). The highest level of specificity is achieved by HRQoL measures designed specifically for brain tumor patients, such as the brain tumor subscale of the FACT-G, called the FACT-BR,135 and the EORTC QLQ Brain Cancer Module (BN 20).136,137 HRQoL and cognitive functioning are strong predictors of survival in glioma patients, both at diagnosis136139 and at recurrence.140 Most large trials of brain metastasis treatment now report HRQoL or cognitive function measures, or both,126 and HRQoL reports in other tumor types such as meningioma,141143 acoustic neuroma,131,133,144 and some types of pituitary adenoma145,146 are becoming increasingly common. Such studies sometimes use disease-specific HRQoL scales that have not been developed or validated with rigorous psychometric methods.

Despite their obvious merits, HRQoL and cognitive function have been tricky end points in brain tumor therapy trials for several reasons. The main obstacle has been the expense and difficulty of administering the instruments themselves. Combined with the declining functional status seen in the late stages of many brain tumor patients’ lives, this causes missing data to be a frequent problem in HRQoL analyses. HRQoL data tend not to be missing at random, but selectively for patients who are doing poorly—thereby falsely inflating the HRQoL of the remaining compliant population. Caregivers can be asked to rate HRQoL for patients under these circumstances (proxy rating). The reliability of proxy ratings as a surrogate for PROs is poor, especially in patients with cognitive deficits, but they are preferable to missing data.147,148 Other problems have included difficulty defining the minimum clinically significant change in HRQoL measures (as opposed to the smallest statistically significant change) and the possibility that some supportive medications commonly used in brain tumor patients (such as antiseizure medications and corticosteroids) can interfere with test validity.102 To date, HRQoL has not been used by the FDA as the primary evidence for approving any anticancer product, although HRQoL data are often included in applications.127,128

Functional status is an individual’s capacity to perform the normal activities of daily living; for adults this includes self-care and work. The oldest and most commonly used measure of functional status in cancer studies is the KPS (Table 113-2);73 a closely allied scale is the ECOG performance scale, which is very similar to the WHO and Zubrod scales (Table 113-3).74 Performance status is an important prognostic factor for survival in brain tumor patients, and most prospective studies include performance status as a baseline variable, usually with a minimum score required as an entry criterion for the study (e.g., KPS score of 70). Examining the KPS itself, it is clear that the difference between some levels is subjective and can be difficult to assign reliably, especially the highest levels of the scale. For example, a history of a single seizure curtails driving in most jurisdictions even if the seizures are controlled; whether such a patient can be assigned a KPS score of 100 (normal) or even 90 (capable of normal activity) is ambiguous. In a general cancer practice setting, interrater agreement on the KPS is generally good; in one study, raters agreed on KPS scores 59% of the time and disagreed by one level (10 points) 32% of the time, with 7% of ratings disagreeing by 20 or 30 points.149 In brain tumor patients the KPS score is highly correlated with age and is sensitive to depression; it is most sensitive to changes in HRQoL in glioma patients for KPS scores in the 50 to 80 range and performs less well for higher scores.150 KPS scores can be difficult to assign in retrospective studies. They are also highly confounded by the extent of resection in glioma studies, probably reflecting a combination of relief of mass effect from extensive resections with deficits caused primarily by unresectable tumors involving eloquent areas. Despite these drawbacks, KPS and other functional status scores are widely used in reporting baseline status in patient cohorts and in expressing immediate improvement or deterioration after brain tumor surgery by comparing presurgical and postsurgical scores.151 In longer term outcome studies, the KPS is used for defining “independent” or “functional” survival, and a 10-point decline in KPS score has been shown to correlate with increased risk for death in glioma patients.10

TABLE 113-2 Karnofsky Performance Scale

100 Normal; no complaints, no signs of disease
90 Capable of normal activity, minor symptoms or signs of disease
80 Normal activity with some difficulty, some symptoms or signs
70 Caring for self, not capable of normal activity or work
60 Requiring some help, can take care of most personal requirements
50 Requires help often, requires frequent medical care
40 Disabled, requires special care and help
30 Severely disabled, hospital admission required but no immanent risk of death
20 Very ill, urgently requiring admission, requires supportive measures or treatment
10 Moribund, rapidly progressive fatal disease processes
0 Dead

From Karnofsky DA, Burchenal JH. The clinical evaluation of chemotherapeutic agents in cancer. In: MacLeod CM, ed. Evaluation of Chemotherapeutic Agents. New York: Columbia University Press; 1949:196.

TABLE 113-3 Eastern Cooperative Oncology Group Performance Scale

0 Asymptomatic (fully active, able to carry out all predisease activities without restriction)
1 Symptomatic but completely ambulatory (restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature)
2 Symptomatic, <50% in bed during waking hours (ambulatory and capable of all self-care but unable to carry out any work activities)
3 Symptomatic, >50% in bed during waking hours, but not bedbound (capable of limited self-care)
4 Bedbound (completely disabled, cannot carry on any self-care, totally confined to bed or chair)
5 Dead

From Oken MM, Creech RH, Tormey DC, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5:649-655.

Cognitive function has already been mentioned in the context of HRQoL140,152 and is most often measured with the Mini-Mental Status Examination (MMSE).153,154 A multitude of other measures are available to express changes in neurological function generally or in specific functions.4 For example, after acoustic neuroma surgery, the Glasgow Benefit Inventory can be used to measure otolaryngologically related health status.155 More specifically, facial nerve function can be graded by using the House-Brackmann scale,76 hearing by using the Gardner-Robertson classification,75 tinnitus by using the Tinnitus Handicap Inventory,156 and vertigo by using the Dizziness Handicap Inventory.157 After glioma surgery, the National Institutes of Health (NIH) Stroke Scale,158161 which measures 15 aspects of neurological function, has been used to detect new neurological deficits.54 Other investigators have used less formal criteria, such as improved/stable/worsened motor function162 or detailed listings of adverse events,163 to express neurological damage from glioma surgery quantitatively. Although the NIH Common Toxicity Criteria (www.fda.gov/cder/cancer/toxicityframe.htm) are widely used to measure the toxicity of chemotherapy, including brain tumor chemotherapy, the scales provided are not readily adapted to neurosurgical complications and are not as widely used as in reports of drug treatment. After pituitary surgery, endocrine end points are often the most useful in measuring cure and recurrence. These specialized tests are covered elsewhere in this textbook.

Tumor Response Rate

The tumor response rate (sometimes called the radiographic or objective response rate) is the proportion of patients whose tumors shrink in size as a direct response to therapy, an end point that is common in oncology studies. In brain tumor trials, tumor response is assessed with imaging studies, although tumors elsewhere in the body (e.g., plexiform neurofibromas) are sometimes measured directly by inspection.164 The response rate has historically been considered to be a valid surrogate variable for improvement in survival or progression-free survival. The advantage of using the response rate as an end point is that it is more rapidly assessed than survival, with the median time to best response for most tumor/drug combinations being in the range of a few months. Assumptions behind the choice of response as an important oncology trial end point are that most anticancer drugs work through a cytotoxic mechanism, more cytotoxicity is reflected by greater macroscopic tumor shrinkage, and spontaneous tumor shrinkage is not part of the natural history of most cancers and must represent an effect of treatment. In addition, some tumor responses in general oncology have direct clinical benefits through reduction of symptoms. The FDA considers the tumor response rate in a single-arm trial to be usable as the major criterion for cancer drug approval under appropriate circumstances, especially if supported by additional evidence of clinical benefit.102 The most common use of response as a major end point in oncology is in phase II trials in which a single-arm design is used to test drugs for preliminary evidence of drug activity; in brain tumor trials, malignant glioma that progresses after initial radiation treatment (with or without adjuvant chemotherapy) is the usual setting for phase II drug testing.

With few exceptions (lymphoma, germinoma, and to a lesser degree, oligodendroglioma), most brain tumors have relatively low response rates to radiation treatment or chemotherapy, and the responses that are observed tend to be partial rather than complete. In general oncology, although drugs that cause higher response rates generally result in greater improvement in survival, large improvements in the tumor response rate tend to correspond to relatively modest survival benefits. For example, in advanced colorectal cancer, a 50% improvement in tumor response rate was associated with only a 6% improvement in survival.165 Few drugs have ever been shown to cause such a large improvement in response rate for any brain tumor.

Tumor response rates have also been shown to be difficult to interpret in malignant glioma trials for several reasons. First, the likelihood of tumor response in drug trials for recurrent malignant glioma has been shown to depend on several patient- and tumor-related factors such as age, KPS score, tumor histology, and previous treatment.166 This makes defining a target response rate that would indicate warranting a drug for further testing difficult.167 Second, some “tumor responses” after surgical and radiation treatment of malignant glioma probably represent spontaneous regression of treatment-related imaging changes rather than the effect of anticancer drugs. Postoperative enhancement of the resection margin is common even after cerebral resection for non-neoplastic pathology unless imaging is performed within 48 to 72 hours after surgery.8991,168170 New enhancement within or adjacent to malignant tumors after radiation therapy may represent treatment-related necrosis rather than progression (“pseudoprogression”); this phenomenon is more common in patients treated with concomitant radiation therapy and temozolomide.171,172 Third, increases in corticosteroid dose have been shown to reduce tumor enhancement, thereby confounding assessment.173 Fourth, newer agents now entering testing may not work through cytotoxic effects or may have other beneficial effects on tumors that are not reflected by decreases in enhancement. Cytostatic agents might cause tumor stability rather than response, and this benefit could be missed if response were the only end point assessed. Some vasoactive drugs may provide direct clinical benefit through reducing peritumoral edema, an effect that is missed when reduction in enhancing volume is the end point.174 Fifth, because enhancing volume itself is only a surrogate for tumor volume (reflecting the volume of blood-brain barrier breakdown characteristic of malignant glioma) and not a direct measure of solid tumor or relatively preserved brain tissue containing infiltrating tumor, vasoactive drugs could potentially cause spurious responses that reflect changes in vascular biology rather than a true antitumor effect.174,175

For these reasons, recent trends in trial design have largely replaced tumor response with 6-month progression-free survival (PFS6) as the major end point in phase II drug trials for recurrent malignant glioma.102 Analysis of two large trial databases showed that PFS6 is a good surrogate for overall 1-year survival.123,124 In addition to being free of several of the problems noted earlier in assessing response, PFS6 is completely assessible by 6 months after the last patient is enrolled in a phase II trial, thus facilitating early reporting.

