Sie sind auf Seite 1von 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/6132727

Recovery from cisplatin-induced ototoxicity: A


case report and review

Article in International Journal of Pediatric Otorhinolaryngology November 2007


DOI: 10.1016/j.ijporl.2007.06.021 Source: PubMed

CITATIONS READS

15 276

3 authors:

Mai Thy Truong Jody Winzelberg


Stanford Medicine Stanford Medicine
23 PUBLICATIONS 956 CITATIONS 1 PUBLICATION 15 CITATIONS

SEE PROFILE SEE PROFILE

Kay W Chang
Stanford University
46 PUBLICATIONS 972 CITATIONS

SEE PROFILE

All content following this page was uploaded by Kay W Chang on 06 August 2017.

The user has requested enhancement of the downloaded file.


International Journal of Pediatric Otorhinolaryngology (2007) 71, 16311638

www.elsevier.com/locate/ijporl

CASE REPORT

Recovery from cisplatin-induced ototoxicity:


A case report and review
Mai Thy Truong a,b,1, Jody Winzelberg a,2, Kay W. Chang a,b,1,*

a
Lucile Packard Childrens Hospital, United States
b
Stanford Hospital and Clinics, United States

Received 4 April 2007; accepted 28 June 2007


Available online 15 August 2007

KEYWORDS Summary We present a pediatric case report of cisplatin-induced ototoxicity with


Cisplatin; subsequent recovery. The patient experienced tinnitus and fluctuating mild high-
Ototoxicity; frequency sensorineural hearing loss (SNHL) with a concomitant decrease in distortion
Pediatric; product otoacoustic emissions (DPOAE). There was recovery of hearing loss and return
Recovery of DPOAE at 1 year after completion of cisplatin therapy. Reports of recovery from
cisplatin-induced ototoxicity in humans are limited in the literature, especially in the
pediatric population. A review of cisplatin ototoxicity and mechanisms of recovery are
discussed, with an emphasis on the particular chemotherapy regimen and dosing
schedule in this case, given at 411 week intervals.
# 2007 Elsevier Ireland Ltd. All rights reserved.

1. Introduction Ototoxic hearing loss has been described as high


frequency, sensorineural with progression to lower
Cisplatin is widely used for the treatment of osteo- frequencies. The incidence of hearing loss has been
sarcoma, hepatoblastoma, neuroblastoma, germ cell reported to be 42% of those receiving higher doses
tumors, head and neck cancers and some central (7085 mg/m2, cumulative dose of 420 mg) with
nervous system tumors. Multiple toxicities have been critical cumulative doses described as low as
reported including, peripheral neuropathy, nephro- 200 mg and up to 400 or 600 mg [2,3].
toxicity, nausea/vomiting and ototoxicity. Ototoxi- Children have been reported to be more suscep-
city has been generally characterized as progressive, tible to ototoxicity, especially with higher dose
irreversible, bilateral hearing loss with tinnitus [1]. regimens (200400 mg/m2) [4,5]. High-frequency
loss is associated with loss of consonant sounds or
* Corresponding author. Tel.: +1 650 736 1314;
fricatives [6]. Hearing loss in children can lead
fax: +1 650 498 2734. to speech and language delay, as well as academic
E-mail addresses: mttruong@stanford.edu (M.T. Truong), difficulties. As more children are surviving their
jwinzelberg@lpch.org (J. Winzelberg), childhood neoplasms, hearing preservation is
kchang@ohns.stanford.edu (K.W. Chang). imperative as well as early identification of hearing
1
801 Welch Road, Stanford, CA 94305, United States.
Tel.: +1 650 736 1314; fax: +1 650 498 2734.
loss for appropriate management [7]. It has
2
1000 Welch Road, Palo Alto, CA 94304, United States. been recommended that close follow-up with pure
Tel.: +1 650 498 2738; fax: +1 650 736 4327. tone audiometry be performed so alterations in

0165-5876/$ see front matter # 2007 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.ijporl.2007.06.021
1632 M.T. Truong et al.

