15
Critical Reviews in Oncology/Hematology, 1993; 14:61-15 0 1993 Elsevier Scientific Publishers Ireland Ltd. All rights reserved. 1040-8428/93/$24.00 61 ONCHEM 00042 Neurotoxicity of antineoplastic agents Maha Hussain, Antoinette J. Wozniak and Mark B. Edelstein Department of Medicine, Division of Hematology and Oncology, Wayne State University School of Medicine, Detroit, MI 48201 and the VAMC Allen Park, MI 48101, USA (Accepted 21 September 1992) Contents I. Introduction .................................................... II. Vinca alkaloids ................................................ A. Pathophysiology and electrophysiology ......................... B. Variables affecting vincristine neurotoxicities .................... C. Treatment of neurotoxicity ................................... III. Cisplatin and platinum analogues ................................ A. Electrophysiology and nerve biopsy ............................ B. Variables effecting cisplatin neurotoxicity ....................... IV. Other neuropathic chemotherapeutic agents ....................... V. Biologic response modifers (BRM) ............................... A. Interferon .................................................. B. Interleukin-2 ................................................ VI. Models for evaluating neurotoxicity of anti-cnacer agents ........... VII. Conclusion .................................................... Summary ............................................................... References ............................................................. 61 62 63 63 64 65 66 66 67 69 69 69 70 70 71 71 I. Introduction Neurotoxicity has been associated with many anti- cancer agents. The vinca alkaloids, in particular vincris- tine and cisplatin have been the drugs most extensively described. Interestingly, neurotoxicity has been a prominent side effect of several of the newest anti-cancer agents that have recently been introduced into clinical practice. Ifosfamide, hexamethylmelamine, and interferon, as well as several of the newer agents screened by the Na- tional Cancer Institute with solid tumor selectivity, have all proven to be neurotoxic and at times this toxicity is Correspondence to: Maha Hussain, M.D., Section of Hematology and Oncology, VAMC, Allen Park, MI 48101, USA. dose limiting. Table 1 provides a comprehensive list of anti-neoplastic agents under the headings of their more common neurotoxicities. Since the previous reviews of this subject [l-4], several analogues of older drugs have been investigated, biologic response modifiers have been more widely in- troduced into clinical practice, high-dose chemotherapy with bone marrow rescue is being more commonly utilized and grades of neurotoxicity have been better defined. This review will focus on the clinical, pathological and laboratory aspects of vinca alkaloid and cisplatin neurotoxicity in order to help in the understanding of the various etiologies of chemotherapy related neurotoxicity and to establish some important guidelines in directing the management of the neurotox- icities that complicate anti-cancer drug therapy in

Neurotoxicity of antineoplastic agents

Embed Size (px)

Citation preview

Page 1: Neurotoxicity of antineoplastic agents

Critical Reviews in Oncology/Hematology, 1993; 14:61-15 0 1993 Elsevier Scientific Publishers Ireland Ltd. All rights reserved. 1040-8428/93/$24.00

61

ONCHEM 00042

Neurotoxicity of antineoplastic agents

Maha Hussain, Antoinette J. Wozniak and Mark B. Edelstein Department of Medicine, Division of Hematology and Oncology, Wayne State University School of Medicine, Detroit, MI 48201 and the VAMC Allen

Park, MI 48101, USA

(Accepted 21 September 1992)

Contents

I. Introduction ....................................................

II. Vinca alkaloids ................................................ A. Pathophysiology and electrophysiology ......................... B. Variables affecting vincristine neurotoxicities .................... C. Treatment of neurotoxicity ...................................

III. Cisplatin and platinum analogues ................................ A. Electrophysiology and nerve biopsy ............................ B. Variables effecting cisplatin neurotoxicity .......................

IV. Other neuropathic chemotherapeutic agents .......................

V. Biologic response modifers (BRM) ............................... A. Interferon .................................................. B. Interleukin-2 ................................................

VI. Models for evaluating neurotoxicity of anti-cnacer agents ...........

VII. Conclusion ....................................................

Summary ...............................................................

References .............................................................

61

62 63 63 64

65 66 66

67

69 69 69

70

70

71

71

I. Introduction

Neurotoxicity has been associated with many anti- cancer agents. The vinca alkaloids, in particular vincris- tine and cisplatin have been the drugs most extensively described.

Interestingly, neurotoxicity has been a prominent side effect of several of the newest anti-cancer agents that have recently been introduced into clinical practice. Ifosfamide, hexamethylmelamine, and interferon, as well as several of the newer agents screened by the Na- tional Cancer Institute with solid tumor selectivity, have all proven to be neurotoxic and at times this toxicity is

Correspondence to: Maha Hussain, M.D., Section of Hematology and Oncology, VAMC, Allen Park, MI 48101, USA.

dose limiting. Table 1 provides a comprehensive list of anti-neoplastic agents under the headings of their more

common neurotoxicities. Since the previous reviews of this subject [l-4],

several analogues of older drugs have been investigated, biologic response modifiers have been more widely in- troduced into clinical practice, high-dose chemotherapy with bone marrow rescue is being more commonly utilized and grades of neurotoxicity have been better defined. This review will focus on the clinical, pathological and laboratory aspects of vinca alkaloid and cisplatin neurotoxicity in order to help in the understanding of the various etiologies of chemotherapy related neurotoxicity and to establish some important guidelines in directing the management of the neurotox- icities that complicate anti-cancer drug therapy in

Page 2: Neurotoxicity of antineoplastic agents

62

TABLE 1 TABLE 2

Neurologic syndromes caused by systemic administration of antineo- plastic agents

Neurotoxicities associated with vincristine

A. Acuie encephalopathy High dose methotrexate

(‘stroke like’) Asparaginases 5-FU (with allopurinol),

Ftorafur High dose thiotepa Procarbazine High dose Ara-C Hexamethylmelamine/

Pentamethylmelamine Spiromustine 5Azacytidine Ifosfamide Interferon

C. Peripheral neuropathies Vinca alkaloid Cis-platinum Procarbazine Hexamethylmelamine Methyl-gag

D. SIADH Vinca alkoloid Cyclophosphamide

Toxicity Incidence References

Peripheral neuropathy Loss deep tendon reflexes Paresthesia in digits Wrist drop and foot drop Slapping gait Mononeuropathy Quadriparesis

Interleukin-2 with or without LAK cells

Fludarabine

Autonomic neuropathy GI symptoms

Constipation, abdom- inal pain Paralytic ileus

Bladder atony Impotence Postural hypotension

B. Acute cerebellar syn- dromes/ataxia

5-FU Procarbazine Hexamethylmelamine High dose Ara-C Ifosfamide

Cranial nerve palsies

Ocular manifestations Ptosis, ophthalmoplegia Optic atrophy

Trigeminal nerve Jaw pain Loss of cornea1 reflex

Hoarseness Facial atrophy

general. We will also review the neurotoxic side effects and high

dose chemotherapy.

con- fusion, seizure, coma

SIADH secretion

II. Vinca alkaloids

The vinca alkaloids

are widely used in cancer therapy for solid tumors and hematologic malignancies. Vincristine the most frequently utilized vinca alkaloid and its primary dose-limiting toxicity many of clinical manifestations of vincristine-related neurotoxicities and their relative incidence. of deep

tendon reflexes the digits are the earliest and common side effects [ 1,5-81. Mus- cle pain, weakness, gait disturbance and sensory impair- ment can ensue with continued drug administration. Usually the sensory and motor neuropathies are bilateral and symmetrical, Many of the manifestations of vincristine neuropathy are slowly reversible with paresthesias being the most readily reversible followed by the motor and other sensory deficits [5,7,11]. The depressed deep tendon reflexes return slowly if at all

WI.

Gastrointestinal symptoms represent the most com- mon autonomic neuropathy and are dose-related par- ticularly with doses larger than 2 mg; however, they are not cumulative with repeated dosages. Cranial nerve palsies are rare, usually bilateral and generally reversible on drug discontinuation with ocular findings occurring most frequently [ 131.

Central nervous system (CNS) toxicity is not clearly established. Vincristine penetration into the CNS is not significant as shown by the very low concentrations detected in the cerebrospinal fluid [ 11. Seizures are rare so that if they occur other causes should be excluded [4,5,12,21].

The syndrome of inappropriate antidiuretic hormone secretion (SIADH) is felt to be secondary to a direct ef- fect of vincristine on the hypothalamus, neurohypo- physial tract, or posterior pituitary [12,23,24].

51%

23-36%

Rare Rare

40%

26% Rare Rare Rare

Rare

Unknown

More frequent in children

1, 5-8

1, 5-7

9 IO

5,7

5,14 1 5,15,16

5,4.17

7,12

18,19 5,20

4,5,12,21,22

12,23,24 3

Page 3: Neurotoxicity of antineoplastic agents

63

Both vindesine and vinblastine are associated with neurotoxicity that is qualitatively similar to vincristine

induced neuropathy. Of the three drugs vinblastine is the least neurotoxic with myelosuppression as the dose- limiting toxicity, while vindesine neurotoxicity is

moderate [25-27,33-381. Vindesine sulfate (desacetyl vinblastineamide sulfate)

is a vinblastine derivative, that manifests neurotoxicity initially as mild paresthesias in 35-60% of patients [29,30] and generally begins after 3-4 courses of weekly bolus dose therapy. In addition, hyporeflexia, muscle weakness, abdominal cramps, constipation, hoarseness, fatigue, myalgias, jaw pain, paralytic ileus, diarrhea, tin- nitus, tremors, SIADH and urinary retention have been

reported [28,3 l-381.

ZZ. A. Pat~o~~ysiol~gy and electruphysiology

All vinca alkaloids bind tubulin preventing its poly- merization into microtubules [39,40]. In addition to their role in mitosis, microtubules are involved in ax-

oplasmic transport and a variety of secretory functions [40,41].

Despite differing degrees of neurotoxicity, the affinity for tubulin has been shown to be much the same for all three vinca compounds 142,431; therefore, mechanisms other than microtubular disruption may be responsible for the variable toxicities [42].

Chan et al. has compared the three vinca alkaloids in

a de-sheathed cat sciatic nerve preparation model. At a low drug concentration (25 pg), he demonstrated that vincristine was more potent than vinblastine which was more potent than vindesine in blocking axoplasmic transport. At higher drug concentrations, axoplasmic block occured so rapidly that the difference between the

three agents was not significant [44]. Using 3H labeled vincas, Iqbal et al. demonstrated that differences in the toxicity of the three vincas may be attributed to dif- ferences in drug uptake by the nerve fibers [45].

