Search This Blog

 


Ifosfamide

Ifosfamide is associated with an encephalopathy thought to result from one of its

metabolites, chloroacetaldehyde. The incidence ranges from 10% to 20%; it presents

hours to days after initiation of treatment with confusion and disorientation and is

generally self-limiting. Methylene blue, albumin, and thiamine have been used for

both prevention and treatment. Conclusive evidence to promote routine prophylaxis

is not available.

158 Reported risk factors for this complication include a history of

ifosfamide-induced encephalopathy, prior cisplatin exposure, concomitant opioids,

concomitant CYP2B6 inhibitors, renal dysfunction, low serum albumin, increased

hemoglobin, and abdominal disease.

159,160

CASE 94-6, QUESTION 2: What is the most likely medication causing A.L.’s numbness?

Peripheral Neuropathy

Paresthesia (numbness and tingling) involving the feet and/or hands is an early

subjective symptom of vincristine neurotoxicity, which often appears within the first

days to weeks of therapy. Because A.L. received vincristine on days 1 and 8, it is

reasonable to assume that her presentation of numbness in her extremities is

secondary to her vincristine. This peripheral nerve toxicity commonly is bilateral and

symmetric and is often referred to as a “stocking-glove” neuropathy. Symptoms

initially consist of paresthesias, loss of ankle jerks, and depression of deep tendon

reflexes. Areflexia (absent reflexes) typically occurs in about 50% to 70% of

patients treated with a cumulative dose greater than 6 to 8 mg. Although older

patients appear to be more susceptible to paresthesias than younger ones, almost all

complain of paresthesias after combination chemotherapy that incorporates

vincristine or vinblastine. Pain and temperature sensory loss are usually more

pronounced than vibration and proprioception sensory loss. Patients also may display

motor weakness with a foot drop or muscle atrophy. Motor weakness, which can

become the most disabling symptom associated with vincristine neurotoxicity, can

occasionally cause muscle wasting. Although some patients exhibit muscle atrophy,

true muscle weakness seldom occurs after treatment with vincristine. Stumbling and

falling that can occur with this peripheral neuropathy is not usually caused by muscle

weakness; instead, it occurs in the dark when patients lose proprioception because

they lack visual orientation. These complications are either partially or completely

reversible, but recovery often takes several months.

161

Other agents that often share the peripheral nerve toxicity of vincristine include

vinblastine, vinorelbine, cisplatin, etoposide, oxaliplatin, paclitaxel, docetaxel,

cabazitaxel, ixabepilone, bortezomib, thalidomide, and lenalidomide among others.

147

Unlike the vinca alkaloids, most of these agents cause numbness only and not a loss

of reflexes, or weakness. Patients may report sensory loss and pain, however. The

incidence may be related to cumulative doses as well as individual risk factors such

as history of diabetic neuropathy.

162–164 Many preventive strategies have been

evaluated including amifostine, glutamine, glutathione, vitamin E, and others, but

many of the studies are limited by small sample sizes and are lacking placebocontrolled randomized designs.

165 The serotonin–norepinephrine reuptake inhibitor

(SNRI), venlafaxine, was evaluated for the prevention of neuropathy in a

randomized, double-blind, placebo-controlled Phase III trial of 48 patients receiving

an oxaliplatin-based regimen. The primary endpoint was the percentage of patients

with no acute neurotoxicity, which was significantly higher in the venlafaxine-treated

group compared with patients receiving placebo (31.3% vs. 5.3%, respectively; p =

0.03). Due to the small study population and ongoing concerns about compromise of

chemotherapy efficacy, this preventive strategy is not routine practice. Treatment

strategies are only palliative and include adjuvant pain medications such as tricyclic

antidepressants, anticonvulsants (pregabalin and gabapentin), and topical agents.

Peripheral neuropathy is usually reversible, although resolution may take months.

Several reviews provide detailed references for this information.

165–167

Oxaliplatin causes peripheral neuropathies that differ from other anticancer agents.

Oxaliplatin-induced neurotoxicity manifests as an acute neurosensory complex as

well as a cumulative sensory neuropathy. Hyperexcitability of peripheral nerves

causes an 85% to 95% incidence of paresthesia and dysesthesias of the hands, feet,

and the perioral region. Laryngeal dysesthesias have been described as well. These

effects are precipitated by exposure to cold. The cumulative dose-limiting chronic

neuropathy is described as a sensory neuropathy that is reversible several months

after completion of therapy. Dose modifications for patients with persistent

neurotoxicities have been developed and typically involves delaying therapy until

their condition improves.

168,169 Prevention of these toxicities with infusions of

magnesium and calcium has been evaluated in a prospective, randomized doubleblind study of patients (n = 102) with colon cancer receiving adjuvant therapy with

oxaliplatin, fluorouracil, and leucovorin. Patients received either calcium gluconate

1 g IV and magnesium sulfate 1 g IV 15 minutes before and immediately after

completion of

p. 1986

p. 1987

the oxaliplatin administration or placebo infusions. Calcium and magnesium

infusions reduced the incidence of grade 2 of greater sensory neurotoxicity

significantly over placebo (22% vs. 41%, respectively).

170 An additional Phase III

randomized trial of 353 patients randomly assigned to receive calcium and

magnesium pre-and postoxaliplatin showed no statistically significant differences in

the incidence of peripheral neuropathy compared to placebo.

171 Decreased efficacy

of this anticancer regimen has been reported with use of the calcium and magnesium

infusions.

172 Therefore, the use of calcium and magnesium infusions remains

controversial.

CASE 94-6, QUESTION 3: What is the significance of A.L.’s lid lag?

Cranial Nerve Toxicity

Cranial nerve toxicity occurs in 1% to 10% of patients receiving vinca alkaloids, and

most patients present with ptosis or ophthalmoplegia,

173,174 probably related to

damage to the third cranial nerve. Toxicity to other cranial nerves can cause

trigeminal neuralgia, facial palsy, depressed corneal reflexes, and vocal cord

paralysis—and may occur in the first few days to weeks after administration.

175 Other

nerve toxicities associated with the vinca alkaloids include jaw pain, which can

occur as early as after the first or second injection

176

; the pain usually resolves

spontaneously and does not recur with subsequent doses. Several of the cranial nerve

toxicities, especially with vincristine, may be dose-limiting because evidence shows

an increased prevalence with increasing doses. A.L.’s eyelid lag probably is caused

by vincristine.

Ifosfamide, vinblastine, and cisplatin have been reported to cause cranial

neuropathies. Intra-arterial administration of chemotherapy agents such as carmustine

may increase the risk of encephalopathy and cranial neuropathies.

Ototoxicity, characterized by a progressive, high-frequency, sensorineural hearing

loss, commonly occurs with cisplatin,

177,178 most likely as a result of a direct toxic

effect on the cochlea. Ototoxicity occurs more frequently at higher dosages, worsens

with concurrent cranial radiation therapy, and appears to be more pronounced in

children. The reversibility of cisplatin ototoxicity is questionable. At some centers,

routine audiometric tests are performed in patients receiving cisplatin; as a result,

these centers have a greater percentage of patients with documented decreases in

audio acuity than others. Early cessation of cisplatin may result in greater hearing

improvement. Although ototoxicity appears to be a major toxicity associated with

cisplatin, it has been reported in patients receiving carboplatin.

178

If ototoxicity is

suspected, a hearing test should be performed and therapy discontinued if alternate

treatments are available.

Autonomic Neuropathy

CASE 94-6, QUESTION 4: What is the cause of A.L.’s constipation, and how might this problem have been

prevented?

Vincristine, as well as vinblastine, commonly causes an autonomic neuropathy. The

earliest symptoms (colicky abdominal pain with or without constipation) are

reported by one-third to one-half of patients receiving these agents.

147,173 Because

severe constipation can progress to or include adynamic ileus, prophylactic laxatives

are recommended on a regular basis for patients receiving vincristine and

vinblastine. Stimulant laxatives such as the senna derivatives or bisacodyl are

believed to be the most effective agents, and stool softeners also may be used

concurrently. No compelling evidence suggests, however, that laxatives prevent

constipation. Other less frequent manifestations of autonomic dysfunction associated

with vinca alkaloids include bladder atony with urinary retention, impotence, and

orthostatic hypotension.

179,180 Patients should be monitored carefully for these signs or

symptoms and receive appropriate management after diagnosis.

CARDIOTOXICITY

Cardiomyopathy

Doxorubicin

CASE 94-7

QUESTION 1: D.A., a 35-year-old man with stage IV Hodgkin lymphoma, is receiving ABVD (doxorubicin

25 mg/m

2

IV days 1, 15, bleomycin 10 units/m

2

IV on days 1, 15, vinblastine 6 mg/m

2

IV on days 1, 15, and

dacarbazine 375 mg/m

2

IV on days 1 and 15) and concurrent radiation therapy to a large mediastinal mass. He

comes to the clinic to receive his fifth cycle of ABVD and complains of tachycardia, SOB, and a nonproductive

cough. Physical examination reveals neck vein distension, pulmonary rales, and ankle edema. Past medical

history is significant for controlled hypertension. What is the most likely cause of D.A.’s current symptoms?

D.A. is experiencing symptoms of congestive heart failure (CHF) most likely

caused by doxorubicin therapy. Doxorubicin, an anthracycline, can cause a dosedependent cardiomyopathy that generally occurs with repeated administration.

Doxorubicin causes myocyte damage by a mechanism that differs from its cytotoxic

effect on tumor cells. Because myocytes stop dividing in infancy, they presumably

would not be affected by an agent whose cytotoxicity relies on actively cycling cells.

Many mechanisms have been proposed to explain the cardiac toxicity associated with

anthracyclines including the formation of reactive oxygen species.

181–184 The

association of anthracycline-induced cardiotoxicity with other agents administered

concomitantly, monitoring techniques, and therapies to prevent and treat this

condition have been reviewed.

181,185,186

D.A.’s presentation is fairly typical of doxorubicin-induced cardiomyopathy,

although he has no significant risk factors usually associated with CHF. The total

cumulative dose of doxorubicin is the most clearly established risk factor for CHF.

187

Patients, such as D.A., who are receiving bolus doses of doxorubicin at the standard

3-week interval face little risk of CHF until a total dose of 450 to 550 mg/m2 has

been reached. After a patient has received a total dose greater than 550 mg/m2

, the

risk of CHF rises rapidly. Patients receiving less than 550 mg/m2 of doxorubicin face

a 0.1% to 1.2% risk of experiencing CHF. Comparatively, patients receiving greater

than 550 mg/m2

face a risk that rises more or less linearly; the probability of CHF in

patients receiving a total dose of 1,000/m2 may be nearly 50%.

