Sordera
Sordera
Sordera
Hearing Loss
Seiji B. Shibata | A. Eliot Shearer | Richard J.H. Smith
Key Points
■ In developed countries, more than 50% of congenital hearing impairments have a genetic origin.
■ Hearing loss can be defined by numerous clinical criteria that include causality, time of onset, age of
onset, clinical presentation, anatomic defect, severity, and frequency of loss.
■ The worldwide rate of profound hearing loss is 4 in every 10,000 infants born.
■ The basic forms of inheritance can be mendelian (single-gene inheritance—autosomal or X-linked),
mitochondrial, or complex (multifactorial inheritance).
■ Nonsyndromic hearing impairment in the absence of other phenotypic manifestations accounts for
70% of hereditary hearing losses.
■ Mutations in GJB2 account for 50% of severe-to-profound autosomal-recessive nonsyndromic
deafness in several world populations.
■ Syndromic hearing impairment refers to deafness that cosegregates with other features and results in
a recognizable constellation of findings known as a syndrome. Sensorineural deafness has been
associated with more than 400 syndromes.
■ The most common syndromic form of hereditary sensorineural hearing loss, Pendred syndrome, is
inherited in an autosomal-recessive fashion; affected individuals also have goiter.
■ After a well-performed patient history, physical examination, and audiologic evaluation, genetic
testing should be the next test ordered in the evaluation of a patient with hearing loss.
■ Prenatal diagnosis of some forms of hereditary hearing loss is technically possible by analysis of
DNA extracted from fetal cells.
■ Cochlear implantation is becoming an increasingly important option for individuals with severe to
profound deafness; in many cases, cochlear implantation can be predicted by genetic testing.
2285
2286 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
FUNDAMENTALS OF GENETICS
Each person has 46 chromosomes—22 pairs of autosomes and
one pair of sex chromosomes (XY in males or XX in females).
According to Mendel’s principle of segregation, in sexually
reproducing organisms, each partner contributes only one
member of each chromosome pair to an offspring. This fact C
means that each individual has inherited one copy of each
chromosome from each parent. Carried on these chromosomes
are genes, estimated to number approximately 30,000. Varia-
tions in these genes impart uniqueness to each individual.
These variations, termed alleles, can sometimes be deleterious.
If a mutation changes the normal or wild-type sequence of
a gene expressed in the inner ear, and if the protein translated
from this new allele variant does not function as well as the
normal protein, deafness may result. However, if the remaining D
normal protein (recall that each gene is represented in dupli-
FIGURE 148-1. Inheritance patterns. A, Pedigree illustrating autosomal-
cate) is able to replace or compensate for the defective mutant
dominant inheritance. The inheritance pattern shows vertical transmission of
protein, deafness will manifest only in individuals who carry two the affected phenotype, including father-to-son transmission. No generations
copies of the abnormal gene. This scenario is an example of are skipped. B, Pedigree of autosomal-recessive inheritance. Note the
autosomal-recessive inheritance. However, if the abnormal protein disease phenotype is not usually seen in the parents or other ancestors.
interferes with normal protein function, deafness will manifest Likelihood of affected males to females is equal. (Refer to the Punnett square
in an individual who carries a single abnormal gene. This sce- in Figure 148-2 for estimating risks for recessive inheritance in heterozygous
nario is an example of autosomal-dominant inheritance. parents.) C, Pedigree of X-linked–recessive inheritance. The disease pheno-
In describing these inheritance patterns, we are describing type is present with one disease allele in the male but requires a homozygous
a person’s genotype or genetic makeup. The term homozygosity genotype to be expressed in the female. D, Pedigree illustrating mitochon-
drial inheritance. Note the maternal transmission of the affected phenotype.
means that a person carries two identical alleles of a gene;
There is no paternal transmission.
