You are on page 1of 6

Treatment of Meningeal Malignancy

STACEY L. BERG, DAVID G. POPLACK


Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas, USA

Key Words. Cancer · Meninges · Chemotherapy · Ommaya · Intrathecal · Brain tumor · Cerebrospinal fluid

A BSTRACT
Meningeal metastasis from cancer has become an increas- requires administration of either very high drug doses or
ingly frequent problem as the treatment of systemic dis- prolonged infusions in order to overcome the poor pene-
ease improves. Leukemia, lymphoma and solid tumors tration of most anticancer agents into the central nervous
may all metastasize to the meninges, where the blood-brain system. Thus, systemic toxicity is often a major drawback
barrier may provide a sanctuary from cytotoxic concen- to this approach. Intrathecal chemotherapy results in
trations of chemotherapeutic agents. Because meningeal delivery of anticancer agents directly into the cerebro-

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


disease may be clinically silent or may present with spinal fluid, usually with minimal systemic toxicity.
unusual signs and symptoms, it is important to maintain a Intrathecal chemotherapy may be administered by lum-
high index of suspicion for this problem. Diagnosis is usu- bar puncture or by use of an Ommaya reservoir with the
ally made by cerebrospinal fluid cytologic testing, mag- tip in the ventricle. Studies are under way to evaluate new
netic resonance imaging, or both. Treatment options agents for both systemic and intrathecal administration.
include radiation therapy, systemic chemotherapy and Further research is required to overcome this difficult
intrathecal chemotherapy. Systemic therapy usually clinical challenge. The Oncologist 1996;1:56-61

INTRODUCTION which flows in the subarachnoid space between the pia and
Meningeal malignancy presents a difficult clinical chal- the arachnoid, may then provide a route for metastasis
lenge for both pediatric and adult oncologists. As improve- along the entire neuraxis. Leukemia, brain tumors and other
ments in systemic therapy have resulted in longer survival for solid tumors may all metastasize to the meninges and
patients with various malignancies, the incidence of meningeal spread in this fashion (Table 1).
metastasis has increased. This is probably due to the “sanctu- Prior to the institution of CNS preventive therapy, meningeal
ary” effect created by the blood-brain barrier, which prevents leukemia occurred in more than 50% of children with acute lym-
many anticancer drugs from achieving cytotoxic concentra- phoblastic leukemia (ALL), and the CNS was the most common
tions in the central nervous system (CNS). In this article, site of relapse in patients who achieved bone marrow remissions.
advances in the treatment of meningeal disease will be pre- The current strategies for preventive therapy have decreased the
sented, including both incidence of CNS leukemia to less than 10%, although relapses
new agents and new still occur and may be difficult to treat. Meningeal leukemia also
Table 1. Some tumors that may metastasize to
the meninges approaches to the use occurs in adults: based on autopsy series, meningeal involvement
of standard agents. may occur in 25% to 81% of cases [1, 2].
▲ Leukemia
Meningeal malig- CNS involvement is common in non-Hodgkin’s lym-
▲ Lymphoma
nancy results from the phomas. Solid tumors also metastasize to the meninges.
▲ Melanoma
metastasis of intracra- In adults, the tumors most commonly responsible are carcino-
▲ Carcinoma (especially breast, lung) nial or extracranial mas of the lung, breast and gastrointestinal tract,
▲ Ewing’s sarcoma tumors to the lep- and melanoma [3]. In children, rhabdomyosarcoma, Ewing’s
▲ Rhabdomyosarcoma tomeninges (the arach- sarcoma and retinoblastoma may metastasize to the CNS. Brain
▲ Retinoblastoma noid membrane and the tumors such as gliomas, medulloblastomas, ependymoblas-
▲ Brain tumors pia mater). The cere- tomas, germinomas and choroid plexus carcinomas, as well
brospinal fluid (CSF), as primary CNS lymphoma, may also spread to the meninges.