Nerve conduction studies in vincristine treated pa- tients show that sensory and motor nerve conduction velocities usually are only slightly impaired even when significant clinical neuropathy is present 158,461.

Although no abnormalities in neuro-muscular trans- mission have been demonstrated [8,19]; electromyo- graphic testing of distal muscles show abnormal pat- terns compatible with denervation [8,46]. These findings as a whole suggest axonal degeneration of the ‘dying- back’ type (centripetal axonal degeneration as a result of metabolic derangement of the neuron).

There is a dose-dependent decrease of action potential amplitude with vincristine, however, there is no change of electroneurographical values noted in vindesine

treated patients who manifest clinical evidence of

neurotoxicity [47]. The electrophysiologic findings of axonal degeneration have been confirmed by nerve biopsies and tissue obtained at autopsy from vincristine treated patients [8,9,48]. This finding is in contrast with segmental demyelination noted in vincristine treated animals [48,49]. Loss of microtubules and accumulation of nemofilaments is revealed by ultra-stuctural studies of neurons from the spinal ganglia, spinal cord and brain stem [12,50].

ZZ. B. Variables affecting vincristine neurotoxicities

Several factors seem to influence the incidence and severity of vincristine neuropathy. The schedule of ad- ministration of vincristine has been noted to influence the incidence of neurotoxicity as illustrated in Table 3. Vincristine neurotoxicity is related to both the in-

dividual dose and the frequency of administration. The majority of patients will develop early symptoms of neuropathy after 5-6 mg of vincristine [51] and signifi- cant toxicity will occur after a total dose of 15-20 mg on a weekly schedule. Higher cumulative doses can be tolerated over a given period of time if the individual dose is smaller than 2 mg and/or the interval between treatments is longer than a week [ 13,52-541.

Gastrointestinal side effects appear to be related to the magnitude of a single dose. Doses greater than 2.5 mg result in significant gastrointestinal motility impair- ment and if the dose is larger than 3 mg, paralytic ileus may occur [7].

It is conceivable that the single dose and the cumul- ative dose effects may be more important than the schedule effect, since reduction in frequency of ad- ministration will result in a smaller cumulative dose over

TABLE 3

Author Schedule and dose Neurotoxicity

Whitelow [55]

Holland [7]

3 mg twice week-

ly x 4-6 week

then 3 mg Q week

vs. 2 mg Q week or

twice weekly

12.5-75 jog/

kg/week

Jackson [54]*

Thant [53]*

1.4 mgIm* (max. 2 mg)

Max 2 mg days 1 & 8

Max 2 mg Q 3 weeks

Higher toxicity

with 3 mg dose

Earlier onset of

paresthesias at

higher dose

55% grade 1-2

36% grade 3

64% grade l-2

36% grade 3

50% grade 1

*Definition of neurotoxicity grade varies.

Page 4: Neurotoxicity of antineoplastic agents

64

a given period of time and hence a lower incidence of neurotoxicity.

Although it is widely believed that there is a signiti-

cant relation between age and the dose of vincristine re- quired to cause neurotoxicity [4,13], available published data do not absolutely support this conclusion [5,55]. There are few reports [56,57] suggesting enhancement of vincristine neurotoxicity by radiation therapy. These data are suggestive but insufficient to support an inter-

action between radiation and vincristine. Vincristine is predominantly excreted via the biliary

system, therefore liver impairment increases the suscep- tibility to neurotoxicity. Sandler et al. [5] reported that patients who had an elevated bilirubin level developed high grade neurotoxicity at a total vincristine dose of 2-6 mg/m2 over 3-4 weeks.

Some reports incriminate several agents such as INH [4], asparaginase [4], VM-26 [58] and vinblastine [59] in potentiating the neurotoxic effect of vincristine. More recent reports suggest an interaction between etoposide (VP16-213) and vincristine resulting in more severe neurotoxicity. Thant et al. [53] described increased neurotoxicity when VP-16 was added to a vincristine containing regimen. In this study, patients with small

cell lung cancer were treated with a schedule that includ- ed giving vincristine on day 1 and day 8 every 3 weeks. The increased toxicity was probably related to the schedule of vincristine administration, in that moditica- tion of dose schedule to once every 3 weeks resulted in marked reduction of neurotoxic side effects. Both

Jackson et al. [54] and Comis [60] reported that there was no difference in neurotoxicity when etoposide was added to a vincristine containing drug regimen.

A pre-existing neurologic dysfunction may make pa- tients more susceptible to vincristine neurotoxicity. Ex- acerbation of the neurologic deficit in patients with

Charcot-Marie-Tooth syndrome has been reported [61]; however, peripheral neuropathy secondary to diabetes mellitus or Parkinson’s disease did not appear to be worsened by vincristine [46,62].

II. C. Treatment of neurotoxicity

Currently there are no effective methods of routinely preventing or decreasing vincristine neurotoxicity other than dose and schedule modification or discontinuation of the drug. Trials of thiamine, Bi2 [2] and folinic acid generally have been unsuccessful.

In murine models pyridoxine has shown some protec-

tion by prolonging survival when lethal doses of vin- cristine were administered, but it did not prevent the gait changes which may represent neurotoxicity in this

species [63]. When tested in humans at doses of 1.5

g/day for 6 weeks orally, Jackson et al. reported no pro- tective effect for pyridoxine [64].

Glutamic acid was also tested by Jackson et al. in a randomized, placebo controlled trial in stage II breast cancer patients receiving vincristine 1 mg/m2/week for 6 weeks. Glutamic acid in a dose of 1.5 g/day in the study

patients resulted in a statistically significant reduction

TABLE 4

Clinical manifestations of cisplatin neurotoxicity

Toxicity Clinical manifestations Incidence References

Ototoxicity

Peripheral neuropathy

Lhermitte’s sign

Other

Tinnitus

Clinical hearing loss

Audiogram abnormality

[4000-8000 Hz)

Paresthesia

Dysesthesia

Vibratory and position

sensation changes

Diminution or loss of

deep tendon reflexes

Gait difficulties

Distal weakness -

Papilledema,

Retrobulbar neuritis,

loss of taste, memory loss,

intention tremors, postural

hypotension, tetany

9% (O-33%)

6%

25% (O-90%)

14/300 patients

Rare

[67-771

[67-781

[68-70,72.

[76,78-801

[78,81-831

(841 14,781

Page 5: Neurotoxicity of antineoplastic agents

65

only in the development of paresthesia and a decreased

achilles reflex [65]. Mechanism of action of glutamic

acid in this setting remains unclear.

III. Cisplatin and platinum analogues

Cisplatin (CDDP) . CDDP is one of the most potent

cytotoxic agents in the present anticancer armamen- tarium with a unique spectrum of anti-tumor activity. Its major dose-limiting toxicity in phase I studies was

renal [66]. Neurotoxicity has been noticed with some frequency, especially with changes in drug administra- tion and hydration methods and more effective an-

tiemetics resulting in a reduction of nephrotoxicity, nausea and vomiting [72-741. The more common neurotoxicities reported with CDDP and their clinical manifestations are listed in Table 4.

Both tinnitus and hearing loss are associated with CDDP therapy. Tinnitus is reversible but not clearly dose-related [73,74]. Less common is clinical hearing loss which is dose-related but not clearly reversible [67-781. The audiogram abnormality with high frequen- cy pure tone hearing loss can occur as early as 4 days after drug administration and is not reversible [68-70,72,76,78-801.

CDDP usually causes sensory and occasionally sen- sory plus motor neuropathy (usually characterized by gait difficulties). Most patients have bilateral sym- metrical neuropathy with vibratory and position sensa- tion being most often affected. EMG studies in these patients showed serious neurogenic involvement with a

reduction of the interference pattern of the motor unit potential and abundant fibrillation at rest, however, clinical motor neuropathy is usually absent or subtle and difficulty with gait or handwriting is presumably

related to a proprioceptive abnormality [4]. Lhermitte’s sign is described as a sensation of electric

shock and tingling of abrupt onset in the legs and/or arms on neck flexion. This sign is a well recognized self- limiting complication of radiation therapy of the spinal cord. Eeles et al. [84] reported that 14/300 patients treated with CDDP containing chemotherapy for metastatic testicular cancer demonstrated this sign. Symptoms appeared during the treatment in 6 patients, within 1 month after starting treatment in 5 patients and in 1 patient at 2 months. The duration of symptoms ranged from 2-8 months. None of the patients had received radiation therapy to the cervical or dorsal spine and the authors did not observe a platinum dose effect. Concomittant peripheral neuropathy was noted in 12 patients.

Neurologic syndromes can be caused by regional ad- ministration of antineoplastic agents (Table 5). Frustaci

TABLE 5

Neurologic syndromes caused by regional administration of antineo- plastic agents

A. Acute encephalopathy Intrathecal methotrexate

- radiotherapy Intracarotid BCNU Intracarotid cisplatin

B. Arachnoiditislmyelopathy Intrathecal methotrexate Intrathecal Ara-C lntrathecal thiotepa

C. Chronic reactions 1. Necrotizing leukoencephalopathy

Radiotherapy - intrathecal methotrexate Intrathecal methotrexate or IV methotrexate Intrathecal Ara-C

2. Mineralizing microangiopathy Radiotherapy + intrathecal methotrexate

3. Cerebra1 atrophy Intrathecal methotrexate f radiotherapy Intrathecal Ara-C f radiotherapy

4. Pontine myelinolysis/somnolence syndrome Radiotherapy f intrathecal methotrexate

D. Peripheral neuropathykranial nerve palsy Cis-platinum

et al. [85] reported on 63 previously untreated patients with head and neck cancer who were treated with intra-

arterial cisplatin (IA-CDDP). Cranial nerve palsies were noted in 4 patients a few days after the infusion on the ipsilateral side. The 12th cranial nerve was involved in I patient, 7th cranial nerve in another and 9th, lOth, 1 lth and 12th cranial nerves in 2 patients and complete recovery occured only in 1 patient. Stewart et al. [86] reported 3 cases of retinal blindness and 5 cases of severe encephalopathy in 11 patients treated with IA-CDDP for brain tumors. Castellanos et al. [87] reported that 9/l 1 patients treated by internal or external iliac artery infusion of CDDP had parasthesias and/or weakness oc- curing within 48 h. Neurologic examination revealed

decreased pain and/or touch sensation in the Ls-SI or the sciatic nerve distribution in 6 patients and decreased joint position and vibratory sensation in the toes in 4 patients; all 9 patients had weakness. EMG/NCV studies in 6 patients were compatible with lower lumbo-sacral plexopathy (axonal in nature) and 2 patients developed mononeuropathy. Only 1 patient had partial recovery. The authors felt that the mechanism of this form of neurotoxicity was chemotherapy-related small vessel damage resulting in infarction of the neural tissues supplied by the perfused vessels. A direct neurotoxic effect of CDDP, however, could not be excluded.

