O To Toxicity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Review Article

Ototoxicity and Cancer Therapy


Wendy Landier PhD, CRNP

Ototoxicity is a well-established toxicity associated with a subgroup of antineoplastic therapies that includes platinum chemother-
apy, radiation or surgery involving the ear and auditory nerve, and supportive care agents such as aminoglycoside antibiotics and
loop diuretics. The reported prevalence of ototoxicity in patients who have received potentially ototoxic therapy ranges from 4% to
90% depending on factors such as age of the patient population, agent(s) used, cumulative dose, and administration techniques.
The impact of ototoxicity on subsequent health-related and psychosocial outcomes in these patients can be substantial, and the
burden of morbidity related to ototoxic agents is particularly high in very young children. Considerable interindividual variability in
the prevalence and severity of ototoxicity has been observed among patients receiving similar treatment, suggesting genetic sus-
ceptibility as a risk factor. The development and testing of otoprotective agents is ongoing; however, to the author’s knowledge, no
US Food and Drug Administration-approved otoprotectants are currently available. Prospective monitoring for ototoxicity allows
for comparison of auditory outcomes across clinical trials, as well as for early detection, potential alterations in therapy, and audi-
tory intervention and rehabilitation to ameliorate the adverse consequences of hearing loss. Cancer 2016;122:1647-58. V C 2016 Amer-

ican Cancer Society.

KEYWORDS: antineoplastic therapy, genetic predisposition, grading scales, management of hearing loss, otoprotection, ototoxicity.

INTRODUCTION
Ototoxicity, manifesting as hearing loss, tinnitus, and/or vertigo, is a well-established toxicity associated with a subgroup
of antineoplastic therapies that include platinum chemotherapy and radiation or surgery involving the ear and auditory
nerve. Supportive care agents, such as aminoglycoside antibiotics and loop diuretics, also may contribute to ototoxicity in
patients with cancer. The impact of ototoxicity on subsequent health-related and social outcomes in these patients can be
substantial. Long-term effects may include permanent, severe to profound hearing loss or deafness, resulting in problems
with communication and social interaction, and impaired health-related quality of life. In young children, ototoxicity also
may significantly impair cognitive performance and the development of language and social skills.

Magnitude of the Problem


The reported prevalence of ototoxicity in patients who have received potentially ototoxic therapy ranges widely, from 4%
to 90%,1-7 depending on many factors, such as agent(s) used, age of the patient population, cumulative dose, and adminis-
tration techniques. A summary of the recent literature reporting the prevalence of ototoxicity associated with various can-
cer populations is presented in Table 1.1-13 It is interesting to note that the burden of morbidity related to ototoxic agents
is substantially higher in very young children2,3,6,14 and in those who receive higher cumulative doses of ototoxic
agents.2,3,13,14 However, interindividual variability in the prevalence and severity of ototoxicity, even in these vulnerable
populations, suggests that genetic susceptibility also may play an important role.15-18

Mechanisms of Ototoxicity
Normal anatomy and physiology of the ear
The ears comprise 3 compartments known as the external, middle, and inner ears (Fig. 1). The external ear serves to
amplify and direct sound toward the middle ear, in which the ossicles transform the sound waves into mechanical energy,
which is transmitted to the inner ear. In the inner ear, sound is transmitted by hydraulic waves in the cochlea that stimulate
the sensory hair cells lining the organ of Corti, releasing neurotransmitters that cause the eighth cranial nerve to fire and
transmit the neural impulses through the brainstem to the auditory cortex in the temporal lobe of the brain (Fig. 2).

Corresponding author: Wendy Landier, PhD, CRNP, Division of Pediatric Hematology/Oncology, Institute for Cancer Outcomes and Survivorship, University of
Alabama at Birmingham, 1600 7th Ave S, Lowder 500, Birmingham, AL 35233; Fax: (205) 623-2121; wlandier@peds.uab.edu

Department of Pediatric Hematology/Oncology, Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama.

DOI: 10.1002/cncr.29779, Received: September 8, 2015; Revised: October 13, 2015; Accepted: October 14, 2015, Published online February 9, 2016 in Wiley
Online Library (wileyonlinelibrary.com)