Correspondence: Stacey L. Berg, M.D., Texas Children’s Hospital, 6621 Fannin Street, MC3-3320, Houston, TX 77030,
USA. Telephone: 713-770-4588; Fax: 713-770-4202. Accepted for publication October 23, 1995. ©AlphaMed Press 1083-
7159/96/$5.00/0

The Oncologist 1996;1:56-61


Berg, Poplack 57

DIAGNOSIS anticancer therapy. Cranial irradiation may produce the


Meningeal cancer may be clinically silent. Therefore, in “somnolence syndrome,” which consists of a prodrome of
diseases like leukemia in which meningeal spread is com- irritability and anorexia followed by a variable period of
mon, “surveillance” of the CSF with periodic lumbar punc- somnolence. Recovery is spontaneous, and the somnolence
tures is often undertaken. Because of such surveillance, syndrome is not predictive of later complications of therapy.
CNS leukemia is now usually diagnosed before it becomes Long-term complications of cranial or craniospinal
symptomatic. In other cases, the clinical picture of irradiation may include second malignancies, short stature,
meningeal metastasis may be nonspecific. The most com- growth hormone abnormalities and hypothyroidism.
mon signs and symptoms of meningeal malignancy are Cortical atrophy, IQ deficits and leukoencephalopathy may
those of increased intracranial pressure. Headache is most also occur. Younger children are more vulnerable to the
common, although cranial nerve and spinal cord symptoms long-term toxicities of radiation therapy than are older chil-
are also frequent [4]. Unusual signs such as sudden hearing dren [9]. The trend in leukemia therapy is toward reduced
loss or complex partial seizures may also occur. Thus, it is doses (e.g., 1800 cGy) of neuraxis radiation. In other child-
important to keep meningeal malignancy in mind when hood tumors, the minimal dose of radiation adequate for
patients with cancer present with any neurologic complaints. tumor control, as well as strategies for delaying radiation

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


The diagnosis of meningeal metastasis is usually made therapy in younger children, are under active study.
by examination of the CSF. For solid tumors, the presence The chemotherapeutic approach to meningeal cancer
of malignant cells in the CSF confirms the diagnosis. For can be divided into systemic (intravenous or oral) and
leukemias, the combination of malignant blasts and an ele- regional (intrathecal or intraventricular) therapy; both have
vated cell count is usually required. The significance of advantages and disadvantages. Systemic administration
leukemic blasts in the presence of a normal CSF white may produce more uniform drug distribution throughout
blood cell count is uncertain [5-7]. In some cases where the CNS, and prolonged intravenous infusion may produce
meningeal malignancy is suspected, repeated lumbar punc- cytotoxic CSF drug concentrations for a prolonged period.
tures may be required to identify malignant cells. This is Most systemically administered agents, however, do not
especially true for solid tumors. Even so, however, in 90% cross the blood-brain barrier well, and systemic toxicity
of cases the diagnosis can be confirmed by three or fewer limits the ability to treat meningeal disease by this route. In
CSF examinations [8]. The diagnostic role of tumor markers contrast, intrathecal drug administration may produce cyto-
in the CSF has not yet been established. toxic concentrations in the CSF even when small doses are
Radiographic evaluation may sometimes be helpful in the used, and systemic toxicity is very rare. However, lumbar
evaluation of patients with suspected meningeal disease. punctures may be difficult, drugs may not be delivered into
Computerized tomography (CT) scanning is relatively insensi- the intrathecal space and drug distribution in the CSF may
tive, but magnetic resonance imaging (MRI), especially with be uneven. The use of an Ommaya reservoir to deliver
gadolinium enhancement, may be more helpful. Many false drugs directly into the ventricular CSF overcomes some of
negative and some false positive results do occur with MRI, these problems, but use of this device requires special
however. In addition, 111Indium-DTPA flow studies may show expertise as well as a neurosurgical procedure for place-
abnormalities of CSF flow, although these studies are usually ment. In addition, neurotoxicity may occur after direct
used as part of treatment planning prior to intrathecal therapy administration of drugs into the CSF.
rather than as a diagnostic tool. A combination approach In the following section, specific agents used in the
using both CSF studies and radiographic modalities is often treatment of CNS malignancy will be discussed. Tables 2
appropriate in patients with suspected meningeal disease. and 3 present some typical doses and schedules for these
agents, but should not be construed as recommendations for
TREATMENT treatment of individual patients.
Both chemotherapy and radiation therapy may be used in
the prevention or treatment of meningeal malignancy. Cranial METHOTREXATE
irradiation is part of many regimens to prevent the develop- Intrathecal methotrexate has been used in the treatment
ment of meningeal leukemia, especially in high-risk patients, of meningeal leukemia since the 1950s. Intrathecal
and craniospinal irradiation is usually employed as part of the methotrexate either given alone or in combination with
therapy for meningeal relapse. Radiation therapy, however, other agents can prevent the development of meningeal
may result in both short- and long-term toxicity. Craniospinal leukemia in 90% of children. Furthermore, intrathecal
radiotherapy often causes significant myelosuppression, methotrexate often provides effective CNS reinduction
which may complicate efforts to administer systemic therapy for meningeal relapse [10-12].
58 Treatment of Meningeal Malignancy

