Sie sind auf Seite 1von 11

Review

Low-grade gliomas: an update on pathology and therapy


Robert Cavaliere, Maria Beatriz S Lopes, David Schiff
Lancet Neurol 2005; 4: 76070 Neuro-oncology Center, University of Virginia, Box 800432, Charlottesville, VA 22908, USA (R Cavaliere MD, M B S Lopes MD, D Schiff MD) Correspondence to: Dr David Schiff ds4jd@virginia.edu

Low-grade gliomas (LGG) are not benign neoplasms. Patients with LGG eventually die as a consequence of this disease. Although the survival of patients with LGG is better than that of patients with higher-grade tumours, many of the treatments can produce or contribute to chronic impairment, particularly radiotherapy. Chemotherapy has emerged as a promising therapy, although denitive ndings are awaited. Breakthroughs in molecular biology have improved our understanding of tumours and have led to the development of novel treatments and better prognoses. Ongoing clinical trials will help to elucidate the optimum management of patients with LGG.

Introduction
Low-grade gliomas (LGG) present a substantial challenge to neuro-oncologists and neurosurgeons. Although more indolent than their high-grade counterparts, they are associated with neurological disability and are fatal. Prognosis is variable; high-risk and low-risk groups have been identied through risk stratication of clinical, tumour, and pathological factors. Optimum treatment is unknown, and the treatment methods routinely used are associated with substantial morbidity. Advances in molecular biology are promising, although more basic and clinical research is needed.

Epidemiology
LGG are a group of tumours with distinct clinical, histological, and molecular characteristics. The WHO classication system classies tumours by predominant cell type and grade.1 Although it is accepted that all grade I and II lesions are low-grade, the clinical diversity of these tumours detracts from a meaningful classication (table 1). A more clinically informative denition of LGG includes only grade II oligodendrogliomas, astocytomas, and oligoastrocytomas, tumours with similar invasive and malignant potential;2 these tumours are the focus of this paper. Although LGG are less common than high-grade gliomas, LGG are not rare (table 2).3 LGG are most common among men and white people and typically affect patients at a younger age than high-grade gliomas (fourth versus sixth decade of life). LGG most commonly involve the cerebral hemispheres, and are typically located in the frontal, parietal, or temporal lobes. Compared with high-grade gliomas, involvement of the insula or supplementary motor area is more
Astrocytic tumours Grade I Pilocytic astrocytoma, subependymal giantcell astrocytoma Diffuse astrocytoma, pleomorphic xanthoastrocytoma Oligodendroglial tumours Mixed tumours Ependymal tumours Subependymoma, myxopapillary ependymoma Ependymoma

common in LGG.4 In adults, LGG rarely involve the cerebellum, brainstem, or spinal cord. Seizure is the presenting symptom in more than 80% of patients.57 Patients also present with focal decits, altered mental state, or headache. Several weak causal associations have been reported in the few epidemiological studies done, including head trauma, mobile-phone use, exposure to power-frequency electromagnetic elds, diet, and occupational exposures including rubber manufacturing, petrochemicals, vinyl chloride, and pesticides.8 The only denite risk factor identied is previous exposure to ionising radiation. Wrensch and colleagues8 review of risk factors is thorough. Hereditary factors do not play a substantial part in development of LGG, although these tumours are more common in patients with neurobromatosis and Li-Fraumeni syndrome.

Neuroimaging
Diagnosis of LGG requires neuroimaging (gure 1). LGG are shown on CT scanning as isointense to hypointense. Evidence of haemorrhage and calcication might be more common in oligodendroglial tumours. On T2-weighted MRI, LGG are hyperintense and either diffuse with indistinct borders or focal with well-circumscribed margins.9 The epicentre of the astrocytoma is typically within the white matter whereas oligodendrogliomas may be more supercial, occasionally expanding the involved gyrus. Lesions extend along white-matter tracts, commonly through the corpus callosum into the contralateral hemisphere. Rarely, large tumours involve three or more cerebral lobes (gliomatosis cerebri). LGG may be multifocal with several non-contiguous regions of involvement (gure 2). LGG are not typically associated with substantial mass effect. 1539% of supratentorial LGG in adults enhance on contrast imaging.1012 Enhancement is typically heterogeneous and faint, and rarely in a ring or nodular pattern. LGG are hypometabolic on PET, a feature that commonly distinguishes them from high-grade gliomas On magnetic resonance spectroscopy, typical spectra include a dominant choline peak (reecting increased membrane synthesis) with low-intensity N-acteylaspartate (reecting decreased neuronal elements) and
http://neurology.thelancet.com Vol 4 November 2005

Grade II

Oligodendroglioma

Oligoastrocytoma

Table 1: WHO classication of LGG1

760

Review

% of all primary Median age brain tumours at diagnosis Diffuse astrocytoma Oligodendroglioma Oligoastrocytoma
*Per 100 000 person-years.

Age adjusted incidence* 010 037 015

07 27 11

45 41 42

Table 2: Age adjusted incidence of LGG in the USA3

tumours lack the classic features of astrocytic or oligodendroglial differentiation and show much morphological overlap. LGG share some common features. Most tumours are inltrative, with microscopic invasion of normal brain distant from the tumour epicentre. Histological features of anaplasia, including microvascular endothelial proliferation and necrosis, are absent, and mitotic activity is low. Proliferative index, assessed by labelling cells with MIB-1 antibody targeting the Ki-67 antigen expressed only in proliferative stages of the cell cycle, is generally low, typically 24%.1316

no quantiable lipid or lactate (suggesting an absence of necrosis, a feature of high-grade gliomas).9 The choline peak may be associated with cellular density and indices of cellular proliferation, thereby improving selection of targets for biopsy. Magnetic resonance spectroscopy may discriminate radiation necrosis from tumour progression and can be used to monitor treatment progress.9

Astrocytomas
Broadly, low-grade astrocytic tumours include tumours with diffusely inltrating (diffuse astrocytoma) and circumscribed (pleomorphic xanthoastrocytoma, pilocytic astrocytoma, subependymal giant cell astrocytoma) growth patterns (table 1).1 Although these tumours are characterised by slow growth and indolent behaviour, those with more circumscribed growth have specic clinicopathological features that distinguish them from the tumours discussed here. Astroctyomas with diffuse histology (WHO grade II) are characterised by low to moderate cellularity and minimum cellular and nuclear atypia (gure 3). Astrocytomas are subclassied into three variants, representing morphological subtypes of astrocytes in normal and reactive brain: brillary, protoplasmic, and gemistocytic astrocytomas. Most tumours are a mixture of the different subtypes. Fibrillary astrocytomas are composed of cells with multipolar cytoplasmic processes that form a rich brillary glial stroma. Protoplasmic astrocytomas are composed of many stellate-shaped cells with small cytoplasmic processes embedded in a microcystic or loose matrix. Gemistocytic astrocytomas are composed of cells with abundant eosinophilic, round to slightly angulated cytoplasm, and odd nuclei. The brillary matrix,

Neuropathology
Pathological criteria for distinguishing low-grade astrocytoma, oligodendroglioma, and oligoastrocytomas are controversial. Oligodendroglial tumours, either pure or mixed with an astrocytic component, were once thought to be rare. Over the past decade, however, they have been diagnosed at a much greater rate. Concomitantly, the incidence of low-grade astrocytomas has declined. Oligodendroglial tumours are more responsive to treatment than astrocytomas and are associated with a better prognosis. Consequently, diagnostic criteria for oligodendrogliomas and oligoastrocytomas have become less rigid, in part owing to neuropathologists awareness of the need to distinguish oligodendroglial tumours from other gliomas, as well as in response to pressure from clinicians. Neuropathologists are challenged to make a precise diagnosis of an LGG when in fact many of these

