Sie sind auf Seite 1von 6

Int. J. Radiation Oncology Biol. Phys., Vol. 64, No. 3, pp.

892– 897, 2006


Copyright © 2006 Elsevier Inc.
Printed in the USA. All rights reserved
0360-3016/06/$–see front matter

doi:10.1016/j.ijrobp.2005.05.067

CLINICAL INVESTIGATION Brain

INTENSITY-MODULATED RADIOTHERAPY IN HIGH-GRADE GLIOMAS:


CLINICAL AND DOSIMETRIC RESULTS

ASHWATHA NARAYANA, M.D.,* JOSH YAMADA, M.D.,* SEAN BERRY, M.S.,† PRITI SHAH, B.S.,*
MARGIE HUNT, M.S.,† PHILIP H. GUTIN, M.D.,‡§ AND STEVEN A. LEIBEL, M.D.储
Departments of *Radiation Oncology, †Medical Physics, and ‡Surgery, Memorial Sloan-Kettering Cancer Center;
§
Department of Neurological Surgery, Weill Medical School and Cornell Medical Center, New York, NY;
and 储Stanford University Cancer Center, Stanford, CA

Purpose: To report preliminary clinical and dosimetric data from intensity-modulated radiotherapy (IMRT) for
malignant gliomas.
Methods and Materials: Fifty-eight consecutive high-grade gliomas were treated between January 2001 and
December 2003 with dynamic multileaf collimator IMRT, planned with the inverse approach. A dose of 59.4 – 60
Gy at 1.8 –2.0 Gy per fraction was delivered. A total of three to five noncoplanar beams were used to cover at least
95% of the target volume with the prescription isodose line. Glioblastoma accounted for 70% of the cases, and
anaplastic oligodendroglioma histology (pure or mixed) was seen in 15% of the cases. Surgery consisted of biopsy
only in 26% of the patients, and 80% received adjuvant chemotherapy.
Results: With a median follow-up of 24 months, 85% of the patients have relapsed. The median progression-free
survival time for anaplastic astrocytoma and glioblastoma histology was 5.6 and 2.5 months, respectively. The
overall survival time for anaplastic glioma and glioblastoma was 36 and 9 months, respectively. Ninety-six
percent of the recurrences were local. No Grade IV/V late neurologic toxicities were noted. A comparative
dosimetric analysis revealed that regardless of tumor location, IMRT did not significantly improve target
coverage compared with three-dimensional planning. However, IMRT resulted in a decreased maximum dose to
the spinal cord, optic nerves, and eye by 16%, 7%, and 15%, respectively, owing to its improved dose
conformality. The mean brainstem dose also decreased by 7%. Intensity-modulated radiotherapy delivered with
a limited number of beams did not result in an increased dose to the normal brain.
Conclusions: It is unlikely that IMRT will improve local control in high-grade gliomas without further dose
escalation compared with conventional radiotherapy. However, it might result in decreased late toxicities
associated with radiotherapy. © 2006 Elsevier Inc.

Intensity-modulated radiotherapy, Glioma, Radiotherapy.

INTRODUCTION The goal of treatment planning is to spare the critical


structures while delivering the complete prescription dose to
The combination of surgery, radiotherapy, and chemotherapy
the target volume. The tumor often lies in close proximity or
represents the standard approach to the treatment of malignant
surrounds several radiosensitive normal tissues, including
gliomas. However, survival remains poor. The median survival
the eyes, optic nerves, chiasm, brainstem, and spinal cord.
time with conventional therapy for patients with glioblastoma
The tolerance of these tissues is less than the desired pre-
multiforme is 10 –12 months, with a 3-year survival rate of
scription dose, and the associated morbidity can be severe.
6 – 8% (1). The median survival time for patients with anaplas-
Any effort to decrease the treatment toxicity might result in
tic astrocytoma is 36 months, and the 3-year survival rate is
compromised target coverage with conventional therapy,
approximately 50%. The pattern of recurrence in high-grade
gliomas is almost always local (2). The inability to deliver thus increasing the risk of local recurrence.
adequate radiation to the target volume often has been cited as Intensity-modulated radiotherapy (IMRT) is a more ad-
the cause for local failure (3). vanced form of three-dimensional conformal radiotherapy
Radiation treatment planning for gliomas can be challenging. (3D-CRT) that relies on advanced accelerator and multileaf