Cancer June 1, 2016 1647


Review Article

TABLE 1. Prevalence of Ototoxicity in Various Cancer Populations

Study Cancer Population Ototoxic Therapy Prevalence of Ototoxicity

Bokemeyer 19988 86 patients with testicular carcinoma Mean cumulative cisplatin doses in Symptomatic ototoxicity was present
who received platinum-based patients with no, transient, and in 20% of patients (tinnitus in 59%,
chemotherapy; median age at persistent ototoxicity: 297 mg/m2, hearing loss in 18%, and both in
diagnosis was 26 y (range, 19-50 337 mg/m2, and 678 mg/m2, 23%); symptoms were bilateral in
y); 100% male. respectively. 81% of patients and 66% of
patients had abnormal audiograms,
42% of which were compatible
with chemotherapy-related hearing
loss. In patients who received
cumulative cisplatin doses >400 mg/
m2, > 50% of patients had sympto-
matic and persistent ototoxicity.
Dean 20087 99 pediatric patients who received Mean cumulative dose of carboplatin Brock grade  1 hearing loss was
platinum-containing regimens; alone: 3987 mg/m2; cisplatin alone: observed in 4% of patients who
median age at diagnosis was 3.6 y 391 mg/m2; combined: cisplatin, received carboplatin alone; 70% of
(range, 1-17 y); 59% male. 401 mg/m2 plus carboplatin, patients who received a
1566 mg/m2; 36% of patients also combination of carboplatin and
underwent cranial radiation. cisplatin; and 57% of patients who
received cisplatin alone.
Gupta 20061 39 patients who received cisplatin for Median cumulative cisplatin dose: Brock grade 1-2 hearing loss was
the treatment of germ cell tumors; 400 mg/m2; all doses were observed in 12.5% of patients;
median age at diagnosis was 9 y administered by continuous 85% of patients had Brock grade
(range not reported); 41% male. infusion at 20 mg/m2/d for 5 d per 0 hearing loss.
course.
Ilveskoski 19969 35 patients who received “8-in-1” Mean cumulative dose of cisplatin: Hearing loss > 25 dB was considered
chemotherapy for treatment of 548 mg/m2 (range, 180-900 mg/m2); significant; 57% had normal
malignant brain tumors; mean age 54% had a VP shunt; all patients hearing, 23% had high-frequency
at diagnosis was 5.3 y (range, also received cranial radiation. hearing loss (4000-8000 Hz), and
birth-15 y); 63% male. 20% had severe hearing loss in the
speech range (500-2000 Hz).
Knight 20053 67 patients who received platinum- Mean cisplatin dose: 493 6 174 mg/ Bilateral hearing loss was observed in
based chemotherapy for the m2; mean carboplatin dose: 61% of patients per the ASHA
treatment of cancer; mean age 4701 mg/m2; 34% had received criteria; 17 patients required
was 9.6 6 6.2 y (range, 8 mo-23 y); cranial radiation prior to platinum. hearing aids.
67% male.
Landier 20142 333 patients with high-risk Cisplatin dose: up to 400 mg/ Prevalence of any postplatinum
neuroblastoma; median age at m2 6 myeloablative carboplatin hearing loss (per Brock, ASHA, NCI
diagnosis was 3.3 y (range, 1700 mg/m2. CTCAE, and Chang scales) ranged
birth-29 y); 56% male. from 64%-90%; prevalence of
severe hearing loss differed by
scale; by NCI CTCAE, the preva-
lence of severe hearing loss was
47% among those not receiving
myeloablative carboplatin and 71%
among those receiving myeloabla-
tive carboplatin; 29% and 58% of
patients, respectively, who did not
and did receive myeloablative
carboplatin required hearing aids.
Lewis 20094 36 children with osteosarcoma were Cisplatin cumulative dose ranged Prevalence of hearing loss of any
treated with cisplatin; median age from 210-480 mg/m2 grade was 42% (Boston scale
at diagnosis was 14 y (range, 3-18 grade 1 in 31% and grade 2 in
y); 39% male. 11%).
Low 200610 115 patients with nasopharyngeal 58 patients received cisplatin; median At 1 y after therapy, hearing
carcinoma; median age was 45 y dose: 160 mg (range, 112-213 mg) thresholds were statistically
(range, 15-74 y); 83% male. combined with radiotherapy; 47 significantly worse at all
patients received radiotherapy frequencies in the
alone; all patients received 70 Gy chemoradiotherapy group
in 35 fractions to the nasopharynx. compared with the
radiotherapy-alone group.
Nitz 20135 129 patients with bone or soft tissue 108 patients received cisplatin at a After treatment, 47% of patients
sarcoma; median age at diagnosis median cumulative dose of demonstrated hearing impairment
was 13.6 y (IQR, 10-16 y); 51% 360 mg/m2; 13 patients received ranging from grade 1-3 on the
male. carboplatin with a median Muenster scale.
cumulative dose of 1500 mg/m2;
8 patients received both cisplatin
and carboplatin.

1648 Cancer June 1, 2016


Ototoxicity and Cancer Therapy/Landier

TABLE 1. Continued

Study Cancer Population Ototoxic Therapy Prevalence of Ototoxicity

Orgel 201211 29 children with brain tumors; median Cisplatin dose: 281 6 88 mg/m2; After treatment, 62% of children had
age at diagnosis was 2.0 y (range, carboplatin dose: 1205 6 277 mg/ abnormal hearing and 38%
birth-9 y); 66% male. m2; no cranial radiation. required hearing aids.
Schell 19896 177 children and young adults with Mean cumulative cisplatin dose: Substantial hearing loss was defined
cancer who received cisplatin (146 407 mg/m2; mean cranial radiation as a hearing threshold of 50 dB;
patients), cranial irradiation (18 dose: 4747 cGy. 11% of patients had substantial
patients), or both (13 patients); hearing loss in the speech
mean age at diagnosis was 10.6 y frequencies and 50% had
(range, 1-27 y); sex not reported. substantial deficits at 4000 Hz; 14
patients required hearing aids.
Sivaprakasam 201112 38 patients with hepatoblastoma who Median cumulative cisplatin dose: Hearing loss was graded as per the
were treated with cisplatin; median 400 mg/m2 (range, 100-600 mg/m2). Brock scale: grade 0: 55.2%;
age at diagnosis was 1.5 y (range, grade 1: 15.8%; grade 2: 18.4%;
birth-11 y); 69% male. grade 3: 5.3%; and grade 4: 5.3%;
4 patients required hearing aids.
Stohr 200513 74 patients with osteosarcoma Median cisplatin dose: 360 mg/m2. After completion of therapy, 51% of
treated with cisplatin; mean age at patients had hearing loss of >20
diagnosis was 14.1 6 5.8 y (range, dB at  4 kHz; 3 patients required
3-38 y); sex not reported. hearing aids.

Abbreviations: ASHA, American Speech-Language-Hearing Association; cGy, centigrays; dB, decibels; Gy, grays; Hz, Hertz; IQR, interquartile range; kHz, kilo-
hertz; NCI CTCAE, National Cancer Institute Common Terminology Criteria for Adverse Events; VP, ventriculoperitoneal.

Although the ears are fully formed at birth, maturation of reactive oxygen species within the cochlea, with resultant
neuronal pathways and auditory structures continues dur- destruction of cochlear hair cells and damage to the stria
ing infancy and early childhood, making young children vascularis and spiral ganglion cells.23 Cochlear hair cell
particularly vulnerable to the ototoxic effects of cancer damage is generally dose-dependent,23 bilateral, and irre-
therapies. versible.6,24 Given the tonotopic arrangement of cochlear
hair cells,25 the initial insult begins at the base of the coch-
Radiation-related toxicity lea, in which high-frequency sounds are processed; addi-
Ototoxicity related to radiation involving the auditory tional drug exposure results in damage that progresses
structures is multifactorial in nature (Fig. 1).19 Patients toward the cochlear apex, in which the processing of lower
receiving higher (30 grays) doses of radiation to the pos- (speech frequency) sounds occurs.6,24 In young children,
terior nasopharynx and mastoid are at risk of developing the implications of even mild to moderate high-frequency
serous otitis media and associated conductive hearing hearing loss are particularly detrimental because they are
loss.20 Radiation involving the external auditory canal generally in the process of acquiring language and com-
may result in an increased susceptibility to soft-tissue munication skills at the time that hearing loss occurs, and
infections as well as excessive cerumen production and/or therefore are dependent on the high-frequency fricative
dry cerumen that may require periodic removal.21 Radia- sounds (eg, “th,” “s”) that are critical for speech discrimi-
tion to the cochlea may lead to sensorineural hearing loss, nation,26 which may be unintelligible to a child with
the exact mechanism of which is unknown to our knowl- high-frequency hearing loss. Once the cochlear sensory
edge, but which is believed to be related to direct damage hair cells are destroyed, they cannot regenerate; therefore,
to cochlear apparatus or damage to small vessels resulting drug-related sensorineural hearing loss is almost always
in hypoxia involving inner ear structures; in addition, bilateral and irreversible, and also can be accompanied by
radiation damage to the brainstem may indirectly contrib- tinnitus and vertigo.27 The ototoxicity of loop diuretics is
ute to hearing loss.21 Sensorineural hearing loss related to believed to be associated with shifts in fluid and electrolyte
radiation is generally permanent and progressive; the concentrations in the inner ear, which may result in
onset may occur during the acute phase of treatment or be edema of cochlear tissue and an associated decrease in
delayed for several years after the completion of therapy.22 endocochlear potential.28 Hearing loss associated with
loop diuretics alone is generally transient28; however, the
Chemotherapy-related and drug-related toxicity administration of these agents concomitantly with plati-
Platinum-induced and aminoglycoside-induced ototoxic- num chemotherapy or aminoglycosides may potentiate
ity is characterized by the production of toxic levels of the ototoxic effects of these drugs.29,30