Figure 1: Neuroimaging of LGG Left: Non-contrasted head CT of a haemorrhagic and calcied midline, grade II oligodendroglioma. Middle: T2-weighted MRI of a grade II astrocytoma with poorly dened margins and diffuse involvement of the cerebral white matter; involvement of three lobes is consistent with gliomatosis cerebri. Right: T2-weighted MRI of a grade II oligodendroglioma with gyral expansion.

http://neurology.thelancet.com Vol 4 November 2005

761

Review

Figure 2: MRI of a multifocal, grade II oligoastrocytoma Left: Fluid-attenuated-inversion-recovery image. Middle: T1 post-gadolinium image. Right: T1 post-gadolinium image done 3 months after the other images, when the patient presented with an increasing number of seizures and left hemiparesis.

common in brillary tumours, is less conspicuous. As a pure histological variant, gemistocytic astrocytomas are rare.17 The presence of substantial gemistocytes suggests a much shorter time to progression than with other grade II astrocytomas (gure 3).18 Astrocytomas are recognised for immunoreactivity for glial brillary acidic protein (GFAP), vimentin, and S-100 protein. The proportion of positive tumour cells varies among diffuse astrocytomas; tumours with more prominent brillary and gemistocytic components show stronger GFAP immunoreactivity than protoplasmic variants. Genetic abnormalities that might be involved in tumour formation and progression have been well

characterised in astrocytic tumours. In low-grade astrocytomas, the most common chromosomal abnormality is loss of heterozygosity on 17p and mutations of TP53 tumour suppressor gene. p53, a transcription factor present in low concentrations in normal cells, regulates cell-cycle arrest, DNA repair, and apoptosis in response to cell stress. Mutation of the TP53 gene is present in 60% of low-grade astrocytomas and 80% of gemistocytic astrocytomas.19 In the absence of a mutation, p53 function may be disrupted by other mechanisms, such as promoter hypermethylation and the resulting inhibition of the tumour suppressor gene p14ARF. This gene encodes a protein that regulates p53.20 Platelet-derived-growth-factor receptor and ligand are commonly overexpressed in low-grade astrocytomas, resulting in autocrine activation of downstream pathways to which they are linked. For example, ras is an intracellular signal transduction network that is commonly overactivated in low-grade astrocytoma. These pathways result in uncontrolled cell growth. Platelet-derived-growth-factor ligands and receptors are expressed about equally in all grades of astrocytomas, suggesting that this pathway is important in the initial steps of astrocytoma pathogenesis. Other chromosomal abnormalities present in low-grade astrocytomas include gains of chromosome 7 and losses of 22q, 13q, 10p, chromosome 6, and sex chromosomes.19,21

Oligodendrogliomas
Oligodendrogliomas (WHO grade II) are soft, gelatinous masses that extend from white to grey matter, obliterating the greywhite junction. Areas of cystic degeneration and calcication are common. The typical oligodendroglioma has a distinct histology: a uniform population of cells with round nuclei and a delicate chromatin pattern and scant cytoplasm (gure 3).22 Artefactual clearing of the cytoplasm gives the cell a characteristic fried-egg appearance. The tumour cells are dispersed in a loose brillary matrix traversed by delicate, branching vessels that occasionally subdivide the tumour into pseudolobules. This distinctive vascular pattern is referred to as a chicken-wire pattern. Oligodendrogliomas diffusely inltrate the cerebral cortex, forming characteristic secondary structures such as perineuronal satellitosis, perivascular accumulations, and subpial inltration. Neoplastic oligodendrocytes commonly have a small rim of eosinophilic cytoplasm and eccentric nuclei, socalled mini-gemistocytes. Immunocytochemical markers that reliably distinguish neoplastic oligodendrocytes do not exist. Most markers expressed by developing and mature oligodendrocytes are inconstantly expressed by neoplastic oligodendrocytes. Therefore, the diagnosis of oligodendrogliomas is primarily made from observations on haematoxylin and eosin stained sections and the absence of reactivity for markers
http://neurology.thelancet.com Vol 4 November 2005

Figure 3: Histopathological features of low-grade gliomas Haematoxylin and eosin staining showing diffuse astrocytoma (A); diffuse astrocytoma with gemistocytic component (B); oligodendroglioma (C); and mixed oligoastrocytoma (D). Magnication 100 (A,C,D) and 200 (B).

762

Review

associated with astroglial differentiation. Most oligodendrogliomas do not express GFAP and vimentin although mini-gemistocytes commonly show intense GFAP immunoreactivity.23,24 In the past 5 years, identication of a new class of oligodendrocyte-specic genes, the basic helix-loop-helix transcription factors Olig1 and Olig2, has given hope for new markers specic to oligodendroglial tumour identication. Olig1 and Olig2 are involved in the specication of oligodendrocyte precursors, but are also expressed by oligodendrocytes in the mature CNS.25,26 Although initial reports conrmed that neoplastic oligodendroglial cells overexpress Olig1 and Olig2,2729 recent studies have shown that these markers are present in high concentrations in inltrating astrocytomas (low and high grade) and pilocytic astrocytomas.30,31 Mixed oligoastrocytomas (WHO grade II) are gliomas with areas that resemble both oligodendroglioma and astrocytoma (gure 3). The distinction between oligodendrogliomas and oligoastrocytomas can be problematic. There are no denitive histological criteria for conrming the mixed oligoastrocytoma diagnosis. The two glial elements may be distributed in distinct areas (biphasic pattern) or in an intermingled pattern, the latter being more common. The main cell component is commonly oligodendroglial. Some researchers have dened mixed glioma as a tumour in which a second minor glial component is present in at least one eld magnied at 100-times magnication.32 Other researchers avoid diagnosing mixed gliomas or oligoastroctyomas, choosing to classify the tumour according to predominant glial morphology.33 At present, the combination of light microscopy, immunohistochemical studies, and cytogenetic or molecular analysis seems to be the best method for identifying these ambiguous gliomas.33 Oligodendroglial tumours have a more distinct genetic prole than astrocytomas. Somatic deletions of chromosomes 1p and 19q are the most important events in tumorigenesis of oligodendrogliomas, occurring in about 4070% of tumours (gure 4).34 The presence of 1p and 19q deletions is highly associated with the oligodendroglial phenotype.35 Point mutations of TP53 in oligodendrogliomas rarely occur and are less common than in astrocytic tumours.36 p53 protein overexpression, as measured by immunohistochemistry, is not uncommon, suggesting mechanisms of p53 pathway disruption other than gene mutation.37 Oligoastrocytomas are genetically heterogeneous. 3070% of oligoastrocytomas have allelic losses on chromosomes 1p and 19q, thus resembling oligodendrogliomas. Another 30% have genetic aberrations commonly found in diffuse astrocytomas, including mutations of TP53 and loss of heterozygosity
http://neurology.thelancet.com Vol 4 November 2005

on chromosome 17p.38 Identical genetic changes have been identied in both astrocytic and oligodendroglial elements in a given tumour, indicating a clonal origin of mixed gliomas.39 The presence of TP53 mutations or 17p loss and 1p and 19q deletions seems to be exclusive in the individual groups.4042

Animal models of LGG


Animal models of glioma have contributed to the understanding of glioma pathogenesis. They also enable preclinical testing of developmental therapies. Tumours can be induced in animals through activation of growth-factor-induced signal transduction pathways or targeting of specic genes that are commonly altered in human gliomas.43 An international consortium recently reviewed the histology of gliomas in genetically engineered mice and provided guidelines for histological classication.44 Most animal models have generated high-grade gliomas. A GFAP/v-src mouse45 and a transgenic mouse with a GFAP/H-ras construction,46 however, have resulted in low-grade astrocytomas. Mice infected with retroviruses expressing platelet-derived-growth-factor B develop either oligodendrogliomas or oligoastrocytomas depending upon the initial expression of plateletderived-growth-factor B in either neural progenitor cells or astrocytes.47 Transfer of polyoma middle T antigen into a Gtv-a mouse induces oligoastrocytomas.48 Deletions of 1p and 19q, commonly seen in human oligodendrogliomas, are not present in animal oligodendroglial tumours induced by platelet-derived growth factor.