187

Other factors that could increase D.A.’s risk of experiencing doxorubicin

cardiomyopathy include mediastinal radiation therapy, preexisting cardiac disease,

and hypertension. Young children, as well as older patients, are likely to experience

CHF at a lower cumulative dose. Concurrent chemotherapy agents (e.g.,

cyclophosphamide, etoposide, mitomycin, melphalan, trastuzumab, paclitaxel,

vincristine, bleomycin) may also potentiate doxorubicin cardiac toxicity.

181,185 When

patients receive paclitaxel and doxorubicin, the risk of cardiac toxicity appears to be

related to the sequence and proximity of the infusions. In a pharmacokinetic study,

paclitaxel increased the AUC of doxorubicin and its active metabolite,

doxorubicinol, when paclitaxel administration immediately preceded doxorubicin.

Therefore, doxorubicin should be given at least 30 minutes before paclitaxel. The

relationship between risk factors and the total cumulative dose of doxorubicin is

sufficiently strong to warrant guidelines restricting the total cumulative dose of

doxorubicin to 450 mg/m2

in patients with one or more identified risk factors

including mediastinal radiation, elderly age, and preexisting cardiovascular

p. 1987

p. 1988

disease (high-risk patients) and to 550 mg/m2

in patients without any of these

aforementioned risk factors (low-risk patients).

It is unusual that D.A., a 35-year-old man who has received a cumulative dose of

only 200 mg/m2 of doxorubicin, would be presenting with symptoms of CHF.

Mediastinal radiation therapy, or an undiagnosed cardiac disease may, however,

have contributed to this event. In addition, Hodgkin lymphoma involving the

myocardium may be responsible for this presentation.

Cardiac Monitoring

CASE 94-7, QUESTION 2: Should D.A. receive routine cardiac monitoring while he is receiving

doxorubicin?

Doxorubicin

Prevention of cardiomyopathy is achieved primarily by limiting the total cumulative

dose. Limiting the total dose, however, cannot entirely prevent the cardiomyopathy

for two reasons. First, individual tolerance to doxorubicin varies such that

cardiotoxicity may occur before the arbitrary dose limit; second, some clinical

situations warrant exceeding the dose limit to achieve positive chemotherapeutic

outcomes.

Early efforts to prevent cardiomyopathy focused on monitoring systolic time

intervals, QRS voltage loss, or ST-T segment changes on an electrocardiogram.

These changes were too nonspecific or occurred too late to be useful; however,

serial echocardiography (ECHO) has been useful. Current monitoring for

anthracycline cardiomyopathy includes assessment of a patient’s left ventricular

ejection fraction (LVEF), which is a measure of the heart’s systolic function by

ECHO, radionuclide cardiac angiography (multiple gated acquisition [MUGA]), or

endomyocardial biopsy. The use of MUGA for early detection of doxorubicininduced cardiac dysfunction has been investigated extensively.

188 A MUGA can

accurately detect functional cardiac status, but it is not particularly sensitive in

detecting patients who have early myocyte damage. Augmenting the MUGA with

exercise appears to give a more accurate picture of functional cardiac reserve.

Because myocyte damage usually occurs days to weeks after treatment with

doxorubicin, the MUGA should be obtained just before, rather than just after, a

course of the agent. Although guidelines vary, most suggest regular cardiac function

assessment by evaluation of LVEF by either ECHO or MUGA.

185

D.A. should have received a baseline assessment of his LVEF either by ECHO or

MUGA before his first cycle of ABVD. During courses of therapy, LVEF monitoring

for D.A. would not have been routinely recommended unless he was approaching his

lifetime cumulative dose or there were clinical signs or symptoms of CHF. Because

D.A. presented before his fifth cycle of ABVD with symptoms of CHF, another

ECHO should be performed, and his doxorubicin should be discontinued.

Additional assessments should be obtained when a patient shows signs or

symptoms of CHF or when low-risk patients receive cumulative doxorubicin doses

greater than 450 mg/m2 or high-risk patients receive greater than 350 mg/m2

, if

additional doses are planned. Most guidelines recommend stopping doxorubicin or

obtaining an endomyocardial biopsy when there is an absolute decrease in the LVEF

of greater than 10% to 20%, the LVEF is less than 40%, or the LVEF fails to increase

greater than 5% with exercise. Endomyocardial biopsies, along with a quantitative

assessment of morphologic changes, provide the most specific evaluation of

myocardial damage induced by anthracyclines. Progressive myocardial pathology is

graded on a scale (the Billingham score) of 0 (no change from normal) to 3 (diffuse

cell damage in >35% of total number of cells with marked change in cardiac

ultrastructure).

189 Abnormal MUGA findings and the appearance of signs and

symptoms of CHF correlate with biopsy scores. Usually, a significant change in

cardiac function is not seen with scores less than 2 to 2.5. Several investigators have

evaluated the predictive value of this technique. With a score of 2, a patient has less

than a 10% chance of experiencing heart failure if 100 mg more of doxorubicin is

given.

190 The most significant risk associated with endomyocardial biopsy is

perforation of the right ventricle with associated tamponade; this occurs rarely and

depends largely on the experience of the individual performing the biopsy.

Other Anthracyclines

Daunorubicin differs structurally from doxorubicin only by hydroxylation of the

fourteenth carbon. Cardiac toxicities are similar for both drugs, although somewhat

higher cumulative doses of daunorubicin are typically tolerated.

191 Although

idarubicin appears less cardiotoxic than doxorubicin in animal models and

daunorubicin in some early clinical trials, other studies show equivalent

myelosuppressive doses can cause cardiotoxicity comparable to that of doxorubicin

and daunorubicin.

192–194 Epirubicin also has an incidence of demonstrated CHF.

195

For all of the agents in the anthracycline class, risk factors for CHF appear to be the

same, and similar assessments should be undertaken to monitor for cardiotoxicity.

Mitoxantrone is an anthracenedione that is structurally similar to the anthracyclines.

Guidelines for monitoring doxorubicin-induced cardiotoxicity should also be

followed with mitoxantrone therapy to minimize the risk for CHF.

185

Prevention

CASE 94-7, QUESTION 3: Could D.A.’s CHF be prevented by the use of a different dose or dosing

schedule or by an agent that protects the myocardium?

Altering the dose schedule of doxorubicin to more frequent, smaller doses while

maintaining dose intensity has consistently resulted in reduction of cardiotoxicity

without obvious compromise of antitumor effects.

196–200 Several reports suggest that

peak plasma levels, as well as cumulative dose, have an important relationship to

doxorubicin cardiotoxicity. Low doses of doxorubicin administered weekly or

prolonged continuous IV infusions (48–96 hours) can be relatively cardiac sparing,

allowing higher cumulative doses to be administered. In a retrospective, uncontrolled

study of 1,000 patients receiving weekly doxorubicin, a total dose of 900 to 1,200

mg/m2 of doxorubicin given in weekly fractions had the equivalent cardiotoxicity of

550 mg/m2 given in every-3-week fractions.

197 Although well-designed studies

comparing cardiac toxicity after bolus doses with fractionated therapy or continuous

infusion are lacking, treatment that incorporates these alternative schedules should be

considered in patients with preexisting risk factors who will be receiving doses

greater than 450 mg/m2 or in patients without risk factors who will be receiving

doses greater than 550 mg/m2

. In patients with preexisting CHF or those who have

exhibited CHF, a continuous-infusion schedule of anthracyclines rather than bolus

doses may be considered. The concurrent use of drugs that might minimize the risk of

cardiotoxicity without compromising efficacy can be considered as well.

Dexrazoxane is a chemoprotectant that reduces the incidence and severity of

cardiomyopathy. It is indicated in women with metastatic breast cancer who have

received a cumulative doxorubicin dose of 300 mg/m2

. The recommended dosing

ratio of dexrazoxane to doxorubicin is 10:1 slow IV push 30 minutes before starting

doxorubicin. Currently, the ASCO guidelines do not support the routine use of

dexrazoxane in patients unless a

p. 1988

p. 1989

plan exists to continue doxorubicin beyond a total cumulative dose greater than

300 mg/m2

.

44 Clinical trials have evaluated the benefits of dexrazoxane in children

and patients receiving other anthracyclines. A meta-analysis including 10 trials with

a total of 1,619 patients evaluated the use of dexrazoxane in anthracycline therapy

and observed a decreased risk of clinical HF (relative risk 0.18, CI 0.1–0.32, p <

0.001), but there was no effect on overall survival.

201 Despite data suggesting

cardioprotection with the use of dexrazoxane, it is not used routinely. Concerns exist

about a possible decrease in efficacy of anthracyclines with its use as well as the

potential for an increase in secondary leukemias. In the meta-analysis, there was no

difference in tumor response rate reported and toxicities attributed to dexrazoxane

only included an increased frequency of risk of neutropenia that resolved with count

recovery.

201

To reduce cardiotoxicity, doxorubicin that is encapsulated in liposomes can be

given instead. A Phase III trial of women (n = 509) with metastatic breast cancer

showed that efficacy with liposomal pegylated doxorubicin may be similar to

conventional doxorubicin with decreased cardiotoxicity.

202 A review and metaanalysis of 55 randomized control trials in patients receiving anthracyclines showed

that the risk of cardiotoxicity was significantly decreased with liposomal

doxorubicin versus conventional doxorubicin (odds ratio, 0.18; 95% confidence

interval, 0.08–0.38).

203 The majority of patients were women with advanced breast

cancer. Despite reduced cardiotoxicity, liposomal doxorubicin has not replaced

standard doxorubicin in current treatment regimens secondary to high cost and lack of

evidence showing equivalency. An established equivalent dose of liposomal

preparations to conventional doxorubicin is not confirmed and is variable depending

on the disease state and regimen.

D.A.’s CHF may have been prevented with continuous-infusion doxorubicin or the

use of dexrazoxane; however, because he had not approached a cumulative dose that

warranted alternative strategies, this would not have been part of the standard

management plan for a patient receiving their first several cycles of ABVD.

Management

CASE 94-7, QUESTION 4: How should D.A.’s doxorubicin-induced CHF be managed clinically?