chemotherapy doses can be made to preserve hear- Table 1 Cisplatin dose schedule
ing. It has also been proposed that monitoring 1. 4/8/04: 216 mg
DPOAE is a more sensitive means of monitoring 2. 2/28/04: 212 mg
ototoxicity, with earlier detection than audiometry. 3. 12/11/03: 212 mg
Evidence shows that the cumulative cisplatin dose 4. 11/03/03: 214 mg
as well as method of infusion play roles in extent of
ototoxicity [8]. In this case we show that dosing
schedule also plays a significant role in ototoxicity. The patient then began chemotherapy according
We present a case of ototoxicity that presented with to the POG Protocol 9351, receiving 120 mg/m2 of
tinnitus, fluctuating high-frequency hearing loss and cisplatin planned to be given once approximately
concomitant fluctuating DPOAE with nearly com- every 5 weeks for four treatments. The patient
plete recovery of both pure tone thresholds and received a total cumulative dose of 854 mg
DPOAE at 1 year post treatment. (Table 1). Baseline audiologic evaluation including
pure tone thresholds and DPOAE were performed
prior to initiation of cisplatin and were normal
2. Case report (Fig. 1). The patient developed tinnitus immedi-
ately following infusion of cisplatin. The first mon-
The patient, a 16-year-old male, was referred to itoring audiogram performed after initiation of
Lucile Packard Childrens Hospital after noting right cisplatin (12/18/03) demonstrated a mild sensori-
shoulder pain while playing basketball. After several neural hearing loss above 2 kHz bilaterally, though
weeks of continued pain and swelling, an AP X-ray of the patient denied any appreciable subjective
the right humerus showed a large soft tissue mass. hearing loss (Fig. 2). DPOAE revealed reduced/
An MRI confirmed a large primary bone tumor, 7 cm absent emissions at 3.5 kHz in the left ear (AS)
in the long axis, and 6.2 cm in diameter. There was and at 33.2, 44.5 and 5.5 kHz in the right ear
no evidence of metastatic disease on further (AD). On follow-up 5 weeks later, (1/22/04) with no
workup. An open biopsy confirmed the diagnosis additional cisplatin treatment, the patients tinni-
of osteosarcoma. tus had stopped and his hearing sensitivity and

Fig. 1 Baseline audiogram. Prior to cisplatin induction, the patients hearing thresholds on pure tone audiometry were
within normal limits. dBHL = decibel hearing level, SRT = speech reception threshold, and WR% = percent word recognition.
Cisplatin-induced ototoxicity 1633

Fig. 2 Audiogram performed after initiation of cisplatin therapy. Pure tone audiometry shows a bilateral mild high-
frequency sensorineural hearing loss. Dashed-dotted line shows previous thresholds for comparison. dBHL = decibel
hearing level, SRT = speech reception threshold, and WR% = percent word recognition.

DPOAE were completely within normal limits bilat- frequency range AU at the end of his treatment
erally (Fig. 3). regimen. However, the patient did not develop
He was subsequently followed 1 week after complete recovery of his pure tone thresholds. Six
administration of his third dose of cisplatin therapy. months post treatment the patient still had a
He again reported significant tinnitus bilaterally. His mild SNHL above 2 kHz AD and above 6 kHz AS.
pure tone thresholds demonstrated a 1015 dB DPOAE were reduced at 4 and 5.56 kHz and absent
decrease from 1 to 8 kHz AD and a 1025 dB at 33.2 kHz in the right ear and absent only at
decrease from 1 to 8 kHz AS. DPOAE were reduced 3.5 kHz in the left ear. One year post treatment the
across all frequencies in both ears (AU). Three patients audiogram improved to a mild hearing loss
weeks later the tinnitus had once again resolved. at 68 kHz AD and at 8 kHz AS. DPOAE amplitudes
There was a very mild residual hearing loss above were also slightly improved. Fig. 4 summarizes
4 kHz AD, however, the DPOAE were now robust and the time course of changes in the 8 kHz pure tone
repeatable from 1 to 6 kHz AU and the other pure threshold and the 6 kHz DPOAE in relation to the
tone thresholds were once again within normal cisplatin infusion times.
limits bilaterally.
After the fourth and final course of treatment,
the above reported pattern of results persisted. 3. Discussion
Tinnitus occurred with sensorineural decrease in
hearing although less of an effect was realized in We present a case of a 16-year-old patient treated
terms of diminished DPOAE. Three weeks later, the with cisplatin for osteosarcoma who reported
tinnitus had subsided with a residual mild loss above bilateral tinnitus immediately following cisplatin
4 kHz and present and robust DPOAE across the infusion, followed by bilateral fluctuating high-
1634 M.T. Truong et al.