Although methotrexate is detectable in plasma after an


Table 2. Some systemically administered agents that have been investigated
for the treatment of meningeal malignancy intrathecal administration, systemic toxicity is not usually a
problem after an intrathecal dose. However, acute or delayed
Standard Agents Dose Schedule Reference neurotoxicity is relatively common after intrathecal methotrex-
Methotrexate 33 g/m2 24-h infusion Balis, 1985 ate administration. Chemical arachnoiditis, manifested by
Cytarabine 1-3 g/m 2 q 12 h × 6 Frick, 1984 headache, back pain, meningismus, fever, vomiting and a CSF
Thiotepa 65 mg/m2 single dose Heideman, 1989 pleocytosis, may occur hours to days after methotrexate
administration and is usually self-limited [15]. In severe cases,
Investigational Agents Dose Schedule Reference oral corticosteroids may improve symptoms. On rare occa-
6-mercaptopurine 50 mg/m2/h 36-h infusion Adamson, 1990 sions, transient or permanent weakness or paraplegia may
Topotecan phase I studies occur following intralumbar administration of methotrexate.
under way This unusual toxicity may be related to delayed clearance of
methotrexate from the CSF resulting in high CSF methotrex-
ate levels [16]. Late neurotoxicity in the form
Table 3. Some intrathecally administered agents that have been investigated for the treatment of of leukoencephalopathy may also occur,

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


meningeal malignancy usually in patients who have received intra-
venous methotrexate and cranial irradiation
Standard Agents Dose Schedule Reference
in addition to intrathecal methotrexate [17].
Methotrexate IT <1 year of age: 6 mg single dose Bleyer, 1977 Inadvertent overdoses of intrathecal
1 year of age: 8 mg Bleyer, 1983
2 years of age: 10 mg methotrexate can be fatal. Immediate treat-
≥3 years of age: 12 mg ment including ventriculostomy with ven-
triculo-lumbar perfusion, administration of
IT systemic corticosteroids and administration
2 mg daily × 3 Bleyer, 1978
(“C × T”)
of systemic leucovorin may be beneficial
[18]. In addition, intrathecal administration of
Cytarabine IT 24-70 mg single dose Blaney, 1991
IT carboxypeptidase-G2 has been shown to res-
15 mg daily × 3 Blaney, 1991
(“C × T”) cue nonhuman primates from experimental
intrathecal methotrexate overdose [19], and
Thiotepa IT 10 mg single dose Grossman, 1993 may play a role in the treatment of accidental
intrathecal overdose in humans.
Investigational Agents Dose Schedule Reference In patients with recurrent meningeal
Diaziquone IT 1 mg twice weekly Berg, 1992 disease, intrathecal methotrexate therapy is
IT often given through an indwelling Ommaya
0.5 mg q6h×3 Berg, 1992
(“C × T”) reservoir. Intraventricular administration
through a reservoir results in a more even
6-mercaptopurine IT 10 mg single dose Adamson, 1991
distribution of drug throughout the CSF
Mafosfamide/4HC IT phase I studies and may prolong the duration of remission
under way in CNS leukemia compared to intralumbar
administration [20, 21]. In addition, the use
Topotecan IT phase I studies of the Ommaya reservoir permits the
under way administration of frequent small doses of
methotrexate instead of single large doses.
This “concentration times time” (C × T)
Because the CSF volume approaches adult size years therapy produces cytotoxic concentrations for a prolonged
before body surface area does, the dose for intrathecal period while avoiding high peak drug levels.
methotrexate is based on patient age, with a constant dose Intravenous administration of methotrexate is also used
administered to all patients over three years of age. This in the treatment of meningeal disease. Although the CSF:
approach both reduces toxicity in older patients and plasma ratio for methotrexate is low, cytotoxic methotrexate
improves outcome in younger patients [13, 14]. Because of concentrations can be attained in the CSF using very high
these observations, most other intrathecal agents in children intravenous doses. One such high-dose methotrexate regimen
are also dosed based on age rather than body size. consisting of a 6000 mg/m2 loading dose given over 1 h,
Berg, Poplack 59