Cancer June 1, 2016 1649


Review Article

Figure 1. Anatomy of the ear. The etiology of ototoxicity related to cancer therapy is multifactorial and may affect the structures
of the external or middle ear (eg, radiation-related toxicity resulting in conductive hearing loss) or the inner ear/cochlea (eg,
toxicity related to platinum-based chemotherapy resulting in sensorineural hearing loss). Tumor growth or surgical resection also
can result in damage to auditory structures with resultant hearing loss.

Surgery-related toxicity Risk Factors for Ototoxicity


Tumors located in or near the auditory structures or au- Chemotherapy
ditory nerve may cause damage by placing pressure on or Platinum-based chemotherapy is widely used and particu-
infiltrating auditory apparatus. Surgical resection has the larly effective against solid tumors involving the head and
potential to result in further damage to the ear and audi- neck, lung, ovary, testicle, and bladder in adults.18 In chil-
tory nerve.31 Patients with central nervous system dren, platinum compounds are commonly used in the treat-
tumors may experience rapid changes in intracranial ment of neuroblastoma, osteosarcoma, hepatoblastoma,
pressure associated with lumbar puncture, tumor resec- germ cell and central nervous system tumors.33 The 3 major
tion, or ventriculostomy, which are associated with hear- platinum compounds (cisplatin, carboplatin, and oxalipla-
ing loss; these patients also often require temporary or tin) differ with regard to their chemical structure and
permanent shunting of cerebrospinal fluid, which is in- adverse effect profiles. A major adverse effect associated with
dependently associated with an increased risk of ototox- cisplatin is irreversible sensorineural hearing loss.34 Younger
icity (Fig. 3).32 age (particularly < 5 years) at the time of therapy, diagnosis

1650 Cancer June 1, 2016


Ototoxicity and Cancer Therapy/Landier

Figure 2. Auditory pathways. Sound waves enter the external ear via the auditory canal; are converted to mechanical energy in
the middle ear; and subsequently are transmitted via hydraulic waves in the inner ear (cochlea), stimulating the release of neuro-
transmitters and causing the eighth cranial (auditory) nerve to fire. These neural impulses are transmitted via the medulla,
midbrain, and thalamus to the auditory cortex in the temporal lobe of the brain.

of a central nervous system tumor, diminished renal func- required to attain curative treatment for many central nerv-
tion, rapid intravenous administration, and treatment with ous system tumors,40 as well as for malignancies involving
multiple potentially ototoxic agents increase the risk of oto- structures in the head, such as rhabdomyosarcoma41 and
toxicity.27,33,35 Carboplatin is generally less ototoxic than nasopharyngeal carcinoma.10 The effects of radiation to
cisplatin, although the risk increases substantially when this the ear appear to be dose-related, with sensorineural hear-
agent is used in infants36 or in myeloablative doses.2,37 ing loss generally occurring at doses >30 grays.40,42 How-
Oxaliplatin-related ototoxicity is rare, although there have ever, the risk of ototoxicity increases in patients who
been some isolated case reports of hearing loss associated require multimodality therapy, such as those receiving
with this agent.38 both radiation and platinum-based chemotherapy.43,44

Radiation Tumor/surgery
Radiation involving the ear or auditory nerve is often asso- Hearing loss may be a presenting symptom of tumors
ciated with ototoxicity.39 Nevertheless, radiation may be located in or near the auditory structures or auditory

Cancer June 1, 2016 1651


Review Article

opurine S-methyltranferase (TPMT) and catechol-O-


methyltransferase (COMT) are less clear.15,27 In what to
the author’s knowledge is the small body of genetic associa-
tion studies of ototoxicity in cancer therapeutics published
to date, genetic variants in TPMT, COMT, and ACYP2
have been associated with platinum-related hearing loss in
children,49,54 variants of the megalin gene have been associ-
ated with platinum-related hearing loss in adults,50 mito-
chondrial mutations have been associated with
Figure 3. Cochlear aqueduct. The cochlear aqueduct provides
a direct connection between cerebrospinal fluid and the peril- aminoglycoside ototoxicity,55 variants in GSTP1 have been
ymphatic space in the cochlea; rapid changes in intracranial associated with increased susceptibility to radiation-related
pressure and shunting of cerebrospinal fluid have been asso-
ciated with hearing loss. hearing loss in children,56 and conflicting results have been
noted regarding associations between GSTs and platinum-
related hearing loss.51,57 A summary of candidate genes
potentially involved in ototoxicity related to cancer therapy
nerve,45 including nasopharyngeal, parameningeal, ves-
is presented in Table 2.49-58
tibular, and base of skull tumors, as well as tumors affect-
ing the temporal bone. The surgical management of
Detection of Ototoxicity
tumors involving essential components of the auditory Types of testing
system46 and the surgical shunting of cerebrospinal fluid32 Given the substantial risk of ototoxicity associated with
also may result in ototoxicity. platinum chemotherapy and radiation involving the ear
and auditory nerve, prospective auditory evaluations to
Other ototoxic agents allow for the early detection of hearing loss are indicated
Additional agents with known ototoxic properties that are for patients receiving therapy with ototoxic potential.
commonly used during therapy for cancer include amino- Pure-tone audiometry is the standard method used for
glycoside antibiotics30 and loop diuretics28; risk increases hearing assessments,59 and generally involves the presen-
with elevated serum trough levels,47 rapid intravenous tation of sounds across a wide range of frequencies (ie, low
administration,28 and the administration of loop diuretics to high pitches, generally 500 to 8000 Hertz at mini-
concurrent with or shortly after the administration of mum). Each sound is presented at increasing intensity (ie,
aminoglycosides.30 loudness, measured in decibels) until the patient is able to
hear it. The lowest level of intensity at which the patient
Genetic predisposition can first hear a sound at a given frequency approximately
Considerable interindividual variability in the prevalence 50% of the time is considered the “pure-tone threshold;”
and severity of ototoxicity has been observed among a normal pure-tone threshold is  20 decibels.60 Thresh-
patients receiving similar treatment with agents of known old shifts in patients with sensorineural hearing loss typi-
ototoxic potential, with some patients remaining unaf- cally begin in the high-frequency range (>4000 Hertz),
fected at high cumulative doses whereas others experience and generally progress to the lower frequencies in a stair-
severe damage at low doses.2,48,49 This interindividual vari- step pattern over time with continued exposure to the oto-
ation is likely to be at least partially explained by genetic toxic agent (Fig. 4).61 The majority of patients aged >5
susceptibility to the ototoxic effects of therapy.15,17 Candi- years are able to complete conventional pure-tone audio-
date genes identified as being potentially involved in oto- metric testing in an acoustically treated (sound-attenuat-
toxicity related to cancer therapy include megalin50 (a ing) test booth. The technique can be adapted for younger
transport protein), glutathione-S-transferase51 (GSTs; children using play (for those aged 3-5 years) or visual
involved in cellular detoxification), excision repair cross- reinforcement (for those aged 9-36 months) techniques.62
complementation groups 1 and 252,53 (ERCC1 and Patients who are too young or unable to cooperate
ERCC2; involved in repairing drug-induced damage), acyl- with behavioral testing can be assessed using electrophy-
phosphatase 254 (ACYP2; involved in calcium and magne- siologic techniques, such as brainstem auditory-evoked
sium transport), and mutations in mitochondrial genes55 response.59 In addition, distortion product otoacoustic
(essential mediators for some apoptotic pathways); the emissions are objective, noninvasive measures that do not
biologic association between ototoxicity and variants in thi- require the child’s active participation and may be more