Reprint requests to: Ashwatha Narayana, M.D., Department of Presented at the 40th Annual Meeting of the American Society
Radiation Oncology, Memorial Sloan-Kettering Cancer Center, of Clinical Oncology, New Orleans, Louisiana, June 5– 8, 2004.
Room SM16, 1275 York Avenue, New York, NY 10021. Tel: (212) Received Jan 13, 2005, and in revised form April 15, 2005.
639-6773; Fax: (212) 639-2417; E-mail: narayana@mskcc.org Accepted for publication May 6, 2005.

892
IMRT in high-grade gliomas ● A. NARAYANA et al. 893

collimator technology to deliver nonuniform beam intensi- immobilized with an Aquaplast face mask (WFR/Aquaplast Cor-
ties. This approach has been demonstrated to improve tumor poration, Wyckoff, NJ). Patients then underwent CT simulation
coverage while decreasing the dose to critical structures in with intravenous contrast, and 3-mm slice images were obtained
the head and neck and other sites (4, 5). At present there is from the vertex to approximately the level of C2. Postresection
magnetic resonance images with 3-mm axial fluid attenuation
very little clinical or dosimetric data regarding the dose
inversion recovery (FLAIR) sequence images obtained within 1
distribution and outcomes from IMRT in the treatment of week before simulation were registered with the CT images, with
malignant gliomas (6). This retrospective analysis presents the image registration software available on the CT simulator
the preliminary data on the potential benefits of IMRT in (AcQSim, Philips Medical Systems, Bothell, WA). The clinical
high-grade gliomas. target volume was defined as the gross disease observed in the
FLAIR sequence MRI series plus a 1.5-cm margin (7). The plan-
ning target volume (PTV) was determined by adding an additional
METHODS AND MATERIALS 0.5-cm margin to the clinical target volume to account for treat-
Fifty-eight consecutive patients with high-grade gliomas received ment uncertainties. The normal tissues delineated included the
radiotherapy with IMRT between January 2001 and December 2003 spinal cord, brainstem, optical structures, and “normal brain,”
at Memorial Sloan-Kettering Cancer Center (MSKCC). Institutional which were defined as all brain tissue outside of the PTV. The dose
review board approval was obtained for retrospective review of the limits that defined an acceptable plan included a maximum point
data. Patient characteristics are shown in Table 1. Glioblastoma dose of 4500 cGy to the spinal cord, 5400 cGy to the optic nerves
accounted for 70% of the cases. Anaplastic astrocytoma and ana- and the chiasm, 4000 cGy to the retina, and 5500 cGy to the
plastic oligodendroglioma histology (pure or mixed) constituted brainstem. An attempt was made to keep the lens dose as low as
15% each of the cases. Surgery was limited to biopsy alone in 26% possible. Acceptable target coverage was defined by a D95 (dose
of patients. Eighty percent of patients received adjuvant chemo- received by 95% of the PTV) of at least 95% of the prescription.
therapy. In all but 2 cases, radiotherapy was started within 1 month As in 3D-CRT, multiple noncoplanar beams were commonly used.
of surgical resection. An attempt was made to limit the number of treatment fields to
The technique of IMRT treatment planning for brain tumors has three to five, in consideration of both treatment complexity and the
been previously described by us (7). Simulation was performed volume of normal brain tissue irradiated to low dose levels. All
generally 2 to 3 weeks after surgical resection. All patients were planning was performed with the MSKCC in-house-developed
treatment-planning system, which relies on the inverse planning
method developed at our institution by Spirou and Chui (8). This
algorithm relies on a gradient minimization technique and a qua-
Table 1. Patient characteristics
dratic objective function. It supports the application of dose con-
Gender (n) straints for targets and both dose and dose–volume constraints for
Male 31 normal tissues. Treatment was delivered with dynamic multileaf
Female 27 collimation. The intensity profiles generated during optimization
Age (y) were converted into leaf motion with the method described by
Median 54 Spirou and Chui (8). All the patients were treated with 6-MV
Range 24–80 X-rays from Varian accelerators (Varian Medical Systems, Palo
Location (n) Alto, CA) equipped with multileaf collimators with either a 1-cm
Temporal 22 or 0.5-cm leaf width. The prescription dose for all patients was
Frontal 20
either 59.4 or 60 Gy, given in 1.8-Gy or 2.0-Gy fractions. The
Parietal 11
Occipital 2 average time from simulation to the start of radiotherapy was 7
Posterior fossa 3 days (range, 5–10 days).
Histology (n) A comparative dosimetric analysis was performed for 20 pa-
Glioblastoma 41 tients treated with IMRT. Conventional 3D-CRT plans were gen-
Anaplastic astrocytoma 9 erated retrospectively with the identical beam arrangement used
Anaplastic 4 for the IMRT, wedges, and static multileaf collimator apertures.
Oligodendroglioma Doses to the critical structures, PTV, and normal brain tissue were
Anaplastic mixed glioma 4 evaluated for both plans through the evaluation of dose–volume
Karnofsky performance status histograms and dose distributions. In addition to maximum normal
Median 80
tissue doses and the PTV D95, the maximum and mean PTV doses
Range 60–90
Radiation dose (Gy) and V18 Gy and V24 Gy (percentage of volume receiving at least
Median 59.4 18 or 24 Gy) for the normal brain were recorded. V18 Gy and V24
Range 59.4–60 Gy were chosen on the basis of neurotoxicity data available in
Surgical resection (n) pediatric populations receiving whole-brain radiotherapy.
Total/near total 27 The baseline evaluations included a complete medical history,
Partial 16 physical examination, determination of performance status, hema-
Biopsy 15 tology and clinical chemistry assessments, and gadolinium-en-
Chemotherapy (n) hanced MRI of the brain. Gadolinium-enhanced MRI was per-
Adjuvant/concomitant 46 formed 2– 4 weeks after the completion of radiotherapy and then
None 12
every 3 months during the first year and every 3 to 4 months
894 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 3, 2006