Page 6: Neurotoxicity of antineoplastic agents

66

III. A. Electrophysiology and nerve biopsy

Peripheral neuropathy. Nerve conduction studies have suggested greater sensory nerve involvement than motor nerve involvement. Of 107 patients treated with CDDP, Von Hoff reported that 3 patients developed peripheral

neuropathy after 210-560 mg/m’ of the drug, and 2 pa- tients showed absent sensory nerve action potentials in the upper and lower extremities. Sural nerve biopsies

revealed scattered myelin damage with intact axons [83]. Thompson et al. [81] noted that the primary damage

was axonal with secondary myelin damage. He did elec- tron microscopic studies of peripheral nerves in 4 pa- tients. Nerve conduction velocity in the right median

nerve, and peroneal nerves remained normal while sural nerve responses abruptly disappeared without previous slowing of conduction velocities in 6 patients at a mean

CDDP dose of (383 mg f 103 mgm’). Cowan et al. 1881 prospectively studied 7 patients treated with CDDP by performing pretreatment and serial electrophysiol-

ogic testing. Two of 7 patients had pretreatment decreased vibratory sensation and 317 had abnormal nerve conduction compatible with peripheral neuro-

pathy. Following CDDP therapy no new clinical peripheral neuropathies were noted but reduced nerve conduction was noted in 3 patients. This suggests that

not all nerve conduction abnormalities in these cancer patients resulted from chemotherapy.

In a report by Carenza et al. [82], 23 of 86 patients with gynecologic neoplasia treated with CDDP based combination chemotherapy were studied serially. None had clinical or neurophysiologic abnormalities prior to therapy. EMG studies post therapy showed reduction of the interference pattern of the motor unit potentials and abundant fibrillation at rest. In 7 symptomatic patients sensory conduction velocity was decreased with slight decrease of the sensory action potential. In 5 patients

maximal motor conduction velocity (MMCV), distal latency and amplitude of the motor potential did not change which is compatible with sensory dysfunction alone. In the other 2 patients no decrease in sensory action potential was noted, but MMCV and motor action potential were reduced and distal latency increas- ed suggesting motor and sensory dysfunction. The authors conclude that this picture is compatible with a degenerative axonal lesion with a secondary myelinic damage similar to the picture of the so called ‘dying- back neuropathies’, the most common morphologic reaction of peripheral nerves to metals like arsenic and thallium [82]. A similar conclusion was drawn by Mabin et al. [89]. In their series patients without any elec- trophysiologic abnormalities had slowing of the motor nerve conduction velocities at 100 mg/m2 of CDDP

with no further worsening at 300 mg/m2 and prolonga- tion of the distal latency of the sensory median nerve after 300 mg/m2. No changes were noted in the elec- trophysiologic studies of patients without clinical abnor- mality but with delayed latency of the Hoffmann reflex.

As in humans, rats treated with CDDP showed evidence of neuropathy in both the CNS and peripheral nervous system, the mechanism of which was shown to be secondary to axonal degeneration [go].

Ototoxicity, demonstrated by loss of hair cells in the organ of corti, has been seen in several animal models

including guinea pigs, rats and monkeys [91]. In monkeys hearing loss was transient with doses as high as the LD,o. The pathology of hearing loss has not been

studied well in man. Clinical studies have suggested a high incidence of cochlear toxicity with a predilection for involvement of the higher frequency range [SO]. In-

itial auditory function testing is recommended as a base line. Recommendations vary for follow up, with some suggesting regular testing in patients committed to long term CDDP therapy to assess the magnitude of potential change [80], while others recommend and we concur, that auditory testing be done when patients develop clinical hearing loss, since changes in therapy may be

warranted [78].

III. B. Variables effecting cisplatin neurotoxicity

Dose and schedule: presently the most effective way to ameliorate CDDP neurotoxicity is to limit the single and/or cumulative dose used. A high incidence of

deafness was reported with single doses of 160-200 mg/m2 [92], suggesting that perhaps a single dose should not exceed 150 mg/m2. Similarly, peripheral neuropathy has been reported with cumulative doses as low as 210 mg/m2, however, clinical tindings usually may not become apparent before a cumulative dose of

at least 350-640 mg/m2 [81,83,93,94]. Pharmacologic modulators of neurotoxicities includ-

ing thiosulfate, WR-2721 (S-2-[3-amino propylamino]

ethylphosphorothioic acid) and ACTH analogues have been used to ameliorate the neurotoxicity of CDDP. The use of these agents has been prompted by preclinical efficacy in animal models. Table 6 summarizes the clinical trials that have been performed. The use of these agents remains investigational.

Hypertonic saline hydration: Legha and Dimery [93] observed significant peripheral neuropathy manifesting as ataxia and difficulty with ambulation in 6/8 patients treated with high dose CDDP [30-40 mg/m2 x 5 days) based chemotherapy. Hydration without hypertonic saline was utilized. The median cumulative dose in pa- tients who developed neurotoxicity was 775 mg/m’.

Page 7: Neurotoxicity of antineoplastic agents

TABLE 6

Pharmacologic modulators of cisplatin neurotoxicity

Agent Mechanism Clincial trials

Thiosulfate Neutralizes toxic effects [95-971

WR-2721 Protects normal tissue from radia- tion and alkyl- ating agent chemotherapy in animal models

t1001

ACTH Animal studies analogues protection from (Org 2766) neurotoxicity

[105,106]

Recombin- Animal studies ant nerve Protection from growth neurotoxicity factor UO81

(I) Low clinical neurotoxicity with high dose intracavitory CDDP (2) No worsening of pre-existing CDDP-induced neurotoxicity

198,991 (I) Glover [loll -7152 mild to moderate peripheral neuropathy at 870 mgim* (cumulative dose) -No ototoxicity (2) Mollman [IO21 - Lower incidence of

neuropathy (25% as compared to 47-100%) -Median dose (635 mgim2) at onset of neuropathy was higher (3) Glover [IO31 -8136 grade 1-2 peripheral neuropathy at median dose of 670 mg/m* -20/36 mild to severe high frequency hearing loss -3136 clinical hearing loss (4) Kish [104] CDDP 120 mg/m’ with or without WR-2721 No protection Randomized, double blind placebo-controlled study

If071 -Fewer neurologic symptoms with high-dose Org 2766 -Increased threshold for vibration perception -No change in cytotoxic effect

When CDDP was administered in hypertonic saline to 47 patients, neurologic toxicity was mild, with signifi- cant peripheral neuropathy seen in only 1 patient. A reduction in auditory acuity was seen in 30% of patients treated at 30 mg/m2/day x 5 or more, but it was dose- limiting in only 2 patients. The authors note that this in- cidence is comparable to trials using CDDP at 120 mg/m2 and the discrepancy in the degree and preval- ence of peripheral neuropathy compared with other studies may be explained by the small cumulative dose in this study and the relatively few patients with prior

CDDP therapy [94]. Haines and colleagues reported a 25% incidence of

67

ototoxicity and 39% incidence of neuropathy in their series of patients treated with 187.5 mg!m2 CDDP ad- ministered over 5 days in hypertonic saline [109]. In animal models salt loading with 3% saline did not result in significant difference in CDDP localization in the

inner ear when compared to non-saline loaded animals.

This is in contrast to the low CDDP localization in the kidneys of the saline loaded animals, explaining the

kidney protective effect of saline hydration [ 1101. Radiation: CDDP administered to 8 children with

recurrent brain tumors and prior cranial radiation was

associated with significant hearing loss in 5 of 6 evaluable patients. Hearing loss progressed on re- challenge with CDDP. In addition, 2 patients developed

profound deterioration of neurologic status within 72 h after CDDP infusion [ 11 I]. A similar observation was

reported by Schell et al. [112]. This effect may be of im-

portance in certain patients such as those with head and neck cancer or others who receive CDDP and radiation therapy to the head and may warrant prospective

evaluation of this phenomenon. Platinum analogues. The potential for dose limiting

and/or permanent gastrointestinal toxicity, renal toxici- ty and/or neuropathy remains despite the present methods of CDDP administration and supportive care, hence the importance of the development of analogues that might demonstrate greater or equivalent clinical ac-

tivity with decreased toxicity.

Recently two platinum analogues have been introduc- ed into clinical trials, carboplatin (CBDCA), which is currently approved for clinical use and iproplatin

(CHIP). In a preclinical trial no significant functional or morphologic cochlear toxicity was reported with CHIP

or CBDCA [ 1 lo]. In phase I and II studies no high frequency sensorineural hearing loss was detected [113-1161. Recently Bishop et al. [117] noted that only 2194 patients treated with CBDCA + VP-16 developed

clinical ototoxicity. Audiograms were not routinely per- formed in these patients. No peripheral neuropathy has been reported.

IV. Other neuropathic chemotherapeutic agents

It is clear that dose, schedule and route of administra-

tion are operational variables influencing the incidence and outcome of neurotoxicity for vinca alkaloids and CDDP. In fact this does not only apply to these drugs but also to some established and newer anti-cancer agents. Table 7 summarizes these agents and their reported neurotoxicities.