followed immediately by an infusion of 1200 mg/m2/h for with meningeal leukemia [29], and 72-h continuous
23 h resulted in cytotoxic steady-state CSF methotrexate intravenous infusions of ≥4 g/m2 also achieved cytotoxic
concentrations and produced complete remissions in 16 of CSF cytarabine concentrations [30]. High-dose systemic
20 children with meningeal relapse of ALL [22]. cytarabine administration is associated with significant
High-dose systemic methotrexate administration toxicity, however. Cerebellar dysfunction requiring dis-
requires both intense hydration and alkalinization of continuation of therapy may be seen in >20% of patients
urine, and leucovorin rescue. Since methotrexate is elimi- following multiple doses of 3 g/m2 every 12 h [31]. Myelo-
nated by the renal tubule, adequate renal function should suppression, nausea, vomiting and mucositis are also
be confirmed prior to therapy, and serum creatinine and common at these doses.
methotrexate concentrations should be monitored during
therapy. If delayed clearance is encountered, intravenous CORTICOSTEROIDS
hydration and leucovorin rescue should be increased. At Prednisone (the orally administered prodrug of pred-
methotrexate concentrations greater than 10-4 mol/l, how- nisolone) and dexamethasone are widely used in the ther-
ever, leucovorin rescue may be ineffective [23]. In the apy of ALL. For both, the CSF concentrations are identical
presence of acute renal failure with severely delayed to the plasma concentrations of free drug. Since at equipo-