Table 2. Progression-free survival

Grade Total 1-y DFS (%) 2-y DFS (%)

GBM (Gr IV) 41 0.0 0.0


All Gr III 17 21.8 10.8
AA 9 27.7 0.0
AO/mixed 8 18.8 18.8

Abbreviations: DFS ⫽ disease-free survival; AA ⫽ anaplastic


astrocytoma; AO ⫽ anaplastic oligodendroglioma; GBM ⫽
glioblastoma.

subsequently. Toxicity was graded according to the Radiation


Therapy Oncology Group grading system and the National Cancer
Institute Common Toxicity Criteria (version 3.0). Overall survival Fig. 1. Progression-free survival in patients with high-grade gli-
omas.
was calculated from the time of diagnosis until death or last
follow-up, according to the Kaplan-Meier method.
changes. Four Grade III late toxicities related to speech or
RESULTS cognitive problems were seen. There were no Grade IV/V
late neurotoxicities. The rate of freedom from late neuro-
With a median follow-up of 24 months (range, 12– 48 toxicity at 24 months was 85% (Fig 3).
months), 49 of the 58 patients (85%) have relapsed. Forty-
seven (96%) of the recurrences were local. Of the remaining Dosimetry
2 patients, 1 recurred in a different lobe of the brain outside A comparison of the IMRT treatment plans with the more
the radiated region, whereas the other had a leptomeningeal conventional 3D-CRT plans was performed in 20 patients
relapse. representative of the overall patient population in terms of
tumor location, size, grade, volume, and the prescription
Local control dose. It showed that regardless of tumor location, IMRT
Overall survival (OS) and progression-free survival does not lead to clinically significant differences in PTV
(PFS) data are shown in Tables 2 and 3 and Figures 1 and coverage. Of all the PTV parameters evaluated, a statisti-
2, respectively. The median PFS for Grade III and IV cally significant difference was observed only for PTV
tumors was 5.6 and 2.5 months, respectively. The median mean dose, with the mean dose from 3D-CRT being, on
OS for Grade III and IV tumors was 36 and 9 months, average, approximately 1% higher than that with IMRT. No
respectively. The subset of patients with anaplastic oligo- differences were observed in PTV maximum dose, mean
dendroglioma histology (pure or mixed) had a similar PFS dose, or D95 coverage (Table 4, Figs. 4 and 5). However,
but a better OS compared with patients with anaplastic the use of IMRT resulted in a significantly lower mean
astrocytomas. brainstem dose (7%) and lower maximum doses to the
spinal cord, optic nerve, and eye by 16%, 7%, and 15%,
Toxicity respectively (Table 4, Fig 6). The use of IMRT did not
Twenty-one patients experienced acute Grade I/II neuro- result in an increase of the total dose received by the normal
toxicities. Four Grade III acute neurotoxicities were seen,
including seizures, altered consciousness, and worsening
confusion. Two Grade IV neurotoxicities occurred because
of brainstem bleed and intractable seizures. Six late Grade
I/II neurotoxicities were noted, all related to white matter