heterozygosity represents the state in which a person carries two
different variants of a given gene. The observable consequence
that derives from an individual’s genotype is his or her pheno- of inheritance, we recommend more appropriate texts.12,13
type. Individuals who carry identical genetic mutations often Punnett squares are used to show inheritance patterns by dis-
exhibit a spectrum of phenotypic features. For instance, not playing the outcomes of crosses with both parents and allowing
every person with Waardenburg syndrome type 1 has synoph- probability estimates for the offspring (Fig. 148-2).
rys, premature graying of the hair, or heterochromia irides, a
phenomenon called variable expressivity. In some cases, the phe- Autosomal Dominant
notype can be so subtle as to be entirely absent, and the caus- In autosomal-dominant diseases, heterozygotes express the
ative genetic mutation is said to exhibit incomplete penetrance or disease phenotype. The affected parent can pass either a disease
to be nonpenetrant. This occurrence can give the impression that allele or a normal allele to offspring, with the likelihood of each
a disease “skips” generations. event being 0.5, or 50%. This means that 50% of offspring
inherit the normal allele and 50% inherit the disease allele. In
the case of dominant diseases, 50% of offspring express the
PATTERNS OF INHERITANCE disease.
The basic forms of inheritance can be mendelian (single-gene Autosomal-dominant transmission implies several things.
inheritance, autosomal or X-linked), mitochondrial, or complex First, no sexual predilection exists for the disease phenotype;
(chromosomal and multifactorial inheritance). Pedigrees for males and females are equally likely to be affected and to trans-
these inheritance patterns are shown in Figure 148-1. Mende- mit the disease allele to their offspring. Second, unless the
lian and mitochondrial forms of inheritance are discussed in disease gene is nonpenetrant, the disease does not skip genera-
this chapter; for the reader who is interested in complex forms tions. This type of transmission is called vertical transmission.
2288 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
process is not random, and inactivation of the mutated X chro-
Male heterozygous carrier mosome is usually much higher than is inactivation of the
normal X chromosome. Males have only a single X chromo-
A a some and always express the disease phenotype.
In a pedigree showing X-linked–recessive inheritance, the trait
is seen more frequently in males; there is no father-son trans-
mission; affected fathers transmit the disease allele to all female
Aa offspring, who may have affected males (“skipped generation”);
AA
Female heterozygous carrier
A
and heterozygous females transmit the disease allele to half of
Not affected
Carrier all sons, who manifest the disease, and half of all daughters,
not affected who are heterozygous and phenotypically normal. In X-linked–
dominant inheritance, affected fathers transmit the disease allele
to all daughters who exhibit the disease phenotype with com-
plete penetrance; there is no father-son transmission. Hetero-
zygous females are affected and transmit the trait to half of all
sons and half of all daughters.
Aa
aa
a Mitochondrial Inheritance
Carrier
Affected The “powerhouses” of the human cell, mitochondria are the
not affected
sites of oxidative phosphorylation, the process that leads to the
production of adenosine triphosphate. Mitochondria possess
their own intrinsic DNA, with several copies of the mitochon-
FIGURE 148-2. Punnett squares are used to estimate inheritance probabili- drial genome in each mitochondrion. The mitochondrial
ties for mendelian inheritance patterns. This Punnett square illustrates the genome is a 16,569 base pair circular molecule that encodes
mating of two individuals who are heterozygous carriers of an autosomal- two ribosomal RNAs, 22 transfer RNAs, and 13 polypeptides.14
recessive gene; the genotypes of all potential offspring show a 25% chance The remaining mitochondrial proteins required for oxidative
of having a child with the recessive phenotype (aa), a 50% chance of having phosphorylation are encoded by the nuclear genome.