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


methotrexate excretion, administration of carboxypepti- tent doses dexamethasone is only 70% protein bound,
dase-G2 may be considered. The use of this enzyme whereas 90% of prednisolone is bound, dexamethasone
results in a greater than 10-fold reduction of serum penetrates much better into the CSF [32]. Patients receiving
methotrexate concentrations within minutes of adminis- dexamethasone rather than prednisone have a significantly
tration [24]. Information about the availability of car- lower rate of CNS relapse [33].
boxypeptidase for emergency use can be obtained from
the National Cancer Institute. THIOTEPA
Even with normal methotrexate clearance and ade- Thiotepa, a lipid-soluble alkylating agent, crosses the
quate hydration and leucovorin rescue, toxicity after high- blood-brain barrier well after systemic administration.
dose methotrexate is frequent. Moderate to severe Furthermore, TEPA, an active metabolite of thiotepa,
mucositis and myelosuppression are common. Hepatic tox- also penetrates very well into the CSF [34, 35]. Thus, sys-
icity with elevated transaminases and bilirubin, as well as temic administration of this agent achieves high concen-
desquamating dermatitis of the hands and feet, can also trations of both parent drug and active metabolite in the
occur. High-dose methotrexate, especially when given in CSF. Systemic administration of thiotepa, however, pro-
association with cranial radiation, has also been associated duces profound bone marrow toxicity, especially throm-
with neurotoxicity [25, 26]. bocytopenia. In contrast, intrathecal administration of
thiotepa is well tolerated [36]. However, intrathecal
CYTARABINE administration of thiotepa may have some disadvantages
Intrathecal cytarabine is used frequently in the treat- compared to the systemic route. The active metabolite
ment of meningeal metastasis. As with methotrexate, high TEPA is not detected in the CSF after intrathecal admin-
CSF cytarabine concentrations with negligible systemic istration. Furthermore, thiotepa diffuses rapidly out of the
toxicity can be achieved by intrathecal cytarabine admin- CSF space [34]. Nonetheless, intrathecal thiotepa may be
istration. Like methotrexate, cytarabine may be given by useful in some settings.
the intralumbar route or through a ventricular reservoir on
a C × T schedule that results in cytotoxic CSF drug con- DIAZIQUONE
centrations for a prolonged period without high peak lev- Diaziquone (AZQ) was specifically developed for its
els [27]. In addition, cytarabine is often combined with property of excellent CSF penetration after systemic
methotrexate for intrathecal administration, especially in administration. Unfortunately, severe myelosuppression
patients with leukemia. limits the utility of this approach. Intrathecal diaziquone,
Intrathecal administration of cytarabine may produce however, is well tolerated at a dose of 1 mg administered
arachnoiditis or, rarely, other forms of neurotoxicity such as twice weekly by either the intralumbar or intraventricular
seizures and paraplegia [28]. Cytarabine therapy, however, route and at a C × T dose of 0.5 mg every 6 h for three
has not been associated with leukoencephalopathy. doses. More than 65% of patients had an objective response
Cytarabine may also be administered systemically for to this therapy, and two of four patients with meningeal
the treatment of meningeal malignancy. A regimen of 3 g/m2 spread of retinoblastoma had complete responses of three
administered every 12 h demonstrated activity in patients months’ duration [37].
60 Treatment of Meningeal Malignancy

6-MERCAPTOPURINE have demonstrated activity in phase I trials against meningeal


6-mercaptopurine is commonly used in low oral doses leukemia [40], medulloblastoma and ependymoma [41].
as an antileukemic agent. Recent studies have shown that a Topotecan, a topoisomerase I inhibitor, achieves a high
48-h intravenous infusion of 6-mercaptopurine at a dose degree of CSF penetration after intravenous administration
rate of 50 mg/m2/h achieves cytotoxic concentrations in the [42]. A wide range of phase II studies of systemically
CSF. The common toxicities include reversible hepatotoxi- administered topotecan in both CNS and non-CNS cancers
city, myelosuppression and mucositis [38]. Intrathecal are now under way. Intrathecally administered topotecan is
administration of 6-mercaptopurine is also being explored. also in the early stages of clinical testing.
In children with refractory meningeal malignancy, a 10 mg The treatment of meningeal disease remains a difficult
intrathecal dose was well tolerated and produced cytotoxic clinical problem. It seems likely that further improvements
concentrations in the CSF for over 12 h. Of nine patients in the therapy of systemic cancer will continue to reveal a
treated at the 10 mg dose, there were four complete and paradoxical increase in the frequency of meningeal metas-
three partial responses [39]. tasis. As this occurs, both the treatment of leptomeningeal
cancer and CNS preventive therapy will require increased
OTHER NEW AGENTS attention from researchers and clinicians alike. Although

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


Studies are under way with intrathecal administration of two therapeutic options are expanding, much remains to be done
preactivated derivatives of cyclophosphamide: mafosfamide to improve current treatment, and ultimately to prevent the
and 4-hydroperoxycyclophosphamide (4HC). These agents development of meningeal malignancy.

R EFERENCES
1 Bojsen-Moller M, Nielsen J. CNS involvement in leukaemia. 11 Whiteside J, Philips F, Dargeon H et al. Intrathecal
Acta Pathol Microbiol Immunol Scand [A] 1983;91:209-216. amethopterin in neurological manifestations of leukemia.
2 Wolk R, Masse S, Conklin R et al. The incidence of central Arch Int Med 1958;101:279-285.
nervous system leukemia in adults with acute leukemia. 12 Sullivan M, Vietti T, Fernbach D et al. Clinical investiga-
Cancer 1974;33:863-869. tions in the treatment of meningeal leukemia: radiation
3 Posner J, Chernik N. Intracranial metastases from systemic therapy regimens vs. conventional intrathecal methotrex-
cancer. Adv Neurol 1978;19:579-592. ate. Blood 1969;34:301-319.