Anthracycline-induced CHF presents similarly to other forms of biventricular CHF

and occurs between 0 and 231 days after the last dose of doxorubicin (mean, 33

days). Anthracycline-induced CHF should be treated with a similar approach to

cardiomyopathy induced by other means. Often these measures are ineffective. The

clinical course varies, with some patients showing stable disease and others showing

improvement. Before cardiotoxicity was a widely recognized toxicity, the course of

anthracycline-induced CHF was characterized by a rapid progression that generally

led to death in a few weeks. The clinical outcome is better now, likely because

anthracycline therapy is promptly discontinued after initial presentation and there are

better treatments for CHF. These include the use of spironolactone, β-blockers,

angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers,

and diuretics, which have decreased morbidity and mortality in non-anthracyclineinduced CHF. Enalapril was evaluated to determine whether it would prevent

cardiac function decline in a randomized, double-blind, placebo-controlled study of

pediatric cancer patients who were at least 2 years out from treatment with

anthracyclines and had evidence of CHF. Patients received enalapril at 0.05

mg/kg/day and this dose was progressively escalated to 0.10 mg/kg/day, and finally

0.15 mg/kg/day if there were no side effects. Although enalapril did not increase

exercise intolerance, it did increase left ventricular end-systolic wall stress in the

first year of treatment. Side effects included dizziness, hypotension, and fatigue.

204,205

An additional trial evaluated 201 patients with anthracycline-induced

cardiomyopathy. Enalapril and carvedilol were initiated as tolerated as soon as

LVEF impairment was observed. Complete resolution of CHF was observed in 85

(42%) of patients, and an additional 26 (13%) demonstrated a partial response.

Patients who had heart failure therapy initiated closer to the time of observation of

their LVEF impairment had better response. No responses were observed in patients

who had heart failure therapy initiated greater than 6 months from the end of their

anthracycline regimen.

206 D.A.’s cardiomyopathy should be managed conservatively

with an ACE inhibitor such as enalapril and fluid restriction with the addition of

diuretics as necessary.

183

Trastuzumab

In addition to anthracyclines, trastuzumab has also been associated with increased

cardiotoxicity, likely through a different mechanism. Signs and symptoms of CHF

(e.g., dyspnea, increased cough, peripheral edema, S3 gallop, and reduced ejection

fraction) have been reported in 3% to 7% of patients receiving trastuzumab singleagent therapy. Of these patients, 5% had New York Heart Association (NYHA) class

III or IV heart failure. The use of trastuzumab in combination with chemotherapy in

patients (n = 469) with metastatic breast cancer was associated with a 27%

incidence of cardiotoxicity compared with an overall 8% incidence in the

anthracycline-alone arm. In these same patients, the incidence of NYHA class III or

IV heart failure was 16% in the trastuzumab and chemotherapy arm versus 3% in

patients receiving anthracyclines alone. Additionally, heart failure occurred in the

combination paclitaxel and trastuzumab arm with an overall incidence of 13% and

2% (classes III and IV, respectively) versus a 1% incidence in the paclitaxel-alone

arm.

207 A meta-analysis of five randomized trials of adjuvant trastuzumab also

showed a 2.5 higher risk of cardiotoxicity following trastuzumab administration.

208

The toxicity seems to be direct and not dependent on cumulative dose or treatment

duration. Trastuzumab-associated cardiac toxicity usually responds to standard

medical treatment or discontinuation of the drug.

181 Before, and periodically during

treatment with trastuzumab, patients should undergo cardiac evaluation to assess left

ventricular ejection fraction. Therapy should be discontinued if patients exhibit a

clinically significant decrease in left ventricular function.

Multitargeted Tyrosine Kinase Inhibitors

The multitargeted tyrosine kinase inhibitors exhibit a range of cardiovascular

toxicities. Because these agents are dosed chronically, toxicities may develop

relatively late in the course of therapy. Imatinib has been associated with the

development of CHF. A single-institution series reviewed all patients ( n = 1,276)

who had received imatinib within their institution. Twenty-two patients (1.2%)

exhibited CHF. Eleven patients continued imatinib with the addition of diuretics, βblockers, and ACE inhibitors. Five of those who continued therapy with imatinib had

dose reductions. The remaining eleven discontinued imatinib (3 secondary to disease

progression, 6 secondary to CHF, and 2 were deaths).

209 Patients receiving imatinib

should be monitored for symptoms and signs of heart failure.

210 Dasatinib has also

been associated with heart failure and ventricular dysfunction.

211 Sorafenib and

sunitinib have been associated with a decline in cardiac ejection fraction in 5% and

14%, respectively, in 86 patients treated for metastatic renal cell cancer.

212 A decline

in cardiac function has also been reported in a pooled analysis of 3,689 patients with

breast cancer receiving lapatinib. The incidence was 1.6% (60 patients). Twelve of

the patients had prior anthracyclines and fourteen had received prior

p. 1989

p. 1990

trastuzumab.

213 There have been additional oral oncolytic agents associated with

CHF including pazopanib and vemurafenib.

185 Patients experiencing cardiomyopathy

on targeted agents are treated similarly to those patients with anthracycline-induced

cardiomyopathy. Discontinuation of the offending agent or reduction of dose with

concurrent pharmacologic management of CHF is warranted. Two reviews of

proposed mechanisms and reported incidence of targeted therapy-induced

cardiomyopathy therapies provide a summary of current evidence.

181,214

Arrhythmias

Electrocardiographic (ECG) changes have been observed during or after treatment

with doxorubicin, other anthracyclines, cisplatin, etoposide, paclitaxel,

cyclophosphamide, mechlorethamine, and arsenic. ST-T segment changes, decreases

in voltage, T-wave flattening, and atrial and ventricular ectopy are most common.

Arrhythmias may occur in 6% to 40% of patients receiving bolus doxorubicin.

215

Paclitaxel also caused significant arrhythmias and conduction defects in Phase I and

II trials

216

; most patients experienced sinus bradycardia. Doxorubicin and paclitaxel

are used in many outpatient regimens, so this toxicity is seen frequently. Arsenic,

used in the treatment of APL, can cause QT interval prolongation and complete

atrioventricular block. Dasatinib, nilotinib, lapatinib, pazopanib, and sunitinib have

also demonstrated QT prolongation.

185 The underlying mechanisms for these

prolongations are not yet understood. Nilotinib, with a reported incidence of 1% to

10%, carries a black box warning regarding prolongation of the QT interval. The

package insert gives specific recommendations for monitoring of ECGs; baseline, 7

days after initiation, after any change of dose, and routinely thereafter.

217 QT

prolongation can lead to a torsade de pointes-type ventricular arrhythmia. Before

initiating therapy, an ECG should be performed, and serum electrolytes, including

potassium and magnesium, should be assessed and corrected. Additionally, all

medications, both anticancer and other supportive care medications that are known to

prolong the QT interval, should be discontinued.

185 Many other anticancer agents not

mentioned previously occasionally cause a rhythm disturbance, but these are limited

to a few scattered reports and should not be considered clinically significant.

Therapy should not be discontinued unless the patient experiences a serious cardiac

arrhythmia.

Hypertension

An increased incidence of hypertension has been observed in patients receiving

VEGF-targeted therapy including bevacizumab, sunitinib, sorafenib, axitinib,

regorafenib, and pazopanib among others. Bevacizumab-related hypertension may be

dose-related; it can occur at any time during therapy and is reported to have a 22% to

32% incidence. Hypertension is usually grade 3 or less and can be controlled with

antihypertensive medications.

218 An increased risk of myocardial ischemia and

infarction has been observed with the use of sorafenib in patients with metastatic

renal cell cancer in a Phase III study.

219 Patients receiving these agents should be

routinely monitored for hypertension and antihypertensives promptly initiated. In

patients with uncontrolled hypertension, anticancer therapy medication may need to

be dose-reduced or discontinued. Several consensus statements and guidelines have

been published to assist in the hypertension management of patients in VEGF

inhibitors.

220,221

Angina and Myocardial Infarction

Fluorouracil and capecitabine have been associated with angina pectoris and

myocardial infarction. In a systematic review including 30 studies, symptomatic

cardiotoxicity occurred in 0% to 20% of the patients treated with fluorouracil and in

3% to 35% with capecitabine. The most common symptom was chest pain (0%–

18.6%) followed by palpitations (0%–23.1%), dyspnea (0%–7.6%), and

hypotension (0%–6%). It appears to occur more frequently in patients receiving

multiple-day continuous infusions.

222 Direct myocyte damage is observed from

animal studies; however, human studies suggest that coronary artery spasm is the

most likely cause of angina. Because the chest pain associated with fluorouracil

responds to nitrates, this problem, theoretically, could be managed prophylactically

or therapeutically with long-acting nitrates or calcium-channel blockers.

221 Other

agents have been associated with myocardial ischemia (including, but not limited to

temsirolimus, docetaxel, paclitaxel, and imatinib) based on case reports in the

literature.

185

NEPHROTOXICITY

Cisplatin

CASE 94-8

QUESTION 1: T.J., a 58-year-old man with nonresectable head and neck cancer, is being treated with

cisplatin 100 mg/m

2

IV on day 1 and fluorouracil 1 g/m

2

/day IV on days 1 to 4. He received 1 L of normal

saline (NS) before and 1 L of NS after his cisplatin on day 1. He presents today for the third cycle of this

regimen. T.J.’s labs reveal an estimated creatinine clearance (CrCl) of 75 mL/minute, decreased from 110

mL/minute at baseline. Other abnormalities include serum magnesium of 1.2 mEq/L; all other electrolyte values

are within normal range. Is cisplatin responsible for T.J.’s decreased glomerular filtration rate (GFR) and serum

magnesium levels?

Cisplatin, a platinum heavy metal complex, is active against many solid tumors and

remains as part of first-line therapy for lung, head and neck, and testicular cancers.

The major dose-limiting toxicity of cisplatin is nephrotoxicity, and various renal and

electrolyte disorders, both acute and chronic, have been associated with cisplatin. In

the early 1970s, before the need for vigorous hydration was recognized, cisplatin

often caused acute renal failure. Today, with the use of vigorous hydration, acute

renal failure is uncommon; however, tubular dysfunction and decreased GFR remain

problematic.