a child who carries the recessive allele but is not affected (Aa), and a 25% Mitochondrial DNA (mtDNA) is inherited solely through
chance of having a child who does not carry the recessive allele (AA). the maternal lineage, reflecting the presence of large amounts
of mtDNA in the cytoplasm of the egg. Therefore, maternal
transmission of the affected phenotype occurs, but no paternal
transmission takes place (see Fig. 148-1, D). If all of the mtDNA
Third, male-to-male (father-son) transmission is seen. This molecules are abnormal, a condition known as homoplasmy, all
observation in a pedigree rules out mitochondrial or X-linked progeny cells contain abnormal mitochondria. If normal and
inheritance. abnormal mtDNA molecules coexist, a condition known as het-
eroplasmy, a wide range of expression of the mutant phenotype
Autosomal Recessive is seen, which reflects the random distribution of mitochondria
In contrast to autosomal-dominant inheritance, in autosomal- to progeny cells. Because mitochondria lack a DNA repair
recessive inheritance, two mutant copies of a gene are necessary mechanism, mtDNA accumulates mutations at a higher rate
for expression of the disease phenotype. An affected parent than nuclear DNA.
transmits a disease allele to all offspring, but offspring do not
show the disease phenotype unless the other parent carries at
least one mutant copy of the same gene. Most frequently, GENETIC HEARING IMPAIRMENT
however, neither parent is affected, but both carry a single
mutant gene, and by chance, each passes this mutant copy on NONSYNDROMIC HEARING IMPAIRMENT
to the affected offspring. In this scenario, 25% of offspring Genetic hearing loss is common in humans. Nonsyndromic
carry two mutant copies of the gene and express the disease hearing impairment in the absence of other phenotypic mani-
phenotype; 50% of offspring are carriers of a mutant copy, festations accounts for 70% of hereditary hearing loss.15 More
similar to the parents; and 25% of offspring have two wild-type than half of SNHL that occurs in neonates is attributable to
copies of the gene. As with autosomal-dominant diseases, there simple mendelian inherited traits (Fig. 148-3). In most cases,
is no sexual predilection; males and females are equally likely the inheritance pattern is recessive (75% to 80% of cases), and
to be affected, but vertical transmission is rarely seen. Recessive consequently, the parents of affected children generally do not
diseases tend to be generation specific. If the disease is excep- exhibit the phenotype. Congenital nonsyndromic hearing
tionally rare, the likelihood of parental consanguinity, albeit loss is inherited in an autosomal-dominant (~20%), X-linked
distant, is high. (2% to 5%), or mitochondrial (~1%) mode. Nomenclature is
based on the prefix “DFN” to designate nonsyndromic DeaFNess.
X-Linked Inheritance DFN followed by an A implies dominant inheritance, whereas
X-linked inheritance may be either recessive or dominant. In B implies recessive inheritance and X implies X-linked inheri-
recessive X-linked inheritance, females are unlikely to be tance. The integer suffix denotes the order of gene locus dis-
affected, because two mutant copies of the gene are needed for covery. DFNA1 and DFNB1 were the first autosomal-dominant
the disease phenotype to manifest. However, heterozygous and recessive nonsyndromic deafness gene loci to be identified.
females do occasionally exhibit subtle aspects of the disease As is described in the following sections, great progress has
phenotype because of the randomness of X-chromosome inac- been made in the scientific study of genetic nonsyndromic
tivation (the Lyon hypothesis). In females, in each cell, only hearing loss (NSHL) since the discovery of the first deafness
one of the two X chromosomes is active, and when X chromo- gene in 1993. These discoveries have improved our understand-
some inactivation is entirely random, 50% of cells in a hetero- ing of the molecular physiology of hearing and deafness and
zygous female will express the disease-carrying X chromosome. have laid the groundwork for clinical genetic testing for deaf-
Through mechanisms that are not clearly understood, this ness, as described under Diagnosis.