The neurotoxicity of high dose methotrexate has been associated with an elevation of myelin basic protein

Page 8: Neurotoxicity of antineoplastic agents

68

TABLE 7

Neuropathic chemotherapeutic agents

Dw Clinical manifestation References

Methotrexate (3-16 g/m2)

Thiotepa (1125-1575

mg/m2)

Reversible stroke-like syndrome Transient encephalopathy Mild cognitive dysfunction Mild to severe behavioral changes Grade 2-4 neurotoxicity Toxicity usually reversible

[118-120)

11211

BCNU Intracarotid High dose (2250-2850

mp/m2) High dose (2 1050 mg/m2) cranial irradiati 5-Fluorouracil (Bolus dosing)

Cytosine Arabinoside (High dose i.e. 3

p/m2 twice daily x 6

days) Ifosfamide (600-4000 mg/m2/day) Hexamethyl- melamine

Acute encephalopathy Encephalomyelopathy leading to coma and death Neuroretinitis

+ on

Taxol

Fludarabine (High dose) Etoposide (VP-16) Cyclophosphamide Suramin (association with peak steady-state, plasma levels of 350 &ml)

Spiromustine

Cerebellar syndrome Gait and limb ataxia Dysarthria Nystagmus Hypotonia Usually reversible Cerebellar symptoms

CNS toxicity approaches

50% at 54 g’m* cumulative dose Acute encephalopathy Acute cerebellar syndrome Incidence < l-30% Depression, hallucination, insomnia, dysphasia, Parkinson’s like tremor, ataxia, peripheral neuropathy Schedule dependent Sensory neuropathy, rapid onset and cumulative, resolves within months after drug discontinuation, motor dysfunction has been reported Altered mental status, Blindness, coma and death peripheral neuropathy SIADH Peripheral neuropathy; generalized flaccid paralysis, variable recovery, distal extremity and peri-oral paresthesias Alteration in cortical integrative function, decreased level of con- sciousness, association with high peak plasma levels, ? cumulative effect

[122-124)

[1251

11251

[I,41 [131-1331

[136,138,139]

[140,141,143]

[4,1441

[147-1501

[151-1531

[154-1561 [157-1591 [160-1621

11631

(MBP) in the CSF suggesting toxic demyelination as a mechnism since MBP is a major component of the pro- tein fraction found in the myelin sheaths [ 1181.

There is an association of neuroretinitis with other in- tensive BCNU series, other than those mentioned in Table 7 and with intra-carotid BCNU suggesting that this complication is indeed related to the high dose

BCNU. Burger et al. (1261 described the histopathologic features of encephalomyelopathy. It is noteworthy that Takvorian et al. [ 1271 noted no unique pattern of CNS

toxicity in his group of patients treated with 600-1400 mg/m* BCNU and bone marrow rescue. Autopsies per-

formed on 15 patients who died subsequent to their

treatment revealed no specific CNS pathology. The incidence of the cerebellar syndrome associated

with 5-FU appears to increase with higher dose. It is almost negligible with doses of 7.5- 15 mg/kg but in- creases in incidence to 3-7% in patients receiving bolus high dose weekly therapy with 15 mg/kg/week or more [l]. However, this toxicity is rare with an infusional 5- FU schedule [128,129]. Attempts to modulate 5-FU metabolism with allopurinol or thymidine has resulted in a higher incidence of the cerebellar syndrome

[ 130,131]. Cerebellar ataxia secondary to this agent has been closely examined. 5-FU readily crosses the blood- brain barrier and achieves significant CNS levels [ 134,135]. Early data suggested that the cerebellar tox- icity may be related to the production of flurocitrate or fluroacetate. This concept has been questioned because thymidine which blocks 5-FU catabolism and presum- ably would reduce flurocitrate and acetate production, may actually enhance the neurotoxicity of 5-FU [ 1311. These compounds may be catabolites of 5-FU and can produce cerebellar lesions in animal models that are similar to those identified in humans [ 136,137].

The mechanism of ifosfamide neurotoxicity is

unclear. Goren et al. suggest that prior therapy with cisplatin resulting in renal tubular damage could lead to alterations in ifosfamide metabolite clearance enhancing the susceptibility of the patient to not only neurotoxicity but also hematologic and renal toxicity [ 1421. Pratt et al. [143] pointed to the development of toxic levels of chloracetaldehyde (an oxidation product of ifosfamide) as a possible cause of the acute neurotoxicity. Chlor- acetaldehyde was felt to be similar to acetaldehyde (ethanol product) in its toxic effect on the brain and the neuro-symptoms of patients treated with ifosfamide resemble many of those seen in patients with alcohol in- toxication. All signs, symptoms and EEG abnormalities were transient.

Some of the new agents introduced by the NC1 have been associated with acute CNS side effects [145,146]. These agents include fludarabine, spiromustine, taxol and suramin. It is interesting to note that new drugs with

Page 9: Neurotoxicity of antineoplastic agents

69

solid tumor selectivity have shown significant neurotox- icity, however, it remains to be seen whether there is a

scientific explanation for such an observation. Taxol, a plant alkaloid, has shown promise in the

treatment of breast and ovarian cancers. Neurotoxicity was not observed in phase I trials using single-dose, multiple-dose or 3-h infusional schedules [ 1471. Neurotoxic effects has been reported with 6 and 24 h

infusions with doses greater than 170-200 mg/m2 [147-1491.

Etoposide (VP-16-213) has been reported to cause

peripheral nerve damage in experimental animals and in humans [154-1561, however; the incriminating data are weak. Littlewood et al. described 9 patients who were treated with VP-16-213 at a cumulative dose of x 2400 mg!m2, 319 were treated with 1400 mg/m2 of VP- 16 + cyclophosphamide. Nerve conduction studies were

either normal or showed no deterioration from pretreat- ment values in 8 patients who were studied [ 1561.

Cyclophosphamide-associated SIADH secretion oc-

curs 4-12 h after the drug infusion and lasts up to 20 h. The mechanism is probably related to direct ADH

release from the posterior pituitary rather than ectopic release or ADH-like action of the metabolite on the kidney.

V. Biologic response modifiers (BRM)

Adoptive immunotherapy has emerged as a promising modality in the treatment of cancer. Several types of this therapy have been introduced into clinical trials; the neurotoxicity associated with interferons and inter- leukin-LAK cells will be reviewed.

V. A. Interferon

Interferon was first characterized in 1957 in England [ 1641. Clinically sufficient quantities of alpha-interferon

were produced in the early 1970s from buffy coat cells and tested in several malignancies. Since 1979, alpha, gamma- and beta-interferons were successfully cloned

and large scale testing begun. Neurotoxicity has been reported with interferon therapy, usually with higher doses of both natural and cloned interferons, irrespec- tive of route of administration [ 165- 17 11.

Interferon related neurotoxicity manifests as overall mental and motor slowing; symptoms include be- havioral, cognitive, affective and personality changes, marked somnolence, lethargy, confusion, loss of taste and smell and expressive dysphasia in extreme instances [ 166,168,169,172]. Death and seizures have also been reported [173,174].

Electroencephalographic studies of patients treated with interferon who experienced neurotoxicity have con- sistently revealed changes of diffuse encephalopathy

with slowing of the alpha-rhythm and the appearance of delta and theta waves predominantly in the frontal lobes [166-169,172]. Rohatiner et al. found no correlation between EEG changes and spinal fluid interferon levels

[168]. Adams et al. [169] reported on 10 patients with metastatic renal cell carcinoma who were serially

evaluated by neuropsychiatric examination to determine the nature of the fatigue-asthenia symptoms associated with alpha-interferon, Prior to therapy all the patients were found to be free of any psychiatric illnesses or cognitive impairment that might indicate cerebral

disease. Interferon produced moderately severe changes in behavior manifesting as psychomotor retardation with loss of incentive, verbal and motor spontaneity and slight to moderate changes in cognition, affect and per-

sonality in some of the patients. These symptoms disap- peared when interferon was discontinued. This toxicity

was suggested to be secondary to frontal lobe dysfunc- tion. Overt depression and episodes of psychotic

behavior have been rare [ 1711. Psychotic reaction and hallucination have been reported by several in-

vestigators when more than 20 munits/m2 of interferon has been used [ 174- 1761. CAT scan of the brain prior to interferon administration has been recommended since preexisting brain metastasis may predispose pa- tients to worse interferon related neurotoxicity [171].

The mechanisms underlying interferon neurotoxicity are unclear, and possibilities include direct effect on frontal lobes, or deeper brain structures, impedance of neurotransmitters, alteration of cholinergic balance or release of other substances. Calvert and Gresser [177] demonstrated enhanced spontaneous neuronal activity and evoked responses when cat or rat cerebral cortex was incubated with interferon. Intracerebral injection of interferon into mice has produced endorphin-like opioid effects including decreased motor activity and catalepsy which could be reversed by naloxone [ 1781.

Adams et al. [169] found both metoclopramide hydrochloride (dopamine antagonist) and methylphen- idate (a dopamine agonist) could reverse some of the neuropsychiatric effects of leukocyte interferon. The hypersomnia with interferon may be a reflection of in- terferon activity in the reticular activating system of the brain stem and/or interference with the frontal lobes extensive reticular activating system connections.

Peripheral nervous system effects, particularly distal paresthesias, have been reported rarely with interferon therapy. At high doses interferon may result in denerva- tion and neurogenic muscle atrophy [ 166,179- 18 11.

V.B. Interleukin-2 (IL-2) with or without lymphokine

activated killer (LAK) cells

Denicoff et al. [182] reported that 15/44 (30%)

Page 10: Neurotoxicity of antineoplastic agents

70

patients with metastatic cancer treated with systemic recombinant IL-2 combined with autologous LAK cells

developed severe behavioral changes that included agitation and combative behavior of variable severity which necessitated acute intervention. Twenty two pa- tients (50%) had severe cognitive changes and 7 developed delusions and hallucinations. These neuro- psychiatric side effects appear to be dose and time

related, more frequent at a higher dose and almost uniform at the end of each treatment.

When only IL-2 was administered 17/24 (70%) pa- tients who received 100 000 units/kg showed severe to

moderate neuropsychiatric changes compared with 6119

(30%) patients who received 30 000 units/kg (P c 0.05). All changes were reversible. Fisher et al. [ 1831 in treating metastatic renal carcinoma with IL-2 and LAK cells noted somnolence or disorientation (grade 3 toxicity) in 31% of patients. Somnolence, coma and disorientation were reported by Rosenberg et al. in patients treated with IL-2 with or without LAK cells. Most patients had appreciable improvement after 2 to 3 days and all returned to baseline behavior and cognitive scores [ 1841.

Thompson et al. [185] in evaluating the influence of dose and duration of infusion of IL-2 on toxicity and immunomodulation noted transient confusion in 315 pa- tients receiving 3 x 10% of IL-2 infused over 24 h. This persisted during IL-2 administration, did not re-

quire medication or psychiatric intervention and resolv- ed after discontinuing IL-2. Confusion was not seen in any of the other treatment groups.