Table 3. Overall survival

Grade Total 1-y OS (%) 2-y OS (%)

GBM (Gr IV) 41 30.0 0.0


All Gr III 17 86.3 61.6
AA 9 88.8 29.6
AO/mixed 8 87.5 87.5

Abbreviation: OS ⫽ overall survival. Other abbreviations as in


Table 2. Fig. 2. Overall survival in patients with high-grade gliomas.
IMRT in high-grade gliomas ● A. NARAYANA et al. 895

Fig. 4. Comparison of intensity-modulated radiotherapy (IMRT)


and three-dimensional (3D) dosimetry in a patient with frontal
Fig. 3. Delayed Grade III neurologic event-free survival in high-
glioma.
grade gliomas.

thermore, IMRT has been shown to improve the dose dis-


brain (Fig 6). On the contrary, the volume of normal brain tribution to the target volume, as well as to reduce the dose
irradiated was significantly less with IMRT. The volume to normal tissues when compared with 3D-CRT techniques
receiving at least 18 Gy and 24 Gy decreased by 7% and (4). The outcomes of clinical studies of IMRT for dose
8%, respectively. escalation and critical organ sparing in prostate and head-
and-neck cancer sites are particularly encouraging (12, 13).
Our hypothesis was that a similar benefit might be achieved
DISCUSSION
in patients with high-grade gliomas.
There has been a dramatic improvement in radiotherapy Our results indicated that neither PFS nor OS improved
techniques over the last 2 decades. Improvements in dose with the use of IMRT in high-grade gliomas. Why did the
distribution and local control have been observed with 3D- improvement in local control noted at other sites not trans-
CRT as compared with conventional two-dimensional treat- late to the brain? The reasons are several. Our PTV was
ment planning for prostate cancer and subsequently for defined as a 1.5–2-cm margin around the FLAIR sequence–
other tumor locations (9, 10). It has also been shown that the defined tumor, which is the conventional volume definition
morbidity of therapy decreased with the use of 3D-CRT (7). Several studies have shown that gliomas can extend
compared with conventional treatment planning (11). Fur- more than 2 cm from the contrast-enhancing lesion (14). In

Table 4. Dosimetric data comparing 3D and IMRT plans

Structure 3D IMRT IMRT/3D p

PTV
Max dose 108.9 ⫾ 2.5 109.1 ⫾ 4.4 1.002 0.86
Min dose 67.3 ⫾ 24.1 65.8 ⫾ 23.2 0.98 0.33
Mean dose 103.6 ⫾ 2.1 102.3 ⫾ 1.7 0.99 0.04
Brainstem max dose 94.1 ⫾ 17.2 92.3 ⫾ 18.6 0.98 0.19
Brainstem mean dose 60.9 ⫾ 25.2 56.4 ⫾ 23.8 0.93 0.001
Spinal cord max dose 29.1 ⫾ 16.2 24.4 ⫾ 13.2 0.84 0.01
Optical structures
Chiasm max dose 81.4 ⫾ 24.8 78.9 ⫾ 24.0 0.97 0.10
Optic nerve max dose 53.9 ⫾ 37.5 49.9 ⫾ 36.5 0.93 0.02
Eye max dose 28.4 ⫾ 31.8 24.0 ⫾ 29.1 0.85 0.04
Lens mean dose 2.8 ⫾ 2.9 3.3 ⫾ 4.1 1.18 0.04
Normal brain*
Mean dose 41.9 ⫾ 12.2 38.9 ⫾ 10.6 0.93 0.0004
% volume receiving ⱖ18 Gy 56.2 ⫾ 18.0 52.0 ⫾ 17.4 0.93 0.01
% volume receiving ⱖ24 Gy 43.3 ⫾ 17.5 39.7 ⫾ 16.8 0.92 0.01