Fig. 3 Audiogram performed 5 weeks after initiation of cisplatin therapy. Pure tone audiometry shows a return of
thresholds to baseline. Dashed-dotted line shows previous thresholds for comparison. dBHL = decibel hearing level,
SRT = speech reception threshold, and DNT = did not test.

frequency sensorineural hearing loss (HFSNHL) with 4. Cisplatin ototoxicity


reduced/absent DPOAE. He received four doses of
cisplatin for a cumulative dose of 854 mg over 23 Platinum compounds were first recognized by Rosen-
weeks, given in 411 week intervals. The patient burg and Calalieri in 1964 for their therapeutic
demonstrated a pattern of tinnitus, hearing loss, potential, though clinical trials of cisplatin as a
and reduced/absent DPOAE that showed recovery chemotherapeutic agent did not begin until 1971.
over the subsequent weeks. This pattern would Final approval for its use in human cancer therapy
repeat itself with the each cycle of cisplatin. After was achieved in 1978. Ototoxicity was recognized
the fourth dose, he maintained a mild HFSNHL at early in case reports in 1972, and cisplatin has been
6 months, with partial recovery at 1-year post identified as the most ototoxic of all platinum com-
treatment. pounds [9]. Predisposing factors for ototoxicity
In this case report, we document the fluctuating reported are dose, duration, mode of administra-
nature of cisplatin-induced ototoxic hearing loss, tion, age extremes, previous or concurrent cranial
with interval periods of recovery, followed by recov- irradiation, previous history of hearing loss, renal
ery at 1-year post treatment. This may represent a disease, concurrent use of other ototoxic agents,
dosing schedule that allows for cochlear recovery, and volume status. Cisplatin ototoxicity has been
demonstrating that ototoxicity is not irreversible, characterized clinically (Table 2) [10].
but may occur as a graduated process, and that dose Cisplatin is a square-planar platinum molecule
and dosing schedule may affect the permanence of with pairs of chlorine atoms and amine groups
the loss and the ability of the cochlea to recover. arranged in a cis position, allowing for binding to
This report also illustrates the potential utility of the N7 atom of purine nucleic acids in DNA. This cross-
DPOAE to monitor ototoxicity. linking inhibits proper DNA replication and transcrip-
Cisplatin-induced ototoxicity 1635

Table 2 Classic clinical presentation of cisplatin ototoxicity


Tinnitus
Bilateral hearing loss
High-frequency sensorineural hearing loss with progression to lower frequencies
Permanent and irreversible loss in humans, with recovery documented in animal studies
Ototoxicity after a total cumulative dose of >200 mg/m2, but reported as low as 60 mg/m 2
Lowered endocochlear potential
Decrease in DPOAE
Decrease in cochlear microphonic