Prognostic factors
Although LGG seem more indolent than high-grade gliomas, they are not benign and eventually prove fatal. Extraneural metastases of LGG are rare. Neoplastic cells, however, can inltrate surrounding brain. Tumour cells acquire genetic defects which result in anaplastic transformation to a high-grade lesion, and 5070% of lesions are high grade at the time of tumour progression.5,12,49 The clinical effects of LGG are

Figure 4: Fluorescence in-situ hybridisation for chromosomes 1 and 19 in an oligodendroglioma showing codeletion of chromosomes 1p and 19q Formalin-xed, parafn-embedded tissues were analysed with four human DNA probes localising 1p, 1q, 19p, and 19q. Left: Analysis of chromosome 1 shows one signal for 1p (rhodamine red), compared with 1q (FITC green). Right: Analysis of chromosome 19 shows one signal for 19q (rhodamine red), compared with 19p (FITC green).

763

Review

variable, with survival ranging from less than 2 years to more than 10 years. Clinical, tumour, and treatmentrelated factors each contribute to the variability.

Clinical factors
Age over 40 years is a predictor of poor outcome. In multivariate analysis of two large, prospective trials, age over 40 years had a hazard ratio for death of 143 (95% CI 117174); this increased risk is unlikely to result from age-related treatment disparity.7 Age-associated physiological and pathological changes and comorbidities may predispose to adverse outcomes. Bahary and colleagues50 noted that tumour volumes in patients older than 35 years were larger than those in younger patients. Alternatively, tumour biology may differ in old patients. Age at diagnosis of low-grade astrocytoma is inversely correlated with time to progression,51 and proliferative index may be higher among those older than 40 years.52,53 Ageing-related impairment of DNA repair and the resulting acquisition of mutations may promote rapid progression after transformation occurs.51,54 Clinical presentation is a strong prognostic factor, whether expressed as the presence of seizures, absence of neurological decits, or good performance status.7,11,55 These factors are inter-relatedeg, neurologically intact patients who present with isolated seizures have a better performance status and prognosis.7,11,55,56 The presence of seizures may select otherwise intact patients for earlier diagnosis than in those without seizures.57 Patients who present with seizures tend to be younger and have smaller tumours than those without seizures.5759 LGG associated with epilepsy might differ biologically from LGG in patients presenting with neurological decits.6,60 Tumours in patients presenting with epilepsy may represent histological variants of other, more benign, low-grade tumours, such as dysembryoplastic neuroepithelial tumours.60 Corticallybased tumours associated with seizures may be derived from a different astrocytic lineage than tumours in white matter.61 Occasional association with cortical dysplasia suggests derivation of these tumours from a developmental abnormality.60,62

with a worse outcome in some11,65 but not all studies.5,10 One explanation is sampling errorie, the reviewed sample may not reect the pathology in the enhancing portion of the tumour. The proportion of LGG with contrast enhancement has not declined substantially in the modern surgical era when enhancing regions can accurately be targeted for biopsy. Thus, one cannot exclude an underlying biological effect to account for this relation.

Treatment
Surgery
Surgery has long been important in the diagnosis and treatment of LGG. Although the presence of a nonenhancing mass lesion on imaging suggests LGG, about 30% of such lesions are higher grade than predicted by imaging, particularly those in old patients.66,67 At a minimum, stereotactic biopsy is required to establish diagnosis, although timing is debatable. A neurosurgeon using image guidance can precisely target a radiographic abnormality via a burr hole to sample a lesion without exposing the patient to unnecessary risk. Compared with craniotomy, stereotactic biopsy is less invasive and requires a shorter period of hospitalisation. However, biopsy provides only a sample of a potentially heterogeneous lesion. Biopsy tissue might be insufcient for additional molecular and genetic testing that could facilitate diagnosis, provide prognostic information, and affect subsequent treatment. Alternatively, cytoreductive surgery allows for rapid decompression of larger lesions associated with mass effect. Debulking may palliate symptoms, improve quality of life, and allow for rapid tapering of corticosteroids. Nonetheless, the anatomic constraints of the CNS may limit the extent to which a lesion is surgically accessible. Furthermore, the inltrative growth pattern of these tumours means that an attempt to resect the tumour places the patient at risk of neurological injury.68 The effect of surgery on overall and progression free survival is uncertain. The inltrative nature of LGG is such that that they are not surgically curable. To date, a prospective randomised surgical trial has not been done. The available data are derived from retrospective series that have substantial limitations. Until recently, the extent of resection was based on the surgeons intraoperative assessment. Currently, postoperative neuroimaging allows for more accurate assessment of extent of resection. The method by which extent of resection was dened and presented has varied between studies.69 For example, some studies compared biopsy with resection, whereas others compared gross total resection with less extensive surgery. Sample size can be insufcient for multivariate analysis. Thus, the benet of surgery may be gained or lost in analysis based on other clinical or treatment variables. Although
http://neurology.thelancet.com Vol 4 November 2005

Tumour factors
The median survival for oligodendrogliomas is 10 years, a better prognosis than for astrocytomas, which have a median survival of 7 years. Gemistocytic astrocytomas, a subtype of grade II astrocytomas, are more aggressive than predicted by grade. Large tumours, non-lobar gliomas, and tumours that cross midline are associated with a short survival (and time to progression) and a high rate of malignant dedifferentiation.7,10,11,63 Such tumours can be less amenable to surgical resection. Proliferative index has occasionally been inversely related with outcome.16,64 Contrast enhancement of LGG has been associated
764

Review

some studies found a survival advantage with more extensive surgery,5,50,70 others did not.10,65,71 In some series, the extent of resection lost signicance in favour of other related variables, such as preoperative7,10 or postoperative tumour volume,72 or the location or distribution of the lesion.7,63,73 Nonetheless, evidence that more extensive surgery may have a positive effect on outcome is growing.69 The timing of surgery is controversial. A large proportion of patients with LGG are young and present with isolated seizures that may be medically well controlled. Potential surgical morbidity may compromise their otherwise intact functional status. Some have advocated deferring surgery in lieu of radiographic surveillance in select populations.74 The risk of deferring surgery includes managing a larger tumour that may have undergone anaplastic transformation in an older patient. Tumour progression may also result in increased symptom burden. Based on limited retrospective data, however, the timing of surgery does not seem to affect overall or progression-free survival, the time to malignant progression or the rate of malignant progression.12,74