1652 Cancer June 1, 2016


Ototoxicity and Cancer Therapy/Landier

TABLE 2. Candidate Genes Potentially Involved in Ototoxicity Related to Cancer Therapy

Gene/Protein Possible Mechanism Study Population/Exposure

Megalin Potential mediator of ototoxic drug Riedemann 200850 Adults/platinum


uptake
51
Glutathione S-transferases (GSTs) Cellular detoxification Peters 2000 Adults/platinum
Oldenburg 200757 Adults/platinum
Rednam 201356 Children/radiation
Excision repair cross- Repair of drug-induced damage Caronia 200952 Children and adults/platinum
complementation groups 1 and 2 Suk 200553 Adults/platinum
(ERCC1 and ERCC2)
Acylphosphatase-2 (ACYP2) Calcium and magnesium transport Xu 201554 Children/platinum
Mitochondrial gene mutations Mediators for some apoptotic Xing 200655 Children and adults/
pathways aminoglycosides
Thiopurine S-methyltransferase Unknown (possibly related to Ross 200949 Children/platinum
(TPMT) and catechol-O- S-adenosyl methionine [SAM])58
methyltransferase (COMT)

and/or cranial radiation have been established previ-


ously,65,66 and current efforts are aimed at standardizing
and refining these guidelines, particularly for young chil-
dren.15,59 For older patients, less frequent testing may be
indicated; however, all patients receiving potentially oto-
toxic therapy should undergo auditory testing at baseline
and at the completion of therapy, at minimum, to deter-
mine whether auditory intervention or rehabilitation is
indicated.

Monitoring during long-term follow-up


Monitoring for ototoxicity after the completion of cancer
Figure 4. Threshold shifts shown in an audiogram of the right
ear from a patient with platinum-related sensorineural hear- therapy is recommended in several standardized long-
ing loss. (A) Hearing loss appears first in the high-frequency term follow-up guidelines.67-69 For patients with a history
range, and (B) progresses to the lower frequencies in a stair-
step pattern after additional exposure to platinum. of treatment with platinum chemotherapy, an auditory
assessment is generally recommended at the baseline long-
term follow-up visit (usually  2 years after the comple-
tion of cancer therapy) and then as clinically indicated.
sensitive to initial changes in auditory function than con- For patients who have received radiotherapy to the head
ventional auditory testing.63,64 or ear, ongoing audiologic monitoring every 5 years (or
more frequently if clinically indicated) is advised due to
Frequency of testing the potential for progression of hearing loss over time.22
The frequency of auditory testing indicated for an indi-
vidual patient depends on several factors, including age, Grading/classification of hearing loss
planned therapy with potentially ototoxic agents, and per- Several ototoxicity grading systems are currently in use
sonal risk profile. Because young children are at the high- (Table 3).15,61,65,70-73 Due to significant variability
est risk of sustaining both ototoxicity and its related between grading scales,2,15 efforts are currently underway
adverse outcomes (ie, delays in linguistic development to develop expert consensus regarding a uniform ototoxic-
and poor psychosocial and cognitive outcomes), baseline ity grading method.15 The accurate classification of hear-
and ongoing auditory monitoring (ie, before initial plati- ing loss has implications for both the clinical management
num dose and every 1-2 courses of platinum-based chem- of patients and for the evaluation and comparison of
otherapy) is often recommended for children to allow for audiologic outcomes on clinical trials.59 Across most
early detection, auditory intervention, and, when possible, scales, hearing loss is assigned a grade ranging from 0 (nor-
modification of ototoxic therapy. Clinical practice guide- mal hearing, or clinically insignificant loss) to 4 (severe or
lines for patients receiving platinum-based chemotherapy profound hearing loss); however, there is significant