tion, ultimately leading to apoptosis [11]. Binding [1921]. It has been proposed that damage to the
also occurs in non-DNA targets such as essential marginal cells of the stria vascularis secondarily
proteins, inducing free radical generation and anti- causes damage to the OHC [22,23]. In a study by
oxidant inhibition [12,13]. It is likely that this anti- Miyasha et al., photochemically induced damage to
neoplastic mechanism is the same mechanism the cochlea with Rose Bengal dye and green light
responsible for the ototoxic, nephrotoxic, neurotoxic irradiation resulted in marginal cell extrusion and
and gastro-intestinal side effects [10]. Cisplatin rupture, followed by OHC damage 12 or more hours
undergoes urinary excretion, concentrates in the after strial insult as seen by TEM. OHCs were noted
liver, and has a low penetration to the CNS. The to have disruption of the cell membrane at the
half-life is variable due to extensive plasma binding, cuticular plate, followed by disappearance of OHCs
ranging from 6 to 47 days [14]. The incidence of by 30 days. The mechanism of this damage is
hearing loss has been reported to be 42% of those reported to be the photochemical production of a
receiving higher doses (7085 mg/m2, cumulative reactive oxygen species leading to endothelial
dose of 420 mg) with critical cumulative doses damage, platelet aggregation, thrombus formation,
described as low as 200, 400, or 600 mg [2,3]. and ischemic changes leading to marginal cell
Cisplatin ototoxicity has clearly been associated damage. In a follow-up study demonstrating delayed
with outer hair cell (OHC) death. Early studies in OHC damage and concurrent increase in compound
rhesus monkeys demonstrated OHC loss in basal action potential (CAP) thresholds after photoche-
turns of the cochlea by histopathology [15]. Animal mically induced strial damage, Ocho et al. proposed
studies have further characterized histopathologi- that hair cell damage is secondarily caused by strial
cal changes in the cochlea. Nakai et al. described degeneration.
changes seen by SEM and TEM in the guinea pig Thomas et al. identified accumulation of plati-
model, summarized in Table 3 [16]. Human cadaver numDNA adducts by immunoflorescence exclu-
studies of the temporal bones have confirmed these sively in the nuclei of marginal cells in the stria
cochlear findings [17]. Low dose cisplatin has also vascularis 448 h after infusion of cisplatin in gui-
been reported to cause damage to OHC stereocilia nea pigs [1921]. This study clearly identifies strial
[17]. OHC damage and loss of OHC function has been marginal cells as the main target of cisplatin-
suggested to trigger hyperactivity in the dorsal induced ototoxicity. No staining was seen in OHCs,
cochlear nucleus, a potential cause for ototoxi- demonstrating that OHCs had no accumulation of
city-related tinnitus [18]. cisplatinDNA adducts. Thomas proposed that mar-
ginal cell damage leads to impaired potassium
secretion into endolymph. This change in cochlear
5. Pathophysiology of cisplatin current is reflected in a decrease in endocochlear
ototoxicity: early damage to the stria potential (EP) after cisplatin infusion, which
vascularis progresses to OHC death directly affects sensorineural transduction by hair
cells. Fluctuating changes in EP following cisplatin
Several studies have shown that damage to the stria infusion in guinea pigs has been described [24].
vascularis precedes OHC loss in cisplatin ototoxicity Similarly, ethacrynic acid, furosemide and ouabain

Table 3 Histopathological characteristics of cisplatin ototoxicity (Nakai et al., 1982)


OHC loss in the first turn of the cochlea, with progression to the 2, 3, and 4th turns with higher doses
OHC loss in a sporadic fashion with lower doses of cisplatin progressing to continuous OHC loss with higher doses
Evidence of damage of remaining OHC: blebs on the surface, enlargement of vacuoles, denaturation of mitochondria
and leakage of cellular contents with surrounding cells
Vacuole enlargement and thinning of stria vascularis
1636 M.T. Truong et al.

to outer hair cell damage and death is also unclear.


However, the progression from strial damage to OHC
loss may be a graduated process that may provide a
mechanism to explain the reversibility of hearing
loss that has been described in animal models as
well as few human case reports including this report
[8,2830]. Marginal cells are capable of DNA repair
and reducing peak platinum adduct burden via
reparative mechanisms as described by Siddik
et al. [11]. Cisplatin-induced reversible acute hear-
ing loss may be due to reversible strial failure,
whereas the survival of the OHC determines the
final degree of functional recovery [31]. Ototoxicity
may be reversible if sufficient time for reparative
processes to occur is provided (Fig. 5).
Recovery of electrophysiological cochlear poten-
tial from cisplatin ototoxicity has been described in
the guinea pig [32,33]. Recovery in humans has been
reported by Aguilar-Markus, Laurell, and Vermorken
[8,28,29]. More recently, Zuur et al. described 27
ears that demonstrated recovery of hearing loss at
1, 2, and 4 kHz after chemoradiation for head and
neck cancer patients [30].

Fig. 4 Summary of audiometric and DPOAE results over


time. Fluctuating hearing loss after cisplatin therapy is
demonstrated in each ear. Hearing loss is shown as changes
from baseline in decibels and can be correlated to time of
cisplatin infusion (red arrows). (For interpretation of the
references to color in this figure legend, the reader is
referred to the web version of the article.)

infusion have been shown to decrease the potassium


concentration in endolymph, with a measured
decrease in EP, and subsequent hair cell degenera-
tion [25,26].
Marginal cells of the stria are hexagonal cells
facing the endolymphatic space containing high
concentrations of sodiumpotassium ATPases and
mitochondria, which are required for pumping
potassium into the endolymph [10,27]. The stria
vascularis is also rich in capillary supply. The high
cellular activity and rich vascular supply may make
these cells more susceptible to ototoxic damage.