Radiotherapy
Although radiotherapy has played an important part in the management of LGG, the efcacy and timing of this treatment has been debated. Until recently, data were derived from retrospective case series that yielded conicting resultsie, some studies found a survival advantage with radiotherapy,75 whereas others failed to do so.70 Non-uniformity of inclusion criteria and the therapeutic regimens used may account for the differences.76 Also, clinical or radiographic factors of prognostic importance are used in treatment selection.5,50 Although it has become practice in some institutions to defer therapy until progression, most patients eventually receive treatment. Several retrospective case series reported similar survival among patients treated postoperatively or at progression.5,50,65,77,78 The European Organisation for Research and Treatment of Cancer randomly assigned patients with LGG to postoperative radiotherapy or observation.79 Patients given radiotherapy had a substantially longer time to progression (48 years) than those who were observed (34 years). 66% of those who were initially observed received radiotherapy at progression. Survival was identical between groups, suggesting that radiotherapy is as effective if given at progression as at initial diagnosis. The results must be interpreted with caution because median follow-up was only 60 months and only 32% of patients died. Radiation treatment elds and doses have varied with time and treatment facility. The inltrative nature of LGG is such that tumour cells may be identied at sites distant from radiographic abnormalities. Early practice was to treat patients with whole brain radiotherapy with
http://neurology.thelancet.com Vol 4 November 2005

or without a boost to the lesion. Recurrences, however, generally occurred in the vicinity of the original lesion. Common practice now is to use partial elds directed at the tumour with a small surrounding margin; although this has not compromised outcome it has reduced treatment toxicity.80 The optimum radiation dose was also debatable because early studies reported conicting data for dose response. Two prospective clinical trials in Europe and the USA assigned patients to low-dose and high-dose regimens (table 3).10,63,79 Neither study found a difference in overall or progression-free survival, and there was more toxicity in high-dose groups.10,81 Radiographic response rate was similar in high-dose and low-dose groups with at least a 50% reduction in tumour size occurring in 32% of patients.10 The adverse effect of radiation on cognition and quality of life is an important part of the debate about radiotherapy and LGG. Of greatest concern to oncologists and patients with LGG is radiation encephalopathy, a neurobehavioural syndrome with some similarities to normal pressure hydrocephalus. Onset is delayed, commonly beginning months to years after the completion of radiotherapy. Patients typically have personality changes, impaired gait and balance, urinary incontinence, and declines in attention, memory, and executive functioning.82 As a result, patients are commonly unable to maintain employment and overall quality of life is diminished. The syndrome is progressive and may culminate in death despite tumour control.82 Although the mechanism of injury has not been elucidated, demyelination and a small-vessel vasculopathy are suspected.82,83 No treatments are available; stimulants and acetylcholinesterase inhibitors are being investigated.8486 Clinical improvement with CSF diversion has been reported87 although the improvement may not be sustained.82 Risk factors for radiation encephalopathy include treatment and patient-related parameters. The larger the volume of brain irradiated, the greater the risk of toxicity. Patients treated with whole-brain radiotherapy have a high incidence of radiation encephalopathy.80,88 Risk of radiation encephalopathy is also associated with high total-radiation dose10,89 and large fraction size.88 Current practice of targeting the tumour with a small
Reference 10 63 79 Treatment (dose/fractions) 504/28 648/36 45/25 594/33 54/30 Number of patients 101 102 171 172 140 150 % 5 year PFS 55 52 47 50 37* 44 % 5 year OS 72 65 58 59 66 63 Median follow-up (months) 768 74 60

PFS=progression-free survival; OS=overall survival. *p 005.

Table 3: Prospective randomised studies of radiation treatment of LGG

765

Review

surrounding margin reduces treatment volumes and minimises the exposure of normal brain to radiation. Unfortunately, the inltrative nature of LGG at times necessitates large treatment elds even when focal strategies are used. Modern regimens that incorporate total doses of less than 60 Gy and fraction sizes of less than 2 Gy have minimised but not eliminated this complication. Children irradiated within the rst decade of life have a higher rate of learning disabilities than those irradiated at an older age. People older than 60 years develop radiographic evidence of injury sooner and more severely than younger patients.88,89 Comorbidities (eg, hypertension, stroke, diabetes, and degenerative brain) and concomitant chemotherapy may predispose patients to radiation encephalopathy.90 The effect of radiotherapy on cognitive status and quality of life in patients with LGG is uncertain because the available data have several limitations. Studies commonly included tumours other than gliomas,80,91 and gliomas of higher or lower grade.89,92,93 Most studies were small and occasionally included paediatric patients and patients treated with chemotherapy. Methods of neuropsychological assessment and grading of imaging abnormalities varied. Few studies included an untreated LGG control group.94,95 Treatment regimens were heterogeneous both within and between studies, with some series including patients treated with whole brain radiotherapy.80,88,92,94,96 Quality-of-life assessment was rarely done. Information about the outcomes of patients with LGG who are treated with radiotherapy continues to grow. Patients with LGG have worse cognition than healthy controls and patients with systemic cancers regardless of treatment.94,95 Most studies failed to identify a statistically signicant, detrimental effect of radiotherapy on outcome. Surma-aho and colleagues92 noted that 28 patients receiving postoperative radiotherapy (19 of whom received whole-brain elds and 9 of whom received focal therapy) did worse on several cognitive tests than those who deferred treatment. Vigliani and colleagues93 noted that patients treated with focal radiotherapy have slower reaction times and a lower rate of employment during the year after treatment than those who deferred treatment. In addition, mean scores in ten of 12 additional test batteries were lower among those treated with radiotherapy postoperatively. By 1 year, the groups were similar. Up to 4 years posttreatment, no additional differences were present. Selective decline in visual memory, beginning at 5 years post-treatment, was noted by Armstrong and colleagues.91 No other differences between pre-treatment and post-treatment cognitive status were found. Other studies failed to show an adverse effect of radiotherapy on cognition. In fact, radiotherapy was associated with improved cognition in some studies.91,93,94,97 Radiotherapy had no effect on quality of life.81

The adverse effects of radiotherapy can also be assessed radiographically. Chronically, radiotherapy is associated with diffuse cerebral atrophy and lesions within the white matter. Hypolucencies on CT and T2 hyperintensities on MRI, typically within the treatment elds, suggest white-matter lesions.89,91 Abnormalities are more common in patients treated with whole-brain radiotherapy. A relation between radiographic abnormalities and cognition was present in studies that included a large proportion of patients treated with whole-brain radiotherapy.88,92 On the contrary, this correlation was not present in studies composed predominantly of patients treated with focal regimens.89,91 Although early radiotherapy does not affect the overall survival of all patients with LGG, some subgroups of patients may benet from early treatment. Several studies noted that patients over age 40 years beneted from radiotherapy more than younger patients.98101 Also, patients with a large volume of residual tumour may benet from postoperative radiotherapy relative to those treated with a gross total resection.98,102 Tumours with a high proliferative index, substantial enhancement, or aggressive histological subtypes, such as the grade II gemistocytic astrocytoma, may benet from early treatment. Prospective study of these factors has not been reported.