Cancer June 1, 2016 1653


Review Article

TABLE 3. Common Ototoxicity Grading Scales

Target
Grading Scale Description Population Features Limitations

Brock (Brock 1991)61 Designed to grade hearing loss Pediatric Widely used; baseline Does not capture
progression from high to low assessment not hearing loss
frequencies in the configuration required <40 dB, misses
commonly associated with significant
ototoxic cancer therapy; hearing functional deficits
loss is graded on 5-point scale
ASHA (1994)65 Hearing is compared with baseline in Pediatric Designed for early Does not classify
absolute terms (ie, presence/ and detection of severity of
absence of hearing loss in adult hearing loss hearing loss;
comparison with baseline) baseline
assessment
is required
Muenster (Schmidt 2007)70 8-point scale accounts for minimal Pediatric Designed for early Complexity of use
hearing loss (>10-20 dB), detection of
subgroups within major hearing loss
classifications, and tinnitus
Chang (Chang & Modification of Brock scale with Pediatric Addresses functional Complexity of use
Chinosornvatana 2010)71 similar configuration and deficits; baseline
expansion to 7-point scale; grades assessment not
hearing loss > 20 dB and measures required
interval frequencies (ie, 3000 and
6000 Hz)
NCI CTCAE, version 4 (2010)72 4-point scale includes both objective Pediatric Familiar to oncologists; Not configured for
and subjective criteria; grades are and widely used in high- to low-
assigned based on threshold shift adult NCI-sponsored frequency hearing
from baseline and not actual clinical trials loss commonly
hearing loss associated with
cancer treat-
ments; baseline
assessment
required
SIOP Boston (Brock 2012)15 5-point scale designed to grade Pediatric Proposed through consensus of Limited reliability
hearing loss progression from high international working group; and validity
to low frequencies; grades hearing potential application across testing to date
loss > 20 dB; uses absolute clinical trials worldwide;
hearing levels baseline assessment not
required
TUNE grading system 7-point scale designed to provide Adults Includes subjective symptoms Time-consuming to
(Theunissen 2014)73 insight into the effect of hearing and threshold shifts at higher use; feasibility
loss on specific daily life situations frequencies (up to 12.5 kHz); testing
(such as speech intelligibility and uses air conduction thresholds completed; needs
ability to appreciate ultrahigh only; designed to represent the external validation
sounds) auditory system’s
real-world functionality

Abbreviations: ASHA, American Speech-Language-Hearing Association; dB, decibels; Hz, Hertz; kHz, kiloHertz; NCI CTCAE, National Cancer Institute Com-
mon Terminology Criteria for Adverse Events; SIOP, International Society of Pediatric Oncology Boston ototoxicity scale.

variability in the definitions of the grades among the however, it is important to understand that although hear-
scales, and some scales are designed for use in children ing aids amplify sound, they do not restore normal hear-
whereas others are designed for use in adult popula- ing. Thus, in patients requiring hearing aids, hearing
tions.2,15,59,61,65,71,73 In addition, there are classification quality will remain distorted to some extent, resulting in a
systems designed to detect early ototoxicity3 that desig- reduced ability to discriminate speech in noisy environ-
nate hearing solely as “normal” versus “impaired,” with- ments. The daily use and care of hearing aids, particularly
out assigning a severity grade.59 in young children, also can be challenging.74 Neverthe-
less, there are many types and models of hearing aids avail-
Management of Ototoxicity able, including behind-the-ear, in-the-ear, and in-the-
Hearing aids canal models, and continued improvements in digital
Hearing aids are a crucial component of the management technology have resulted in increased programmability
of significant hearing loss in both children and adults; and advanced speech processing in newer models.75

1654 Cancer June 1, 2016


Ototoxicity and Cancer Therapy/Landier

TABLE 4. Summary of Approaches to the Management of Ototoxicity

Approach Benefits Limitations

Hearing aids Amplification of sound; numerous models and Hearing quality remains distorted to some extent;
features available; increased programmability reduced ability to discriminate speech in noisy
and advanced speech processing in newer environments; daily care required
models
Cochlear implants Direct stimulation of auditory neural pathway in the Requires ongoing audiology and speech therapy
cochlea provides a pathway for the rehabilitation program
transmission of sound to the brain in patients
with severely damaged sensory hair cells
Assistive devices Provide augmentation to hearing aids or Some devices must be compatible with the
(eg, auditory trainers, supplementary communication; particularly particular model of hearing aid; devices may
telephone amplifiers, useful in noisy environments become outdated and need to be replaced as
audio streamers, use technologies continue to rapidly evolve
of text messaging and
social media)
Special accommodations Provision of specialized services at public Requires awareness of applicable laws and
expense; particularly helpful for children, completion of appropriate applications and
adolescents, and young adults attending evaluative procedures; often requires
school; free of charge to the patient/family reevaluation and renewal of service
authorizations on an annual basis

Cochlear implants Education Act Amendments of 1997, and related provi-


In patients with severe to profound hearing loss who are sions in “Part B” of this law)80 and Public Law 101-336
unable to benefit from hearing aids, a cochlear implant (Americans With Disabilities Act of 1990), provide for
may provide significant benefit.76 Cochlear implants are equal access and free and appropriate public education for
surgically placed devices that directly stimulate auditory those with special needs such as hearing loss. This includes
neural pathways in the cochlea. By circumventing the the provision of specialized services and devices, provided
damaged sensory hair cells, cochlear implants provide a at public expense.81
pathway for the transmission of sound waves to the A summary of approaches to the management of
brain.77 Hybrid cochlear implants are specifically ototoxicity is presented in Table 4.
designed for patients with severe to profound hearing loss
limited to the high frequencies and combine both acoustic Prevention of Ototoxicity
and electrical stimulation; these hybrid implants may be Monitoring during treatment
particularly useful in adults with steeply sloping high- Implementation of standardized audiologic monitoring
frequency sensorineural hearing loss typically noted after protocols has the potential to allow for the early detection
exposure to ototoxic agents.78 of ototoxicity in patients receiving therapy for cancer, and
thus also may provide an opportunity for treatment modi-
Assistive devices fication, if possible, before auditory damage becomes
In addition to hearing aids, devices such as auditory train- severe. Even when no reasonable alternative is available
ers, telephone amplifiers, telephone devices for the deaf, and therapy with the ototoxic agent must continue, moni-
audio streamers, and more recently, the wide availability toring may still be of value in allowing for early interven-
of text messaging and social media provide alternate com- tion and auditory rehabilitation.
munication methods for patients with severe hearing
loss.79 These assistive devices can be used with compatible Otoprotective agents
hearing aids in difficult listening environments to Much preclinical work has been done to develop otopro-
improve the delivery of sound directly to the affected tective agents, some of which have moved into clinical
patient. studies. Trials of amifostine, an antioxidant, in children
receiving cisplatin for hepatoblastoma82 and germ cell
Special accommodations tumors83 failed to demonstrate otoprotection; however, a
Hearing loss is generally recognized as a disability under more recent trial of amifostine in children with medullo-
the law. For example, in the United States, 2 public laws, blastoma demonstrated otoprotection in those with
Public Law 105-17 (Individuals With Disabilities average-risk but not high-risk disease.84 Several clinical