Morphologic damage is greatest in the straight segment of the proximal renal

tubules where the highest concentration of platinum occurs. Acute and cumulative

renal tubular damage has been demonstrated by increased urinary excretion of

proximal tubular enzymes, such as β2

-microglobulin, alanine aminopeptidase, and Nacetyl glucosamine. Acute renal failure occurs from acute proximal tubular damage

and presents as polyuria in the first 24 to 48 hours when urine osmolality declines,

but GFR is normal. Polyuria will decline, and then 72 to 96 hours later polyuria

increases again, urine osmolality declines, and the GFR declines as well and is

persistent. Increases in proximal tubular enzymes correlate well with urinary

excretion of protein and magnesium as well as decreased reabsorption of salt and

water in the proximal tubules. T.J. has hypomagnesemia, which is the most common

electrolyte abnormality caused by cisplatin. Hypomagnesemia appears to be doserelated and may occur after a single treatment. Despite replacement with oral

magnesium, renal losses of magnesium and decreased serum magnesium levels can

persist for months or even years after completion of cisplatin therapy. Hypocalcemia,

hyponatremia, and hypokalemia occur less frequently. The cause of these electrolyte

abnormalities is thought to be similar to that of hypomagnesemia in that a proximal

tubular defect occurs that interferes with reabsorption of these electrolytes.

223,224

p. 1990

p. 1991

Chronic renal toxicity associated with cisplatin presents as a decrease in the GFR.

Published reports suggest that the GFR decreases by 12% to 25% in most patients

receiving multiple courses.

224 The decrease appears to be persistent and only

partially reversible. An increase in serum creatinine or a decrease in CrCl does not

necessarily reflect the decline in GFR. T.J. is at risk for chronic renal toxicity

because he has previously received two other cycles. The renal function of patients

receiving cisplatin therapy should be evaluated, because dosage reductions of

cisplatin may be necessary if the CrCl decreases. T.J.’s decreased GFR and low

serum magnesium likely are caused by repeated cycles of cisplatin therapy. Although

a dose reduction of cisplatin generally is not recommended for creatinine clearances

that are greater than 60 mL/minute, the clinician should provide T.J. with adequate

and aggressive hydration to prevent cisplatin nephrotoxicity. Despite preventive

methods, many patients will still experience declines in GFR as seen in T.J. In

addition, T.J. should receive an oral magnesium supplement. When large doses of

magnesium are necessary, diarrhea often limits the use of oral supplementation. IV

administration should be used if higher magnesium doses are required. Patients

should undergo frequent measurements of their electrolytes, including magnesium, to

minimize potential complications.

Prevention

CASE 94-8, QUESTION 2: What measures should be taken to prevent further cisplatin nephrotoxicity in

T.J.?

Several measures are used to minimize or prevent cisplatin- induced

nephrotoxicity, including hydration with saline and prophylactic magnesium.

Incorporation of hydration with saline and magnesium is the standard of care for all

patients receiving cisplatin. The patient should be vigorously hydrated with 2 to 3 L

of normal saline over 8 to 12 hours to maintain a urine output of 100 to 200 mL/hour

for at least 6 hours after treatment with cisplatin.

223–225 A loop diuretic (e.g.,

furosemide) may be required in elderly patients to eliminate excess sodium or in

patients with compromised cardiac reserve, but these diuretics should not be used

routinely to prevent nephrotoxicity. Additionally, mannitol (25–50 g) may be

administered just before chemotherapy to prevent cisplatin-induced renal artery

vasoconstriction, which can increase the concentration of platinum in the renal

tubules. The use of mannitol is controversial and is not given in all practice settings.

Most patients may also benefit from prophylactic magnesium supplementation.

Patients who received prophylactic magnesium 16 mEq IV daily during a 5-day

course of cisplatin followed by 60 mEq orally (20 mEq 3 times daily) between

courses experienced less nephrotoxicity compared with those who received no

supplements in a prospective trial of 16 patients with testicular carcinoma.

226 T.J.

should be encouraged to increase his oral hydration to 2 to 3 L/day for several days

after this cycle of chemotherapy. Additionally, he should take oral magnesium

supplementation between courses. For cycle 4 of his chemotherapy, he should

receive 3 to 4 L of saline the day of cisplatin administration. He may also receive 25

to 50 g of IV mannitol before cisplatin to reduce his risk of further nephrotoxicity.

Patients who experience renal dysfunction commonly have the dose of cisplatin

reduced. Guidelines to modify the dosage of cisplatin in patients with decreased

renal function are available. Most suggest a 50% dosage reduction when the GFR

decreases to 30 to 60 mL/minute and discontinuation when the GFR falls to less than

10 to 30 mL/minute. Percentage dose reductions are relative to the recommended

dose for a specific cancer in a given combination chemotherapy regimen.

227 Because

the cisplatin dose ranges from 50 to 120 mg/m2

, the precise dose for a patient with a

GFR of less than 60 mL/minute must be individualized to the situation. T.J. should

have another SCr drawn to estimate CrCl before his next cycle. As long as his CrCl

is maintained above 60 mL/minute, then no dose reductions of his cisplatin are

necessary. If a tumor type is known to be responsive to carboplatin and efficacy will

not likely be compromised, substitution of carboplatin should be considered because

it does not cause nephrotoxicity. Carboplatin is excreted primarily by the kidneys and

the dose is calculated by the Calvert equation,

228 which accounts for decreased GFR

(discussed further in Chapter 98, Lung Cancer). Therefore, patients who have

decreased renal function receive lower doses than those with normal function. Other

agents requiring dose adjustments or omission for renal dysfunction are shared in

Table 94-9.

Other Nephrotoxic Agents

Proximal Tubule Dysfunction

Other agents reported to cause renal tubular defects include lomustine, carmustine,

ifosfamide, pemetrexed, and azacytidine.

223,225 Nephrotoxicity appears to be related

to the total cumulative dose for carmustine and lomustine, but not necessarily for

ifosfamide. Renal abnormalities associated with high doses of bolus ifosfamide have

led to the use of fractionated doses and a reduced incidence of toxicity.

223 Most

patients show signs and symptoms consistent with proximal tubular dysfunction.

The primary renal lesion associated with each of these agents occurs in the

proximal renal tubule and patients experience several electrolyte imbalances, such as

loss of protein, glucose, bicarbonate, and potassium. Serum creatinine, bicarbonate,

potassium, urinary pH, protein, and glucose should be monitored closely in patients

receiving these agents. Because the reversibility of the lesions is reported to be

highly variable and a significant number of patients who exhibit severe renal toxicity

with these agents require dialysis,

223 patients should discontinue treatment with these

agents if they show any changes in serum creatinine or electrolytes.

Proteinuria

Bevacizumab, an anti-VEGF monoclonal antibody, is associated with proteinuria and

the reported incidence ranges from 21% to 46% of patients.

229 While bevacizumab is

the anti-VEGF agent most noted with this toxicity, oral tyrosine kinase inhibitors that

affect the VEGF receptor such as axitinib have also been associated with

proteinuria.

230 Mechanisms for this toxicity include microcirculatory angiogenesis

and inhibition of nitric oxide synthesis. This may lead to an increase in peripheral

resistance and endothelial dysfunction. Glomerular injury from VEGF inhibition may

also lead to renal thrombotic microangiopathy and glomerulonephritis. Severe

nephrotic syndrome has been observed in 1% to 2% of patients.

221,229,231 Patients

receiving bevacizumab should be monitored routinely for proteinuria by dipstick

urinalysis. The manufacturer recommends that patients with a 2+ or greater urine

dipstick reading undergo further assessment with a 24-hour urine collection.

Additionally, it is recommended to delay further administration of bevacizumab when

greater than 2 g of proteinuria in 24 hours is observed. Therapy may be reinitiated

when the proteinuria observed is less than 2 g in 24 hours. Proteinuria is most often

mild in patients and is usually reversible upon discontinuation of the agent. The

highest incidence of severe proteinuria requiring permanent discontinuation has been

seen in patients with metastatic renal cell carcinoma.

232

Table 94-9

Anticancer Agents Requiring Dosage Modifications or Dosage Omissions in

Renal Insufficiency

Bleomycin Lenalidomide

Capecitabine Lomustine

Carboplatin Melphalan

Carmustine Methotrexate

Cisplatin Mitomycin

Cytarabine Pemetrexed

Dacarbazine Pentostatin

Fludarabine Topotecan

Ifosfamide

Sources: Kintzel PE, Dorr RT. Anticancer drug renal toxicity and elimination: dosing guidelines for altered renal

function. Cancer Treat Rev. 1995;21:33; Launay-Vacher V et al. Prevalence of renal insufficiency in cancer

patients and implications for anticancer drug management: the renal insufficiency and anticancer medications

(IRMA) study. Cancer. 2007;110:1376; Li YF et al. Systemic anticancer therapy in gynecologic cancer patients

with renal dysfunction. Int J Gynecol Cancer. 2007;17:739.

p. 1991

p. 1992

CASE 94-9

QUESTION 1: J.R., a 15-year-old boy with osteogenic sarcoma of the right knee, is to be treated with

chemotherapy consisting of high-dose methotrexate with leucovorin rescue, doxorubicin, and cisplatin. The dose

of methotrexate is 12 g/m

2

IV administered over 4 hours. What precautions are necessary to prevent the renal

and other toxicities associated with high-dose methotrexate therapy in J.R.?

Methotrexate normally is not nephrotoxic, although 90% of the agent is excreted

unchanged in the urine; however, acute tubular obstruction can occur with high-dose

methotrexate if appropriate precautions are not taken. Acute tubular obstruction is

caused by tubular precipitation of methotrexate, which is poorly soluble at a pH less

than 7.0. To prevent this, J.R. should receive hydration and brisk diuresis to produce

urine output of 100 to 200 mL/hour for at least 24 hours after administration. A urine

pH greater than 7.0 usually can be ensured by administration of 25 to 150 mEq/L

sodium bicarbonate within the hydration fluid. J.R.’s urine output and pH must be

monitored closely to prevent acute tubular obstruction during this period.

233

In

addition, intrapatient and interpatient variability in methotrexate clearance is

considerable, particularly with high doses of methotrexate therapy. Renal excretion

of methotrexate is a complex process involving glomerular filtration, tubular

reabsorption, and secretion. Acute tubular obstruction associated with high-dose

methotrexate therapy can be prevented only by appropriate attention to optimal

urinary output before, and for at least 24 hours after, high-dose methotrexate

administration and urinary alkalization.

233

If J.R. has existing renal insufficiency, methotrexate excretion will be decreased,

leading to higher systemic exposure. As a result, myelosuppression and mucositis can

become more problematic. Leucovorin (folinic acid) is a reduced form of folic acid

given after methotrexate administration to selectively rescue normal cells from

adverse effects such as myelosuppression and mucositis. Because leucovorin is

already in reduced form, it can bypass the action of dihydrofolate reductase and not

interfere with methotrexate’s inhibition of this enzyme. Therefore, it is important that

leucovorin rescue is initiated 24 hours after the high-dose methotrexate infusion.