Abbreviations: 3D ⫽ three-dimensional conformal radiotherapy; IMRT ⫽ intensity-modulated


radiotherapy; PTV ⫽ planning target volume; Max ⫽ maximum; Min ⫽ minimum.
Values are percentages of total prescription dose, unless otherwise noted.
* “Normal brain” includes all the brain tissue, excluding the PTV.
896 I. J. Radiation Oncology ● Biology ● Physics Volume 64, Number 3, 2006

compared with conventional 3D-CRT planning. For many


gliomas, target coverage and dose uniformity are excellent
with standard 3D techniques, owing to the nearly spherical
or cylindrical shape of the lesion. Therefore, it was not
surprising that significant further improvement was not ob-
served with IMRT. Target coverage and uniformity im-
provements with IMRT have been primarily reported in
sites such as the head and neck or prostate (4, 12, 13), where
the target is concave, surrounding normal tissues with dose
limits much less than that of the tumor. Gliomas can be
highly irregular but typically exhibit few concavities. When
Fig. 5. Dose–volume histogram of planning target volume (PTV) concavities do exist, such as when the tumor surrounds the
with three-dimensional (3D) and intensity-modulated radiotherapy chiasm, the required dose gradient between tumor and nor-
(IMRT) treatment. mal tissue is often less than that observed in other sites. As
a result, very good target coverage is often achieved with
addition, the radiation dose used in this study was only 3D planning. However, as we escalate the prescription dose
59.4 – 60 Gy, which, although certainly adequate by con- for these tumors, even if only to areas of suspected high
ventional standards, might be insufficient for long-term tumor density, the benefit of IMRT might increase because
control in a relatively radioresistant tumor like glioma. As in steeper dose gradients and more concave dose distributions
conventional techniques, IMRT was used in this study to will be necessary. Indeed, our group and others have already
deliver a uniform dose throughout the target volume. It is shown the clear benefit of IMRT in concave brain lesions
clear that the density of tumor throughout the volume could (15, 16).
vary, and failure to give a higher dose to areas with highest The use of IMRT did result in a moderate decrease of
tumor density might have resulted in tumor recurrence. In dose to the critical structures in the brain. This might be
addition, the use of conventional fractionation of 1.8 –2.0 responsible for the decreased acute and late neurotoxicity
Gy per fraction might not be the optimal fractionation we observed compared with historical results. Contrary to
regimen for these aggressive tumors. the conventional thinking (17), the use of IMRT with three
Our dosimetric analysis confirmed that IMRT did not to five beams did not result in increased dose to the normal
lead to significant improvements in target coverage when brain. In fact, the volume of the brain receiving 18 and 24

Fig. 6. Dose–volume histogram of critical structures and normal brain tissue with three-dimensional (3D) and
intensity-modulated radiotherapy (IMRT) treatment volume (%).
IMRT in high-grade gliomas ● A. NARAYANA et al. 897

Gy decreased with the use of IMRT, owing to optimal beam result in decreased acute and late toxicities associated
modulation. This has obvious implications for reducing with radiotherapy. There is a need to better define the
neurocognitive deficits in long-term survivors after radio- target volume and then to perform selective dose inten-
therapy. Decreasing the toxicity would also enable possible sification according to the tumor density to optimize the
dose escalation, although the potential results of this ap- radiation technique. A prospective phase I/II dose-esca-
proach are uncertain (18). lation clinical trial with the use of multivoxel magnetic
In summary, it is unlikely that the use of IMRT with resonance spectroscopy to define the target volume and
conventional doses and volume definition will improve then use IMRT dose painting on the basis of tumor cell
local control in high-grade gliomas. However, it can density is planned.