Cancer June 1, 2016 1655


Review Article

trials evaluating additional agents, including sodium thio- REFERENCES


sulfate, another antioxidant, currently are ongoing. 1. Gupta AA, Capra M, Papaioannou V, et al. Low incidence of oto-
toxicity with continuous infusion of cisplatin in the treatment of pe-
Because the mechanism of platinum-related cytotoxicity diatric germ cell tumors. J Pediatr Hematol Oncol. 2006;28:91-94.
to normal tissues, including the cochlea, may be similar to 2. Landier W, Knight K, Wong FL, et al. Ototoxicity in children with
the platinum-related cytotoxicity aimed at the tumor cells high-risk neuroblastoma: prevalence, risk factors, and concordance of
grading scales–a report from the Children’s Oncology Group. J Clin
(ie, interaction with nuclear DNA), the potential for oto- Oncol. 2014;32:527-534.
protectant interference with the efficacy of the chemother- 3. Knight KR, Kraemer DF, Neuwelt EA. Ototoxicity in children
receiving platinum chemotherapy: underestimating a commonly
apy has been raised as a concern.27 To the author’s occurring toxicity that may influence academic and social develop-
knowledge to date, there are no otoprotectants approved ment. J Clin Oncol. 2005;23:8588-8596.
4. Lewis MJ, DuBois SG, Fligor B, Li X, Goorin A, Grier HE. Oto-
by the US Food and Drug Administration that currently toxicity in children treated for osteosarcoma. Pediatr Blood Cancer.
are available for use in pediatric or adult oncology.27 2009;52:387-391.
5. Nitz A, Kontopantelis E, Bielack S, et al. Prospective evaluation of
cisplatin- and carboplatin-mediated ototoxicity in paediatric and
Identification of patients at highest risk adult soft tissue and osteosarcoma patients. Oncol Lett. 2013;5:311-
Risk-prediction models for platinum-related ototoxicity, 315.
6. Schell MJ, McHaney VA, Green AA, et al. Hearing loss in children
based on age and cumulative dose, have been developed14; and young adults receiving cisplatin with or without prior cranial
however, these models do not accurately predict risk for irradiation. J Clin Oncol. 1989;7:754-760.
7. Dean JB, Hayashi SS, Albert CM, King AA, Karzon R, Hayashi RJ.
individual patients.27 Although there currently is not suf- Hearing loss in pediatric oncology patients receiving carboplatin-
ficient evidence to identify genetic predictors of suscepti- containing regimens. J Pediatr Hematol Oncol. 2008;30:130-134.
8. Bokemeyer C, Berger CC, Hartmann JT, et al. Analysis of risk fac-
bility to ototoxicity before the implementation of therapy, tors for cisplatin-induced ototoxicity in patients with testicular can-
continued progress is being made in ototoxicity-related cer. Br J Cancer. 1998;77:1355-1362.
pharmacogenomics, making the prospect of personalized 9. Ilveskoski I, Saarinen UM, Wiklund T, et al. Ototoxicity in children
with malignant brain tumors treated with the “8 in 1” chemotherapy
therapy based on the identification of high-risk genotypes protocol. Med Pediatr Oncol. 1996;27:26-31.
a future possibility for the prevention of ototoxicity. 10. Low WK, Toh ST, Wee J, Fook-Chong SM, Wang DY. Sensorineu-
ral hearing loss after radiotherapy and chemoradiotherapy: a single,
blinded, randomized study. J Clin Oncol. 2006;24:1904-1909.
Conclusions 11. Orgel E, Jain S, Ji L, et al. Hearing loss among survivors of child-
Ototoxicity is a serious consequence associated with sev- hood brain tumors treated with an irradiation-sparing approach.
Pediatr Blood Cancer. 2012;58:953-958.
eral agents commonly used in contemporary cancer ther- 12. Sivaprakasam P, Gupta AA, Greenberg ML, Capra M, Nathan PC.
apy, including platinum-based chemotherapy, radiation Survival and long-term outcomes in children with hepatoblastoma
treated with continuous infusion of cisplatin and doxorubicin.
and surgery involving the ear, and supportive care drugs J Pediatr Hematol Oncol. 2011;33:e226-e230.
including aminoglycoside antibiotics and loop diuretics. 13. Stohr W, Langer T, Kremers A, et al; German Late Effects Working
Group in the German Society of Pediatric Oncology and Hematol-
Progress has been made toward the early identification of ogy. Cisplatin-induced ototoxicity in osteosarcoma patients: a report
ototoxicity, and rehabilitative strategies are available to from the late effects surveillance system. Cancer Invest. 2005;23:201-
help ameliorate the associated adverse social and educa- 207.
14. Li Y, Womer RB, Silber JH. Predicting cisplatin ototoxicity in chil-
tional consequences. Progress is also being made toward dren: the influence of age and the cumulative dose. Eur J Cancer.
the identification of those individuals at highest risk of 2004;40:2445-2451.
15. Brock PR, Knight KR, Freyer DR, et al. Platinum-induced ototoxic-
developing toxicity as a result of genetic susceptibility, as ity in children: a consensus review on mechanisms, predisposition,
well as the development of otoprotective agents to dimin- and protection, including a new International Society of Pediatric
Oncology Boston ototoxicity scale. J Clin Oncol. 2012;30:2408-
ish toxicities associated with current regimens. Future 2417.
directions include the development of reliable profiles to 16. Travis LB, Fossa SD, Sesso HD, et al; Platinum Study Group.
Chemotherapy-induced peripheral neurotoxicity and ototoxicity: new
identify at-risk individuals, and the implementation of paradigms for translational genomics. J Natl Cancer Inst. 2014;
personalized strategies such as the use of otoprotective 106(5). pii: dju044.
agents or avoidance of ototoxic drugs in susceptible indi- 17. Bhatia S. Role of genetic susceptibility in development of treatment-
related adverse outcomes in cancer survivors. Cancer Epidemiol Bio-
viduals to decrease the prevalence of ototoxicity and markers Prev. 2011;20:2048-2067.
improve health-related quality of life in these patients. 18. Mukherjea D, Rybak LP. Pharmacogenomics of cisplatin-induced
ototoxicity. Pharmacogenomics. 2011;12:1039-1050.
19. Young YH, Lu YC. Mechanism of hearing loss in irradiated ears: a
FUNDING SUPPORT long-term longitudinal study. Ann Otol Rhinol Laryngol. 2001;110:
904-906.
No specific funding was disclosed. 20. Walker GV, Ahmed S, Allen P, et al. Radiation-induced middle ear
and mastoid opacification in skull base tumors treated with radio-
therapy. Int J Radiat Oncol Biol Phys. 2011;81:e819-e823.
CONFLICT OF INTEREST DISCLOSURES 21. Parsons JA. The effect of radiation on normal tissues of the head
The author made no disclosures. and neck. In: Million RR, Cassisi NJ, eds. Management of Head