VI. Models for evaluating neurotoxicity of anti-cancer

agents

It is important to be able to evaluate the potential of anti-cancer agents for producing neurotoxicity prior to their introduction into clinical trials. For example, a system that is suitable for studying factors contributing to the differences in the neurotoxic potential between certain drugs and their analogues might result in the discovery of a novel agent that exhibits low neurotoxic potential.

Animal models, used for this evaluation, have not been useful. In the case of the vinca alkaloids, only cer- tain species manifested the neurotoxic side effects. Chicken, monkeys and cats developed neurotoxicity when treated with vincristine but not with the other two vincas. The mouse, rat, dog and guinea pig were not useful models for evaluating vinca alkaloid neurotoxi- city [50]. Another barrier is that the neurotoxicity that is exhibited by animals is not necessarily pathologically similar to the toxicity noted in humans. The peripheral

neuropathy seen with vincristine is primarily secondary to axonal degeneration, while variable degrees of toxic

myopathy are noted in animals [50]. As a result, some investigators have introduced alternative systems utiliz- ing cultures of new-born rat mid-brain [ 1861 or fetal rat hypothalamic neurons [ 1871. Such systems eliminate

pharmacokinetics as a factor in the pathogenesis of neurotoxicity, they are rapid and potentially easier to

quantitate. Some of the initial available studies indicate that the order of neurotoxicity induced by different agents appears to correlate well with their known

relative clinical toxicities [ 186,187]. Earlier models of experimental toxic neuropathy

evaluated morphologic and physiologic parameters, more recent models tend to evaluate biochemical and behavioral parameters. These may prove to be better predictors of early neurotoxicity [ 1881.

VII. Conclusion

In recent years more attention has been paid to the quality of life of cancer patients, not only when their disease is in remission but more importantly while on therapy, hence the importance of preventing and/or

modifying the neurotoxic side effects of the anti-cancer agents.

From the previous review it is clear that the dose, schedule and route of administration determine significantly the incidence and outcome of some of the anti-cancer agents’ neurotoxicity. Nevertheless, at- tempts at utilizing modifiers of neurototoxicity and ac- quiring a better understanding of the underlying mechanisms of this toxicity need to be made in an effort to develop an efficient and representative model that may help exclude unacceptable neurotoxic agents.

Because of the progress that has been made in ameliorating some of the distressing side effects of anti- cancer therapy, more attention has been focused on neurotoxicity. This necessitates a more detailed grading

system so that the extent of the neurotoxicity can be ap- propriately evaluated. Clinical trials should prospective- ly evaluate the potential for neurotoxicity of anti-cancer agents.

The WHO neurotoxicity grading system [189] is presently used. Castellanos and Field [190] introduced 14 criteria to evaluate neurotoxicity. By using a similar evaluation system, Pazdur et al. [ 1631 were able to better define and quantitate the neurotoxicity of spiromustine.

In addition to a uniform grading system it is clearly important that a reliable and predictive model is established for screening new drugs prior to the in- troduction of such agents into clinical trials.

Page 11: Neurotoxicity of antineoplastic agents

71

Summary

Purpose: to review the neurotoxicity associated with antineoplastic agents.

Methods: four hundred articles, abstracts and book

chapters were selected for review. One hundred and ninety (articles, book chapters and abstracts) were iden- tified as representative of the important aspects of neurotoxicity to be presented in this review.

Results: in general the dose, schedule and route of ad- ministration significantly determine the incidence and outcome of antineoplastic agents neurotoxicity. An up- dated and detailed review of neurotoxicity is provided with special attention to vinca alkaloids, cisplatin and

biologic response modifiers. The neurotoxic side effects

of some of the new approaches in cancer therapy and some of the investigational agents are discussed. Guidelines for the prevention and management of this toxicity are presented. In addition, suggestions are made in regard to the preclinical and clinical screening of new agents for neurotoxicity.

Conclusion: quality of life issues have become a focal point in many clinical trials. Neurotoxicity associated with antineoplastic therapy clearly has an impact on the short and long term quality of the life of cancer patients.

A better understanding of this toxicity requires develop- ing reliable and predictive models to screen new agents prior to their introduction into clinical trials; a more detailed and uniform grading system; and the prospec- tive evaluation of neurotoxicity in clinical trials of new antineoplastic agents.

Acknowledgment

The authors thank Ms. Donna Bennett for secret- arial assistance.

Biographies

Dr. Muha Hussain is a staff Oncologist at the VA Medical Center, Allen Park, MI and an Assistant Pro- fessor of Medicine, Dept. of Internal Medicine, Division Hematology/Oncology, Wayne State University,

Detroit, MI. Dr. Wozniak is a staff Oncologist at Harper Hospital, Detroit, MI and an Assistant Professor of Medicine, Dept. of Internal Medicine, Division of Hematology/Oncology, Wayne State University, Detroit, MI. Dr. Edelstein is Chief of the Medical Ser- vice at the VA Medical Center, Allen Park, MI and an

Associate Professor of Medicine, Dept. of Internal Med- icine, Division of Hematology/Oncology, Wayne State University, Detroit, MI.

Reviewer

This paper was reviewed by Monica Spaulding, M.D.,

Buffalo VA Medical Center, 3495 Bailey Avenue, Buf- falo, New York 14215, USA.

References

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of common-

ly used antineoplastic agents. N Engl J Med 291:75-81,127-133.

1974.

Kaplan RS, Wiernik PH. Neurotoxicity of antineoplastic drugs.

Semin Oncol 82:9,1;103-130, 1982. Shapiro WR, Young DF. Neurological complications of antineo-

plastic therapy. Acta Neurol Stand 70 (suppl 100):125-132,

1984.

Kaplan RS, Wiernik PH. Neurotoxicity of antitumor agents. In

Perry MC, Yarho JW, eds. Toxicity of Chemotherapy. Clincial

Oncology Monographs. Orlando, FL: Grune and Straton,

1984;365-43 1.

Sandler SG, Tobin W, Henderson ES. Vincristine-induced

neuropathy. A clinical study of fifty leukemic patients.

Neurology (Minn) 19:367-374, 1969.

Watkins SM. Griffin JP. High incidence of vincristine

neuropathy in lymphomas. Br Med J 1:610-612, 1978.

Holland JF, Scharlau C, Gailani S et al. Vincristine treatment of

advanced cancer. A cooperative study of 392 cases. Cancer Res

33:1258-1264, 1973.

Bradley WB, Lassman LP, Pearce GW et al. The neuromyopathy

of vincristine in man. Clinical electrophysiological and

pathological studies. J Neurol Sci 10: 107-131. 1970.

Moress GR, D’Agostino AN, Jarcho LW. Neuropathy in lym-

phoblastic leukemia treated with vincristine. Arch Neurol 16:

377-384, 1967.

Levitt LP, Prager D. Mononeuropathy due to Vincristine toxici-

ty. Neurology (Minn) 25: 894-895, 1975.

Berenson MP. Recovery after inadvertant massive over dosage of

vincristine. Cancer Chemother Rep 55: 525-526, 1971.

Rosenthal S, Kaufman S. Vincristine neurotoxicity. Ann Intern

Med 80: 733-737, 1974.

Legha SS. Vincristine neurotoxcity pathophysiology and

management. Toxicology management review. Medical Tox-

icology 1:421-427, 1986.

Gottlieb RJ, Cuttner J. Vincristine-induced bladder atony.

Cancer 28:674-675, 1971.

Dibella NJ. Vincristine-induced orthostatic hypotension. A pro-

spective study. Cancer Treat Rep 64:359-360, 1980.

Carmichael SM, Eagleton L, Ayers CR et al. Orthostatic

hypotension during vincristine therapy. Arch Intern Med

126:290-293, 1970.

Albert DM, Wong VG, Henderson ES. Ocular complications of

vincristine therapy. Arch Ophthalmol 78:709-713, 1967.

Johnson IS. Armstrong JG, Gorman M et al. The vinca

alkaloids. A new class of oncolytic agents. Cancer Res

23:1390-1427, 1963.

Bohannon RA, Miller DG, Diamond HD. Vincristine in the

treatment of lymphomas and leukemias. Cancer Res 23:613-625, 1963.

Haggard ME, Fernbach DJ, Holcomb TM et al. Vincristine in

acute leukemia of childhood. Cancer 22:438-&t, 1968.

Hildebrand J. Lesions of the nervous system in cancer patients.

New York: Raven Press, 1978;49-70.

Whittaker JA, Parry DH, Bunch C et al. Coma associated with

vincristine therapy. Br Med J 4:335-337, 1973.

Slatter LM, Wiener RA, Set-pick AA. Vincristine neurotoxicity

with hyponatremia. Cancer 23:122-125, 1969.

Suskind RM, Brusilow SW, Zehr J. Syndrome of inappropriate

Page 12: Neurotoxicity of antineoplastic agents

12

secretion of antidiuretic hormone produced by vincristine toxici- ty (with Bioassay of ADA level). J Pedia 81:90-92, 1972.

25 Hertz R, Lipsett MB, Moy RH. Effect of vincaleukoblastin on metastatic choriocarcinoma and related trophoblastic tumors in women. Cancer Res 20:1050-1053, 1960.

26 Frei E, Franzino A, Shnider BI et al. Clinical studies of vinblastine. Cancer Chemother Rep 12:125-129, 1961.

27 Brook J, Schreiber W. Vocal cord paralysis; a toxic reaction to vinblastine (NSC-49842) therapy. Cancer Chemother Rep 55591-593, 1971.

28 Cersosimo RJ, Bromer R, Licciardello JTW et al. Pharmacology, clinical efIicacy and adverse effects of vindesine sulfate. A new vinca alkaloid. Pharmacotherapy 3:259-74, 1983.

29 Brade WP. Critical review of pharmacology, toxicology phar- macokinetics of vincristine, vindesine, vinblastine. In: Pro- ceedings of the International Vinca Alkaloid Symposium - Vindesine. Frankfurt: 1981;95-123.

30 Bayssas M, Gouveia J, de Vassal F et al. Vindesine. A new vinca alkaloid. Ret Results Cancer Res 79:91-97, 1980.

31 Dyke RW, Nelson RL. Phase I anti-cancer agents. Vindesine (desacetyl vinblastine sulfate amide). Cancer Treat Rev 4:135-142, 1977.