4 Bleyer W. Central nervous system leukemia. Pediatr Clin 13 Bleyer W. Clinical pharmacology of intrathecal methotrexate.
North Am 1988;35:789-814. II. An improved dosage regimen derived from age-related
pharmacokinetics. Cancer Treat Rep 1977;61:1419-1425.
5 Mahmoud H, Rivera G, Hancock M et al. Low leukocyte
counts with blast cells in cerebrospinal fluid of children 14 Bleyer W, Coccia P, Sather H et al. Reduction in the inci-
with newly diagnosed acute lymphoblastic leukemia. N dence of central nervous system leukemia with a pharmaco-
Engl J Med 1993;329:314-319. kinetically derived intrathecal methotrexate dosage regimen.
J Clin Oncol 1983;1:317-325.
6 Mahmoud H, Relling M, Liu Q et al. Presence of leukemic
blasts in cerebrospinal fluid is predictive of central nervous 15 Ochs J. Neurotoxicity due to central nervous system therapy
system relapse in childhood lymphoblastic leukemia. Proc for childhood leukemia. Am J Pediatr Hematol Oncol
Am Soc Clin Oncol 1994;13:1048a. 1989;11:93-105.
7 Tubergen D, Cullen J, Boyett J et al. Blasts in CSF with a 16 Bleyer W, Drake J, Chabner B. Neurotoxicity and elevated
normal cell count do not justify alteration of therapy for cerebrospinal fluid methotrexate concentration in meningeal
acute lymphoblastic leukemia in remission: a Children’s leukemia. N Engl J Med 1973;289:770-773.
Cancer Group study. J Clin Oncol 1994;12:272-278. 17 Price R. Therapy related central nervous system diseases in
8 Wasserstrom W, Glass J, Posner J. Diagosis and treatment children with acute lymphocytic leukemia. In: Mastrangelo
of leptomeningeal metastases from solid tumors: experience R, Poplack D, Riccardi R, eds. Central Nervous System
with 90 patients. Cancer 1982;49:759-772. Leukemia. Boston: Martinus Nijhoff Publishers, 1983:71-81.
9 Mulhern R, Kun L. Neuropsychologic function in children 18 Addegio J, Ridgway D, Bleyer W. The acute management
with brain tumors. III. Interval changes in the six months of intrathecal methotrexate overdose: pharmacologic ration-
following treatment. Med Pediatr Oncol 1985;13:318-324. ale and guidelines. J Pediatr 1981;98:825-828.
10 Pochedly C. Prevention of meningeal leukemia. Review of 19 Adamson PC, Balis FM, McCully CL et al. Rescue of
20 years of research and current recommendations. Hematol experimental intrathecal methotrexate overdose with car-
Oncol Clin North Am 1990;4:951-969. boxypeptidase-G2. J Clin Oncol 1991;9:670-674.
Berg, Poplack 61