Chemotherapy
Until recently, chemotherapy was thought to be of only small benet in the treatment of patients with gliomas. In the late 1980s, anaplastic oligodendrogliomas, with or without an astrocytic component, were recognised to be chemosensitive.103 Identication of genetic markers that predicted a better response to chemotherapy further increased interest in this type of treatment.104 Several phase II trials of procarbazine, lomustine, and vincristine (PCV) or temozolomide in the treatment of new, progressive, or recurrent LGG have been done (table 4).105112 Follow-up was short and radiographic response was the primary endpoint. Comparison between studies was difcult because inclusion criteria varied. Patients with enhancing lesions, possibly reecting high-grade pathology, were more responsive to chemotherapy than those without.105,111 A similar response rate was noted for astrocytomas and oligodendroglial tumours.106,111,112 Resolution of focal neurological decits and improvement in seizure frequency and quality of life were reported in responders and those with stable disease.105,109,111114 Chemotherapy was effective in previously treated and untreated patients.107,108,112 Although LGG with deletion of chromosome 1p and 19q may be more sensitive to treatment,109,115 tumours without these deletions were also responsive to chemotherapy.114 The most extensively studied drugs are PCV and singleagent temozolomide. Lomustine is associated with
http://neurology.thelancet.com Vol 4 November 2005

766

Review

Study 105 106 107 108 109 110 111 112

Pathology Oligodendroglioma, oligoastrocytoma Astrocytoma, oligodendroglioma, oligoastrocytoma Oligodendroglioma, oligoastrocytoma Oligodendroglioma, oligoastrocytoma Oligodendroglioma, oligoastrocytoma Oligodendroglioma, oligoastrocytoma Astrocytoma, Oligodendroglioma, oligoastrocytoma Astrocytoma, Oligodendroglioma, oligoastrocytoma

Chemotherapy PCV Temozolomide Temozolomide Temozolomide Temozolomide PCV; radiotherapy Temozolomide Temozolomide

% previous radiotherapy 42 15 100 100 0 0 0 70

% previous CT 0 22 0 100 0 0 0 37

% contrast enhancement 73 70 100 100 11 43 0 61

1-year PFS 80* 76 40 11 73 91 92* 39

% radiographic response 62 61 53 22 17 29 10 47

PFS=progression-free survival. *Estimated; regression.

Table 4: Phase II studies of chemotherapy in LGG

substantial cumulative myelotoxicty and uncommonly irreversible pulmonary injury. Procarbazine produces putative weak monoamine-oxidase-inhibitor activity, and patients taking this drug should be given dietary restrictions. Vincristine, an intravenous chemotherapy, may result in neuropathy and ileus. Patients are commonly unable to complete scheduled treatment despite tumour stability.110,114 Temozolomide, an oral alkylating drug with good bioavailability and CNS penetration, is generally well tolerated.116 Substantial toxicity occurs in less than 10% of patients and cumulative toxicity has not been reported. A standard regimen, adopted from use in high-grade gliomas, is dosing on days 15 of a 28 day cycle. Alternatively, a protracted course of low-dose temozolomide, given on days 121 of a 28 day cycle, has been used. This regimen provides 158 times greater drug exposure over 4 weeks than the standard regimen, and it depletes the DNA repair enzyme, O6-alkylguanine-DNA alkyltransferase, thereby theoretically decreasing drug resistance.117 A protracted course of low-dose temozolomide may also interfere with tumour angiogenesis.118 The favourable side-effect prole and ease of use of temozolomide have made it the drug of choice. Although comparative studies have not been reported, the efcacy of the regimens seems similar.

The future
Many issues are unresolved in the management of LGG. Chemotherapy is emerging as an alternative to radiotherapy as a neoadjuvant treatment. Whether chemotherapy is more effective than up-front radiotherapy is unknown, as is optimum timing of intervention. Ideally, risk stratication from clinical, radiographic, and tumour factors would identify patients who would benet from early treatment. Although this approach is used in practice, it has yet to be proven. Molecular markers may allow oncologists to tailor treatment to a particular patient. For example, promotor hypermethylation of O6-methylguanine-DNA methyltransferase, a gene that encodes a DNA repair enzyme, has been associated with better response to temozolomide in high-grade gliomas.119 Whether it confers the same benet in LGG is to be determined. Other markers of tumour behaviour and responsiveness, such as 1p/19q status, need to be investigated further. Therapies that specically target molecular and genetic abnormalities within neoplastic cells are emerging. The advantage of non-cytotoxic strategies is that they specically target tumour cells while minimising toxicity. Currently, such strategies are being explored in high-grade gliomas. Alternative modes of chemotherapy delivery, such as delivery of drugs via catheters surgically placed in the vicinity of
Stratication End points 3 year overall survival; PFS; toxicity PFS

Inclusion RTOG Phase II trial of previously untreated high-risk* patients with grade II inltrating gliomas Phase III trial of adults with untreated progressive or recurrent grade II gliomas Proposed phase III trial of adults with symptomatic (with exception of wellcontrolled seizures) or progressive grade II gliomas

Treatment Radiation (5400 cGy in 30 fractions) with concomitant daily temozolomide (75 mg/m2) followed by 12 cycles standard temozolomide Patients randomised to radiation (5040 cGy in 28 fractions) or temozolomide (75 mg/m2) on days 1 through 21 of 28 day cycle Patients randomised to radiation (5040 cGy in 28 fractions) with or without concomitant temozolomide (75 mg/m2 during radiotherapy) followed by standard temozolomide

EORTC

1p status; age ( 40 years or 40 years); contrast enhancement; performance status 1p/19q status; age ( 40 years or 40 years); postoperative tumour size ( or 5 cm); performance status

ECOG/ NCCTG

PFS; survival; quality of life

RTOG=radiation therapy oncology group; EORTC=European Organisation for Research and Treatment of Cancer; ECOG/NCCTG=Eastern Cooperative Oncology Group/North Central Cancer Treatment Group; PFS=progression-free survival. *High risk=three of these features: age 40; tumour 6 cm or crossing midline; astrocytoma or astrocytic predominant mixed glioma; zubrod 02; preoperative NFS 1.

Table 5: Ongoing and proposed large phase II and III trials of LGG

http://neurology.thelancet.com Vol 4 November 2005

767

Review

15

Search strategy and selection criteria We identied references by searching MEDLINE (1966 to 2005) in March 2005 for low-grade glioma, radiotherapy, chemotherapy, surgery, and molecular genetics. Although all studies that included patients treated after the introduction of CT were considered, preference was given to studies published after 1995 and to prospective studies. Only studies published in English and focusing on LGG were considered. References were also identied from relevant articles and through searches of our les.

16

17 18

19

20

the tumour, and the targeting of tumour cells via a viral vector are also being explored in high-grade gliomas. Whether these approaches may benet patients with LGG is unknown. Several clinical trials designed to explore some of these issues are ongoing (table 5).
Authors contributions We all contributed equally to the preparation of the paper. Conicts of interest We have no conicts of interest. References 1 Kleihues P, Cavenee WK. Pathology and genetics of tumors of the nervous system. France: IARC Press, 2000. 2 Perry A. Pathology of low-grade gliomas: an update of emerging concepts. Neuro-oncol 2003; 5: 16878. 3 CBTRUS. Statistical report: primary brain tumors in the United States, 19951999. USA: Central Brain Tumor Registry of the United States, 2002. 4 Duffau H, Capelle L. Preferential brain locations of low-grade gliomas. Cancer 2004; 100: 262226. 5 Leighton C, Fisher B, Bauman G, et al. Supratentorial low-grade glioma in adults: an analysis of prognostic factors and timing of radiation. J Clin Oncol 1997; 15: 1294. 6 Piepmeier J, Christopher S, Spencer D, et al. Variations in the natural history and survival of patients with supratentorial lowgrade astrocytomas. Neurosurgery 1996; 38: 87278. 7 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: 207684. 8 Wrensch M, Minn Y, Chew T, Bondy M, Berger MS. Epidemiology of primary brain tumors: current concepts and review of the literature. Neuro-oncol 2002; 4: 27899. 9 Henson JW, Gaviani P, Gonzalez RG. MRI in treatment of adult gliomas. Lancet Oncol 2005; 6:16775. 10 Shaw E, Arusell R, Scheithauer B, et al. Prospective randomized trial of low- versus high-dose radiation therapy in adults with supratentorial low-grade glioma: initial report of a North Central Cancer Treatment Group/Radiation Therapy Oncology Group/Eastern Cooperative Oncology Group study. J Clin Oncol 2002; 20: 226776. 11 Kreth FW, Faist M, Rossner R, Volk B, Ostertag CB. Supratentorial World Health Organization grade 2 astrocytomas and oligoastrocytomas: a new pattern of prognostic factors. Cancer 1997; 79: 37079. 12 Olson JD, Riedel E, DeAngelis LM. Long-term outcome of lowgrade oligodendroglioma and mixed glioma. Neurology 2000; 54: 144248. 13 Karamitopoulou E, Perentes E, Diamantis I, Maraziotis T. Ki-67 immunoreactivity in human central nervous system tumors: a study with MIB 1 monoclonal antibody on archival material. Acta Neuropathol (Berl) 1994; 87: 4754. 14 Montine TJ, Vandersteenhoven JJ, Aguzzi A, et al. Prognostic signicance of Ki-67 proliferation index in supratentorial brillary astrocytic neoplasms. Neurosurgery 1994; 34: 67478.