6. Recovery of ototoxicity Fig. 5 Proposed mechanism of reversible and irreversi-


ble cisplatin ototoxicity. Marginal cell death leads to
Why cisplatin accumulates in strial marginal cells in fluctuations in endocochlear potential that result in hear-
not clear, though it is hypothesized that cisplatin is ing loss which may be reversible. Cisplatin given without
actively transported into these cells and trapped sufficient time for cell reparative mechanisms will lead to
[21]. The exact mechanism of strial damage leading outer hair cell death resulting in permanent hearing loss.
Cisplatin-induced ototoxicity 1637

7. Monitoring for ototoxicity: pure cisplatin is a feasible treatment option: analysis on prog-
nostic factors for toxicity in 400 patients, Br J Cancer 88
tone audiometry, ABR, DPOAE (2003) 1199.
[3] C. Bokemeyer, C.C. Berger, J.T. Hartmann, C. Kollmanns-
Pure tone behavioral audiometry remains the stan- berger, H.J. Schmoll, M.A. Kuczyk, et al., Analysis of risk
dard means to monitor for ototoxicity and hearing factors for cisplatin-induced ototoxicity in patients with
testicular cancer, Br J Cancer 77 (1998) 1355.
loss. Since cisplatin ototoxicity preferentially
[4] P. Bertolini, M. Lassalle, G. Mercier, M.A. Raquin, G. Izzi, N.
affects the basal end of the cochlea, high-frequency Corradini, et al., Platinum compound-related ototoxicity in
audiometry is a more sensitive method of detecting children: long-term follow-up reveals continuous worsening
early ototoxicity [34,35]. The pediatric oncology of hearing loss, Pediatr Hematol Oncol 26 (2004) 649.
population is frequently difficult to test behavio- [5] V.A. McHaney, G. Thibadoux, F.A. Hayes, A.A. Green, Hear-
rally, and therefore auditory brainstem response ing loss in children receiving cisplatin chemotherapy, J
Pediatr 102 (1983) 314.
(ABR) and DPOAE are sometimes useful objective [6] P.G. Stelmachowicz, A.L. Pittman, B.M. Hoover, D.E. Lewis,
studies that can be obtained. Tone burst threshold M.P. Moeller, The importance of high-frequency audibility in
ABR studies are preferred, since the broadband click the speech and language development of children with
stimulus may miss isolated high-frequency loss hearing loss, Arch Otolaryngol Head Neck Surg 130 (2004)
which is typical for cisplatin ototoxicity. High-fre- 556.
[7] K.R.G. Knight, D.F. Kraemer, E.A. Neuwelt, Ototoxicity in
quency tone-burst-evoked ABR have been found to children receiving platinum chemotherapy: underestimating
be even more useful in identifying early ototoxicity a commonly occurring toxicity that may influence academic
changes [36,37]. A number of researchers have and social development, J Clin Oncol 23 (2005) 8588.
found DPOAE, a specific measure of OHC activity, [8] J.B. Vermorken, T.S. Kapteijn, A.A. Hart, H.M. Pinedo,
Ototoxicity of cis-diamminedichloroplatinum (II): influence
a particularly useful and more easily administered
of dose, schedule and mode of administration, Eur J Cancer
modality to monitor for early cisplatin otoxicity 19 (1983) 53.
[3840]. As seen in this case (Fig. 4), high-frequency [9] A.H. Rossof, R.E. Slayton, C.P. Perlia, Preliminary clinical
(6 kHz) DPOAE amplitudes correlate well with experience with cis-diamminedichloroplatinum (II) (NSC