Measurement of response for malignant glioma has changed over the past 2 decades and is still in the course of evolution. The earliest response scale (“Levin criteria”), based on computed tomography, used investigators’ subjective assessment of progression, stability, and response to define a five-level scale ranging from definite response to definite progression, with a sixth possible score indicating development of a new lesion.176 The 1990 “Macdonald criteria” used the area calculated by multiplying the largest cross-sectional diameter of the tumor by the largest diameter perpendicular to it as the major determinant of response, in conjunction with neurological status and corticosteroid dose.177 A 50% reduction in area was necessary to achieve a response, and a 25% increase in area indicated tumor progression. Corticosteroid dose was included in the criteria because of evidence that dose changes can affect tumor enhancement.173 Although in general oncology a unidimensional response criterion based on changes in the greatest linear dimension of the tumor or tumors has recently been adopted,178,179 widespread use of this method for primary brain tumors would have obvious drawbacks because of the highly irregular shape of most gliomas. Most scales for measuring response also require that the response be durable, specifically, that it be maintained over some minimum period, such as 2 months.

Current trends in brain tumor response assessment favor measurements based on MRI using semiautomated segmentation software, edited manually as needed by the radiologist, to define the measured volume because inter- and intrarater variability is reduced in comparison to linear- or area-based measurements.180,181 Although no standard change in volume to define progression or response has yet emerged, a 50% change in volume to define both thresholds has been suggested.182 Volumetric assessment is particularly attractive for radiosurgery studies because the treatment planning process typically supplies a baseline volume that can easily be related to volumes at follow-up. Although novel imaging methods that reflect changes in tumor metabolism or vascular biology, such as perfusion or permeability, are in early stages of testing as possible response measures in brain tumor trials,183 none has yet reached the level of routine clinical use at the time of writing. Volumetric methods also hold promise for assessment of benign tumor responses, such as in meningiomas or acoustic neuromas.184 Whether unidimensional methods will gain future currency in brain metastasis trials remains to be seen, although to date response assessment has not been a major end point in these studies.

Special Considerations in Specific Phases of Drug or Technology Testing

Some types of study design seem to be consistently associated with certain important statistical issues or types of bias. These are discussed first with respect to the classic stages in new drug or technology development (i.e., phase I, II, and III trials) and then by studies describing certain tumor histologies and types of therapy.

Early-Phase-of-Development Studies (Phase I, Phase II, “Phase 0”)

The characteristics of early- and late-phase studies in drug or product development are covered in depth in Chapter 11. In brief, a phase I trial is intended to prove the basic safety of a new treatment and, when applicable, to find the maximal tolerated dose of a new drug. Phase II trials are designed to detect the first evidence of a new treatment’s activity against tumors in patients and to select promising treatments for definitive proof of efficacy in phase III testing. Detailed guidelines for reporting phase I and II trials of brain tumor drugs185 and surgically implanted brain tumor treatments186 have been published.

Phase I trials of interest to surgeons primarily involve surgically administered treatments, such as drug-impregnated polymer wafers,49,163,187189 gene190 or oncolytic virus therapy,191 convection-enhanced delivery (CED) of targeted toxins or other drugs,192194 or brachytherapy.195197 In phase I studies the main goal is to define the maximal safe dose of the novel agent or, for some agents, the maximal feasible dose (when toxicity is negligible and cost or other factors prevent delivery of the agent above a certain practical ceiling). The number of patients treated in phase I studies is sharply limited to expose the fewest possible patients to harm. Evidence of efficacy is not expected in phase I trials, although tumor response is typically monitored and many drugs that eventually prove effective do show responses in phase I testing.

The typical structure of a phase I trial is a dose escalation model with three to six patients treated per dose stratum and escalation to a higher dose as each successive stratum proves safe. Either the dose of the implanted therapy187,198 or the dose of a concomitant systemic therapy199,200 may be escalated. Dose escalation phase I trials of surgically implanted brain tumor treatments have typically reported major toxicities thought to be at least potentially related to the intracranial implantation, including seizures, confusion, focal neurological deficits, cerebral edema, wound-healing problems, and fatigue. Infection is another important concern in these studies, particularly when foreign bodies are implanted or percutaneous catheters must remain in place for several days while treatments are being infused, although experience to date has not shown increased clinical infection rates to be a common problem.

Perhaps the two major challenges for phase I trials of surgically implanted treatments are related to the small number of patients treated, which makes it possible to miss uncommon but important toxicities, and the frequent difficulty distinguishing an elevated toxicity profile from the range of possible outcomes after routine craniotomy. In general oncology trials, well-established dose escalation and reduction rules have been developed to guide the course of a phase I trial and have been shown to be good predictors of the experience expected when a larger cohort is treated in phase II or III trials or in general use.6 Most toxicities in these trials are hematologic and readily reversible. In contrast, toxicity from implanted brain treatments may not be reversible, and an expected level of one such toxicity per three to six patients treated may exceed what would be acceptable in practice. Brain tumor surgery trials, then, may sometimes be better structured as phase I/II trials in which a larger cohort of patients are treated at the projected maximal tolerated dose under the same conditions of rigorous toxicity monitoring that characterize phase I trials. Alternatively, initial phase II trials may incorporate a pilot phase in which special attention is paid to toxicity in the first small cohort treated while further accrual is placed on hold.

In addition, when thrombocytopenia or neutropenia occur in phase I trials of systemic therapies, they are usually readily attributable to the experimental drug, whereas the normal range of postoperative morbidity after tumor craniotomies that do not use novel therapies includes some patients who have seizures, new neurological deficits, and transient confusion. Some trials, therefore, not only define dose-limiting toxicities by the NIH Common Toxicity Criteria level but also specify that seizures that are similar in character and frequency to preoperative patterns should not count as dose-limiting toxicity.192 Additional ad hoc toxicities not included in the NIH Common Toxicity Criteria may be monitored, such as increases in intracerebral pressure, Glasgow Coma Scale deterioration, or persistent (>2 weeks) new focal deficits.192 For some drug delivery methods, such as CED, reversible mass effect symptoms related to local volume infusion have been described and characterized.194 In some cases, investigators have chosen phase II doses based on phase I trials in unconventional ways, such as that certain dose cohorts had longer or shorter survival. In one such example,187 a later phase I trial using more standard dose escalation (made possible by drug manufacturing improvements) demonstrated that much higher doses of the agent could be safely delivered than had originally been thought possible.198

Phase II trials are designed to detect the first evidence of an anticancer drug’s activity against tumors in patients. They are typically single-arm open-label studies enrolling 40 to 80 patients, although other designs are occasionally used. The purpose of a phase II trial is not to provide definitive evidence of efficacy, that being the role of phase III testing. In general oncology, the typical end point in phase II drug trials is the tumor response rate, but survival or progression-free survival can be used if measurement of response is problematic. Phase II trial results are compared with a historical control group to make the decision whether to proceed to phase III testing of the drug.167 Many single-arm brain tumor surgical case series can informally be considered to be phase II trials as a way of examining the strength or weakness of the study design.

The major problem specific to brain tumor surgery phase II trial design is defining an appropriate historical control group. The major ways of constructing a standard-treatment historical control group were mentioned earlier, such as using results from previously conducted large clinical trials stratified by a validated prognostic index, such as the RTOG RPA for malignant glioma treatment.56 The difficulty in using RPA to predict patient survival in surgical studies is that surgical patients are highly selected based on tumor features that favor surgical resection and the RPA classification does not fully account for these features. Similar bias is seen when using RPA to match patients selected for brachytherapy, radiosurgery, or intra-arterial chemotherapy.201 For each of these treatments, initial phase II trials showed highly favorable response or survival results.197,202,203 However, RCTs subsequently showed each of the treatments to be ineffective.195,196,204,205 For each treatment, later study showed that the historical control patients selected for eligibility for the treatment (but who did not receive the treatment) had a survival advantage based solely on their eligibility, which explained the overoptimistic phase II testing results.206210

For surgically implanted therapies and high-dose radiation studies (using brachytherapy or radiosurgery), imaging changes in or around the tumor site that mimic tumor progression but actually represent treatment-related brain necrosis are frequent.211214 This prevents the use of response or time to tumor progression as reliable phase II end points for these types of studies; survival end points are a better choice.102 Similarly, when gross total resection is a criterion for trial entry, response cannot be used as an end point, and progression-free survival or overall survival is used in phase II trials.66 Some therapies progress directly from phase I to phase III testing because of the difficulty of designing a phase II trial.187,215

The “phase 0” trial216 is a new addition to the standard phases of drug development described in Chapter 11. Phase 0 trials enroll a small number of patients (typically <15), with limited drug exposure, and have no therapeutic or diagnostic intent. They clarify drug pharmacokinetics or pharmacodynamics during first-in-human use of new agents. In brain tumor studies, assessment of whether the novel agent achieves the desired modulation of its intended target is commonly the goal of phase 0 studies. In a typical design, a patient with recurrent glioma receives a dose of a novel, usually molecularly targeted agent immediately before planned surgical resection. After surgery, the resected tissue is submitted for analysis of tissue drug levels and the drug’s biologic target. This tests the drug’s penetration into the tumor, as well as drug activity. Problems with this design include ethical barriers (because of the lack of intended benefit to the patient, combined with concrete risks) and the possibility that novel agents could increase the risk associated with surgery (e.g., because of a direct effect on tumor vasculature217). This trial design has been used to measure gene transduction after gene therapy,218 inactivation of methylguanine-deoxyribonucleic acid methyltransferase (MGMT) after systemic O6-benzylguanine administration,199 and epidermal growth factor receptor inhibitor effects on recurrent malignant gliomas.219 It will probably gain wider currency because of the expense of testing molecularly targeted agents and the difficulty of submitting the many available new agents to extensive clinical testing in glioma, a relatively rare disease.220