1656 Cancer June 1, 2016


Ototoxicity and Cancer Therapy/Landier

and Neck Cancer. A Multidisciplinary Approach. Philadelphia: Lip- 46. Wang AC, Chinn SB, Than KD, et al. Durability of hearing preser-
pincott; 1984:173-207. vation after microsurgical treatment of vestibular schwannoma using
22. Mujica-Mota M, Waissbluth S, Daniel SJ. Characteristics of the middle cranial fossa approach. J Neurosurg. 2013;119:131-138.
radiation-induced sensorineural hearing loss in head and neck cancer: 47. Lerner SA, Schmitt BA, Seligsohn R, Matz GJ. Comparative study
a systematic review. Head Neck. 2013;35:1662-1668. of ototoxicity and nephrotoxicity in patients randomly assigned to
23. Ding D, Allman BL, Salvi R. Review: ototoxic characteristics of plat- treatment with amikacin or gentamicin. Am J Med. 1986;80:98-104.
inum antitumor drugs. Anat Rec (Hoboken). 2012;295:1851-1867. 48. Kushner BH, Budnick A, Kramer K, Modak S, Cheung NK. Oto-
24. McHaney VA, Thibadoux G, Hayes FA, Green AA. Hearing loss in toxicity from high-dose use of platinum compounds in patients with
children receiving cisplatin chemotherapy. J Pediatr. 1983;102:314- neuroblastoma. Cancer. 2006;107:417-422.
317. 49. Ross CJ, Katzov-Eckert H, Dube MP, et al; CPNDS Consortium.
25. Mann ZF, Kelley MW. Development of tonotopy in the auditory Genetic variants in TPMT and COMT are associated with hearing
periphery. Hear Res. 2011;276:2-15. loss in children receiving cisplatin chemotherapy. Nat Genet. 2009;
26. Stelmachowicz PG, Pittman AL, Hoover BM, Lewis DE, Moeller 41:1345-1349.
MP. The importance of high-frequency audibility in the speech and 50. Riedemann L, Lanvers C, Deuster D, et al. Megalin genetic poly-
language development of children with hearing loss. Arch Otolaryngol morphisms and individual sensitivity to the ototoxic effect of cispla-
Head Neck Surg. 2004;130:556-562. tin. Pharmacogenomics J. 2008;8:23-28.
27. Langer T, Am Zehnhoff-Dinnesen A, Radtke S, Meitert J, Zolk O. 51. Peters U, Preisler-Adams S, Hebeisen A, et al. Glutathione S-
Understanding platinum-induced ototoxicity. Trends Pharmacol Sci. transferase genetic polymorphisms and individual sensitivity to the
2013;34:458-469. ototoxic effect of cisplatin. Anticancer Drugs. 2000;11:639-643.
28. Rybak LP. Ototoxicity of loop diuretics. Otolaryngol Clin North Am. 52. Caronia D, Patino-Garcia A, Milne RL, et al. Common variations
1993;26:829-844. in ERCC2 are associated with response to cisplatin chemotherapy
29. Stringer SP, Meyerhoff WL, Wright CG. Ototoxicity. In: Paparella and clinical outcome in osteosarcoma patients. Pharmacogenomics J.
MM, Shumrick DA, eds. Otolaryngology. Philadelphia: Saunders; 2009;9:347-353.
1991:1653-1669. 53. Suk R, Gurubhagavatula S, Park S, et al. Polymorphisms in ERCC1
30. Bates DE. Aminoglycoside ototoxicity. Drugs Today (Barc). 2003;39: and grade 3 or 4 toxicity in non-small cell lung cancer patients. Clin
277-285. Cancer Res. 2005;11:1534-1538.
31. Simon MV. Neurophysiologic intraoperative monitoring of the vesti- 54. Xu H, Robinson GW, Huang J, et al. Common variants in ACYP2
bulocochlear nerve. J Clin Neurophysiol. 2011;28:566-581. influence susceptibility to cisplatin-induced hearing loss. Nat Genet.
32. Guillaume DJ, Knight K, Marquez C, Kraemer DF, Bardo DM, 2015;47:263-266.
Neuwelt EA. Cerebrospinal fluid shunting and hearing loss in 55. Xing G, Chen Z, Wei Q, et al. Mitochondrial 12S rRNA A827G
patients treated for medulloblastoma. J Neurosurg Pediatr. 2012;9: mutation is involved in the genetic susceptibility to aminoglycoside
421-427. ototoxicity. Biochem Biophys Res Commun. 2006;346:1131-1135.
33. Ruggiero A, Trombatore G, Triarico S, et al. Platinum compounds 56. Rednam S, Scheurer ME, Adesina A, Lau CC, Okcu MF. Glutathi-
in children with cancer: toxicity and clinical management. Anticancer one S-transferase P1 single nucleotide polymorphism predicts perma-
Drugs. 2013;24:1007-1019. nent ototoxicity in children with medulloblastoma. Pediatr Blood
34. Rybak LP, Mukherjea D, Jajoo S, Ramkumar V. Cisplatin ototoxic- Cancer. 2013;60:593-598.
ity and protection: clinical and experimental studies. Tohoku J Exp 57. Oldenburg J, Kraggerud SM, Cvancarova M, Lothe RA, Fossa SD.
Med. 2009;219:177-186. Cisplatin-induced long-term hearing impairment is associated with
35. Grewal S, Merchant T, Reymond R, McInerney M, Hodge C, specific glutathione s-transferase genotypes in testicular cancer survi-
Shearer P. Auditory late effects of childhood cancer therapy: a report vors. J Clin Oncol. 2007;25:708-714.
from the Children’s Oncology Group. Pediatrics. 2010;125:e938- 58. Aebi S, Christen R, Naredi P, et al. Synergy between cisplatin and
e950. an inhibitor of S-adenosylmethionine dependent transmethylation in
36. Qaddoumi I, Bass JK, Wu J, et al. Carboplatin-associated ototoxicity human ovarian adenocarcinoma cells. Int J Oncol. 1997;11:869-874.
in children with retinoblastoma. J Clin Oncol. 2012;30:1034-1041. 59. Bass JK, Bhagat SP. Challenges in ototoxicity monitoring in the pe-
37. Punnett A, Bliss B, Dupuis LL, Abdolell M, Doyle J, Sung L. Oto- diatric oncology population. J Am Acad Audiol. 2014;25:760-774;
toxicity following pediatric hematopoietic stem cell transplantation: a quiz 782-783.
prospective cohort study. Pediatr Blood Cancer. 2004;42:598-603. 60. Stach BA. Clinical Audiology: An Introduction. San Diego: Singular;
38. Oh SY, Wasif N, Garcon MC, Rodriguez G, Saif MW. Ototoxicity 1998.
associated with oxaliplatin in a patient with pancreatic cancer. JOP. 61. Brock PR, Bellman SC, Yeomans EC, Pinkerton CR, Pritchard J.
2013;14:676-679. Cisplatin ototoxicity in children: a practical grading system. Med
39. Gamble JE, Peterson EA, Chandler JR. Radiation effects on the Pediatr Oncol. 1991;19:295-300.
inner ear. Arch Otolaryngol. 1968;88:156-161. 62. Smits C, Swen SJ, Theo Goverts S, Moll AC, Imhof SM, Schouten-
40. Merchant TE, Gould CJ, Xiong X, et al. Early neuro-otologic effects van Meeteren AY. Assessment of hearing in very young children receiv-
of 3-dimensional irradiation in children with primary brain tumors. ing carboplatin for retinoblastoma. Eur J Cancer. 2006;42:492-500.
Int J Radiat Oncol Biol Phys. 2004;58:1194-1207. 63. Lonsbury-Martin BL, Martin GK. Evoked otoacoustic emissions as
41. Schoot RA, Theunissen EA, Slater O, et al. Hearing loss in survivors objective screeners for ototoxicity. Semin Hear. 2001;22:337-391.
of childhood head and neck rhabdomyosarcoma; a long-term follow- 64. Knight KR, Kraemer DF, Winter C, Neuwelt EA. Early changes in
up study [published online ahead of print August 21, 2015]. Clin auditory function as a result of platinum chemotherapy: use of
Otolaryngol. doi: 10.1111/coa.12527. extended high-frequency audiometry and evoked distortion product
42. Hua C, Bass JK, Khan R, Kun LE, Merchant TE. Hearing loss after otoacoustic emissions. J Clin Oncol. 2007;25:1190-1195.
radiotherapy for pediatric brain tumors: effect of cochlear dose. Int J 65. American Speech-Language-Hearing Association. Guidelines for the
Radiat Oncol Biol Phys. 2008;72:892-899. audiologic management of individuals receiving cochleotoxic drug
43. Warrier R, Chauhan A, Davluri M, Tedesco SL, Nadell J, Craver R. therapy. ASHA. 1994;36(suppl 12):11-19.
Cisplatin and cranial irradiation-related hearing loss in children. 66. American Academy of Audiology. Ototoxicity monitoring 2009.
Ochsner J. 2012;12:191-196. http://audiology-web.s3.amazonaws.com/migrated/OtoMonGuide-
44. Rivelli TG, Mak MP, Martins RE, da Costa E Silva VT, de Castro lines.pdf_539974c40999c1.58842217.pdf Accessed September 7,
G Jr. Cisplatin based chemoradiation late toxicities in head and neck 2015
squamous cell carcinoma patients. Discov Med. 2015;20:57-66. 67. Children’s Oncology Group. Children’s Oncology Group Long-
45. Van Abel KM, Carlson ML, Link MJ, et al. Primary inner ear Term Follow-Up Guidelines for Survivors of Childhood, Adolescent,
schwannomas: a case series and systematic review of the literature. and Young Adult Cancers. Version 4.0. Monrovia, CA: Children’s
Laryngoscope. 2013;123:1957-1966. Oncology Group; 2013.