Blood concentrations of methotrexate obtained within 24 hours after the infusion

often are not predictive of concentrations at 48 hours. Therefore, methotrexate

concentrations between 24 and 48 hours after infusion must be monitored in J.R. and

in all patients receiving high-dose therapy. Methotrexate levels are necessary to

guide leucovorin dosing. Leucovorin rescue does not affect the renal clearance of

methotrexate. A rescue agent, glucarpidase, is indicated for the treatment of toxic

methotrexate concentrations in patients with delayed methotrexate clearance due to

impaired renal function. Glucarpidase is a recombinant bacterial carboxypeptidase

enzyme that converts methotrexate to its inactive metabolites, and provides an

alternative route for methotrexate elimination in patients with renal dysfunction and

signs or symptoms of methotrexate toxicity.

234






ion in patients with renal dysfunction and

signs or symptoms of methotrexate toxicity.

234

HEMORRHAGIC CYSTITIS

Ifosfamide

CASE 94-9, QUESTION 2: After J.R.’s surgery, it is decided to add ifosfamide to his treatment. What

bladder toxicity occurs with ifosfamide that requires attention before its administration?

Pathogenesis

Ifosfamide is a structural analog of cyclophosphamide belonging to the

oxazaphosphorine class of antitumor alkylating agents, which must be hydroxylated

and activated by the cytochrome P-450 3A4/3A5 and 2B6 enzymes in the liver. The

4-hydroxy metabolite spontaneously liberates acrolein, which is excreted in high

concentrations in the urine. Acrolein is responsible for urotoxicity causing a direct

irritation of the bladder mucosa. Both ifosfamide and cyclophosphamide can produce

cystitis, which ranges from mild-to-severe bladder damage and hemorrhage. Cystitis

is characterized by tissue edema and ulceration followed by sloughing of mucosal

epithelial cells, necrosis of smooth muscle fibers and arteries, and culminating in

focal hemorrhage.

Clinical Presentation

Patients with acrolein-induced hemorrhagic cystitis initially go through an

asymptomatic stage characterized by complaints of brief episodes of painful

urination, frequency, and hematuria. The symptoms may subside over a period of

several days or weeks after discontinuing the agent. The course of acrolein-induced

hemorrhagic cystitis usually is relatively benign, although death from massive

refractory hemorrhage has occurred.

235 The primary factors that may predispose J.R.

to hemorrhagic cystitis include the dose of ifosfamide he is receiving.

Prevention

Historically, forced hydration was the primary method used to prevent hemorrhagic

cystitis in patients treated with cyclophosphamide therapy. Theoretically, hydration

flushes the toxic acrolein metabolite out of the bladder so that insufficient contact

time is available to set up the tissue reaction. The more urotoxic agent, ifosfamide,

was introduced to the market with a uroprotective agent, mesna. This agent contains a

free thiol group, which can neutralize acrolein in the bladder. When administered in

an appropriate dosing schedule, mesna can prevent the bladder toxicity completely,

and thus use of mesna is the current standard of care.

225,235

The ASCO guidelines recommend a parenteral mesna dose of 20% of the

ifosfamide dose given at zero, 4, and 8 hours after ifosfamide (for a total mesna dose

of 60% of the ifosfamide dose).

44 The goal is to maintain prolonged mesna

concentrations within the urinary tract that are uroprotective. Repeated administration

is required because mesna has a much shorter elimination half-life (<1 hour) than

ifosfamide. If patients receive a continuous infusion of ifosfamide, a different dosing

strategy for mesna is required. To prolong mesna’s protective effects, ASCO

guidelines recommend an IV bolus mesna dose that is 20% of the ifosfamide dose,

followed by a mesna continuous infusion that is an additional equivalent of 40%

given during and for 12 to 24 hours after the end of the ifosfamide infusion.

46 This

regimen ensures that mesna remains

p. 1992

p. 1993

in the bladder for an extended amount of time after the end of the ifosfamide

infusion.

Various other mesna dosing schedules are clinically used, but no trials have

compared the different regimens. Many clinicians use a 1:1 mg dose of mesna to

ifosfamide when administered by continuous infusion. The dosing guidelines become

less well defined, however, when patients receive higher dosages of ifosfamide

(>2.5 g/m2

). The lack of data and the unique pharmacokinetic properties of

ifosfamide have caused some concerns about the current dosing guidelines. The

pharmacokinetics of ifosfamide are nonlinear. For example, the elimination half-life

associated with doses of 2.5 g/m2

is 6 to 8 hours, whereas with doses of 3.5 to 5

g/m2

, it is 14 to 16 hours. The current recommendations for mesna administration

enable protection for approximately 12 hours after an IV bolus; thus, with higher

dosages of ifosfamide, mesna should be infused beyond the recommended 8 hours

after ifosfamide to maintain bladder protection.

235 Also, concern exists that the 4-hour

dosing interval used with lower doses may be inadequate to maintain sufficient

mesna concentrations within the bladder. To ensure maximal protection against

urotoxicity, ASCO currently recommends more frequent or prolonged mesna dosage

regimens to account for its short half-life.

44

Ifosfamide and mesna are compatible in

solution; therefore, they can be infused together, offering greater patient convenience.

Because mesna works in the bladder, frequent urination may diminish its efficacy.

Patients may be reminded to try to empty their bladder every few hours. Although

forced hydration has been the mainstay for prevention of cyclophosphamide-induced

hemorrhagic cystitis, it is unnecessary and potentially disadvantageous when mesna

is used. This is because forced hydration can increase urination and thus the

evacuation of mesna from the bladder.

Mesna is usually given IV but an oral formulation is available. The oral

bioavailability of mesna is approximately 50%; therefore, patients should receive

twice the standard IV dose (e.g., oral mesna 40% of the IV ifosfamide dose) 2 hours

before and 4 and 8 hours after ifosfamide.

236 Others have recommended that an oral

dose also be given with the ifosfamide dose. Many centers administer the first dose

of mesna IV followed by oral doses at 4 and 8 hours, particularly in the outpatient

clinic setting.

44 All patients receiving cyclophosphamide should receive saline

diuresis or forced saline diuresis to protect urothelial tissue. When patients receive

cyclophosphamide for an HCT, it is at high dose; therefore, they receive mesna and

hydration. Other practices to prevent this complication include hyperhydration and

the use of continuous bladder irrigation. Data comparing these methods are

controversial and report varying rates of hematuria and severe hemorrhagic cystitis.

These recommendations are currently supported by the ASCO consensus guideline

44

(see Chapter 101, Hematopoietic Cell Transplantation). J.R. will receive mesna at a

dose of 20% of the ifosfamide dose given immediately before ifosfamide, and at 4

and 8 hours after ifosfamide (for a total mesna dose of 60% of the ifosfamide dose).

Treatment

CASE 94-9, QUESTION 3: If J.R. exhibits hemorrhagic cystitis, how should it be treated?

Once hemorrhagic cystitis develops, the agent causing the disorder must be

discontinued and vigorous hydration started. If gross hematuria occurs, a large-bore

urinary catheter should be inserted to avoid obstruction of the urethra by clots. Some

clinicians also use continuous silver nitrate irrigation, local instillation of formalin

or alum, or electrocauterization of bladder blood vessels to control bleeding. There

is no consensus as to which of these methods is superior. If these measures fail,

surgical intervention may be necessary to divert urine flow away from the

bladder.

235,237

PULMONARY TOXICITIES

Bleomycin and Other Agents

CASE 94-10

QUESTION 1: J.A., a 54-year-old man with stage III Hodgkin lymphoma, has received ABVD (doxorubicin

25 mg/m

2

IV days 1 and 15, bleomycin 10 units/m

2

IV on days 1 and 15, vinblastine 6 mg/m

2

IV on days 1 and

15, and dacarbazine 375 mg/m

2

IV on days 1 and 15) for six cycles. He presents to the clinic 6 months after his

last cycle with dyspnea, a nonproductive cough, and fever. Chest radiograph showed diffuse bilateral infiltrates;

his respiratory rate was 36 breaths/minute; and his arterial blood gases (ABG) were as follows:

pH, 7.50

PO2

, 62 mm Hg

PCO2

, 28 mm Hg

O2

saturation, 92%

What are the possible causes of his new pulmonary findings?

J.A. is at risk for several processes that could produce diffuse pulmonary

infiltrates and dyspnea. He is immunosuppressed secondary to his lymphoma and the

therapy; therefore, J.A. has an increased risk for infection and may have pneumonia.

In addition, the infiltrates may represent a relapse of his disease. Pulmonary

infiltrates also may represent toxicity from one or more of the cytotoxic agents he

received. Further diagnostic workup is necessary to establish the cause.

CASE 94-10, QUESTION 2: A bronchoscopy with bronchoalveolar lavage and a biopsy with pathologic and

microbiologic evaluations were performed. Bacterial, fungal, and viral cultures were negative, and the biopsy

revealed inflammation and fibrosis with no evidence of lymphoma. These results are highly suggestive of

chemotherapy-induced pulmonary damage. Which of the agents that J.A. received is associated with pulmonary

toxicity?

J.A. has received bleomycin and that places him at risk for pulmonary toxicity. As

part of initial workup before initiating chemotherapy with ABVD, J.A. had

pulmonary function tests, which were normal. Many chemotherapy agents have been

associated with pulmonary toxicity and the varying types of mechanisms and clinical

presentations have been reviewed (Table 94-10).

238–250 Several reviews discuss the

different types of pulmonary toxicities associated with anticancer agents.

238–240,250

Among all chemotherapy agents, bleomycin is associated with the highest

incidence of pulmonary toxicity. Although several types have been reported, the most

frequent is interstitial pneumonitis followed by pulmonary fibrosis.

238,243,251 Patients

generally present with a nonproductive cough and dyspnea. Clinicians may detect

only fine crackling bibasilar rales that often progress to coarse rales. The chest

radiograph may be normal in the early stages, but patients can exhibit bilateral

alveolar and interstitial infiltrates. Arterial blood gases show hypoxia and pulmonary

function tests generally reveal a progressive fall in the diffusing capacity without a

significant decrease in the forced vital capacity.

243,251 The most significant factor

associated with the development of pulmonary toxicity is the cumulative dose of

bleomycin. At total doses less than 400 units, fewer than 10% of patients may

experience pulmonary toxicity. When the cumulative dose reaches 450 to 500 units,

the incidence is higher. A rarer, hypersensitivity reaction produces fever,

eosinophilia, and diffuse infiltrates, and this pulmonary toxicity is not dose-related.