Phase III Clinical Trials

The validity of phase III trial results is critical because it is these trials that most often cause a change in general clinical practice. Details of assessing the reliability of RCTs is covered in Chapter 11 of this textbook, and special points to consider in surgical RCTs are reviewed there. Among the most important problems for designing and conducting surgical RCTs are difficulty defining a patient population for which there is community equipoise (uncertainty in the medical community about the best treatment),221223 overcoming preexisting patient preferences in enrolling patients to an RCT,224,225 difficulty blinding patients and outcome assessors,226,227 dependence on the technical skill of the operator and related issues such as the learning curve,192194228 and the ongoing evolution characteristic of new technologies.229,230

In surgical RCTs on brain tumor treatment, besides these issues that confront internal validity, comparability of the trial population to patients in general practice and the feasibility (and cost) of widespread adoption of complex procedures are special problems. RCT entry criteria are typically restrictive in comparison to general practice, and some examples indicate that relatively few patients in unselected series would actually qualify for surgical brain tumor RCTs. Whittle and coauthors231 reported that only 25% of malignant glioma patients seen in an academic neuro-oncologic surgical practice met the entry criteria for an RCT of drug-impregnated wafers that was open at their center. Trial-eligible patients had better prognostic factors, including younger age, better clinical grade, and more extensive resections, and were more likely to undergo postoperative adjuvant radiation therapy. Generalizing the results of the RCT to the larger ineligible population is therefore problematic. Similarly, when therapies include resection of recurrent malignant glioma or are tested specifically in this population, it must be remembered that only 15% of patients will qualify for resection at time of glioma recurrence and that these patients likewise have relatively favorable prognostic factors when compared with patients who do not undergo second operations.232

Surgical treatments of malignant glioma are becoming increasingly complex with the introduction of CED, treatment plans that demand imaging-complete resections, and other technically demanding procedures. Proper training and credentialing may be necessary for surgeons who participate in trials of some complex therapies, such as CED, to prevent high rates of unsatisfactory technical performance from threatening the validity of the trial.192,194 Technical aspects of performing the treatment may require study before or during the trial: investigators found that more CED catheters placed at a second, separate procedure met quality criteria than did catheters placed at the time of resection.194 Ideally, these matters would be straightened out during phase I or II testing. Outcomes in patients undergoing complex surgical procedures, including craniotomy for tumor,109111233 are better with high-volume providers (hospitals and surgeons), and specialist surgeons have higher rates of complete tumor resection and fewer neurological complications.234,235 However, with pretrial credentialing, the effects of surgeon volume on technical outcomes can be mitigated or prevented,236 and such trials offer an educational opportunity to begin the diffusion of advanced surgical technologies into the broader community.237,238

Special Considerations in Specific Types of Brain Tumor Study Design

Some study designs specific to brain tumor surgery are especially prone to certain design problems or types of bias, including studies on the extent of resection as a prognostic factor, studies of technologies intended to improve the extent of resection, and special problems in survival studies involving recurrent and metastatic brain tumors. Finally, health services research (such as volume outcome studies and disparities studies) are addressed briefly.

Extent of Surgical Resection as a Prognostic Factor for Survival

One of the oldest and most popular study designs in brain tumor surgery is a comparison of survival in patients who receive extensive resection and those who do not. Nazzaro and Neuwelt published an influential rebuttal of much of this research in 1990 in which the flaws in trial design and statistical analysis nearly universal in these studies at that time were pointed out: failure to adjust the analyses for other important prognostic factors that might not be equally distributed between biopsied and resected patients (age, functional status, tumor location, tumor pathology); differential use of adjuvant therapies after biopsy or resection, such as radiation therapy, chemotherapy, and resection at recurrence; and general design flaws, such as consistent use of retrospective design and, frequently, failure to use “any form of statistical analysis.”239 Recent reviews confirm that this literature is still deeply flawed.83,240

The main limitation of biopsy-versus-resection studies is that they compare “apples to oranges.” Comparisons of results of randomized and nonrandomized studies on the same treatments have shown that nonrandomized studies often give similar answers as RCTs on treatment questions, but only when the nonrandomized studies closely mimic the RCTs with which they are being compared.241243 Specifically, the control group in nonrandomized studies should be treated concurrently with the treatment group, and all patients in both the treatment and control groups should be equally eligible for the treatment being studied. This criterion is almost never met in studies that compare patients with different degrees of surgical resection: patients undergo gross total resection, less extensive resection, or biopsy based largely on the resectability of their tumors rather than by randomization. When eligibility for treatment (i.e., resectability) is a prognostic factor for outcome, confounding by indication prevents a valid nonrandomized comparison between treatments.

Some alternative trial designs are available that can avoid or adjust for this bias. First, an actual RCT can be performed, such as that reported by Vuorinen and associates on resection of glioblastoma.53 Second, randomized studies of the addition of technical adjunctive treatments to surgery that are intended to improve the extent of resection (such as neuronavigation or intraoperative imaging) can provide a relatively pure means of randomizing patients to different chances for total resection. In the three such trials reported to date, when the adjunctive treatment improved the degree of resection, survival was improved as well.54,162,244 The third design, which has not yet been used in a published study, would be to start with patients who were eligible for complete resection but in whom the initial resection proved to be subtotal. These patients, all of whom would have imaging-detected residual disease that was thought to be resectable, would then be randomized to second-look surgery or to immediate treatment with adjuvant therapies (radiation therapy and chemotherapy). A fourth design would be a nonrandomized comparison of subtotal versus total resection adjusted for “resectability” by using stratification or a propensity score model. Although several glioma resectability scales have been published that could be used for this purpose,151,245248 the design has not yet been used. Fifth, studies could use a cohort247 or case-control249 design for patients who were considered eligible for gross total resection to compare those who did or did not undergo total removal. In a variant on this design, Shaw and coworkers used the well-known unreliability of the surgeon’s grading of the extent of resection in a constructive manner: in a cohort of prospectively observed, low-grade glioma patients in whom surgeons graded surgical excision as total, better progression-free survival was seen when the resection was deemed truly complete by imaging than when imaging showed measurable residual disease.85

Studies on Technologic Adjuncts for Improving the Extent of Resection

Another popular research theme in brain tumor surgery is technologic advances to improve the extent of resection. Examples include intraoperative imaging techniques (MRI, ultrasound) to detect residual tumor, fluorescent dye visualization of residual tumor through specially adapted microscopes, and stereotactic neuronavigation based on preoperative imaging. The strongest design for studying these methods is an RCT in which patients eligible for resection (or for gross total resection) are randomized to receive or not receive the surgical adjunctive treatment being tested, with an end point measuring the extent of resection achieved.54,162,244 A problem with such studies is the impossibility of blinding surgeons to the type of resection to be used. For intraoperative imaging or fluorescent dye studies, the best time for randomization would be after completion of the best possible resection via conventional surgery, although this is not usually done. For any technique promoting more aggressive surgery, careful and objective documentation of any additional risk for permanent neurological deficits is mandated.

A common, but much weaker study design is the routine use of an intraoperative tumor visualization technique at the end of conventional resection, with an end point of whether residual tumor is demonstrated (in which case additional resection is performed).250253 This design obviously biases toward artificially conservative initial efforts at resection with consequently high rates of “usefulness” of the technique in detecting residual tumor. An unrelated, but characteristic weakness of these nonrandomized studies is comparison against a historical control group demonstrating shorter length of stay as a purported benefit of the technique. Confounding by the strong temporal trend toward shorter length of stay for all neurosurgical treatments over the past 2 decades,254 primarily driven by insurance considerations, cannot be adjusted away by using statistical techniques.

Health Services Research: Volume Outcome and Disparities Studies

General considerations relevant to these types of studies are given in Chapter 11 of this textbook. With specific relevance to brain tumor studies, special consideration should be given to the influence of cultural differences on the detection and treatment of cancer among socially defined groups, which is particularly strong because cancer care is so expensive, invasive, and potentially toxic.257 In addition, the underrepresentation in cancer clinical trials of patients who are underinsured or uninsured, members of racial or ethnic minorities, older, female, less well educated, and unmarried68,69,258 limits the confident assumption that new treatments will be equally effective in these patient groups.

Suggested Reading

Aldape K, Simmons ML, Davis RL, et al. Discrepancies in diagnoses of neuroepithelial neoplasms: the San Francisco Bay Area Adult Glioma Study. Cancer. 2000;88:2342-2349.

Brandsma D, Stalpers L, Taal W, et al. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9:453-461.

Chang S, Vogelbaum M, Lang FF, et al. GNOSIS: guidelines for neuro-oncology: standards for investigational studies—reporting of surgically based therapeutic clinical trials. J Neurooncol. 2007;82:211-220.

Chang SM, Lamborn KR, Kuhn JG, et al. Neurooncology clinical trial design for targeted therapies: lessons learned from the North American Brain Tumor Consortium. Neuro Oncol. 2008;10:631-642.

Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med. 2000;342:1887-1892.

Curran WJJr, Scott CB, Horton J, et al. Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst. 1993;85:704-710.

Florell RC, Macdonald DR, Irish WD, et al. Selection bias, survival, and brachytherapy for glioma. J Neurosurg. 1992;76:179-183.