32 Todd GC, Gibson WR, Grifing WJ et al. The preclinical study of desacetyl vinblastine amide (DVA). Proc Am Assoc Cancer Res 16:279, 1975 (abstr).

33 Obrist R, Paravicini U, Hartmann D et al. Vindesine. A clinical trial with special reference to neurological side effects. Cancer Chemother Pharmacol 2:233-237, 1979.

34 Gralla RJ, Tan CT, Young CW. Vindesine. A review of Phase-II trials. Cancer Chemother Pharmacol 2:271-274, 1979.

35 Ohnuma T, GreenSpan EM. Holland JF. Initial clinical study with Vindesine. Tolerance to weekly IV bolus and 24-hour infu- sion. Cancer Treat Rep 64:25-30, 1980.

36 Carroll DS. Gralla RJ, Kemeny NE. Phase II evaluation of vindesine in patients with advanced colorectal carcinoma. Cancer Treat Rep 63:2097-2098, 1979.

37 Bedikian AY, Valdivieso M, Maroun J et al. Evaluation of vindesine and MER in colorectal cancer. Cancer 46:463-467, 1980.

38 Smith IE, Powles TJ. Vindesine in treatment of breast cancer. Cancer Chemother Pharmacol 2:261-262, 1979.

39 Wilson L, Anderson KA, Chin D. Nonstoichiometric poisoning of microtubule polymerization. a model for the mechanism of the vinca alkaloids, podophyilotoxin and colchicine. In: Goldman et al., eds. Cold Spring Harbor Conference on Cell Proliferation III, Cell Motility. New York: Cold Spring Harbor Laboratory, 1976;1051-1064.

40 Pratt WB, Ruddon RW. The anticancer drugs. Oxford Universi- ty Press. New York, 1974; 221-233

41 Iqbal 2, Ochs S. Uptake of vinca alkaloids into mammalian nerve and its subcellular components. J Neurochem 3459-68, 1980.

42 Himes RH, Kersey RN, Heller-Bettinger I et al. Action of the vinca alkaloids vincristine, vinblastine and desacetyl vinblastine amide on microtubules in vitro. Cancer Res 36:3798-3802, 1976.

43 Alejandro Donoso J, Green LS, Heller-Bettinger IE et al. Action of the vinca alkaloids vincristine, vinblastine and desacetyl vinblastine amide on axonal fibrillar organelles in vitro. Cancer Res 37:1401-1407, 1977.

44 Chan SY, Worth R and Ochs S. Block of axoplasmic transport in vitro by vinca alkaloids. J Neurobiol ll,3:251-264, 1980.

45 Iqbal Z, Ochs S. Fast axoplasmic transport of a calcium-binding protein in mammalian nerve. J Neurochem 31:409-418, 1979.

46 Casey EB, Jellife AM, Le Quesne PM et al. Vincristine neuropathy. clinical and electrophysiological observations. Brain 96:69-86, 1973.

47 Weber W, Tachmann W, Freund HJ et al. The evaluation of

neurotoxicity in cancer patients treated with vinca alkaloids with special reference to vindesine. Anticancer Res I :3 I-34. 198 I,

48 Gottschalk PG, Dyck PJ, Kiely JM. Vinca alkaloid neuropathy. Nerve biopsy studies in rats and in man. Neurology 18:875-882, 1968.

49 Bradley WG. Neuropathy of vincristine in the guinea pig. An electrophysiological and pathological study. J Neurol Sci 10:133-162, 1970.

50 Todd GC, Griffng WJ, Gibson WR et al. Animal models for the comparative assessment of neurotoxicity following repeated ad- ministration of vinca alkaloids. Cancer Treat Rep 63:35-41. 1979.

51 Korbitz BC, Davis HL Jr, Ramirez G et al. Low doses of vin- cristine (NSC-67574) for malignant disease. Cancer Chemother Rep 53~249-254, 1969.

52 Grinberg R, Nemoto T, Dao TL. Vincristine (NSC-67574): Dosage and response in advanced breast cancer. Cancer Chemother Rep 45:57-61, 1969.

53 Thant M, Hawley RJ, Smith MT et al. Possible enhancement of vincristine neuropathy by VPl6. Cancer 49:859-864, 1982.

54 Jackson DV. Wells HB, White DR et al. Lack of potentiation of vincristine induced neurotoxicity by VP-16-213. Am J Clin On- co1 (CCT) 6:327-330, 1983.

55 Whitelaw DM. Coman DH, Cassidy FR et al. Clinical experience with vincristine. Cancer chemother Rep 30:13-20, 1963.

56 Cassady JR, Tonnesen GL, Wolfe LC et al. Augmentation of vincristine neurotoxicity by irradiation of peripheral nerves. Cancer Treat Rep 64:963-965, 1980.

57 Byfield JE. Ionizing radiation and vincristine. Possible neurotox- ic synergism. Radio1 Clin Biol 4!:129-138, 1972.

58 Grifliths JD, Stark RJ, Ding JC et al. Vincristine neurotoxicity enhanced in combination chemotherapy including both teniposide and vincistine. Cancer Treat Rep 70:519-521. 1986.

59 Stewart DJ, Maroun JA, Lefebvre B et al. Neurotoxicity and ef- ficacy of combined vinca alkaloids in breast cancer. Cancer Treat Rep 70:57 I-573, 1986.

60 Comis RL. Clinical trials of cyclophosphamide, etoposide, and vincristine in the treatment of small-cell lung cancer. Semin On- col l3:3, suppl 3,40-44. 1986.

61 Weiden PL, Wright SE. Vincristine neurotoxicity. New Engl J Med 286: I369- 1370, 1972.

62 Tobin W, Sandler SG. Neurophysiologic alterations induced by vincristine. Cancer Chemother Rep 52:519-526, 1968.

63 Jackson D, Long T, Rich C et al. Pyridoxine modification of vin- cristine toxicity. Proc Am Assoc Cancer Res 25:313,1984 (abstr).

64 Jackson DV, Pope EK, McMahan RA et al. Clinical trial of pyridoxine to reduce vincristine neurotoxicity. J Neuro Oncol 4:37-41, 1986.

65 Jackson DV, Wells HB, Atkins JN et al. Amelioration of vincristine-induced neurotoxicity by glutamic acid. Results of a randomized study of the Piedmont Oncology Association. Proc Am Assoc Cancer Res 28:744, 1987 (abstr).

66 Krakoff IH. Nephrotoxicity of cis-dichlorodiammineplatinum (II). Cancer Treat Rep 63: 1523-1525, 1979.

67 Young RC, Van Hoff DD. Gormley P et al. Cis-Dichloro- diammine-platinum (II) for the treatment of advanced ovarian cancer. Cancer Treat Rep 63~1539-1544, 1979.

68 Higby DJ, Wallace HJ, JR, Albert D et al. Diamminodichloro- platinum in the chemotherapy of testicular tumors. J Urol 112:100-104, 1974.

69 Yagoda A, Watson RC, Gonzalez-Vitale JC et al. Cis-dichlor- diammineplatinum (II) in advanced bladder cancer. Cancer Treat Rep 60:917-923, 1976.

70 Kovach JS, Moertel CG, Schutt AJ et al. Phase II Study of cis- diamminedichloroplatinum (NSC-119875) in advanced car- cinoma of the large bowl. Cancer Chemother Rep 57:357-359, 1973.

Page 13: Neurotoxicity of antineoplastic agents

13

71 Wiltshaw E and Kroner T. Phase II study of cis-dichlorodiam- mine platinum (II) NSC- I 19875) in advanced adenocarcinoma of the ovary. Cancer Treat Rep 60:55-60, 1976.

72 Hayes DM, Cvitkovic E, Golbey RB et al. High-dose cis-

platinum diamminedicholoride. Ameliorition of renal toxicity by mannitol diuresis. Cancer 39: 1372- 138 1, 1977.

73 Wittes RE, Cvitkovic E, Shah J et al. Cis-dichlorodiamine- platinum (II) in the treatment of epidermoid carcinoma of the head and neck. Cancer Treat Rep 61:359-366, 1977.

74 Chary KK, Higby DJ, Henderson ES et al. Phase I study of high- dose cis-dichlorodiammineplatinum (II) with forced diuresis. Cancer Treat Rep 61:367-370, 1977.

75 Hill JM, Loeb E, MacLellan A et al. Clinical studies of platinum coordination compounds in the treatment of various malignant diseases. Cancer Chemother Rep 59647-659, 1975.

76 Rossof AH, Bearden JD III, Coltman CA, JR. Phase II evalu- ation of cis-diamminedichloroplatinum (II) in lung cancer. Cancer Treat Rep 60:1679-1680, 1976.

77 Yap HY, Salem P, Hortobagyi GN et al. Phase II study of cis- dichlorodiammineplatinum (II) in advanced breast cancer. Cancer Treat Rep 62405-408, 1978.

78 VonHoff DD, Schilsky R, Reichert CM et al. Toxic effects of cis- dichlorodiammineplatinum (II) in man. Cancer Treat Rep 63:1527-1531. 1979.

79 Kamalakar P, Freeman AI, Higby DJ et al. Clinical response and toxicity with cis-dichlordiammine platinum (II) in children. Cancer Treat Rep 61:835-839, 1977.

80 Fausti SA, Schechter MA, Rappaport BZ et al. Early detection of cisplatin ototoxicity. Cancer 53:224-231, 1984.

81 Thompson SW, Davis LE, Kornfeld M et al. Cisplatin neuropathy clinical, electrophysiologic, morphologic and tox- icologic studies. Cancer 54: 1269- 1275, 1984.

82 Carenza L, Villani C, Framarino Dei Malatesta ML et al. Peripheral neuropathy and ototoxicity of dichlorodiam- mineplatinum: Instrumental evaluation, preliminary results. Gyn Oncol 25:244-249, 1986.

83 Von Hoff DD, Reichert CM, Cuneo R et al. Demyelination of peripheral nerves associated with cis-diamminedichloroplatinum (II) (DDP) therapy. Proc Am Assoc Cancer Res 20:91, 1979 (abstr).

84 Eeles R, Tait DM and Peckham MJ. Lhermitt’s sign as a com- plication of cisplatin-containing chemotherapy for testicular cancer. Cancer Treat Rep 70;7:905-907, 1986.

