Table of Contents Table of Contents
Previous Page  1522 / 1708 Next Page
Information
Show Menu
Previous Page 1522 / 1708 Next Page
Page Background

the recurrence was local. The nature and timing of the local

failure may provide helpful clues about the risk of neuraxis

dissemination and move caregivers to recommend focal or

craniospinal treatment. The specter of metastatic disease

may be decreased in a patient who experiences disease pro-

gression where residual tumor was known to remain after ini-

tial surgery or in a patient who develops obvious local failure

relatively late, longer than 3 years after RT. Clinical condi-

tion and age of a patient also require consideration; very

young or debilitated patients may not fare well with CSI.

We do not propose a lower age cutoff for CSI, but consider

it to be an option in children older than 3 years because pa-

tients of a similar age with medulloblastoma continue to un-

dergo high-dose CSI as a front-line treatment option.

The role of radiosurgery is difficult to define from our

series because of the small number of patients and prepon-

derance of treatment sites that involve the brainstem. Al-

though it might be considered a better option for patients

with supratentorial local or metastatic disease, these patients

tend to have operable disease that may be removed and sim-

plify follow-up, which, after radiosurgery, is often com-

plicated by changes in the treated volume and normal

tissues. Even with radiosurgery, some normal brain is irra-

diated. High-dose single-fraction treatment can be harmful,

especially when such a critical structure as the brainstem is

involved.

Reirradiation for recurrent primary brain tumors has been

a long-standing treatment option, with investigators cogni-

zant of the attendant risks of necrosis or neurologic compli-

cation

(9)

. One published series reported a 9% risk of

necrosis and overall complication rate of 29% in 34 patients

with primary brain tumors, including children, undergoing

fractionated reirradiation to a median combined dose of

79.7 Gy (range, 43.2–111 Gy)

(1) .

This series showed only

a modest palliative and survival benefit in a diverse group

of patients. A more specific evaluation of combined reirradia-

tion and lomustine therapy was conducted in a small cohort

of patients with high-grade glioma, showing a median overall

survival of 13.7 months. The reirradiation dose was limited to

34.5 Gy in 23 fractions, with a median interval between first

and second courses of irradiation of 14 months

(10) .

With the

advent of conformal RT, investigators attempted to minimize

the dose to normal tissues when reirradiation was attempted.

One series included 20 patients with primary brain tumors

unsuitable for brachytherapy or radiosurgery, predominantly

high-grade glioma. With a median reirradiation dose of 36

Gy (range, 30.6–59.4 Gy) and combined dose range of

80.6–119.4 Gy, neurologic improvement and stabilization

of disease was observed in more than 67% of patients

(11) .

Different dose and fractionated schemes were attempted for

similar patients. For example, low-dose (36 Gy) fractionated

reirradiation was applied successfully to predominantly adult

patients with low- and high-grade astrocytoma. The lack of

observed toxicity might be attributable to the long interval

between courses (median, 50 months) for patients with low

grade and relatively short time to progression for patients

with high grade

(12, 13) .

Similar low hypofractionated doses

were applied in patients with high-grade glioma

(14)

and EP

(15)

with modest results. The FFRT and radiosurgery for

medulloblastoma appears to be safe, provided doses are rela-

tively low, and locally effective. However, overall results are

poor in a tumor system prone to metastatic failure, not unlike

EP

(2) .

The patients in this report continue to be followed up for

treatment-related side effects involving neurologic, endo-

crine, and cognitive function. None was lost to follow-up.

Of the 23 patients for whom salvage therapy did not fail, 4

have notable disabilities, including the 2 patients alive and

without disease progression after necrosis (1 radiosurgery pa-

tient and 1 patient treated with CSI), 1 patient who was func-

tionally disabled by surgery before reirradiation, and the

patient who is the longest survivor in our series (>20 years)

who lives with parents and is simply employed. The rest of

the children continue to be followed up, and the magnitude

of side effects has been greatest in children treated with

CSI. Given the very small volume targeted for FFRT, barring

structure damage to the brainstem, the risks of endocrinop-

athy, ototoxicity, and cognitive decline for these patients

do not appear to be significantly greater than those observed

after their initial treatment course.

In summary, reirradiation with curative intent should be

considered for patients with recurrent EP after previous adju-

vant focal irradiation. Aggressive attempts to resect local and

metastatic disease are favored in this approach. Patients

treated in this manner require careful surveillance for side

effects of this combined salvage treatment approach.

REFERENCES

1. BaumanGS, Sneed PK,WaraWM,

et al

. Reirradiation of primary

CNS tumors.

Int J Radiat Oncol Biol Phys

1996;36:433–441.

2. Milker-Zabel S, Zabel A, Thilmann C,

et al

. Results of three-

dimensional stereotactically-guided radiotherapy in recurrent

medulloblastoma.

J Neurooncol

2002;60:227–233.

3. Weprin BE, Hall WA, Cho KH,

et al

. Stereotactic radiosurgery

in pediatric patients.

Pediatr Neurol

1996;15:193–199.

4. Lo SS, Chang EL, Sloan AE. Role of stereotactic radiosurgery

and fractionated stereotactic radiotherapy in the management

of intracranial ependymoma.

Expert Rev Neurother

2006;6:

501–507.

5. Merchant TE, Mulhern RK, Krasin MJ,

et al

. Preliminary results

from a phase II trial of conformal radiation therapy and evalua-

tion of radiation-related CNS effects for pediatric patients with

localized ependymoma.

J Clin Oncol

2004;22:3156–3162.

6. Nieder C, Milas L, Ang KK. Tissue tolerance to reirradiation.

Semin Radiat Oncol

2000;10:200–209.

7. Sminia P, van der Kleij AJ, Carl UM,

et al

. Prophylactic hyper-

baric oxygen treatment and rat spinal cord re-irradiation.

Cancer

Lett

2003;191:59–65.

8. Nieder C, GrosuAL, Andratschke NH,

et al

. Update of human spi-

nal cord reirradiation tolerance based on additional data from 38

patients.

Int J Radiat Oncol Biol Phys

2006;66:1446–1449.

9. Dritschilo A, Bruckman JE, Cassady JR,

et al

. Tolerance of

brain to multiple courses of radiation therapy. I. Clinical expe-

riences.

Br J Radiol

1981;54:782–786.

96

I. J. Radiation Oncology

d

Biology

d

Physics

Volume 71, Number 1, 2008