Glantz MJ, Burger PC, Herndon JE2nd, et al. Influence of the type of surgery on the histologic diagnosis in patients with anaplastic gliomas. Neurology. 1991;41:1741-1744.

Green S, Benedetti J, Crowley J. Clinical Trials in Oncology, 2nd ed. London: Chapman & Hall/CRC; 2002.

Kelly PJ, Daumas-Duport C, Kispert DB, et al. Imaging-based stereotaxic serial biopsies in untreated intracranial glial neoplasms. J Neurosurg. 1987;66:865-874.

Lamborn KR, Yung WK, Chang SM, et al. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol. 2008;10:162-170.

Lang FF, Asher A. Prospective clinical trials of brain tumor therapy: the critical role of neurosurgeons. J Neurooncol. 2004;69:151-167.

Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97-109.

Macdonald DR, Cascino TL, Schold SCJr, et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277-1280.

Mauer M, Stupp R, Taphoorn MJ, et al. The prognostic value of health-related quality-of-life data in predicting survival in glioblastoma cancer patients: results from an international randomised phase III EORTC Brain Tumour and Radiation Oncology Groups, and NCIC Clinical Trials Group study. Br J Cancer. 2007;97:302-307.

Pazdur R, Rock EP, Fine H, et al. FDA/AACR/ASCO Public Workshop on Brain Tumor Clinical Trial Endpoints, January 20, 2006. Available at http://www.fda.gov/cder/drug/cancer_endpoints/brain_summary.pdf, 2008. Accessed January 5

Ryken TC, Frankel B, Julien T, et al. Surgical management of newly diagnosed glioblastoma in adults: role of cytoreductive surgery. J Neurooncol. 2008;89:271-286.

Smith JS, Cha S, Mayo MC, et al. Serial diffusion-weighted magnetic resonance imaging in cases of glioma: distinguishing tumor recurrence from postresection injury. J Neurosurg. 2005;103:428-438.

Sorensen AG, Batchelor TT, Wen PY, et al. Response criteria for glioma. Nat Clin Pract Oncol. 2008;5:634-644.

Spilker B. Guide to Clinical Trials. New York: Raven Press; 1991.

Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7:392-401.

Vuorinen V, Hinkka S, Farkkila M, et al. Debulking or biopsy of malignant glioma in elderly people—a randomised study. Acta Neurochir (Wien). 2003;145:5-10.

Weitzner MA, Meyers CA, Gelke CK, et al. The Functional Assessment of Cancer Therapy (FACT) scale. Development of a brain subscale and revalidation of the general version (FACT-G) in patients with primary brain tumors. Cancer. 1995;75:1151-1161.

Wong ET, Hess KR, Gleason MJ, et al. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17:2572-2578.

Zia MI, Siu LL, Pond GR, et al. Comparison of outcomes of phase II studies and subsequent randomized control studies using identical chemotherapeutic regimens. J Clin Oncol. 2005;23:6982-6991.

References

1 Crowley J, editor. Handbook of Statistics in Clinical Oncology. New York: Marcel Dekker, 2001.

2 Girling D, Parmar M, Stenning S, et al. Clinical Trials in Cancer: Principles and Practice. Oxford: Oxford University Press; 2003.

3 Green S, Benedetti J, Crowley J. Clinical Trials in Oncology, 2nd ed. London: Chapman & Hall/CRC; 2002.

4 Guiloff RJ, editor. Clinical Trials in Neurology. Berlin: Springer, 2001.

5 Lipscomb J, Gotay CG, Snyder C, editors. Outcomes Assessment in Cancer. Cambridge, UK: Cambridge University Press, 2005.

6 Piantadosi S. Clinical Trials: a Methodologic Perspective, 2nd ed. New York: John Wiley & Sons; 2005.

7 Spilker B. Guide to Clinical Trials. New York: Raven Press; 1991.

8 Lang FF, Asher A. Prospective clinical trials of brain tumor therapy: the critical role of neurosurgeons. J Neurooncol. 2004;69:151-167.

9 Fontaine D, Bauchet L, Capelle L. [French neurosurgical practice in neurooncology (national survey—part II). Census of current research protocols on brain tumors in France. Neurochirurgie. 2005;51:136-141.

10 Barker FG2nd, Huhn SL, Prados MD. Clinical characteristics of long-term glioma survivors. In: Berger MS, Wilson MS, editors. The gliomas. Philadelphia: W.B. Saunders; 1999:710-722.

11 Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 2005;64:479-489.

12 Wisoff JH, Boyett JM, Berger MS, et al. Current neurosurgical management and the impact of the extent of resection in the treatment of malignant gliomas of childhood: a report of the Children’s Cancer Group trial no. CCG-945. J Neurosurg. 1998;89:52-59.

13 Saran FH, Driever PH, Thilmann C, et al. Survival of very young children with medulloblastoma (primitive neuroectodermal tumor of the posterior fossa) treated with craniospinal irradiation. Int J Radiat Oncol Biol Phys. 1998;42:959-967.

14 Packer RJ, Rood BR, MacDonald TJ. Medulloblastoma: present concepts of stratification into risk groups. Pediatr Neurosurg. 2003;39:60-67.

15 Woodward ER, Wall K, Forsyth J, et al. VHL mutation analysis in patients with isolated central nervous system haemangioblastoma. Brain. 2007;130:836-842.

16 Mohyuddin A, Neary WJ, Wallace A, et al. Molecular genetic analysis of the NF2 gene in young patients with unilateral vestibular schwannomas. J Med Genet.. 2002;39:315-322.

17 Cushing H. Intracranial Tumors: Notes upon a Series of Two Thousand Verified Cases with Surgical-Mortality Percentages Pertaining Thereto. Springfield, IL: Charles C. Thomas; 1932.

18 Louis DN, Ohgaki H, Wiestler OD, et al, editors. WHO Classification of Tumours of the Central Nervous System, 4th ed, Lyon: International Agency on Research on Cancer, 2007.

19 Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97-109.

20 Brat DJ, Parisi JE, Kleinschmidt-DeMasters BK, et al. Surgical neuropathology update: a review of changes introduced by the WHO classification of tumours of the central nervous system, 4th edtion. Arch Pathol Lab Med. 2008;132:993-1007.

21 Nelson JS, Tsukada Y, Schoenfeld D, et al. Necrosis as a prognostic criterion in malignant supratentorial, astrocytic gliomas. Cancer. 1983;52:550-554.

22 Burger PC, Vogel FS, Green SB, et al. Glioblastoma multiforme and anaplastic astrocytoma. Pathologic criteria and prognostic implications. Cancer. 1985;56:1106-1111.

23 Barker FG2nd, Davis RL, Chang SM, et al. Necrosis as a prognostic factor in glioblastoma multiforme. Cancer. 1996;77:1161-1166.

24 Morita M, Rosenblum MK, Bilsky MH, et al. Long-term survivors of glioblastoma multiforme: clinical and molecular characteristics. J Neurooncol. 1996;27:259-266.

25 Scott JN, Rewcastle NB, Brasher PM, et al. Which glioblastoma multiforme patient will become a long-term survivor? A population-based study. Ann Neurol. 1999;46:183-188.

26 Ullen H, Mattsson B, Collins VP. Long-term survival after malignant glioma. A clinical and histopathological study on the accuracy of the diagnosis in a population-based cancer register. Acta Oncol. 1990;29:875-878.

27 Komotar RJ, Burger PC, Carson BS, et al. Pilocytic and pilomyxoid hypothalamic/chiasmatic astrocytomas. Neurosurgery. 2004;54:72-79.

28 Tihan T, Fisher PG, Kepner JL, et al. Pediatric astrocytomas with monomorphous pilomyxoid features and a less favorable outcome. J Neuropathol Exp Neurol. 1999;58:1061-1068.

29 Biegel JA, Rorke LB, Emanuel BS. Monosomy 22 in rhabdoid or atypical teratoid tumors of the brain. N Engl J Med. 1989;321:906.

30 Kaderali Z, Lamberti-Pasculli M, Rutka JT. The changing epidemiology of paediatric brain tumours: a review from the Hospital for Sick Children. Childs Nerv Syst. 2009;25:787-793.

31 Cairncross JG, Macdonald DR. Successful chemotherapy for recurrent malignant oligodendroglioma. Ann Neurol. 1988;23:360-364.

32 McCarthy BJ, Propp JM, Davis FG, et al. Time trends in oligodendroglial and astrocytic tumor incidence. Neuroepidemiology. 2008;30:34-44.

33 Pearson BE, Markert JM, Fisher WS, et al. Hitting a moving target: evolution of a treatment paradigm for atypical meningiomas amid changing diagnostic criteria. Neurosurg Focus. 2008;24(5):E3.

34 Simon M, Bostrom J, Koch P, et al. Interinstitutional variance of postoperative radiotherapy and follow up for meningiomas in Germany: impact of changes of the WHO classification. J Neurol Neurosurg Psychiatry. 2006;77:767-773.

35 Smith SJ, Boddu S, Macarthur DC. Atypical meningiomas: WHO moved the goalposts? Br J Neurosurg. 2007;21:588-592.

36 Davis FG, Malmer BS, Aldape K, et al. Issues of diagnostic review in brain tumor studies: from the Brain Tumor Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev. 2008;17:484-489.

37 Castillo MS, Davis FG, Surawicz T, et al. Consistency of primary brain tumor diagnoses and codes in cancer surveillance systems. Neuroepidemiology. 2004;23:85-93.

38 Aldape K, Simmons ML, Davis RL, et al. Discrepancies in diagnoses of neuroepithelial neoplasms: the San Francisco Bay Area Adult Glioma Study. Cancer. 2000;88:2342-2349.

39 Glantz MJ, Burger PC, Herndon JE2nd, et al. Influence of the type of surgery on the histologic diagnosis in patients with anaplastic gliomas. Neurology. 1991;41:1741-1744.