The mortality associated with bleomycin pulmonary toxicity is about 50%.

238,251

If

bleomycin is discontinued while symptoms are minimal and before pulmonary

function has decompensated significantly, the damage may not progress. In contrast,

patients with prominent physical and radiographic findings generally die because of

pulmonary complications. Other anticancer agents can potentially exacerbate the

pulmonary toxicity associated with bleomycin. J.A.’s pulmonary findings are most

likely suggestive of bleomycin toxicity. Unfortunately, there are no methods for

reversing the pulmonary toxicity seen with bleomycin and treatment consists of

supportive measures such as oxygen and steroids.

p. 1993

p. 1994

 


Table 94-3

Chemotherapeutic Drugs Reported to Produce Local Toxicities

Potential Vesicants

Dactinomycin Epirubicin

Daunorubicin Streptozocin

Doxorubicin Vinblastine

Idarubicin Vincristine

Mechlorethamine Paclitaxel

Mitomycin Oxaliplatin

Potential Irritants

Carmustine Etoposide

Cisplatin Mitoxantrone

Dacarbazine Melphalan

Vinorelbine Vindesine

Cyclophosphamide Teniposide

Source: Doellman D et al. Infiltration and extravasation: update on prevention and management. J Infus Nurs.

2009;32:203; Boulanger J et al. Management of the extravasation of antineoplastic agents. Support Care Cancer.

2015;23(5):1459.

p. 1979

p. 1980

CASE 94-4, QUESTION 6: What factors in C.W. increase her risk of extravasation, and what administration

techniques and precautions can minimize these risks?

Several factors have been associated with an increased risk of extravasation and

subsequent tissue damage after administration of cytotoxic chemotherapy. Risk

factors include generalized vascular disease commonly found in elderly and

debilitated patients or in patients who have undergone frequent venipuncture and

treatment with irritating chemotherapy (the latter causes venous fragility and

instability or decreased local blood flow); elevated venous pressure, which typically

occurs in patients with an obstructed superior vena cava or venous drainage after

axillary dissection; prior radiation therapy to the injection site; recent venipuncture in

the same vein; use of injection sites over joints, which increases the risk of needle

dislodgement; and others.

68,103

Tissue damage may be more severe if extravasation occurs in areas with only a

small amount of subcutaneous tissue (e.g., the back of the hand or wrist) because

wound healing is more difficult and exposure of deeper structures, such as the

tendons, is increased.

103 These risks have led to the increased use of central catheters

in patients receiving vesicant chemotherapy.

C.W. has several risks for extravasation. She had an axillary lymph node

dissection for her breast cancer, which places her at higher risk for obstructed

venous drainage. Additionally, she has had multiple venous punctures, and she is a

thin woman with relatively small amounts of subcutaneous tissue.

Extravasations of agents with vesicant properties can produce devastating tissue

damage that can potentially cause loss of an extremity or death. To prevent significant

morbidity or mortality, major emphasis must be placed on prevention. All caretakers

who administer agents with vesicant or irritant properties should be skilled in IV

drug administration and receive special instruction before administering these agents.

The patient also must be told how agent administration should feel and to report

1.

2.

3.

4.

5.

6.

7.

8.

immediately any change in sensation, including pain, burning, or itching.

CASE 94-4, QUESTION 7: C.W.’s oncology nurse believes that the doxorubicin may have extravasated

during administration. How should this be managed? Do management strategies differ for other vesicant

agents?

Immediate management of a potential vesicant extravasation should include

stopping the injection if the entire agent has not been administered. Various other

recommended measures may minimize vesicant exposure and subsequent tissue

damage (Table 94-4). These include application of cold compresses to the

extravasation site and elevation of the extremity. Cold compresses have been shown

to cause vasoconstriction, which can help to localize the extravasation and allow

time for local vessels to displace the extravasated agent, whereas warm compresses

are thought to induce vasodilation, increase drug distribution and absorption,

therefore decreasing the concentration of the offending agent around the immediate

site. Warm compresses are recommended for vinca alkaloids and

epipodophyllotoxins.

103,104 With the exception of these two classes of agents, cooling

has been shown to be more effective than warm compresses. Specific antidotes

thought to inactivate the extravasated chemotherapy have been suggested; however,

many of these antidotes are based on observations in a few patients or animal

models, and their effectiveness, in many cases, is unsubstantiated. Antidotes

recommended in some guidelines may actually worsen tissue damage (e.g., sodium

bicarbonate for doxorubicin). Recommended treatments for suspected extravasation

of vesicant agents are outlined in Table 94-5.

103,104

Table 94-4

Suggested Procedures for Management of Suspected Extravasation of Vesicant

Drugs

Stop the infusion immediately, but do not remove the needle. Any drug remaining in the tubing or needle, as

well as the infiltrated area, should be aspirated

Contact a physician as soon as possible

If deemed appropriate, instill an antidote in the infiltrated areas (via the extravasated intravenous (IV) needle if

possible)

Remove the needle

Apply ice to the site and elevate the extremity for the first 24–48 hours (if vinca or epipodophyllotoxin, use

warm compresses)

Document the drug, suspected volume extravasated, and the treatment in the patient’s medical record

Check the site frequently for 5–7 days

Consult a surgeon familiar with extravasations early so that the surgeon can periodically review the site, and, if

ulceration begins, the surgeon can rapidly assess if surgical debridement or excision is necessary

Source: Doellman D et al. Infiltration and extravasation: update on prevention and management. J Infus Nurs.

2009;32:203; Boulanger J et al. Management of the extravasation of antineoplastic agents. Support Care Cancer.

2015;23(5):1459.

Dexrazoxane has been established as a reliable antidote for anthracycline

extravasations. Dexrazoxane, an iron chelator, was studied based on evidence that it

protects cardiac tissue from anthracycline-induced toxicities. Two prospective,

multicenter, single-arm trials were conducted in a total of 54 evaluable patients with

anthracycline extravasations. In 98.2% of patients, surgical intervention was avoided

and 71% of patients were able to continue treatment regimen without any delays.

Hospitalization of 41% of patients was necessary secondary to their extravasation.

Toxicities included myelosuppression, increased liver function tests, nausea, and

pain at the dexrazoxane infusion site.

105 Based on these results, dexrazoxane is

approved for the treatment of anthracycline extravasations. Dexrazoxane is given

once daily for 3 days at a dose of 1,000 mg/m2

IV on days 1 and 2, and 500 mg/m2

IV

on day 3. Administration should start within 6 hours of extravasation.

106

HYPERSENSITIVITY REACTIONS

Almost all anticancer agents have produced at least an isolated instance of a

hypersensitivity reaction. All types of hypersensitivity reactions can occur with

anticancer agents, although type I is the most common reaction documented. Type I

hypersensitivity reactions are immediate reactions that are most often

immunologically mediated, although there are other possible mechanisms for type I

hypersensitivities. Anaphylactic or immunoglobulin E (IgE)-mediated reactions

occur when an antigen interacts with IgE bound to a mast cell membrane, causing

degranulation of mast cells. Major signs and symptoms of type I reactions include

urticaria, angioedema, rash, bronchospasm, abdominal cramping, and hypotension.

Although many reactions associated with anticancer agents probably are

immunologically mediated, other mechanisms may cause type I reactions. Those

include the degranulation of mast cells and basophils through a direct effect on the

cell surface that releases histamine and other vasoactive substances. Activation of the

alternative complement pathway can also release vasoactive substances from mast

cells. When non-IgE-mediated mechanisms account for the symptoms of a type I

reaction, it is called an anaphylactoid reaction (see Chapter 32, Drug

Hypersensitivity Reactions).

p. 1980

p. 1981

Table 94-5

Recommended Extravasation Antidotes

Class/Specific

Agents

Local/Systemic Antidote

Recommended Specific Procedure

Alkylating Agents

Cisplatin

a

Oxaliplatin

Mechlorethamine

1/6-M solution sodium thiosulfate Mix 4 mL 10% sodium thiosulfate USP with 6 mL

of sterile water for injection, USP for a 1/6-M

solution. Into site, inject 2 mL for each mg of

mechlorethamine or 100 mg of cisplatin

extravasated

Mitomycin-C Dimethylsulfoxide 99% (w/v) Apply 1–2 mL to the site every 6 hours for 14 days.

Allow to air dry; do not cover

Anthracyclines Cold compresses Apply immediately for 30–60 minutes on first day

Doxorubicin

Daunorubicin

Dexrazoxane Once daily for 3 days. First dose should be given

within the first 6 hours. Day 1: 1,000 mg/m

2

IV

Day 2: 1,000 mg/m

2

IV

Day 3: 500 mg/m

2

IV

Vinca alkaloids

Vinblastine

Vincristine

Warm compresses

Hyaluronidase

Apply immediately for 30–60 minutes, then alternate

off/on every 15 minutes for 1 day

Inject 150 units into site

Epipodophyllotoxins

a Warm compresses Apply immediately for 30–60 minutes, then alternate

off/on every 15 minutes for 1 day

Etoposide Hyaluronidase Inject 150 units into site

Taxanes Cold compresses Apply immediately for 30–60 minutes every 6 hours

for 1 day

Docetaxel Hyaluronidase Inject 150 units into site

Paclitaxel

aTreatment indicated only for large extravasations (e.g., doses one-half or more of the planned total dose for the

course of therapy).

IV, intravenous; w/v, weight per volume.

Source: Goolsby TV, Lombardo FA. Extravasation of chemotherapeutic agents: prevention and treatment. Semin

Oncol. 2006;33:139; Doellman D et al. Infiltration and extravasation: update on prevention and management. J

Infus Nurs. 2009;32:203; Totect (dexrazoxane injection) [package insert]. Rockaway NTTU, Inc.; 2011.

Many of the type I hypersensitivity reactions produced by anticancer medications

appear to be mediated by non-IgE mechanisms. Although little research has been

conducted on the mechanism of these reactions, two features suggest that they are not

mediated by IgE. First, many reactions occur during or immediately after

administration of the first dose. This is in contrast to immunologic reactions that

require prior exposure (i.e., one must be sensitized before becoming

hypersensitized). In addition, certain symptoms or symptom complexes are more

diagnostic of immunologically mediated disorders. These symptoms include

urticaria, angioedema, bronchospasm, laryngeal spasm, cytopenias, arthritis,

mucositis, vasculitic syndromes, and vesicular dermatitis. Although the spectrum of

symptoms and their severity vary widely in the case reports, most hypersensitivity

reactions that occur with anticancer agents are classified as grade 1 (transient rash,

mild) or grade 2 (mild bronchospasm, moderate) by the NCI Common Terminology

Criteria for Adverse Events.