changes in high-frequency audiometric thresholds, 119875 CACP), Cancer 30 (1972) 1451.
while the low frequency (1 kHz) DPOAE amplitudes [10] P.S.R.J. Roland, Ototoxicity, BC Decker Inc., London, 2004,
p. 212.
are stable and can be used as a verification of the [11] Z.H. Siddik, Cisplatin: mode of cytotoxic action and mole-
validity of the OAE measure. cular basis of resistance, Oncogene 22 (2003) 7265.
[12] P. Evans, B. Halliwell, Free radicals and hearing. Cause,
consequence, and criteria, Ann NY Acad Sci 884 (1999) 19.
8. Conclusions [13] M.J. McKeage, Comparative adverse effect profiles of pla-
tinum drugs, Drug safety 13 (1995) 228.
[14] R. Safirstein, M. Daye, J.B. Guttenplan, Mutagenic activity
In this case report, we present a pediatric case of and identification of excreted platinum in human and rat
recovery of mild high-frequency sensorineural hear- urine and rat plasma after administration of cisplatin, Can-
ing loss after cisplatin ototoxicity. The patient cer Lett 18 (1983) 329.
reported tinnitus immediately following the initia- [15] S.W. Stadnicki, R.W. Fleischman, U. Schaeppi, P. Merriam,
tion of cisplatin therapy, followed by a dosing sche- Cis-dichlorodiammineplatinum (II) (NSC-119875): hearing
loss and other toxic effects in rhesus monkeys, Cancer
dule of 411 week intervals between doses, allowing Chemother Rep 59 (1975) 467.
for appropriate time for reparative processes in the [16] Y.Y. Nakai, K.K. Konishi, K.C. Chang, K. Ohashi, N. Morisaki,
cochlea. In this case, DPOAE did not provide earlier Y. Minowa, et al., Ototoxicity of the anticancer drug cispla-
detection of hearing loss, however correlated very tin. An experimental study, Acta Oto-Laryngol 93 (1982) 227.
closely to audiometric threshold changes. Following [17] M. Strauss, J. Towfighi, S. Lord, A. Lipton, H.A. Harvey, B.
Brown, Cis-platinum ototoxicity: clinical experience and tem-
documented hearing loss or clinical complaints of poral bone histopathology, Laryngoscope 93 (1983) 1554.
tinnitus, delay of the next dose of cisplatin should [18] J.A. Kaltenbach, R.J. Mathog, T.A. Zhang, J. Falzarano, P.R.
be considered until clinical improvement is shown. Lewandowski, Cisplatin-induced hyperactivity in the dorsal
Allowing the stria vascularis to recover may result in cochlear nucleus and its relation to outer hair cell loss:
less OHC loss, and thus lesser degree of permanent relevance to tinnitus, J Neurophysiol 88 (2) (2002) 699714.
[19] S. Sluyter, S.F. Klis, J.C. de Groot, G.F. Smoorenburg, Altera-
irreversible hearing loss. tions in the stria vascularis in relation to cisplatin ototoxicity
and recovery, Hearing Res 185 (2003) 49.
[20] R.A. Tange, V.D. Vuzevski, Changes in the stria vascularis of
the guinea pig due to cis-platinum, Archives of Oto-Rhino-
References Laryngology 239 (1984) 41.
[21] J.P. Thomas, J. Lautermann, B. Liedert, F. Seiler, J. Tho-
[1] P. Brock, S. Bellman, Ototoxicity of cisplatinum, Br J Cancer male, High accumulation of platinum-DNA adducts in strial
63 (1991) 159. marginal cells of the cochlea is an early event in cisplatin
[2] F.E. de Jongh, R.N. van Veen, S.J. Veltman, R. de Wit, M.E. but not carboplatin ototoxicity, Mol Pharmacol. 70 (2006)
van der Burg, M.J. van den Bent, et al., Weekly high-dose 2329.
1638 M.T. Truong et al.