40 Muragaki Y, Chernov M, Maruyama T, et al. Low-grade glioma on stereotactic biopsy: how often is the diagnosis accurate? Minim Invasive Neurosurg. 2008;51:275-279.

41 Woodworth G, McGirt MJ, Samdani A, et al. Accuracy of frameless and frame-based image-guided stereotactic brain biopsy in the diagnosis of glioma: comparison of biopsy and open resection specimen. Neurol Res. 2005;27:358-362.

42 Jackson RJ, Fuller GN, Abi-Said D, et al. Limitations of stereotactic biopsy in the initial management of gliomas. Neuro Oncol. 2001;3:193-200.

43 Perry A, Jenkins RB, O’Fallon JR, et al. Clinicopathologic study of 85 similarly treated patients with anaplastic astrocytic tumors. An analysis of DNA content (ploidy), cellular proliferation, and p53 expression. Cancer. 1999;86:672-683.

44 Aghi M, Batchelor TT, Henson J, et al. Increased diagnosis of oligodendroglioma with resection in low-grade cerebral tumors: sampling error or differential resectability [abstract]? J Neurosurg. 2007;106:A779-A780.

45 Carter BS, Aghi M, Curry WTJr, et al. Supratentorial low-grade glioma: diagnostic trends and temporal and geographic variation in practice patterns—a population-based study [abstract]. Neurosurgery. 2005;57:412.

46 Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med. 1990;322:494-500.

47 Noordijk EM, Vecht CJ, Haaxma-Reiche H, et al. The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age. Int J Radiat Oncol Biol Phys. 1994;29:711-717.

48 Mintz AH, Kestle J, Rathbone MP, et al. A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis. Cancer. 1996;78:1470-1476.

49 Ewend MG, Brem S, Gilbert M, et al. Treatment of single brain metastasis with resection, intracavity carmustine polymer wafers, and radiation therapy is safe and provides excellent local control. Clin Cancer Res. 2007;13:3637-3641.

50 Barker FG2nd, Chang SM, Huhn SL, et al. Age and the risk of anaplasia in magnetic resonance-nonenhancing cerebral tumors. Cancer. 1997;80:936-941.

51 McCormack BM, Miller DC, Budzilovich GN, et al. Treatment and survival of low-grade astrocytoma in adults—1977-1988. Neurosurgery. 1992;31:636-642.

52 Tofts PS, Benton CE, Weil RS, et al. Quantitative analysis of whole-tumor Gd enhancement histograms predicts malignant transformation in low-grade gliomas. J Magn Reson Imaging. 2007;25:208-214.

53 Vuorinen V, Hinkka S, Farkkila M, et al. Debulking or biopsy of malignant glioma in elderly people—a randomised study. Acta Neurochir (Wien). 2003;145:5-10.

54 Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7:392-401.

55 McLendon RE, Halperin EC. Is the long-term survival of patients with intracranial glioblastoma multiforme overstated? Cancer. 2003;98:1745-1748.

56 Curran WJJr, Scott CB, Horton J, et al. Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst. 1993;85:704-710.

57 Akman F, Cooper RA, Sen M, et al. Validation of the Medical Research Council and a newly developed prognostic index in patients with malignant glioma: how useful are prognostic indices in routine clinical practice? J Neurooncol. 2002;59:39-47.

58 Prognostic factors for high-grade malignant glioma: development of a prognostic index. A Report of the Medical Research Council Brain Tumour Working Party. J Neurooncol. 1990;9:47-55.

59 Lamborn KR, Chang SM, Prados MD. Prognostic factors for survival of patients with glioblastoma: recursive partitioning analysis. Neuro Oncol. 2004;6:227-235.

60 Pignatti F, van den Bent M, Curran D, et al. Prognostic factors for survival in adult patients with cerebral low-grade glioma. J Clin Oncol. 2002;20:2076-2084.

61 Gaspar LE, Scott C, Murray K, et al. Validation of the RTOG recursive partitioning analysis (RPA) classification for brain metastases. Int J Radiat Oncol Biol Phys. 2000;47:1001-1006.

62 Gaspar L, Scott C, Rotman M, et al. Recursive partitioning analysis (RPA) of prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain metastases trials. Int J Radiat Oncol Biol Phys. 1997;37:745-751.

63 Sperduto PW, Berkey B, Gaspar LE, et al. A new prognostic index and comparison to three other indices for patients with brain metastases: an analysis of 1,960 patients in the RTOG database. Int J Radiat Oncol Biol Phys. 2008;70:510-514.

64 Colman H, Berkey BA, Maor MH, et al. Phase II Radiation Therapy Oncology Group trial of conventional radiation therapy followed by treatment with recombinant interferon-beta for supratentorial glioblastoma: results of RTOG 9710. Int J Radiat Oncol Biol Phys. 2006;66:818-824.

65 Barker FG2nd, Prados MD, Chang SM, et al. Radiation response and survival in patients with glioblastoma multiforme. J Neurosurg. 1996;84:442-448.

66 Butowski N, Lamborn KR, Berger MS, et al. Historical controls for phase II surgically based trials requiring gross total resection of glioblastoma multiforme. J Neurooncol. 2007;85:87-94.

67 Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987-996.

68 Gross CP, Herrin J, Wong N, et al. Enrolling older persons in cancer trials: the effect of sociodemographic, protocol, and recruitment center characteristics. J Clin Oncol. 2005;23:4755-4763.

69 Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials: race-, sex-, and age-based disparities. JAMA. 2004;291:2720-2726.

70 Lamont EB, Hayreh D, Pickett KE, et al. Is patient travel distance associated with survival on phase II clinical trials in oncology? J Natl Cancer Inst. 2003;95:1370-1375.

71 Zia MI, Siu LL, Pond GR, et al. Comparison of outcomes of phase II studies and subsequent randomized control studies using identical chemotherapeutic regimens. J Clin Oncol. 2005;23:6982-6991.

72 George SL. Selection bias, phase II trials, and the FDA accelerated approval process. J Natl Cancer Inst. 2003;95:1351-1352.

73 Karnofsky DA, Burchenal JH. The clinical evaluation of chemotherapeutic agents in cancer. In: MacLeod CM, editor. Evaluation of Chemotherapeutic Agents. New York: Columbia University Press; 1949:196.

74 Oken MM, Creech RH, Tormey DC, et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5:649-655.

75 Gardner G, Robertson JH. Hearing preservation in unilateral acoustic neuroma surgery. Ann Otol Rhinol Laryngol. 1988;97:55-66.

76 House JW, Brackmann DE. Facial nerve grading system. Otolaryngol Head Neck Surg. 1985;93:146-147.

77 Sheehan JP, Sun MH, Kondziolka D, et al. Radiosurgery for non–small cell lung carcinoma metastatic to the brain: long-term outcomes and prognostic factors influencing patient survival time and local tumor control. J Neurosurg. 2002;97:1276-1281.

78 Christakis NA, Lamont EB. Extent and determinants of error in doctors’ prognoses in terminally ill patients: prospective cohort study. BMJ. 2000;320:469-472.

79 Glare P, Virik K, Jones M, et al. A systematic review of physicians’ survival predictions in terminally ill cancer patients. BMJ. 2003;327:195-198.

80 Chow E, Harth T, Hruby G, et al. How accurate are physicians’ clinical predictions of survival and the available prognostic tools in estimating survival times in terminally ill cancer patients? A systematic review. Clin Oncol (R Coll Radiol). 2001;13:209-218.

81 Albert FK, Forsting M, Sartor K, et al. Early postoperative magnetic resonance imaging after resection of malignant glioma: objective evaluation of residual tumor and its influence on regrowth and prognosis. Neurosurgery. 1994;34:45-60.

82 de Tribolet N, Fankhauser H. Surgery in the treatment of malignant glioma: current status and future prespectives. In: Karim ABMF, Laws ERJr, editors. Glioma. Berlin: Springer, 1991.

83 Sanai N, Berger MS. Glioma extent of resection and its impact on patient outcome. Neurosurgery. 2008;62:753-764.

84 Keles GE, Lamborn KR, Berger MS. Low-grade hemispheric gliomas in adults: a critical review of extent of resection as a factor influencing outcome. J Neurosurg. 2001;95:735-745.

85 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;109:835-841.

86 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;26:1338-1345.

87 Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg. 2001;95:190-198.

88 Keles GE, Chang EF, Lamborn KR, et al. Volumetric extent of resection and residual contrast enhancement on initial surgery as predictors of outcome in adult patients with hemispheric anaplastic astrocytoma. J Neurosurg. 2006;105:34-40.

89 Ulmer S, Braga TA, Barker FG2nd, et al. Clinical and radiographic features of peritumoral infarction following resection of glioblastoma. Neurology. 2006;67:1668-1670.

90 Laohaprasit V, Silbergeld DL, Ojemann GA, et al. Postoperative CT contrast enhancement following lobectomy for epilepsy. J Neurosurg. 1990;73:392-395.

91 Smith JS, Cha S, Mayo MC, et al. Serial diffusion-weighted magnetic resonance imaging in cases of glioma: distinguishing tumor recurrence from postresection injury. J Neurosurg. 2005;103:428-438.

92 Kelly PJ, Daumas-Duport C, Kispert DB, et al. Imaging-based stereotaxic serial biopsies in untreated intracranial glial neoplasms. J Neurosurg. 1987;66:865-874.

93 Hochberg FH, Pruitt A. Assumptions in the radiotherapy of glioblastoma. Neurology. 1980;30:907-911.

94 Gehan EA, Walker MD. Prognostic factors for patients with brain tumors. Natl Cancer Inst Monogr. 1977;46:189-195.

95 Simpson D. The recurrence of intracranial meningiomas after surgical treatment. J Neurol Neurosurg Psychiatry. 1957;20:22-39.