85 Frustaci S, Barzan L, Comoretto R et al. Local neurotoxicity after intra-arterial cisplatin in head and neck cancer. Cancer Treat Rep 71:257-259, 1987.

86 Stewart D, Wallace S, Leavens M et al. Phase I study of in- tracarotid cis-diamminedichloroplatinum in patients with in- tracerebral tumors. Proc Am Assoc Cancer Res 22:189, 1981.

87 Castellanos AM, Glass JP and Yung WKA. Regional nerve in- jury after intra-arterial chemotherapy. Neurology 37:834-837, 1987.

88 Cowan JD, Kies MS, Roth JL et al. Nerve conduction studies in patients treated with cis-diamminedichloroplatinum (II): A preliminary report. Cancer Treat Rep 64: 1119-l 122. 1980.

89 Mabin D, Borsotti JP, Tea S et al. Neuropathies peripheriques au tours des traitements au cisplatine. Etude clinique et elec- trophysiologique de (II) cas. Rev E.E.G. Neurophysiol Clin 15:341-348, 1985.

90 Clark AW, Parhad IM, Griffin JW et al. Neurotoxicity of cis platinum. Pathology of the central and peripheral nervous sys Neurology 30:429. 1980.

91 Stadnicki SW, Fleischman RW, Schaeppi U et al. Cis-dichloro- diammine platinum (II) (NSC-119875): Hearing loss and other toxic effects in Rhesus monkeys. Cancer Chemother Rep 59~467-480, 1975.

92 Blumenreich MS, Woodcock TM, Jone M et al. High dose

cisplatin in patients with advanced malignancies. Cancer 55:1118-1122, 1985.

93 Legha SS and Dimery IW. High-dose cisplatin administration without hypertonic saline. observation of disabling neurotoxici- ty. J Clin Oncol 3:10,1373-1378, 1985.

94 Bajorin D, Bosl GJ and Fein R. Phase I trial of escalating doses of cisplatin in hypertonic saline. J Clin Oncol 5.10:1589-1593, 1987.

95 Burchenal JH, Kalaher K. Lokys L et al. Studies of cross- resistance, synergistic combination and blocking activity of platinum derivitives. Biochemie 60:961-965, 1978.

96 Howe1 SB, Taetle R. Effect of sodium thiosulfate on cis-dichloro- diammine platinum (II) toxicity and anti-tumor activity in L1210 leukemia. Cancer Treat Rep 64:61 l-616, 1980.

97 Filipski J, Kohn KW, Prather R et al. Thiourea reverses cross links and restores biological activiity in DNA treated with dichloro diaminoplatinum (II). Science 204: 181-183, 1979.

98 PIiefle CE, Howell SB, Felthouse RD et al. High-dose cisplatin with sodium thiosulfate. J Clin Oncol 3:237-244, 1985.

99 Markman M, Cleary S, Pfeifle CE et al. High-dose intracavitary cisplatin with intravenous thiosulfate, low incidence of serious neurotoxicity. Cancer 56:2364-2368, 1985.

100 Yahas JM, Spellman JM, Culo F. The role of WR-2721 in radiotherapy and/or chemotherapy. In: Brady L, ed. Radiation Sensitizers. New York, Masson Press, 1980; 303-308.

101 Glover D, Glick JH, Weiler C et al. Phase I/II trials of WR-2721 and &platinum. Int J Rad Oncol Biol Phys 12: 1509- I5 12, 1986.

102 Mollman JE, Glover DJ, Hogan WM et al. Cisplatin neuropathy. Risk factors, prognosis and protection by WR-2721. Cancer 61:2192-2195, 1988.

103 Glover D, Glick JH, Weiler C et al. WR-2721 and high-dose cisplatin. An active combination in the treatment of metastatic melanoma. J Clin Oncol 5,4:574-578. 1987.

104 Kish JA, Ensley JF, Tilchen E et al. Evaluation of high dose WR2721 + high dose cisplatin + 5-Fluorouracil infusion in recurrermmetastatic head and neck cancer. Proc Am Sot Clin Oncol 10:681, 1991 (abstr).

10.5 Idem. Org. 2766 protects from cisplatin-induced neurotoxicity in rats. Exp Neural 97:746-50, 1987.

106 Gerritsen van der Hoop R, De Koning P, Boven E et al. Efficacy of the neuropeptide Org 2766 in the prevention and treatment of cisplatin-induced neurotoxicity in rats. Eur J Cancer Clin Oncol 241637-642, 1988.

107 Gerritsen van der Hoop R, Vecht CJ, Van Der Burg MEL et al. Prevention of cisplatin neurotoxicity with an ACTH[4-91 analogue in patients with ovarian cancer. N Engl J Med 322:89-94, 1990.

108 Apfel SC? Arezzo JC, Lipson L et al. Nerve growth factor prevents experimental cisplatin neuropathy. Ann Neurol 31:76-80, 1992.

109 Haines I, Bosl G, Pfister D et al. Very-high-dose cisplatin with bleomycin infuion as initial treatment of advanced head and neck cancer. J Clin Oncol 5,10:1594-1600, 1987.

110 Schweitzer VG, Rarey KE, Dolan D et al. Ototoxicity of cisplatin vs. platinum anologs CBDCA (JM-87) and CHIP (JM-9) Otolaryngol Head Neck Surg 94:458, 1986.

111 Granowetter L, Rosenstock JG and Packer RJ. Enhanced cis- platinum neurotoxicity in pediatric patients with brain tumors. J Neuro Oncol 1:293-297, 1983.

112 Schell MJ, McHaney VA, Green AA et al. Hearing loss in children and young adults receiving cisplatin with or without prior cranial irradiation. J Clin Oncol 7,6:754-760, 1989.

113 Harland SJ, Newell DR, Siddik ZH et al. Pharmacokinetics of cis-diammine-l,l-cyclobutane dicarboxylate platinum (II) in pa- tients with normal and impaired renal function. Cancer Res 441693-1697, 1984.

114 Calvert A, Harland S, Siddik Z et al. Early clinical studies with

Page 14: Neurotoxicity of antineoplastic agents

74

cis-diammine-I,1 cyclobutane dicarboxylate platinum (II). Cancer Chemother Pharmacol 9: 140- 147, 1982.

115 Creavern PJ, Mittelman A, Pendyala L et al. Phase I study of a new antineoplastic platinum analogue cis-dichlorotransdihydr- oxy-bisisopropylamine platinum IV (CHIP). Clin Pharm 22, 1982.

116 Creaven P, Madajewicz S, Pendyala L et al. Phase I clinical trial of cis-dichloro-transdihydroxybisisopropylamine platinum (IV) (CHIP). Cancer Treat Rep 67:795-800, 1983.

117 Bishop JF, Raghavan D, Stuart-Harris R. et al. Carboplatin (CBDCA, JM-8) and VP-l 6-213 in previously untreated patients with small-cell lung cancer. J Clin Oncol 5:10,1574-1578. 1987.

118 Neijstrom E, Gabriel DA. High-dose methotrexate-induced neurotoxicity associated with elevation of CSF myelin basic pro- tein J Clin Oncol 3:593-594, 1985.

119 Allen JC, Rosen GR. Transient cerebral dysfunction following chemotherapy for osteogenic sarcoma. Ann Neurol 3441-444, 1978.

120 Fritsch G, Urban C. Transient encephalopathy during the late course of treatment with high-dose methotrexate. Cancer 53:1849-1861, 1984.

121 Herzig RH, Fay JW, Herzig GP et al. Phase I-II studies with high-dose thiotepa and autologous marrow transplantation in patients with refractory malignancies. Advances in cancer chemotherapy. High-dose thiotepa and autologous marrow transplantation. Proceedings of Advances in Cancer chemotherapy Symposium held Ott 25, 1986; in Dallas, TX, 17-23.

122 West CR, Avellanosa AM, Barua NR et al. Phase II study on malignant gliomas of the brain treated with intra-arterial BCNU in combination with vinicristine and procarbazine. Proc Am Assoc Cancer Res 21: 482, 1980.

123 Shapiro WR and Shapiro JR. Principles of brain tumor chemotherapy. Semin Oncol 3,1:56-69, 1986.

124 Madajewicz S, West CR, Park HC et al. Phase II study intra- arterial BCNU therapy for metastatic brain tumors. Cancer 47:65-657, 1981.

125 Phillips GL, Fay JW, Herzig GP et al. Intensive 1,3-bis [2-chloro ethyl -l-nitrosourea] (BCNU). NSC#366 650 and cryopreserved autologous marrow transplantation for refractory cancer. Cancer 52:1792-1802, 1983.

126 Burger PC, Kamenar E, Schold CS, Fay JW, Phillips GL, Herzig GP. Encephalomyelopathy follow high-dose BCNU therapy. Cancer 48:1318-1327, 1981.

127 TakVorian T, Parker LM, Hochberg FH et al. Autologous bone marrow transplantation. Host effects of high dose BCNU. J Clin Oncol 1,10:610-620, 1983.

128 Siefert P, Baker LH, Reed ML et al. Comparison of continuously infused 5-fluorouracil with bolus injection in treatment of pa- tients with colorectal adenocarcinoma. Cancer 36: 123-128, 1975.

129 Lokich JJ, Ahlgren JD, Gullo JJ et al. A prospective randomized comparsion of continuous infusion fluorouracil with a conven- tional bolus schedule in metastatic colorectal carcinoma. A mid- Atlantic Oncology program study. J Clin Oncol 7,4:425-432. 1989.

130 Kroener JF, Saleh F, Anderson RE et al. 5-Fluorouracil and allopurinol. Toxicity modulation and phase II results in colon cancer. Proc Am Assoc Cancer Res 22:459, 1981 (abstr).

131 Cheng E, Woodcock T. Young C et al. Enhanced neurotoxicity of 5-Fluorouracil (FU) by thymidine (TdR). Proc Am Assoc Cancer Res 21:350, 1980.

132 Moertel CG, Reitemeier RJ, Bolton CF et al. Cerebellar ataxia associated with fluorinated pyrimidine therapy. Cancer Chemotherapy Rep 41:15-18, 1964.

133 Boileau G, Piro AJ, Lahiri SR et al. Cerebellar ataxia during 5-Fluorouracil (NSL-19893) therapy. Cancer Chemotherapy Rep 55595-598, 1971.