[22] H. Miyashita, S. Iwasaki, T. Hoshino, Photochemically [32] C.H. Stengs, S.F. Klis, E.H. Huizing, G.F. Smoorenburg,
induced focal cochlear lesions in the guinea pig. Part II. A Cisplatin-induced ototoxicity; electrophysiological evi-
transmission electron microscope study, Microsc Res Tech 41 dence of spontaneous recovery in the albino guinea pig,
(1998) 334. Hearing Res 111 (1997) 103.
[23] S. Ocho, S. Iwasaki, K. Umemura, T. Hocino, A new model for [33] R.M. Cardinaal, J.C. de Groot, E.H. Huizing, J.E. Veldman,
investigating hair cell degeneration in the guinea pig follow- G.F. Smoorenburg, Cisplatin-induced ototoxicity: morpholo-
ing damage of the stria vascularis using a photochemical gical evidence of spontaneous outer hair cell recovery in
reaction, Eur Arch Otorhinolaryngol 257 (2000) 182. albino guinea pigs? Hearing Res 144 (2000) 147.
[24] S.F. Klis, S.J. OLeary, F.P. Hamers, J.C. de Groot, G.F. [34] S.A. Fausti, V.D. Larson, D. Noffsinger, R.H. Wilson, D.S.
Smoorenburg, Reversible cisplatin ototoxicity in the albino Phillips, C.G. Fowler, High-frequency audiometric monitor-
guinea pig, Neuroreport 11 (2000) 623. ing strategies for early detection of ototoxicity, Ear Hear 15
[25] S.K. Bosher, C. Smith, R.L. Warren, The effects of ethacrynic (1994) 232.
acid upon the cochlear endolymph and stria vascularis. A [35] S.A. Fausti, J.A. Henry, H.I. Schaffer, D.J. Olson, R.H. Frey,
preliminary report, Acta Otolaryngol 75 (1973) 184. G.C. Bagby Jr., High-frequency monitoring for early detec-
[26] T. Konishi, M. Mendelsohn, Effect of ouabain on cochlear tion of cisplatin ototoxicity, Arch Otolaryngol Head Neck
potentials and endolymph composition in guinea pigs, Acta Surg 119 (1993) 661.
Oto-Laryngol 69 (1970) 192. [36] S.A. Fausti, R.H. Frey, J.A. Henry, D.J. Olson, H.I. Schaffer,
[27] A.N. Salt, I. Melichar, R. Thalmann, Mechanisms of endoco- Early detection of ototoxicity using high-frequency, tone-
chlear potential generation by stria vascularis, Laryngo- burst-evoked auditory brainstem responses, J Am Acad
scope 97 (1987) 984. Audiol 3 (1992) 397.
[28] N.V. Aguilar-Markulis, S. Beckley, R. Priore, C. Mettlin, [37] S.A. Fausti, R.H. Frey, J.A. Henry, D.J. Olson, H.I. Schaffer,
Auditory toxicity effects of long-term cis-dichlorodiammi- High-frequency testing techniques and instrumentation for
neplatinum II therapy in genitourinary cancer patients, J early detection of ototoxicity, J Rehabil Res Dev 30 (1993) 333.
Surg Oncol 16 (1981) 111. [38] R. Toral-Martinon, P. Shkurovich-Bialik, M.A. Collado-Cor-
[29] G. Laurell, U. Jungnelius, High-dose cisplatin treatment: ona, I. Mora-Magana, S. Goldgrub-Listopad, M. Shkurovich-
hearing loss and plasma concentrations, Laryngoscope 100 Zaslavsky, Distortion product otoacoustic emissions test is
(1990) 724. useful in children undergoing cisplatin treatment, Arch Med
[30] C.L. Zuur, Y.J. Simis, P.E. Lansdaal, C.R. Rasch, R.A. Tange, Res 34 (2003) 205.
A.J. Balm, et al., Audiometric patterns in ototoxicity of [39] B.D. Ress, K.S. Sridhar, T.J. Balkany, G.M. Waxman, B.B.
intra-arterial cisplatin chemoradiation in patients with Stagner, B.L. Lonsbury-Martin, Effects of cis-platinum che-
locally advanced head and neck cancer, Audiol Neurootol motherapy on otoacoustic emissions: the development of an
11 (2006) 318. objective screening protocol. Third placeResident Clinical
[31] F.P. Hamers, J. Wijbenga, F.L. Wolters, S.F. Klis, S. Sluyter, Science Award 1998, Otolaryngol Head Neck Surg 121 (1999)
G.F. Smoorenburg, Cisplatin ototoxicity involves organ 693.
of Corti, stria vascularis and spiral ganglion: modulation [40] O. Ozturan, J. Jerger, H. Lew, G.R. Lynch, Monitoring of
by alphaMSH and ORG 2766, Audiol Neurootol 8 (2003) cisplatin ototoxicity by distortion-product otoacoustic emis-
305. sions, Auris Nasus Larynx 23 (1996) 147.

View publication stats

Das könnte Ihnen auch gefallen