96 Kinjo T, al-Mefty O, Kanaan I. Grade zero removal of supratentorial convexity meningiomas. Neurosurgery. 1993;33:394-399.

97 Bloch DC, Oghalai JS, Jackler RK, et al. The fate of the tumor remnant after less-than-complete acoustic neuroma resection. Otolaryngol Head Neck Surg. 2004;130:104-112.

98 Sakaki S, Nakagawa K, Hatakeyama T, et al. Recurrence after incompletely resected acousticus neurinomas. Med J Osaka Univ. 1991;40:59-66.

99 Seol HJ, Kim CH, Park CK, et al. Optimal extent of resection in vestibular schwannoma surgery: relationship to recurrence and facial nerve preservation. Neurol Med Chir (Tokyo). 2006;46:176-180.

100 McDowell I. Measuring Health: a Guide to Rating Scales and Questionnaires, 3rd ed. New York: Oxford University Press; 2006.

101 Ramsden RT. The bloody angle: 100 years of acoustic neuroma surgery. J R Soc Med. 1995;88:464P-468P.

102 Pazdur R, Rock EP, Fine H, et al. FDA/AACR/ASCO Public Workshop on Brain Tumor Clinical Trial Endpoints, January 20, 2006. Available at http://www.fda.gov/cder/drug/cancer_endpoints/brain_summary.pdf, 2008. Accessed January 5

103 Johnson JR, Williams G, Pazdur R. End points and United States Food and Drug Administration approval of oncology drugs. J Clin Oncol. 2003;21:1404-1411.

104 Pazdur R. Endpoints for assessing drug activity in clinical trials. Oncologist. 2008;13(Suppl 2):19-21.

105 Tirakotai W, Lapanich S, Riegel T, et al. Present status of known and possible outcomes in neurosurgery: a survey of outcome assessment. Childs Nerv Syst. 2007;23:779-785.

106 Mathoulin-Pelissier S, Gourgou-Bourgade S, Bonnetain F, et al. Survival end point reporting in randomized cancer clinical trials: a review of major journals. J Clin Oncol. 2008;26:3721-3726.

107 Doody MM, Hayes HM, Bilgrad R. Comparability of national death index plus and standard procedures for determining causes of death in epidemiologic studies. Ann Epidemiol. 2001;11:46-50.

108 Cowper DC, Kubal JD, Maynard C, et al. A primer and comparative review of major US mortality databases. Ann Epidemiol. 2002;12:462-468.

109 Barker FG2nd. Craniotomy for the resection of metastatic brain tumors in the U.S., 1988-2000: decreasing mortality and the effect of provider caseload. Cancer. 2004;100:999-1007.

110 Barker FG2nd, Curry WTJr, Carter BS. Surgery for primary supratentorial brain tumors in the United States, 1988 to 2000: the effect of provider caseload and centralization of care. Neuro Oncol. 2005;7:49-63.

111 Curry WT, McDermott MW, Carter BS, et al. Craniotomy for meningioma in the United States between 1988 and 2000: decreasing rate of mortality and the effect of provider caseload. J Neurosurg. 2005;102:977-986.

112 Quality of care in the Medicare program. In: A Data Book: Healthcare Spending and the Medicare Program. Washington, D.C.: MEDPAC; 2005:21-35.

113 Baser ME, Friedman JM, Aeschliman D, et al. Predictors of the risk of mortality in neurofibromatosis 2. Am J Hum Genet. 2002;71:715-723.

114 Barker FG2nd, Jannetta PJ, Bissonette DJ, et al. The long term outcome of microvascular decompression for trigeminal neuralgia. N Engl J Med. 1996;334:1077-1083.

115 Swearingen B, Barker FG2nd, Katznelson L, et al. Long term mortality after transsphenoidal surgery and adjunctive therapy for acromegaly. J Clin Endocrinol Metab.. 1998;83:3419-3426.

116 Swearingen B, Biller BMK, Barker FG2nd, et al. Long-term mortality after transsphenoidal surgery for Cushing’s disease. Ann Intern Med. 1999;130:821-824.

117 Arrighi HM, Hertz-Picciotto I. The evolving concept of the healthy worker survivor effect. Epidemiology. 1994;5:189-196.

118 McCarthy BJ, Davis FG, Freels S, et al. Factors associated with survival in patients with meningioma. J Neurosurg. 1998;88:831-839.

119 Mendenhall WM, Morris CG, Amdur RJ, et al. Radiotherapy alone or after subtotal resection for benign skull base meningiomas. Cancer. 2003;98:1473-1482.

120 Clark TG, Altman DG, De Stavola BL. Quantification of the completeness of follow-up. Lancet. 2002;359:1309-1310.

121 Pontiroli AE, Fossati A, Vedani P, et al. Post-surgery adherence to scheduled visits and compliance, more than personality disorders, predict outcome of bariatric restrictive surgery in morbidly obese patients. Obes Surg. 2007;17:1492-1497.

122 Edwards P, Fernandes J, Roberts I, et al. Young men were at risk of becoming lost to follow-up in a cohort of head-injured adults. J Clin Epidemiol. 2007;60:417-424.

123 Ballman KV, Buckner JC, Brown PD, et al. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol. 2007;9:29-38.

124 Lamborn KR, Yung WK, Chang SM, et al. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol. 2008;10:162-170.

125 Patchell RA, Tibbs PA, Regine WF, et al. Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA. 1998;280:1485-1489.

126 Wong J, Hird A, Kirou-Mauro A, et al. Quality of life in brain metastases radiation trials: a literature review. Curr Oncol. 2008;15:25-45.

127 Rock EP, Scott JA, Kennedy DL, et al. Challenges to use of health-related quality of life for Food and Drug Administration approval of anticancer products. J Natl Cancer Inst Monogr. 2007;37:27-30.

128 Rock EP, Kennedy DL, Furness MH, et al. Patient-reported outcomes supporting anticancer product approvals. J Clin Oncol. 2007;25:5094-5099.

129 Brown PD, Ballman KV, Rummans TA, et al. Prospective study of quality of life in adults with newly diagnosed high-grade gliomas. J Neurooncol. 2006;76:283-291.

130 Williams G, Pazdur R, Temple R. Assessing tumor-related signs and symptoms to support cancer drug approval. J Biopharm Stat. 2004;14:5-21.

131 Farace E, Marshall LF. Quality of life in acoustics. J Neurosurg. 2003;99:807-808.

132 Gilbert M, Armstrong T, Meyers C. Issues in assessing and interpreting quality of life in patients with malignant glioma. Semin Oncol. 2000;27:20-26.

133 Bateman N, Nikolopoulos TP, Robinson K, et al. Impairments, disabilities, and handicaps after acoustic neuroma surgery. Clin Otolaryngol Allied Sci. 2000;25:62-65.

134 Fayers P, Hays R, editors. Assessing Quality of Life in Clinical Trials: Methods and Practice, 2nd ed, New York: Oxford University Press, 2005.

135 Weitzner MA, Meyers CA, Gelke CK, et al. The Functional Assessment of Cancer Therapy (FACT) scale. Development of a brain subscale and revalidation of the general version (FACT-G) in patients with primary brain tumors. Cancer. 1995;75:1151-1161.

136 Mauer M, Stupp R, Taphoorn MJ, et al. The prognostic value of health-related quality-of-life data in predicting survival in glioblastoma cancer patients: results from an international randomised phase III EORTC Brain Tumour and Radiation Oncology Groups, and NCIC Clinical Trials Group study. Br J Cancer. 2007;97:302-307.

137 Mauer ME, Taphoorn MJ, Bottomley A, et al. Prognostic value of health-related quality-of-life data in predicting survival in patients with anaplastic oligodendrogliomas, from a phase III EORTC brain cancer group study. J Clin Oncol. 2007;25:5731-5737.

138 Klein M, Postma TJ, Taphoorn MJ, et al. The prognostic value of cognitive functioning in the survival of patients with high-grade glioma. Neurology. 2003;61:1796-1798.

139 Sehlen S, Lenk M, Hollenhorst H, et al. Quality of life (QoL) as predictive mediator variable for survival in patients with intracerebral neoplasms during radiotherapy. Onkologie. 2003;26:38-43.

140 Meyers CA, Hess KR, Yung WK, et al. Cognitive function as a predictor of survival in patients with recurrent malignant glioma. J Clin Oncol. 2000;18:646-650.

141 Neil-Dwyer G, Lang D, Garfield J. The realities of postoperative disability and the carer’s burden. Ann R Coll Surg Engl. 2001;83:215-218.

142 Neil-Dwyer G, Lang DA, Davis A. Outcome from complex neurosurgery: an evidence based approach. Acta Neurochir (Wien). 2000;142:367-371.

143 Akagami R, Napolitano M, Sekhar LN. Patient-evaluated outcome after surgery for basal meningiomas. Neurosurgery. 2002;50:941-948.

144 Myrseth E, Moller P, Wentzel-Larsen T, et al. Untreated vestibular schwannomas: vertigo is a powerful predictor for health-related quality of life. Neurosurgery. 2006;59:67-76.

145 van der Klaauw AA, Kars M, Biermasz NR, et al. Disease-specific impairments in quality of life during long-term follow-up of patients with different pituitary adenomas. Clin Endocrinol (Oxf). 2008;69:775-784.

146 Webb SM, Badia X, Barahona MJ, et al. Evaluation of health-related quality of life in patients with Cushing’s syndrome with a new questionnaire. Eur J Endocrinol. 2008;158:623-630.

147 Moinpour CM, Lyons B, Schmidt SP, et al. Substituting proxy ratings for patient ratings in cancer clinical trials: an analysis based on a Southwest Oncology Group trial in patients with brain metastases. Qual Life Res. 2000;9:219-231.

148 Brown PD, Decker PA, Rummans TA, et al. A prospective study of quality of life in adults with newly diagnosed high-grade gliomas: comparison of patient and caregiver ratings of quality of life. Am J Clin Oncol. 2008;31:163-168.