20 Bleyer W, Poplack D. Intraventricular versus intralumbar 32 Balis F, Lester C, Chrousos G et al. Differences in cere-
methotrexate for central nervous-system leukemia: pro- brospinal fluid penetration of corticosteroids: possible rela-
longed remission with the Ommaya reservoir. Med Pediatr tionship to the prevention of meningeal leukemia. J Clin
Oncol 1979;6:207-213. Oncol 1987;5:202-207.
21 Bleyer W, Poplack D, Simon R. “Concentration × Time” 33 Jones B, Freeman AI, Shuster JJ et al. Lower incidence of
methotrexate via a subcutaneous reservoir: a less toxic regi- meningeal leukemia when prednisone is replaced by dexam-
men for intraventricular chemotherapy of central nervous ethasone in the treatment of acute lymphocytic leukemia.
system neoplasms. Blood 1978;51:835-842. Med Pediatr Oncol 1991;19:269-275.
22 Balis F, Savitch J, Bleyer W et al. Remission induction of 34 Strong J, Collins J, Lester C et al. Pharmacokinetics of intra-
meningeal leukemia with high-dose intravenous methotrexate. ventricular and intravenous N,N′,N″-triethylenethiophospho-
J Clin Oncol 1985;3:485-489. ramide (thiotepa) in rhesus monkeys and humans. Cancer
Res 1986;46:6101-6104.
23 Pinedo H, Zaharko D, Bull J et al. The reversal of methotrex-
ate cytotoxicity to mouse bone marrow cells by leucovorin 35 Heideman R, Cole D, Balis F et al. Phase I and pharmacoki-
and nucleosides. Cancer Res 1976;36:4418-4424. netic evaluation of thiotepa in the cerebrospinal fluid and
plasma of pediatric patients: evidence for dose-dependent
24 Widemann B, Hetherington M, Smithson W et al. plasma clearance of thiotepa. Cancer Res 1989;49:736-741.

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 17, 2017


Carboxypeptidase-G2 as a rescue agent following methotrex-
ate induced renal failure or intrathecal methotrexate overdose. 36 Grossman SA, Finkelstein DM, Ruckdeschel JC et al.
Proc Am Assoc Cancer Res 1995;36:1384. Randomized prospective comparison of intraventricular
methotrexate and thiotepa in patients with previously
25 Walker R, Allen J, Rosen G et al. Transient cerebral dys- untreated neoplastic meningitis. Eastern Cooperative
function secondary to high-dose methotrexate. J Clin Oncol Oncology Group. J Clin Oncol 1993;11:561-569.
1986;4:1845-1850.
37 Berg SL, Balis FM, Zimm S et al. Phase I/II trial and phar-
26 Bleyer W. Neurologic sequelae of methotrexate and ionizing macokinetics of intrathecal diaziquone in refractory
radiation: a new classification. Cancer Treat Rep 1981;65(suppl meningeal malignancies. J Clin Oncol 1992;10:143-148.
1):89-98.
38 Adamson P, Zimm S, Ragab A et al. A phase II trial of con-
27 Blaney SM, Balis FM, Poplack DG. Current pharmacologi- tinuous-infusion 6-mercaptopurine for childhood solid
cal treatment approaches to central nervous system tumors. Cancer Chemother Pharmacol 1990;26:343-344.
leukaemia. Drugs 1991;41:702-716.
39 Adamson PC, Balis FM, Arndt CA et al. Intrathecal 6-mer-
28 Saiki J, Thompson S, Smith F et al. Paraplegia following captopurine: preclinical pharmacology, phase I/II trial, and
intrathecal chemotherapy. Cancer 1972;29:370-374. pharmacokinetic study. Cancer Res 1991;51:6079-6083.
29 Frick J, Rich P, Hansen R et al. Successful treatment of 40 Blaney S, Balis F, Murphy R et al. A phase I study of intrathe-
meningeal leukemia using systemic high-dose cytosine ara- cal mafosfamide in patients with refractory meningeal
binoside. J Clin Oncol 1984;2:365-368. malignancy. Proc Am Soc Clin Oncol 1992;11:274.
30 Donehower R, Karp J, Burke P. Pharmacology and toxicity 41 Friedman H, Ochs J, Findaly J et al. Phase I trial of intrathecal
of high-dose cytarabine by 72-hour continuous infusion. 4-hydroperoxycyclophosphamide for neoplastic meningitis.
Cancer Treat Rep 1986;70:1059-1065. Proc Am Assoc Cancer Res 1993;34:1598.
31 Nand S, Messmore H, Patel R et al. Neurotoxicity associated 42 Blaney S, Cole D, Balis F et al. Plasma and cerebrospinal fluid
with systemic high-dose cytosine arabinoside. J Clin Oncol pharmacokinetic study of topotecan in nonhuman primates.
1986;4:571-575. Cancer Res 1993;53:725-727.

You might also like