Cancer June 1, 2016 1657


Review Article

68. Dutch Childhood Oncology Group. Guidelines for follow-up in sur- 77. Papsin BC, Gordon KA. Cochlear implants for children with severe-
vivors of childhood cancer 5 years after diagnosis. Den Haag/Am- to-profound hearing loss. N Engl J Med. 2007;357:2380-2387.
sterdam: SKION; 2010. 78. Woodson EA, Reiss LA, Turner CW, Gfeller K, Gantz BJ. The
69. United Kingdom Children’s Cancer Study Group Late Effects Hybrid cochlear implant: a review. Adv Otorhinolaryngol. 2010;67:
Group. Therapy-Based Long-Term Follow-Up Practice Statement. 125-134.
London: United Kingdom Children’s Cancer Study Group; 2011. 79. Brookhouser PE, Beauchaine KL, Osberger MJ. Management of the
70. Schmidt CM, Bartholomaus E, Deuster D, Heinecke A, Dinnesen child with sensorineural hearing loss. Medical, surgical, hearing aids,
AG. The “Muenster classification” of high frequency hearing loss fol- cochlear implants. Pediatr Clin North Am. 1999;46:121-141.
lowing cisplatin chemotherapy [in German]. HNO. 2007;55:299- 80. Kreisman BM, John AB. A case law review of the individuals with
306. disabilities education act for children with hearing loss or auditory
71. Chang KW, Chinosornvatana N. Practical grading system for evalu- processing disorders. J Am Acad Audiol. 2010;21:426-440.
ating cisplatin ototoxicity in children. J Clin Oncol. 2010;28:1788- 81. US Department of Justice, Civil Rights Division, Disability Rights
1795. Section. ADA requirements: effective communication. http://www.
72. National Cancer Institute. Common Terminology Criteria for ada.gov/effective-comm.htm. Accessed September 7, 2015.
Adverse Events. Version 4.0. http://evs.nci.nih.gov/ftp1/CTCAE/ 82. Katzenstein HM, Chang KW, Krailo M, et al; Children’s Oncology
About.html. Accessed September 7, 20015. Group. Amifostine does not prevent platinum-induced hearing loss
73. Theunissen EA, Dreschler WA, Latenstein MN, et al. A new grading associated with the treatment of children with hepatoblastoma: a
system for ototoxicity in adults. Ann Otol Rhinol Laryngol. 2014; report of the Intergroup Hepatoblastoma Study P9645 as a part of
123:711-718. the Children’s Oncology Group. Cancer. 2009;115:5828-5835.
74. Munoz K, Olson WA, Twohig MP, Preston E, Blaiser K, White 83. Marina N, Chang KW, Malogolowkin M, et al; Children’s Oncol-
KR. Pediatric hearing aid use: parent-reported challenges. Ear Hear. ogy Group. Amifostine does not protect against the ototoxicity of
2015;36:279-287. high-dose cisplatin combined with etoposide and bleomycin in pedi-
75. Kulkarni K, Hartley DE. Recent advances in hearing restoration. J R atric germ-cell tumors: a Children’s Oncology Group study. Cancer.
Soc Med. 2008;101:116-124. 2005;104:841-847.
76. Iseli C, Buchman CA. Management of children with severe, severe- 84. Gurney JG, Bass JK, Onar-Thomas A, et al. Evaluation of amifos-
profound, and profound sensorineural hearing loss [published online tine for protection against cisplatin-induced serious hearing loss in
ahead of print August 18, 2015]. Otolaryngol Clin North Am. doi: children treated for average-risk or high-risk medulloblastoma. Neuro
10.1016/j.otc.2015.06.004. Oncol. 2014;16:848-855.

1658 Cancer June 1, 2016

You might also like