149 Schag CC, Heinrich RL, Ganz PA. Karnofsky performance status revisited: reliability, validity, and guidelines. J Clin Oncol. 1984;2:187-193.

150 Mackworth N, Fobair P, Prados MD. Quality of life self-reports from 200 brain tumor patients: comparisons with Karnofsky performance scores. J Neurooncol. 1992;14:243-253.

151 Sawaya R, Hammoud M, Schoppa D, et al. Neurosurgical outcomes in a modern series of 400 craniotomies for treatment of parenchymal tumors. Neurosurgery. 1998;42:1044-1055.

152 Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol. 2006;24:1305-1309.

153 Folstein M. Mini-mental and son. Int J Geriatr Psychiatry. 1998;13:290-294.

154 Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189-198.

155 Robinson K, Gatehouse S, Browning GG. Measuring patient benefit from otorhinolaryngological surgery and therapy. Ann Otol Rhinol Laryngol. 1996;105:415-422.

156 Newman CW, Jacobson GP, Spitzer JB. Development of the Tinnitus Handicap Inventory. Arch Otolaryngol Head Neck Surg. 1996;122:143-148.

157 Jacobson GP, Newman CW. The development of the Dizziness Handicap Inventory. Arch Otolaryngol Head Neck Surg. 1990;116:424-427.

158 Wityk RJ, Pessin MS, Kaplan RF, et al. Serial assessment of acute stroke using the NIH Stroke Scale. Stroke. 1994;25:362-365.

159 Goldstein LB, Bertels C, Davis JN. Interrater reliability of the NIH stroke scale. Arch Neurol. 1989;46:660-662.

160 Josephson SA, Hills NK, Johnston SC. NIH Stroke Scale reliability in ratings from a large sample of clinicians. Cerebrovasc Dis. 2006;22:389-395.

161 Goldstein LB, Samsa GP. Reliability of the National Institutes of Health Stroke Scale. Extension to non-neurologists in the context of a clinical trial. Stroke. 1997;28:307-310.

162 Wu JS, Zhou LF, Tang WJ, et al. Clinical evaluation and follow-up outcome of diffusion tensor imaging-based functional neuronavigation: a prospective, controlled study in patients with gliomas involving pyramidal tracts. Neurosurgery. 2007;61:935-948.

163 Westphal M, Hilt DC, Bortey E, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro Oncol. 2003;5:79-88.

164 Babovic-Vuksanovic D, Widemann BC, Dombi E, et al. Phase I trial of pirfenidone in children with neurofibromatosis 1 and plexiform neurofibromas. Pediatr Neurol. 2007;36:293-300.

165 Buyse M, Thirion P, Carlson RW, et al. Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis. Meta-Analysis Group in Cancer. Lancet. 2000;356:373-378.

166 Wong ET, Hess KR, Gleason MJ, et al. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17:2572-2578.

167 Vickers AJ, Ballen V, Scher HI. Setting the bar in phase II trials: the use of historical data for determining “go/no go” decision for definitive phase III testing. Clin Cancer Res. 2007;13:972-976.

168 Henegar MM, Moran CJ, Silbergeld DL. Early postoperative magnetic resonance imaging following nonneoplastic cortical resection. J Neurosurg. 1996;84:174-179.

169 Elster AD, DiPersio DA. Cranial postoperative site: assessment with contrast-enhanced MR imaging. Radiology. 1990;174:93-98.

170 Sato N, Bronen RA, Sze G, et al. Postoperative changes in the brain: MR imaging findings in patients without neoplasms. Radiology. 1997;204:839-846.

171 Brandsma D, Stalpers L, Taal W, et al. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9:453-461.

172 Peca C, Pacelli R, Elefante A, et al. Early clinical and neuroradiological worsening after radiotherapy and concomitant temozolomide in patients with glioblastoma: Tumour progression or radionecrosis? Clin Neurol Neurosurg. 2009;111:331-334.

173 Cairncross JG, Macdonald DR, Pexman JH, et al. Steroid-induced CT changes in patients with recurrent malignant glioma. Neurology. 1988;38:724-726.

174 Ananthnarayan S, Bahng J, Roring J, et al. Time course of imaging changes of GBM during extended bevacizumab treatment. J Neurooncol. 2008;88:339-347.

175 Vredenburgh JJ, Desjardins A, Herndon JE2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25:4722-4729.

176 Levin VA, Crafts DC, Norman DM, et al. Criteria for evaluating patients undergoing chemotherapy for malignant brain tumors. J Neurosurg. 1977;47:329-335.

177 Macdonald DR, Cascino TL, Schold SCJr, et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277-1280.

178 Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205-216.

179 Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228-247.

180 Sorensen AG, Patel S, Harmath C, et al. Comparison of diameter and perimeter methods for tumor volume calculation. J Clin Oncol. 2001;19:551-557.

181 Suzuki C, Jacobsson H, Hatschek T, et al. Radiologic measurements of tumor response to treatment: practical approaches and limitations. Radiographics. 2008;28:329-344.

182 Sorensen AG, Batchelor TT, Wen PY, et al. Response criteria for glioma. Nat Clin Pract Oncol. 2008;5:634-644.

183 Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83-95.

184 Plotkin SR, Halpin C, Blakeley JO, et al. Suggested response criteria for phase II antitumor drug studies for neurofibromatosis type 2 related vestibular schwannoma. J Neurooncol. 2009;93:61-77.

185 Chang SM, Reynolds SL, Butowski N, et al. GNOSIS: guidelines for neuro-oncology: standards for investigational studies—reporting of phase 1 and phase 2 clinical trials. Neuro Oncol. 2005;7:425-434.

186 Chang S, Vogelbaum M, Lang FF, et al. GNOSIS: guidelines for neuro-oncology: standards for investigational studies—reporting of surgically based therapeutic clinical trials. J Neurooncol. 2007;82:211-220.

187 Brem H, Mahaley MSJr, Vick NA, et al. Interstitial chemotherapy with drug polymer implants for the treatment of recurrent gliomas. J Neurosurg. 1991;74:441-446.

188 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 Neurochir (Wien). 2006;148:269-275.

189 Sabel M, Giese A. Safety profile of carmustine wafers in malignant glioma: a review of controlled trials and a decade of clinical experience. Curr Med Res Opin. 2008;24:3239-3257.

190 Chiocca EA. Gene therapy: a primer for neurosurgeons. Neurosurgery. 2003;53:364-373.

191 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;10:958-966.

192 Sampson JH, Akabani G, Archer GE, et al. Intracerebral infusion of an EGFR-targeted toxin in recurrent malignant brain tumors. Neuro Oncol. 2008;10:320-329.

193 Vogelbaum MA, Sampson JH, Kunwar S, et al. Convection-enhanced delivery of cintredekin besudotox (interleukin-13–PE38QQR) followed by radiation therapy with and without temozolomide in newly diagnosed malignant gliomas: phase 1 study of final safety results. Neurosurgery. 2007;61:1031-1037.

194 Kunwar S, Prados MD, Chang SM, et al. Direct intracerebral delivery of cintredekin besudotox (IL13-PE38QQR) in recurrent malignant glioma: a report by the Cintredekin Besudotox Intraparenchymal Study Group. J Clin Oncol. 2007;25:837-844.

195 Laperriere NJ, Leung PM, McKenzie S, et al. Randomized study of brachytherapy in the initial management of patients with malignant astrocytoma. Int J Radiat Oncol Biol Phys. 1998;41:1005-1011.

196 Selker RG, Shapiro WR, Burger P, et al. The Brain Tumor Cooperative Group NIH Trial 87-01: a randomized comparison of surgery, external radiotherapy, and carmustine versus surgery, interstitial radiotherapy boost, external radiation therapy, and carmustine. Neurosurgery. 2002;51:343-355.

197 Prados MD, Gutin PH, Phillips TL, et al. Interstitial brachytherapy for newly diagnosed patients with malignant gliomas: the UCSF experience. Int J Radiat Oncol Biol Phys. 1992;24:593-597.

198 Olivi A, Grossman SA, Tatter S, et al. Dose escalation of carmustine in surgically implanted polymers in patients with recurrent malignant glioma: a New Approaches to Brain Tumor Therapy CNS Consortium trial. J Clin Oncol. 2003;21:1845-1849.

199 Friedman HS, Kokkinakis DM, Pluda J, et al. Phase I trial of O6-benzylguanine for patients undergoing surgery for malignant glioma. J Clin Oncol. 1998;16:3570-3575.

200 Gururangan S, Cokgor L, Rich JN, et al. Phase I study of Gliadel wafers plus temozolomide in adults with recurrent supratentorial high-grade gliomas. Neuro Oncol. 2001;3:246-250.

201 Haines SJ. Moving targets and ghosts of the past: outcome measurement in brain tumour therapy. J Clin Neurosci. 2002;9:109-112.

202 Shrieve DC, Alexander E3rd, Black PM, et al. Treatment of patients with primary glioblastoma multiforme with standard postoperative radiotherapy and radiosurgical boost: prognostic factors and long-term outcome. J Neurosurg. 1999;90:72-77.

203 Greenberg HS, Ensminger WD, Chandler WF, et al. Intra-arterial BCNU chemotherapy for treatment of malignant gliomas of the central nervous system. J Neurosurg. 1984;61:423-429.

204 Shapiro WR, Green SB, Burger PC, et al. A randomized comparison of intra-arterial versus intravenous BCNU, with or without intravenous 5-fluorouracil, for newly diagnosed patients with malignant glioma. J Neurosurg. 1992;76:772-781.

205 Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet. 2004;363:1665-1672.

206 Irish WD, Macdonald DR, Cairncross JG. Measuring bias in uncontrolled brain tumor trials—to randomize or not to randomize? Can J Neurol Sci. 1997;24:307-312.