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

Giannini C, Scheithauer BW, Burger PC, et al. Cellular proliferation in pilocytic and diffuse astrocytomas. J Neuropathol Exp Neurol 1999; 58: 4653. Coons SW, Johnson PC, Pearl DK. The prognostic signicance of Ki-67 labeling indices for oligodendrogliomas. Neurosurgery 1997; 41: 87884. Krouwer HG, Davis RL, Silver P, Prados M. Gemistocytic astrocytomas: a reappraisal. J Neurosurg 1991; 74: 399406. Watanabe K, Tachibana O, Yonekawa Y, Kleihues P, Ohgaki H. Role of gemistocytes in astrocytoma progression. Lab Invest 1997; 76: 27784. Cavanee WK, Furnari FB, Nagane M, Huang HJS, Newcomb EW, Bigner DD. Diffusely inltrating astrocytomas. In: Kleihues P, Cavanee WK, eds. Pathology and genetics of tumours of the central nervous system. Lyon: IARC, 2000: 1021. Nakamura M, Watanabe T, Klangby U, et al. p14ARF deletion and methylation in genetic pathways to glioblastomas. Brain Pathol 2001; 11: 15968. Wessels PH, Twijnstra A, Kessels AG, et al. Gain of chromosome 7, as detected by in situ hybridization, strongly correlates with shorter survival in astrocytoma grade 2. Genes Chromosomes Cancer 2002; 33: 27984. Burger PC, Minn AY, Smith JS, et al. Losses of chromosomal arms 1p and 19q in the diagnosis of oligodendroglioma: a study of parafn-embedded sections. Mod Pathol 2001; 14: 84253. Kros JM, Van Eden CG, Stefanko SZ, Waayer-Van Batenburg M, van der Kwast TH. Prognostic implications of glial brillary acidic protein containing cell types in oligodendrogliomas. Cancer 1990; 66: 120412. Herpers MJ, Budka H. Glial brillary acidic protein (GFAP) in oligodendroglial tumors: gliobrillary oligodendroglioma and transitional oligoastrocytoma as subtypes of oligodendroglioma. Acta Neuropathol (Berl) 1984; 64: 26572. Lu QR, Yuk D, Alberta JA, et al. Sonic hedgehogregulated oligodendrocyte lineage genes encoding bHLH proteins in the mammalian central nervous system. Neuron 2000; 25: 31729. Zhou Q, Wang S, Anderson DJ. Identication of a novel family of oligodendrocyte lineage-specic basic helix-loop-helix transcription factors. Neuron 2000; 25: 33143. Lu QR, Park JK, Noll E, et al. Oligodendrocyte lineage genes (OLIG) as molecular markers for human glial brain tumors. Proc Natl Acad Sci USA 2001; 98: 1085156. Hoang-Xuan K, Aguirre-Cruz L, Mokhtari K, Marie Y, Sanson M. OLIG-1 and 2 gene expression and oligodendroglial tumours. Neuropathol Appl Neurobiol 2002; 28: 8994. Marie Y, Sanson M, Mokhtari K, et al. OLIG2 as a specic marker of oligodendroglial tumour cells. Lancet 2001; 358: 298300. Bouvier C, Bartoli C, Aguirre-Cruz L, et al. Shared oligodendrocyte lineage gene expression in gliomas and oligodendrocyte progenitor cells. J Neurosurg 2003; 99: 34450. Ligon KL, Alberta JA, Kho AT, et al. The oligodendroglial lineage marker OLIG2 is universally expressed in diffuse gliomas. J Neuropathol Exp Neurol 2004; 63: 499509. Coons SW, Johnson PC, Scheithauer BW, Yates AJ, Pearl DK. Improving diagnostic accuracy and interobserver concordance in the classication and grading of primary gliomas. Cancer 1997; 79: 138193. McLendon RE, Rasheed A, Wiltshire R, Herndon J. Correlation of 1p-19q-defects in human gliomas with the light microscopic appearance of oligodendroglioma. Mod Pathol 2004; 17: 60405. Reifenberger G, Kros JM, Burger PC. Oligodendroglioma. In: Kleihues P, Cavanee WK, eds. Pathology and genetics of tumours of the central nervous system. Lyon: IARC, 2000: 5661. Fuller CE, Perry A. Fluorescence in situ hybridization (FISH) in diagnostic and investigative neuropathology. Brain Pathol 2002; 12: 6786. Ohgaki H, Eibl RH, Wiestler OD, Yasargil MG, Newcomb EW, Kleihues P. p53 mutations in nonastrocytic human brain tumors. Cancer Res 1991; 51: 620205. Reifenberger G, Louis DN. Oligodendroglioma: toward molecular denitions in diagnostic neuro-oncology. J Neuropathol Exp Neurol 2003; 62: 11126.