1 Furthermore, a patient who has had a reaction to an

agent that is not immunologically mediated can safely receive future courses of

anticancer therapy if he or she receives appropriate premedication. For example,

appropriate premedication allows many (>60%) patients who have previously

experienced a hypersensitivity reaction secondary to paclitaxel to continue therapy;

this also reduces the incidence of hypersensitivity reactions associated with shortduration infusions (i.e., 3 hours). Some agents can commonly cause hypersensitivity

reactions after the first and subsequent doses of therapy.

The other types of hypersensitivity reactions are less commonly documented with

cytotoxic and targeted therapy administration. Type II is hemolytic anemia. Type III

results from deposition of antigen–antibody complexes that form intravascularly and

in tissues that can result in tissue injury. Sensitized T lymphocytes that react with

antigens causing a release of lymphokines are responsible for type IV reactions.

107

Anticancer agents most frequently reported to produce hypersensitivity reactions and

their characteristic reactions are listed in Table 94-6.

108–134 Most valuable

information stems from patient series and case reports. However, they often provide

conflicting and contradictory information, particularly with respect to incidence,

severity, characteristic symptoms, time course, and the success of rechallenge. If a

patient experiences a hypersensitivity reaction and the clinician decides to continue

therapy with this regimen, a full review of all of the relevant literature as well as

manufacturer’s data is advised. Several reviews are available to assist in this

effort.

107,135,136

Monoclonal Antibodies

CASE 94-5

QUESTION 1: S.R., a 58-year-old man with metastatic colorectal cancer, previously progressed after four

cycles of FOLFOX (oxaliplatin 85 mg/m

2

IV on day 1, leucovorin 100 mg/m

2

IV on days 1 and 2, and

fluorouracil 400 mg/m

2

IV bolus, followed by 600 mg/m

2

IV for 22 hours on days 1 and 2) plus bevacizumab 5

mg/kg. He also recently progressed after two cycles of second-line FOLFIRI (irinotecan 180 mg/m

2 on day 1,

leucovorin 100 mg/m

2

IV on days 1 and 2, and fluorouracil 400 mg/m

2

IV bolus, followed by 600 mg/m

2

IV for

22 hours on days 1 and 2). S.R. now presents to the clinic for his first weekly dose of cetuximab (400 mg/m

2

IV load, followed by 250 mg/m

2

IV weekly). Discuss the toxicities that S.R. may expect and when they might

appear. How should these side effects be managed? S.R. asks how these side effects can be prevented.

p. 1981

p. 1982

Table 94-6

Cancer Chemotherapeutic Agents Commonly Causing Hypersensitivity

Drug Frequency Risk Factors Manifestations Mechanism Comments

Asparaginase

107 10%–20% Increasing

doses; interval

(weeks to

months)

between doses;

IV

administration;

history of atopy

or allergy; use

without

Pruritus,

dyspnea,

agitation,

urticaria,

angioedema,

laryngealspasm

Type I Substitute

pegaspargase,

but up to 32%

may demonstrate

mild

hypersensitivity

prednisone,

mercaptopurine

and/or

vincristine

Paclitaxel

107,108 Up to 10%

first or

second dose

None known Rashes,

dyspnea,

bronchospasm,

hypotension

Nonspecific

release of

mediators;

Cremophor

EL

Premedicate

with

diphenhydramine

corticosteroids,

and H2

receptor

antagonists

Paclitaxel

protein-bound

particles

(Abraxane) may

be substituted

and better

tolerated in some

patients

Cisplatin

109–113 Up to 20%

intravesicular,

5%–10%

systemic;

case reports

of hemolytic

anemia

Increasing

number of

doses (typically

> dose 6)

Anemia: none

known

Rash, urticaria,

bronchospasm

Anemia:

hemolytic

anemia

Type I

Anemia: type

III

Carboplatin may

be substituted in

some cases but

cross-reactivity

has been

reported

Procarbazine

116–118 Up to 15%,

case reports

None known Urticaria

pneumonitis

Type I

Type III

All patients

rechallenged

have prompt

return of

symptoms

Anthracyclines

119–123,125 1%–15%

depending on

anthracycline

None known Dyspnea,

bronchospasm,

angioedema

Unknown;

nonspecific

release

Cross-reactivity

documented, but

incidence and

likelihood

unknown

Bleomycin

126–128 Common Lymphoma Fever (up to

42°C),

tachypnea

Endogenous

pyrogen

release

Not technically

classified as

HSR;

premedicate with

acetaminophen

and

diphenhydramine

Rituximab

129 First

treatment

80%;

subsequent

treatments

40%

Female sex,

pulmonary

infiltrates, CLL

or mantle cell

lymphoma

Fevers, chills,

occasional

nausea, urticaria,

fatigue, HA,

pain, pruritus,

bronchospasm,

SOB,

angioedema,

rhinitis, vomiting,

↓ BP, flushing

Unknown;

related to

manufacturing

process

Stop or ↓ infusion

rate by 50%;

provide

supportive care

with IV fluids,

acetaminophen,

diphenhydramine,

vasopressors

PRN

Trastuzumab

130 First None known Chills, fever, Unknown, Manage with

treatment

40%;

subsequent

treatments

rare

occasional

nausea or

vomiting; pain,

rigors, HAs,

dizziness, SOB,

↓ BP, rash,

asthenia

related to

manufacturing

process

acetaminophen,

diphenhydramine,

meperidine

p. 1982

p. 1983

Cetuximab

131 First treatment,

15%–20%;

grades 3–4, 3%;

subsequent

treatments

uncommon

None known Airway

obstruction

(bronchospasm,

stridor,

hoarseness),

urticaria,

hypotension, or

cardiac arrest

Premedicate with

diphenhydramine;

stop or decrease

infusion rate;

provide

supportive care

with epinephrine,

corticosteroids,

IV

antihistamines,

bronchodilators,

and oxygen PRN

Alemtuzumab

132 ∼90% with IV

administration in

first week

None known Hypotension,

rigors, fever,

SOB,

bronchospasm,

chills, rash

Unknown Dose titration

during several

days; substitute

with SC

administration

rather than IV;

premedicate with

acetaminophen,

diphenhydramine,

meperidine

Docetaxel

133 0.9% with

premedication

None known ↓ BP,

bronchospasm,

rash, flushing,

pruritus, SOB,

pain, fever, chills

Unknown Premedicate with

acetaminophen,

dexamethasone,

and

diphenhydramine

Doxorubicin

134

liposomal

6.8% None known Flushing, SOB,

angioedema, HA,

chills, ↓ BP

Unknown,

related liposomal

components

Stop infusion;

restart at a lower

rate

Type I:Antigen interaction with IgE bound to mast cell membrane causes degranulation. Drug binding to mast cell

surface causes degranulation. Activation of classic or alternative complement pathways produces anaphylatoxins.

Neurogenic release of vasoactive substances. Type III: Antigen–antibody complexes form intravascularly and

deposit in or on tissues.

BP, blood pressure; CLL, chronic lymphocytic leukemia; HA, headache; HSR, hypersensitivity reaction; IV,

intravenous; PRN, as needed; SC, subcutaneous; SOB, shortness of breath.

p. 1983

p. 1984

The most common toxicities observed in patients receiving cetuximab include

rash, diarrhea, hypomagnesemia, headache, nausea, and hypersensitivity reactions.

Infusion-related reactions occur in 15% to 20% of patients receiving their first

infusion. However, severe hypersensitivity reactions (including allergic and

anaphylactic reactions) occur in 1% to 3% of patients. The reactions are related to

the infusion of cetuximab and generally occur during or within 1 hour of completing

the first dose. Patients should be premedicated with diphenhydramine before the

infusion. The infusion can be stopped or the rate decreased if S.R. begins

experiencing these effects. The skin rash and dry skin occurring after cetuximab

administration are related to the inhibition of EGFR and were the most common side

effect seen in clinical trials. The rash occurred in approximately 80% of patients and

appeared in the first 1 to 3 weeks of therapy. Grade 3 or 4 skin rashes occurred in

5% to 10% of patients.

137

Several of the monoclonal antibodies (e.g., rituximab, trastuzumab, cetuximab,

ofatumumab) are associated with a higher incidence of hypersensitivity reactions than

traditional cytotoxic agents. These agents are genetically engineered humanized

monoclonal antibodies containing foreign proteins that can trigger the reaction.

During the first infusion with trastuzumab, approximately 40% of patients experience

a symptom complex, mild to moderate in severity, which consists of chills, fever, or

both. These symptoms usually do not recur with subsequent injections.

135

In

comparison, approximately 80% of patients receiving rituximab may experience an

infusion-related reaction ranging from fever, chills, and rigors to severe reactions

(7%) characterized by hypoxia, pulmonary infiltrates, adult respiratory distress

syndrome, myocardial infarction, ventricular fibrillation, or cardiogenic shock with

the first dose. Approximately 40% of patients receiving rituximab experience

infusion-related reactions with subsequent infusions (5%–10% severe).

135 Treatment

of these reactions follows the recommendations for treatment of hypersensitivity

reactions that occur with more traditional agents.

Treatment

Recommended treatment of hypersensitivity reactions is reviewed in Table 94-7. If a

patient experiences a severe type I hypersensitivity reaction to any anticancer agent,

the treatment should be stopped. If a structural analog or another agent in the same

chemical class is an effective treatment for the same cancer, subsequent therapy

should use the analog or other agent to minimize the risk of future reactions. If the

reaction is mild or moderate, the patient may continue with the same therapy if

treatment is preceded by methods to prevent or minimize hypersensitivity reactions.

General recommendations for preventing hypersensitivity reactions are found in

Table 94-7. Pretreatment with corticosteroids and diphenhydramine significantly

decreases the frequency and severity of hypersensitivity reactions; however, the

effect of H2

receptor antagonists and epinephrine remains controversial. Because the

success of these preventive measures depends on the cause of the reaction

(immunologic or anaphylactoid), the aforementioned characteristics of type I

reactions should be used to assess the underlying pathogenesis. In addition, other

chemicals present in the formulation or other agents administered concomitantly with

the chemotherapy can cause the hypersensitivity reaction. Potential allergens

included in the diluent or formulation of chemotherapy agents include Cremophor EL

(present in paclitaxel), polysorbate 80 (present in docetaxel and etoposide), benzyl

alcohol (present in the parenteral form of methotrexate, cytarabine, and etoposide),

and methoxypolyethylene glycol (present in liposomal doxorubicin). Recognizing

potential allergens can significantly affect treatment of the current reaction and

minimize the risk of future reactions.