REFERENCES
1. Medical Research Council Brain Tumor Working Party. Ran- distributions for 3D conformal boost treatments in carcinoma
domized trial of procarbazine, lomustine, and vincristine in the of the nasopharynx. Int J Radiat Oncol Biol Phys 1991;20:
adjuvant treatment of high grade astrocytoma: A medical 823– 833.
research council trial. J Clin Oncol 2001;19:509 –518. 11. Skwarchuk MW, Jackson A, Zelefsky MJ, et al. Late rectal
2. Wallner KE. Radiation therapy treatment planning for malig- toxicity after conformal radiotherapy of prostate cancer (I):
nant astrocytomas. Semin Radiat Oncol 1991;1:17–25. Multivariate analysis and dose-response. Int J Radiat Oncol
3. Hochberg FH, Pruitt A. Assumptions in the radiotherapy of Biol Phys 2000;47:103–113.
glioblastomas. 9 Neurology 1980;30:907–911. 12. Zelefsky MJ, Fuks Z, Hunt M, et al. High-dose intensity
4. Hunt MA, Zelefsky MJ, Wolden S, et al. Treatment planning modulated radiation therapy for prostate cancer: Early toxicity
and delivery of intensity-modulated radiation therapy for pri- and biochemical outcome in 772 patients. Int J Radiat Oncol
mary nasopharynx cancer. Int J Radiat Oncol Biol Phys 2001; Biol Phys 2002;53:1111–1116.
49:623– 632. 13. Lee N, Xia P, Fischbein NJ, et al. Intensity-modulated radia-
5. Huang E, Teh BS, Strother DR, et al. Intensity-modulated tion therapy for head-and-neck cancer: The UCSF experience
radiation therapy for pediatric medulloblastoma: Early report focusing on target volume delineation. Int J Radiat Oncol Biol
on the reduction of ototoxicity. Int J Radiat Oncol Biol Phys Phys 2003;57:49 – 60.
2002;52:599 – 605. 14. Chamberlain MC, Murovic JA, Levin VA. Absence of con-
6. Shaw EG, Stieber V, Tatter S, et al. A phase I dose escalating trast enhancement on CT brain scans of patients with supra-
study of intensity modulated radiation therapy (IMRT) for the tentorial malignant gliomas. Neurology. 1988;38:1371–1374.
treatment of glioblastoma multiforme (GBM) [abstract]. Int J 15. Narayana A, Yenice KM, Chang J, et al. Intensity-modulated
Radiat Oncol Biol Phys 2002;54:206. stereotactic radiotherapy for the treatment of skull base
7. Narayana A, Leibel SA. Primary and metastatic brain tumors meningioma—a novel approach. Skull Base 2004;14:18–19.
in adults. In: Leibel S, Philips TL, editors. Textbook of radi- 16. Baumert BH, Norton IA, Davis JB. Intensity-modulated ste-
ation oncology. 2nd ed. Philadelphia: Saunders; 2004. 471– reotactic radiotherapy vs. stereotactic conformal radiotherapy
495. for the treatment of meningioma located predominantly in the
8. Spirou SV, Chui CS. A gradient inverse planning algorithm skull base. Int J Radiat Oncol Biol Phys 2003;57:580 –592.
with dose-volume constraints. Med Phys 1998;25:321–333. 17. Glatstein E. Intensity-modulated radiation therapy: The in-
9. Zelefsky MJ, Marion C, Fuks Z, et al. Improved biochemical verse, the converse, and the perverse. Semin Radiat Oncol
disease-free survival of men younger than 60 years with 2002;12:272–281.
prostate cancer treated with high dose conformal external 18. Chan JL, Lee SW, Fraass BA, et al. Survival and failure
beam radiotherapy. J Urol 2003;170:1828 –1832. pattern of high-grade gliomas after three-dimensional confor-
10. Leibel SA, Kutcher GJ, Harrison LB, et al. Improved dose mal radiotherapy. J Clin Oncol 2002;20:1635–1642.

Das könnte Ihnen auch gefallen