768

http://neurology.thelancet.com Vol 4 November 2005

Review

38

39

40

41

42

43 44

45

46

47

48

49

50

51

52

53

54 55

56

57

58

59

Reifenberger G, Kros JM, Burger PC, Louis DN, Collins VP. Oligoastrocytoma. In: Kleihues P, Cavanee WK, eds. Pathology and genetics of tumours of the central nervous system. Lyon: IARC, 2000: 6567. Kraus JA, Koopmann J, Kaskel P, et al. Shared allelic losses on chromosomes 1p and 19q suggest a common origin of oligodendroglioma and oligoastrocytoma. J Neuropathol Exp Neurol 1995; 54: 9195. Fuller CE, Schmidt RE, Roth KA, et al. Clinical utility of uorescence in situ hybridization (FISH) in morphologically ambiguous gliomas with hybrid oligodendroglial/astrocytic features. J Neuropathol Exp Neurol 2003; 62: 111828. Reifenberger J, Reifenberger G, Liu L, James CD, Wechsler W, Collins VP. Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p. Am J Pathol 1994; 145: 117590. Maintz D, Fiedler K, Koopmann J, et al. Molecular genetic evidence for subtypes of oligoastrocytomas. J Neuropathol Exp Neurol 1997; 56: 1098104. Begemann M, Fuller GN, Holland EC. Genetic modeling of glioma formation in mice. Brain Pathol 2002; 12: 11732. Weiss WA, Israel M, Cobbs C, et al. Neuropathology of genetically engineered mice: consensus report and recommendations from an international forum. Oncogene 2002; 21: 745363. Weissenberger J, Steinbach JP, Malin G, Spada S, Rulicke T, Aguzzi A. Development and malignant progression of astrocytomas in GFAP-v-src transgenic mice. Oncogene 1997; 14: 200513. Ding H, Roncari L, Shannon P, et al. Astrocyte-specic expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas. Cancer Res 2001; 61: 382636. Dai C, Celestino JC, Okada Y, Louis DN, Fuller GN, Holland EC. PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev 2001; 15: 191325. Holland EC, Li Y, Celestino J, et al. Astrocytes give rise to oligodendrogliomas and astrocytomas after gene transfer of polyoma virus middle T antigen in vivo. Am J Pathol 2000; 157: 103137. Berger MS, Deliganis AV, Dobbins J, Keles GE. The effect of extent of resection on recurrence in patients with low grade cerebral hemisphere gliomas. Cancer 1994; 74: 178491. Bahary JP, Villemure JG, Choi S, et al. Low-grade pure and mixed cerebral astrocytomas treated in the CT scan era. J Neurooncol 1996; 27: 17377. Shafqat S, Hedley-Whyte ET, Henson JW. Age-dependent rate of anaplastic transformation in low-grade astrocytoma. Neurology 1999; 52: 86769. Franzini A, Leocata F, Cajola L, Servello D, Allegranza A, Broggi G. Low-grade glial tumors in basal ganglia and thalamus: natural history and biological reappraisal. Neurosurgery 1994; 35: 81720. McKeever PE, Junck L, Strawderman MS, et al. Proliferation index is related to patient age in glioblastoma. Neurology 2001; 56: 121618. Evans DA, Burbach JP, van Leeuwen FW. Somatic mutations in the brain: relationship to aging? Mutat Res 1995; 338: 17382. Schuurman PR, Troost D, Verbeeten B Jr, Bosch DA. 5-year survival and clinical prognostic factors in progressive supratentorial diffuse low-grade astrocytoma: a retrospective analysis of 46 cases. Acta Neurochir (Wien) 1997; 139: 27. Baumann F, Bjeljac M, Kollias SS, et al. Combined thalidomide and temozolomide treatment in patients with glioblastoma multiforme. J Neurooncol 2004; 67: 191200. Whittle IR, Beaumont A. Seizures in patients with supratentorial oligodendroglial tumours: clinicopathological features and management considerations. Acta Neurochir (Wien) 1995; 135: 1924. Lote K, Stenwig AE, Skullerud K, Hirschberg H. Prevalence and prognostic signicance of epilepsy in patients with gliomas. Eur J Cancer 1998; 34: 98102. Smith DF, Hutton JL, Sandemann D, et al. The prognosis of primary intracerebral tumours presenting with epilepsy: the

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74 75

76 77

78

79

80 81

outcome of medical and surgical management. J Neurol Neurosurg Psychiatry 1991; 54: 91520. Bartolomei JC, Christopher S, Vives K, Spencer DD, Piepmeier JM. Low-grade gliomas of chronic epilepsy: a distinct clinical and pathological entity. J Neurooncol 1997; 34: 7984. Piepmeier JM, Fried I, Makuch R. Low-grade astrocytomas may arise from different astrocyte lineages. Neurosurgery 1993; 33: 62732. Prayson RA, Estes ML, Morris HH. Coexistence of neoplasia and cortical dysplasia in patients presenting with seizures. Epilepsia 1993; 34: 60915. Karim AB, Maat B, Hatlevoll R, et al. A randomized trial on doseresponse in radiation therapy of low-grade cerebral glioma: European Organization for Research and Treatment of Cancer (EORTC) study 22844. Int J Radiat Onco Biol Phys 1996; 36: 549. McKeever PE, Strawderman MS, Yamini B, Mikhail AA, Blaivas M. MIB-1 proliferation index predicts survival among patients with grade II astrocytoma. J Neuropathol Exp Neurol 1998; 57: 93136. Bauman G, Lote K, Larson D, et al. Pretreatment factors predict overall survival for patients with low-grade glioma: a recursive partitioning analysis. Int J Radiat Oncol Biol Phys 1999; 45: 92329. Barker FG 2nd, Chang SM, Huhn SL, et al. Age and the risk of anaplasia in magnetic resonance-nonenhancing supratentorial cerebral tumors. Cancer 1997; 80: 93641. Kondziolka D, Lunsford LD, Martinez AJ. Unreliability of contemporary neurodiagnostic imaging in evaluating suspected adult supratentorial (low-grade) astrocytoma. J Neurosurg 1993; 79: 53336. Kelly PJ, Daumas-Duport C, Scheithauer BW, Kall BA, Kispert DB. Stereotactic histologic correlations of computed tomography and magnetic resonance imaging dened abnormalities in patients with glial neoplasms. Mayo Clin Proc 1987; 62: 45059. Keles GE, Lamborn KR, Berger MS. Low-grade hemispheric gliomas in adults: a critical review of extent of resection as a factor inuencing outcome. J Neurosurg 2001; 95: 73545. Nicolato A, Gerosa MA, Fina P, Iuzzolino P, Giorgiutti F, Bricolo A. Prognostic factors in low-grade supratentorial astrocytomas: a unimultivariate statistical analysis in 76 surgically treated adult patients. Surg Neurol 1995; 44: 20821. Grabenbauer GG, Roedel CM, Paulus W, et al. Supratentorial lowgrade glioma: results and prognostic factors following postoperative radiotherapy. Strahlenther Onkol 2000; 176: 25964. Wood JR, Green SB, Shapiro WR. The prognostic importance of tumor size in malignant gliomas: a computed tomographic scan study by the Brain Tumor Cooperative Group. J Clin Oncol 1988; 6: 33843. Shibamoto Y, Kitakabu Y, Takahashi M, et al. Supratentorial lowgrade astrocytoma: correlation of computed tomography ndings with effect of radiation therapy and prognostic variables. Cancer 1993; 72: 19095. Recht LD, Lew R, Smith TW. Suspected low-grade glioma: is deferring treatment safe? Ann Neurol 1992; 31: 43136. Jeremic B, Bamberg M. Radiation therapy for incompletely resected supratentorial low-grade glioma in adults. J Neurooncol 2001; 55: 10112. Morantz RA. Radiation therapy in the treatment of cerebral astrocytoma. Neurosurgery 1987; 20: 97582. van Veelen ML, Avezaat CJ, Kros JM, van Putten W, Vecht C. Supratentorial low grade astrocytoma: prognostic factors, dedifferentiation, and the issue of early versus late surgery. J Neurol Neurosurg Psychiatry 1998; 64: 58187. Knisely JP, Haffty BG, Christopher SR. Early vs delayed radiotherapy in a small cohort of patients with supratentorial low grade glioma. J Neurooncol 1997; 34: 2329. Karim AB, Afra D, Cornu P, et al. Randomized trial on the efcacy of radiotherapy for cerebral low-grade glioma in the adult: European Organization for Research and Treatment of Cancer study 22845 with the Medical Research Council study BRO4, an interim analysis. Int J Radiat Oncol Biol Phys 2002; 52: 31624. Gregor A, Cull A. Radiotherapy for malignant glioma. BMJ 1996; 313: 150001. Kiebert GM, Curran D, Aaronson NK, et al. Quality of life after radiation therapy of cerebral low-grade gliomas of the adult: results of a randomised phase III trial on dose response (EORTC trial 22844). Eur J Cancer 1998; 34: 190209.