134 Myers CE, Diasio R, Eliot HM et al. Pharmacokinetics of fluoropyrimidines. Implication for their clinical use. Cancer Treat Rev 3:175-183, 1976.

135 Bourke RS, West CR, Chheda G et al. Kinetics of entry and distribution of 5-fluorouracil in cerebrospinal fluid and brain following intravenous infection in a primate. Cancer Res 33:1735-1746, 1973.

136 Koenig H. Pate1 A. The acute cerebellar syndrome in 5-fluorouracil chemotherapy. A manifestation of fluoroacetate intoxication. Neurology (Minn) 20:416, 1970.

137 Riehl JL, Brown WJ. Acute cerebellar syndrome secondary to 5-fluorouracil therapy. Neurology (Minn) 14:254. 1964.

138 Barnet MJ, Richards MA, Ganesan TS et al. Central nervous system toxicity of high-dose cytosine arabinoside. Semin Oncol 2, Suppl 31227-232, 1985.

139 Herzig RH, Lazarus HM, Herzig GP et al. Central nervous system toxicity with cytosin arabinoside. Semin Oncol 2. Suppl 3:233-236. 1985.

140 Rodriguez V, Bodey GP, Freireich EJ et al. Reduction of ifosfamide toxicity using dose fractionation. Cancer Res 36:2945-2948, 1976.

141 Brade WP, Herdrich K, Varini M. Ifosfamide-pharmacology, safety and therapeutic potential. Cancer Treat Rev 12:1-47, 1985.

142 Goren MP, Wright RK, Pratt CB et al. Potentiation of ifosfamide neurotoxicity, hematotoxicity and tubular nephrotox- icity by prior cis-diamminedichloro-platinum (II) therapy. Cancer Res 47:1457-1460, 1987.

143 Pratt CB, Green AA, Horowitz ME et al. Central nervous system toxicity following the treatment of pediatric patients with ifosfamide-mesna. J Clin Oncol 4,8:1253-1261, 1986.

144 Hahn DA and Black C. Hexamethylmelamine. A review. Drug Intel Clin Pharm 14591-598, 1980.

145 Minutes, Phase I working group meeting, National Cancer In- stitute, Bethesda, MD, November. 1984;19-20.

146 Minutes, Phase I working group meeting, National Cancer In- stitute, Bethesda, MD, March. 1985;20-21.

147 Rowinsky EK, Cazenave L, Donehower RC. Taxol. A novel in- vestigational antimicrotubule agent. J Nat1 Cancer Inst 82:1247-1259, 1990.

148 Holmes FA, Walters RS, Theriault RL et al. Phase II trial of tax- 01, an active drug in the treatment of metastatic breast cancer. J Nat1 Cancer Inst 83: 1797- 1805, 1991.

149 Rowinsky EK, Gilbert MR, McGuire WP et al. Sequences of tax- 01 and cisplatin. A phase I and pharmacologic study. J Clin On- co1 9,9:1692-1703, 1991.

150 Lipton RB, Apfel SC, Dutcher JP et al. Taxol produces a predominantly sensory neuropathy. Neurology 39:368-373. 1989.

151 Von Hoff DD. Phase I clinical trials with fludarabine phosphate. Semin Oncol 17.5:33-38. 1990.

152 Grever M. Leiby J, Kraut E et al. A comprehensive phase I and II clinical investigation of fludarabine phosphate. Semin Oncol 17.5:39-48, 1990.

153 Hood MA, Finley RS. Fludarabine. A review. DICP Ann Phar- macother 25:518-524, 1991.

154 Achterrath W, Niederle N, Raettig R et al. Etoposide chemistry, preclinical and clinical pharmacology. Cancer Treat Rev. 9 (suppl A):3-13, 1982.

155 Falkson G, Van Dyke JJ, Van Eden EB et al. A clinical trial of the oral form of 4-demethyl-epipodophyllotoxin-b-ethylidene glucoside (NSC-141540) (VP-16-213). Cancer 35:1141-l 144. 1975.

156 Littlewood TJ, Bentley DP, McQueen INF. High dose etopside does not cause peripheral neuropathy. Cancer Chemother Phar- ,,E+COl. 19:180-181, 1987.

157 Steel TH, Serpick AA, Block JB. Anti-diuretic response to

Page 15: Neurotoxicity of antineoplastic agents

75

cyclophosphamide in man. J Pharm Exp Therap 185245-253, 1973.

158 DeFronzo RA, Braine H, Colvin OM et al. Water intoxication in man after cyclophosphamide therapy. Time course and rela- tion to drug activation. Ann Intern Med 78:861-869, 1973.

159 Harlow PJ, DeClerck YA, Shore NA et al. A fatal case of inap- propriate ADH secretion induced by cyclophosphamide therapy. Cancer 44:896-898, 1979.

160 LaRocca RV, Cooper MR, Uhrich M et al. Use of suramin in the treatment of prostatic carcinoma refractory to conventional hor- monal manipulation. Urol Clin North Am 18:123-129, 1991.

161 La Rocca RV, Stein CA, Danesi R et al. A pilot study of suramin in the treatment of metastatic renal cell carcinoma. Cancer 67:1509-1513, 1991.

162 Stein CA, LaRocca RV, Thomas R et al. Suramin. An anticancer drug with a unique mechnism of action. J Chn Oncol7:499-508, 1989.

163 Pazdur R. Redman BG, Corbett T et al. Phase I trial of spiromustine (NSC 172112) and evaluation of toxicity and schedule in a murine model. Cancer Res 47:4213-4217, 1987.

164 Isaacs H and Lindemann J. Virus interference 1. The interferon. Proc R Sot Lond. (Biol) 147:257-262, 1957.

165 Goldstein D and Las.210 J. Interferon therapy in cancer. From imaginon to interferon. Cancer Res 46:4315-4329, 1986.

166 Smedley H, Katrak M, Sikora K et al. Neurological effects of recombinant human interferon. Br Med J 286:262-264, 1983.

167 Cantell K, Mattson K, Niiranen A et al. Neurotoxicity of IFN. Read before the Third International Congress of Interferon Research, Miami, 1982, Nov. l-3.

168 Rohatiner AZS, Prior PF, Burton AC et al. Central nervous of system toxicity of interferon. Br J Cancer 47:419-422, 1983.

169 Adams F, Quesada JR. Neuropsychiatric manifestations of human leukocyte interfon therapy in patients with cancer. JAMA 252,7:938-941, 1984.

170 Volberding PA, Mitsuyasu RT, Golando JP et al. Treatment of Kaposi’s sarcoma with interferon alfa-2b (IntronRA). Cancer 59:620-625, 1987.

171 Quesada JR, Talpaz M, Rios A et al. Clinical toxicity of in- terferons in cancer patients. A review. J Clin Oncol 4:234-243, 1986.

172 Mattson K, Niiranen A, Iivanainen M et al. Neurotoxicity of in- terferon. Cancer Treat Rep 67:958-961, 1983.

173 Nethersell A, Smedley H, Katrak M et al. Recombinant in- terferon in advanced breast cancer. Br J Cancer 49:615-620, 1984.

174 Kirkwood JM, Ernstoff MS, Davis CA et al. Comparison of in- tramuscular and intravenous recombinant alpha-2 interferon in melanoma and other cancers. Ann Intern Med 103:32-36, 1985.

175 Creagan ET, Ahmann DL, Green SJ et al. Phase II study of recombinant leukocyte A interferon (rIfN alpha-A) in disseminated malignant melanoma. Cancer 54:2844-2849, 1984.

176 Eggermont AM, Weimar W, Marquet RL et al. Phase II trial of high-dose recombinant leukocyte alpha-2 interferon for metastatic colorectal cancer without previous systemic treatment. Cancer Treat Rep 69:185-187, 1985.

177 Calvert MC, Gresser I. Interferon enhances the excitability of cultured neurons. Nature 178:558-560, 1979.

178 Blalock JE, Smith EM. Human leukocyte interferon. potent endorphin-like opioid activity. Biochem Biophys Res Commun 101:472-478, 1981.

179 Gutterman JU. Fein S, Quesada J et al. Recombinant leukocyte A interferon. Pharmacokinetics, single-dose tolerance, and biologic effects in cancer patients. Ann Intern Med 96:549, 1982.

180 Quesada JR, Gutterman JU. Clinical study of recombinant DNA produced leukocyte interferon in an intermittent schedule in cancer patients. JNCI 70:1041-1046, 1983.

181 Bernsen PLJA, Wong-Chung RD, Janssen JT. Neurologic amyotrophy and polyradiculopathy during interferon therapy. Lancet i:50. 1985.

182 Denicoff KD, Rubinow DR, Papa MZ et al. The Neurop- sychiatric effects of treatment with interleukin-2 and lymphokine-activated killer cells. Ann Intern Med 107:293-300, 1987.

183 Fisher RI, Coltman CA Jr, Doroshow JH et al. Metastatic renal cancer treated with interleukin-2 and lymphokine-activated clinical trial. Ann Intern Med 108:518-523, 1988.

184 Rosenberg SA, Lotze MT, Muul LM et al. A progress report on the treatment of 157 patients wtih advanced cancer using lym- phokine -activated killer cells and interleukin-2 or high-dose interleukin-2 alone. N Engl J Med 316:889-97, 1987.

185 Thompson JA, Lee DJ, Lindgren CG et al. Influence of dose and duration of infusion of interleukin-2 on toxicity and im- munomodulation. J Clin Oncol 6:669-678, 1988.

186 King KL and Boder GB. Correlation of the clinical neurotoxicity of the vinca alkaloids vincristine, vinblastine, and vindesine with their effects on cultured rat midbrain cells. Cancer Chemother Pharmacol 2:239-242, 1979.

187 Whatley SA and Hill BT. In vitro neurotoxic assessment of an- titumor drugs. Cancer Letter 20:75-82, 1983.

188 Apfel SC, Lipton RB, Arezzo JC et al. Nerve growth factor prevents toxic neuropathy in mice. Ann Neural 29:87-89, 1991.

189 Miller AB, Hoogstraten B, Staquet M et al. Reporting results of cancer treatment. Cancer 47:207-214, 198 I

190 Castellanos AM and Fields WS. Grading of neurotoxicity in cancer therapy (correspondence). J Clin Oncol 4: 1277-1278, 1986.