To reduce the hypersensitivity reactions observed with paclitaxel, paclitaxel

protein-bound particles (Abraxane), an albumin-bound formulation of paclitaxel, has

been created. Because paclitaxel protein-bound particles formulation is Cremophor

EL-free and less likely to cause hypersensitivity than traditional paclitaxel, it is not

necessary to premedicate patients with steroids and antihistamines. Doses between

the two agents are not comparable. Although fewer hypersensitivity reactions are

associated with this formulation, myelosuppression remains a dose-limiting

toxicity.

108,138

Table 94-7

Prophylaxis and Treatment of Hypersensitivity Reactions from Anticancer

Drugs

Prophylaxis

IV access must be established

BP monitoring must be available

Premedication

Dexamethasone 20 mg PO and diphenhydramine 50 mg PO 12 and 6 hours before treatment, then the same

dose IV immediately before treatment

Consider addition of H2

antagonist with schedule similar to dexamethasone

Have epinephrine and diphenhydramine readily available for use in case of a reaction

Observe the patient up to 2 hours after discontinuing treatment

Treatment

Discontinue the drug (immediately if being administered IV)

Administer epinephrine 0.3 mg IM or SC minutes until reaction subsides

Administer diphenhydramine 50 mg IV

If hypotension is present that does not respond to epinephrine, administer IV fluids

If wheezing is present that does not respond to epinephrine, administer nebulized albuterolsolution

Although corticosteroids have no effect on the initial reaction, they can block late allergic symptoms. Thus,

administer methylprednisolone 125 mg (or its equivalent) IV to prevent recurrent allergic manifestations

BP, blood pressure; IV, intravenous; PO, orally.

Specific Organ Toxicities

NEUROTOXICITY

Specific Agents

CASE 94-6

QUESTION 1: A.L., a 39-year-old woman with acute lymphocytic leukemia, has been admitted to the hospital

for induction chemotherapy. Methotrexate 3 g/m

2

IV once on day 1, cytarabine 2 g/m

2

IV every 12 hours on

days 2 and 3 for four doses, vincristine 2 mg IV on days 1 and 8 for two doses, and dexamethasone 20 mg

orally daily for on days 1 through 5 are ordered. Laboratory data obtained on admission include a WBC count of

120,000 cells/μL with 9% neutrophils, 11% lymphocytes, and 80% blasts. On day 3, A.L. is confused and she

has difficulty performing a finger-to-nose neurologic examination. On day 10, she complains of numbness in her

hands and feet. In addition, the clinician notes an eyelid lag and ataxia. A.L. also complains of severe

constipation. What signs and symptoms of neurotoxicity is A.L. experiencing? Should the leukemia regimen be

modified for future courses?

p. 1984

p. 1985

Methotrexate, Cytarabine, and Vincristine

Methotrexate causes little or no neurotoxicity when administered orally or

intravenously in doses less than 1 g/m2

; however, high-dose IV methotrexate (usually

>1 g/m2

) can occasionally cause acute encephalopathy. The encephalopathy that

occurs after therapy with methotrexate is usually transient and reversible. Some

patients may experience a progressive leukoencephalopathy after high-dose IV

methotrexate. The risk of leukoencephalopathy increases with higher cumulative

doses of methotrexate and concomitant cranial radiation therapy.

139,140 Posterior

reversible encephalopathy syndrome has also been associated with high-dose

methotrexate and intrathecal methotrexate. Chemical meningitis can occur with

intrathecal administration of methotrexate and, less frequently, myelopathy or

paraplegia may be observed

141

(see Chapter 95, Pediatric Malignancies). Patients

receiving intrathecal therapy or high-dose methotrexate should be carefully

monitored for signs and symptoms associated with neurotoxicity.

High doses of cytarabine (>1 g/m2

in multiple doses) are associated with CNS

toxicity in 8% to 37% of patients.

142,143 These neurotoxicities are dose-related and

schedule-related. Doses greater than 18 g/m2 per course increase the frequency of

neurotoxicity. Older patients are more susceptible than younger patients, and the

prevalence seems higher in subsequent versus initial courses of therapy. As

illustrated by A.L., neurotoxicity may become evident within a few days after

treatment with cytarabine and, most commonly, the neurotoxicity is manifested by a

generalized encephalopathy with symptoms such as confusion, obtundation, seizures,

and coma. Cerebellar dysfunction, presenting as ataxia, gait and coordination

difficulties, and dysmetria (inability to arrest muscular movement when desired and

lack of harmonious action between muscles when executing voluntary movement), is

also commonly observed in patients receiving high-dose cytarabine therapy. These

neurologic symptoms may partially resolve over days to weeks after discontinuation

of therapy. Other neurologic toxicities reported with cytarabine include progressive

leukoencephalopathy and chemical meningitis. Intrathecal administration of

cytarabine, including the liposomal formulation, may also cause a chemical

meningitis or arachnoiditis.

141,144 Leukoencephalopathy typically presents with

progressive personality and intellectual decline, dementia, hemiparesis, and,

sometimes, seizures. These neurotoxicities also can occur after treatment with other

chemotherapy agents.

Asparaginase and pegaspargase can cause encephalopathy, which presents most

commonly as lethargy and confusion. These agents are used in acute lymphocytic

leukemia regimens. Severe cerebral dysfunction occurs occasionally, and patients

may present with stupor, coma, excessive somnolence, disorientation, hallucination,

or severe depression. Symptoms can occur early (within days of administration of

asparaginase) or late, depending on the treatment schedule.

145,146 The suspected

mechanism is the direct neurocytotoxic effect of aspartic acid, glutamic acid, and

ammonia. The neurotoxicity is usually reversible with the acute syndrome clearing

rapidly and a delayed syndrome lasting several weeks.

A.L.’s symptoms most likely are the result of CNS toxicity caused by both highdose methotrexate and cytarabine. A decision regarding further treatment with these

agents is complicated because omitting a dose or decreasing the dose of either of

these agents could compromise the likelihood of a complete remission. High-dose

cytarabine cerebellar toxicity may be irreversible. Therefore, the clinician may

decide to discontinue cytarabine in A.L.’s future regimens. Additionally,

modifications of methotrexate including dose reductions may be necessary in future

therapy for A.L.

Multiple other anticancer agents including fluorouracil, fludarabine, nelarabine,

procarbazine, and ifosfamide produce an encephalopathic toxicity (Table 94-8).

Recognition of neurotoxicity resulting from cytotoxic chemotherapy is often difficult

because of comorbid conditions such as metastatic disease and other paraneoplastic

syndromes, but it is important in assessing the need for potential dose modifications

or even discontinuation of the agent. Several reviews provide detailed explanations

of signs and symptoms, mechanisms, and potential treatments for chemotherapyinduced neurotoxicities.

145,147 When a patient presents with any signs or symptoms of

neurotoxicity, the patient should receive a neurologic examination followed by a

dose reduction or discontinuation of therapy.

Table 94-8

Neurotoxicity of Selected Chemotherapeutic Agents

Acute

Chronic

Encephalopathic Cerebellar Peripheral Cranial

Arachnoiditis

(Intrathecal Autonomic

Encephalopathy Syndrome Neuropathy Neuropathy Neuropathy Therapy) Neuropathy

Asparaginase Cytarabine Cytarabine Bortezomib Fluorouracil Cytarabine Vinblastine

Cisplatin Methotrexate Cisplatin Brentuximab Ifosfamide Methotrexate Vincristine

Cytarabine Nelarabine Fludarabine Cisplatin Thiotepa Vinorelbine

Fludarabine Thiotepa Fluorouracil Docetaxel

Ifosfamide Ifosfamide Fluorouracil

Methotrexate Ifosfamide

Nelarabine Lenalidomide

Procarbazine Nelarabine

Paclitaxel

Thalidomide

Vinblastine

Vincristine

Vinorelbine

SIADH, syndrome of inappropriate secretion of antidiuretic hormone.

Sources: Newton HB. Neurological complications of chemotherapy to the central nervous system. Handbook of Clinica2012;105:903–916; Magge RS, DeAngelis LM. The double-edged sword: Neurotoxicity of chemotherapy. Blood Rev.

p. 1985

p. 1986

Fluorouracil

Fluorouracil can cause acute cerebellar dysfunction characterized by the rapid onset

of gait ataxia, limb incoordination, dysarthria, and nystagmus. Cerebellar dysfunction

occurs in approximately 5% to 10% of patients receiving fluorouracil at all treatment

schedules in common use and can present weeks to months after beginning therapy. A

more diffuse encephalopathy presenting as headache, confusion, disorientation,

lethargy, and seizures can also occur. These symptoms can be reversed if

fluorouracil is discontinued or the dose is reduced. Reports of cerebellar ataxia have

also been reported with capecitabine, an oral prodrug of fluorouracil.

148,150

Fludarabine and Nelarabine

Fludarabine can cause severe neurotoxicity when used at doses greater than 90 mg/m2

for 5 to 7 days.

145,151,152 Symptoms include altered mental status, photophobia,

amaurosis (blindness that usually is temporary without change in the eye itself),

generalized seizures, spastic or flaccid paralysis, quadriparesis, and coma. Patients

may progress to death even when therapy is discontinued. This neurotoxicity,

however, is not common with the current recommended dosage of 20-30 mg/m2

/day

for 5 days. Mild neurologic symptoms are typically reported, but severe

neurotoxicity,

145,151 and optic demyelination occurs only occasionally.

153 Patients with

signs or symptoms suggestive of significant neurotoxicity should receive a neurologic

examination and, if warranted, therapy should be discontinued without rechallenging

with a dose reduction. Nelarabine, another purine analog has dose-limiting

neurotoxicity, and 18% to 37% of patients in Phase II trials showed severe grade 3

or 4 neurotoxicies.

154,155 The clinical presentation includes severe somnolence,

convulsions, and peripheral neuropathy ranging from paresthesias to motor weakness.

Several cases of ascending peripheral neuropathies and demyelination have been

reported.

156 Therapy should be stopped if grade 2 toxicity is present because some

cases have been irreversible.

157

mcq general

 

Search This Blog