http://neurology.thelancet.com Vol 4 November 2005

769

Review

Vigliani MC, Duyckaerts C, Hauw JJ, Poisson M, Magdelenat H, Delattre JY. Dementia following treatment of brain tumors with radiotherapy administered alone or in combination with nitrosoureabased chemotherapy: a clinical and pathological study. J Neurooncol 1999; 41: 13749. 83 Schultheiss TE, Kun LE, Ang KK, Stephens LC. Radiation response of the central nervous system. Int J Radiat Oncol Biol Phys 1995; 31: 1093112. 84 Meyers CA, Weitzner MA, Valentine AD, Levin VA. Methylphenidate therapy improves cognition, mood, and function of brain tumor patients. J Clin Oncol 1998; 16: 252227. 85 Kaleita T, Cloughesy T, Ford J, et al. A pilot study of Modanil for treatment of fatigue and neurobehavioral dysfunctions in adult brain tumor patients. Neuro-oncol 2004; 6: 356. 86 Rapp SR, DAgostini RB, Lovato J, Naughton M, Robbins ME, Shaw EG. Improving cognitive functioning in brain irradiated patients: a phase II trial of an acetylcholinesterase inhibitor. Neuro-oncol 2004; 6: 358. 87 Asai A, Matsutani M, Kohno T, et al. Subacute brain atrophy after radiation therapy for malignant brain tumor. Cancer 1989; 63: 196274. 88 Swennen MH, Bromberg JE, Witkamp TD, Terhaard CH, Postma TJ, Taphoorn MJ. Delayed radiation toxicity after focal or whole brain radiotherapy for low-grade glioma. J Neurooncol 2004; 66: 33339. 89 Corn BW, Yousem DM, Scott CB, et al. White matter changes are correlated signicantly with radiation dose: observations from a randomized dose-escalation trial for malignant glioma. Cancer 1994; 74: 282835. 90 Keime-Guibert F, Napolitano M, Delattre JY. Neurological complications of radiotherapy and chemotherapy. J Neurol 1998; 245: 695708. 91 Armstrong CL, Hunter JV, Ledakis GE, et al. Late cognitive and radiographic changes related to radiotherapy: initial prospective ndings. Neurology 2002; 59: 4048. 92 Surma-aho O, Niemela M, Vilkki J, et al. Adverse long-term effects of brain radiotherapy in adult low-grade glioma patients. Neurology 2001; 56: 128590. 93 Vigliani MC, Sichez N, Poisson M, Delattre JY. A prospective study of cognitive functions following conventional radiotherapy for supratentorial gliomas in young adults: 4-year results. Int J Radiat Oncol Biol Phys 1996; 35: 52733. 94 Klein M, Heimans JJ, Aaronson NK, et al. Effect of radiotherapy and other treatment-related factors on mid-term to long-term cognitive sequelae in low-grade gliomas: a comparative study. Lancet 2002; 360: 136168. 95 Reijneveld JC, Sitskoorn MM, Klein M, Nuyen J, Taphoorn MJ. Cognitive status and quality of life in patients with suspected versus proven low-grade gliomas. Neurology 2001; 56: 61823. 96 Postma TJ, Klein M, Verstappen CC, et al. Radiotherapy-induced cerebral abnormalities in patients with low-grade glioma. Neurology 2002; 59: 12123. 97 Brown PD, Buckner JC, OFallon JR, et al. Effects of radiotherapy on cognitive function in patients with low-grade glioma measured by the folstein mini-mental state examination. J Clin Oncol 2003; 21: 251924. 98 Philippon JH, Clemenceau SH, Fauchon FH, Foncin JF. Supratentorial low-grade astrocytomas in adults. Neurosurgery 1993; 32: 55459. 99 Shaw EG, Daumas-Duport C, Scheithauer BW, et al. Radiation therapy in the management of low-grade supratentorial astrocytomas. J Neurosurg 1989; 70: 853. 100 Laws ER Jr, Taylor WF, Clifton MB, Okazaki H. Neurosurgical management of low-grade astrocytoma of the cerebral

82

hemispheres. J Neurosurg 1984; 61: 66573. 101 Garcia DM, Fulling KH, Marks JE. The value of radiation therapy in addition to surgery for astrocytomas of the adult cerebrum. Cancer 1985; 55: 91927. 102 Shimizu KT, Tran LM, Mark RJ, Selch MT. Management of oligodendrogliomas. Radiology 1993; 186: 56972. 103 Cairncross JG, Macdonald DR. Successful chemotherapy for recurrent malignant oligodendroglioma. Ann Neurol 1988; 23: 36064. 104 Cairncross JG, Ueki K, Zlatescu MC, et al. Specic genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J Natl Cancer Inst 1998; 90: 1473. 105 Sofetti R, Ruda R, Bradac GB, Schiffer D. PCV chemotherapy for recurrent oligodendrogliomas and oligoastrocytomas. Neurosurgery 1998; 43: 106673. 106 Quinn JA, Reardon DA, Friedman AH, et al. Phase II trial of temozolomide in patients with progressive low-grade glioma. J Clin Oncol 2003; 21: 64651. 107 van den Bent MJ, Taphoorn MJ, Brandes AA, et al. Phase II study of rst-line chemotherapy with temozolomide in recurrent oligodendroglial tumors: the European Organization for Research and Treatment of Cancer Brain Tumor Group Study 26971. J Clin Oncol 2003; 21: 252528. 108 van den Bent MJ, Chinot O, Boogerd W, et al. Second-line chemotherapy with temozolomide in recurrent oligodendroglioma after PCV (procarbazine, lomustine and vincristine) chemotherapy: EORTC Brain Tumor Group phase II study 26972. Ann Oncol 2003; 14: 599602. 109 Hoang-Xuan K, Capelle L, Kujas M, et al. Temozolomide as initial treatment for adults with low-grade oligodendrogliomas or oligoastrocytomas and correlation with chromosome 1p deletions. J Clin Oncol 2004; 22: 313338. 110 Buckner JC, Gesme D Jr, OFallon JR, et al. Phase II trial of procarbazine, lomustine, and vincristine as initial therapy for patients with low-grade oligodendroglioma or oligoastrocytoma: efcacy and associations with chromosomal abnormalities. J Clin Oncol 2003; 21: 25155. 111 Brada M, Viviers L, Abson C, et al. Phase II study of primary temozolomide chemotherapy in patients with WHO grade II gliomas. Ann Oncol 2003; 14: 171521. 112 Pace A, Vidiri A, Galie E, et al. Temozolomide chemotherapy for progressive low-grade glioma: clinical benets and radiological response. Ann Oncol 2003; 14: 172226. 113 Mason WP, Krol GS, DeAngelis LM. Low-grade oligodendroglioma responds to chemotherapy. Neurology 1996; 46: 203. 114 Stege EM, Kros JM, de Bruin HG, et al. Successful treatment of low-grade oligodendroglial tumors with a chemotherapy regimen of procarbazine, lomustine, and vincristine. Cancer 2005; 103: 80209. 115 Chahlavi A, Kanner A, Peereboom D, Staugaitis SM, Elson P, Barnett G. Impact of chromosome 1p status in response of oligodendroglioma to temozolomide: preliminary results. J Neurooncol 2003; 61: 26773. 116 Danson SJ, Middleton MR. Temozolomide: a novel oral alkylating agent. Expert Rev Anticancer Ther 2001; 1: 1319. 117 Tolcher AW, Gerson SL, Denis L, et al. Marked inactivation of O6-alkylguanine-DNA alkyltransferase activity with protracted temozolomide schedules. Br J Cancer 2003; 88: 100411. 118 Kurzen H, Schmitt S, Naher H, Mohler T. Inhibition of angiogenesis by non-toxic doses of temozolomide. Anticancer Drugs 2003; 14: 51522. 119 Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benet from temozolomide in glioblastoma. N Engl J Med 2005; 352: 9971003.

770

http://neurology.thelancet.com Vol 4 November 2005

Das könnte Ihnen auch gefallen