207 Florell RC, Macdonald DR, Irish WD, et al. Selection bias, survival, and brachytherapy for glioma. J Neurosurg. 1992;76:179-183.

208 Kirby S, Brothers M, Irish W, et al. Evaluating glioma therapies: modeling treatments and predicting outcomes. J Natl Cancer Inst. 1995;87:1884-1888.

209 Curran WJJr, Scott CB, Weinstein AS, et al. Survival comparison of radiosurgery-eligible and -ineligible malignant glioma patients treated with hyperfractionated radiation therapy and carmustine: a report of Radiation Therapy Oncology Group 83-02. J Clin Oncol. 1993;11:857-862.

210 Lustig RA, Scott CB, Curran WJ. Does stereotactic eligibility for the treatment of glioblastoma cause selection bias in randomized studies? Am J Clin Oncol. 2004;27:516-521.

211 Parney IF, Kunwar S, McDermott M, et al. Neuroradiographic changes following convection-enhanced delivery of the recombinant cytotoxin interleukin 13-PE38QQR for recurrent malignant glioma. J Neurosurg. 2005;102:267-275.

212 Valk PE, Dillon WP. Radiation injury of the brain. AJNR Am J Neuroradiol. 1991;12:45-62.

213 Constine LS, Konski A, Ekholm S, et al. Adverse effects of brain irradiation correlated with MR and CT imaging. Int J Radiat Oncol Biol Phys. 1988;15:319-330.

214 Kumar AJ, Leeds NE, Fuller GN, et al. Malignant gliomas: MR imaging spectrum of radiation therapy- and chemotherapy–induced necrosis of the brain after treatment. Radiology. 2000;217:377-384.

215 Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer–brain Tumor Treatment Group. Lancet. 1995;345:1008-1012.

216 Kummar S, Doroshow JH, Tomaszewski JE, et al. Phase 0 clinical trials: recommendations from the task force on methodology for the development of innovative cancer therapies. Eur J Cancer. 2009;45:728-729.

217 Genentech Inc. Bevacizumab (Avastin) package insert. Available at www.gene.com/gene/products/information/pdf/avastin-prescribing.pdf, 2008.

218 Lang FF, Bruner JM, Fuller GN, et al. Phase I trial of adenovirus-mediated p53 gene therapy for recurrent glioma: biological and clinical results. J Clin Oncol. 2003;21:2508-2518.

219 Lassman AB, Rossi MR, Raizer JJ, et al. Molecular study of malignant gliomas treated with epidermal growth factor receptor inhibitors: tissue analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01. Clin Cancer Res. 2005;11:7841-7850.

220 Chang SM, Lamborn KR, Kuhn JG, et al. Neurooncology clinical trial design for targeted therapies: lessons learned from the North American Brain Tumor Consortium. Neuro Oncol. 2008;10:631-642.

221 Young JM, Solomon MJ, Harrison JD, et al. Measuring patient preference and surgeon choice. Surgery. 2008;143:582-588.

222 Young J, Harrison J, White G, et al. Developing measures of surgeons’ equipoise to assess the feasibility of randomized controlled trials in vascular surgery. Surgery. 2004;136:1070-1076.

223 Freedman B. Equipoise and the ethics of clinical research. N Engl J Med. 1987;317:141-145.

224 Arega A, Birkmeyer NJ, Lurie JD, et al. Racial variation in treatment preferences and willingness to randomize in the Spine Patient Outcomes Research Trial (SPORT). Spine. 2006;31:2263-2269.

225 King M, Nazareth I, Lampe F, et al. Conceptual framework and systematic review of the effects of participants’ and professionals’ preferences in randomised controlled trials. Health Technol Assess. 2005;9:1-186. iii-iv

226 Miller FG. Sham surgery: an ethical analysis. Sci Eng Ethics. 2004;10:157-166.

227 London AJ, Kadane JB. Placebos that harm: sham surgery controls in clinical trials. Stat Methods Med Res. 2002;11:413-427.

228 O’Malley BWJr, Grady MS, Gabel BC, et al. Comparison of endoscopic and microscopic removal of pituitary adenomas: single-surgeon experience and the learning curve. Neurosurg Focus. 2008;25(6):E10.

229 Lilford R, Braunholtz D, Harris J, et al. Trials in surgery. Br J Surg. 2004;91:6-16.

230 McLeod RS. Issues in surgical randomized controlled trials. World J Surg. 1999;23:1210-1214.

231 Whittle IR, Lyles S, Walker M. Gliadel therapy given for first resection of malignant glioma: a single centre study of the potential use of Gliadel. Br J Neurosurg. 2003;17:352-354.

232 Barker FG2nd, Chang SM, Gutin PH, et al. Survival and functional status after resection of recurrent glioblastoma multiforme. Neurosurgery. 1998;42:709-720.

233 Barker FG2nd, Klibanski A, Swearingen B. Transsphenoidal surgery for pituitary tumors in the United States, 1996-2000: mortality, morbidity, and the effects of hospital and surgeon volume. J Clin Endocrinol Metab. 2003;88:4709-4719.

234 Ciric I, Ragin A, Baumgartner C, et al. Complications of transsphenoidal surgery: results of a national survey, review of the literature, and personal experience. Neurosurgery. 1997;40:225-236.

235 Albright AL, Sposto R, Holmes E, et al. Correlation of neurosurgical subspecialization with outcomes in children with malignant brain tumors. Neurosurgery. 2000;47:879-885.

236 Larson DW, Marcello PW, Larach SW, et al. Surgeon volume does not predict outcomes in the setting of technical credentialing: results from a randomized trial in colon cancer. Ann Surg. 2008;248:746-750.

237 Leitch AM, Beitsch PD, McCall LM, et al. Patterns of participation and successful patient recruitment to American College of Surgeons Oncology Group Z0010, a phase II trial for patients with early-stage breast cancer. Am J Surg. 2005;190:539-542.

238 Posther KE, McCall LM, Blumencranz PW, et al. Sentinel node skills verification and surgeon performance: data from a multicenter clinical trial for early-stage breast cancer. Ann Surg. 2005;242:593-599.

239 Nazzaro JM, Neuwelt EA. The role of surgery in the management of supratentorial intermediate and high-grade astrocytomas in adults. J Neurosurg. 1990;73:331-344.

240 Ryken TC, Frankel B, Julien T, et al. Surgical management of newly diagnosed glioblastoma in adults: role of cytoreductive surgery. J Neurooncol. 2008;89:271-286.

241 Benson K, Hartz AJ. A comparison of observational studies and randomized, controlled trials. N Engl J Med. 2000;342:1878-1886.

242 Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. N Engl J Med. 2000;342:1887-1892.

243 Ioannidis JPA, Haidich AB, Pappa M, et al. Comparison of evidence of treatment effects in randomized and nonrandomized studies. JAMA. 2001;286:821-830.

244 Willems PWA, Taphoorn MJ, Burger H, et al. Effectiveness of neuronavigation in resecting solitary intracerebral contrast-enhancing tumors: a randomized controlled trial. J Neurosurg. 2006;104:360-368.

245 Talos IF, Zou KH, Ohno-Machado L, et al. Supratentorial low-grade glioma resectability: statistical predictive analysis based on anatomic MR features and tumor characteristics. Radiology. 2006;239:506-513.

246 Chang EF, Smith JS, Chang SM, et al. Preoperative prognostic classification system for hemispheric low-grade gliomas in adults. J Neurosurg. 2008;109:817-824.

247 Shinoda J, Sakai N, Murase S, et al. Selection of eligible patients with supratentorial glioblastoma multiforme for gross total resection. J Neurooncol. 2001;52:161-171.

248 Vorster SJ, Barnett GH. A proposed preoperative grading scheme to assess risk for surgical resection of primary and secondary intraaxial supratentorial brain tumors. Neurosurg Focus. 1998;4(6):e2.

249 Krex D, Klink B, Hartmann C, et al. Long-term survival with glioblastoma multiforme. Brain. 2007;130:2596-2606.

250 Knauth M, Wirtz CR, Tronnier VM, et al. Intraoperative MR imaging increases the extent of tumor resection in patients with high-grade gliomas. AJNR Am J Neuroradiol. 1999;20:1642-1646.

251 Senft C, Seifert V, Hermann E, et al. Usefulness of intraoperative ultra low-field magnetic resonance imaging in glioma surgery. Neurosurgery. 2008;63:257-266.

252 Nimsky C, Fujita A, Ganslandt O, et al. Volumetric assessment of glioma removal by intraoperative high-field magnetic resonance imaging. Neurosurgery. 2004;55:358-370.

253 Bohinski RJ, Kokkino AK, Warnick RE, et al. Glioma resection in a shared-resource magnetic resonance operating room after optimal image-guided frameless stereotactic resection. Neurosurgery. 2001;48:731-742.

254 Barker FG2nd, Amin-Hanjani S. Changing neurosurgical workload in the United States, 1988-2001: craniotomy other than trauma in adults. Neurosurgery. 2004;55:506-517.

255 Forsyth PA, Kelly PJ, Cascino TL, et al. Radiation necrosis or glioma recurrence: is computer-assisted stereotactic biopsy useful? J Neurosurg. 1995;82:436-444.

256 Tihan T, Barletta J, Parney I, et al. Prognostic value of detecting recurrent glioblastoma multiforme in surgical specimens from patients after radiotherapy: should pathology evaluation alter treatment decisions? Hum Pathol. 2006;37:272-282.

257 Haynes MA, Smedley BD, editors. The Unequal Burden of Cancer: An Assessment of NIH Research and Programs for Ethnic Minorities and the Medically Underserved. Washington, DC: National Academies Press. 1999:352.

258 Avis NE, Smith KW, Link CL, et al. Factors associated with participation in breast cancer treatment clinical trials. J Clin Oncol. 2006;24:1860-1867.