148 | GENETIC SENSORINEURAL HEARING LOSS 2289
50% Genetic
2%-5% X-linked, <1% mitochodrial
Congenital
hearing loss 30% Syndromic
50% Environmental
Autosomal-Recessive Nonsyndromic DFNB1. In 1994, Guilford and coworkers17 mapped the first
Hearing Impairment autosomal-recessive nonsyndromic deafness locus to 13q12-13
Autosomal-recessive nonsyndromic hearing impairment is and called it DFNB1. Three years later, Kelsell and colleagues18
usually prelingual and severe to profound across all frequen- identified the DFNB1 gene as a gap junction gene called GJB2.
cies.16 To date, 95 loci have been mapped, and 41 causative The encoded protein, connexin 26, oligomerizes with five
genes have been cloned (Table 148-3).11 Expression patterns of other connexin proteins to form a connexon; the docking of two
autosomal-recessive nonsyndromic deafness-related genes in connexons in neighboring cells results in a gap junction (see
the cochlea are demonstrated in Figure 148-4. Fig. 148-4, B). These gap junctions are thought to be conduits
Continued
2290 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
through which potassium ions are recycled from the outer hair
cells back through the supporting cells and spiral ligament to
Autosomal-Dominant Nonsyndromic
the stria vascularis.19 The ions are pumped into the endolymph Hearing Impairment
to perpetuate the mechanosensory transduction of hair cells. To date, 64 loci for autosomal-dominant nonsyndromic deafness
Mutations in GJB2 may disrupt this recycling process and may have been mapped, and 27 causative genes have been cloned
prevent normal mechanosensory transduction.18 This role is (Table 148-4).11 Generally, the onset of deafness is postlingual,
consistent with the expression of GJB2 in the stria vascularis, progressive, and milder than recessive forms, and several loci
nonsensory epithelial cells, spiral ligament, and spiral limbus have characteristic audioprofiles.28,29 Several common forms
of the inner ear.18 of dominant hearing impairment have unique or characteristic
Mutations in GJB2 are found in 50% of people with audioprofiles. One of the most common types of autosomal-
severe to profound congenital autosomal-recessive nonsyn- dominant nonsyndromic deafness is high-frequency hearing
dromic deafness in many world populations.20 More than 100 loss as a result of KCNQ4 mutations at the DFNA2 locus.30 Expres-
different deafness-causing mutations are described, and several sion patterns of autosomal-dominant nonsyndromic deafness–
are common in specific ethnic groups.21 For example, the related genes in the cochlea are demonstrated in Figure 148-4.
35delG mutation predominates in populations of European
descent22; this mutation has a carrier frequency in the Midwest- DFNA2 (High-Frequency Hearing Loss). Autosomal-dominant
ern United States of 2.5%.23 For comparison, in the Ashkenazi high-frequency hearing loss can be the consequence of muta-
Jewish population, the most common mutation is the 167delT; tions in a variety of genes that include KCNQ4 (DFNA2), DFNA5
its carrier frequency is approximately 4%.24 In Japanese popula- (DFNA5), COCH (DFNA9), and POU4F3 (DFNA15). Mutations
tions, the 235delC mutation is the most common deafness- in many of these genes cause autosomal-dominant deafness
causing allele variant of GJB2.25 through a dominant-negative mechanism of action.30-32 One
The DFNB1 auditory phenotype is varied, although homo- example is deafness at the DFNA2 locus, which is caused by
zygosity for protein-truncating mutations is generally associated dominant-negative mutations in KCNQ4.30,31 KCNQ4 is orga-
with impaired gap junction activity and moderate to profound nized into 14 exons that encode a protein with six transmem-
deafness.21-26 With missense mutations, the degree of residual brane domains and a P-loop to confer Κ+ ion selectivity to the
hearing can be substantially greater.21 Most frequently, the loss channel pore.31 A voltage sensor in the fourth transmembrane
is symmetric between ears and does not progress over the long domain drives a conformational change that leads to channel
term. Temporal bone anomalies are not part of the DFNB1 opening. KCNQ4 subunits are typically organized into homotet-
phenotype, which obviates the need for routine temporal bone ramers to form functional channels.31
imaging. The G285S allele was the first DFNA2 mutation identified,
Genetic testing is available to diagnose GJB2-related deaf- and a mouse model carrying the orthologous mutation (equiva-
ness and is warranted, because the relative contribution of this lent to the human G285S allele variant) has been generated.33
gene to the total genetic deafness mutation load is high. Muta- The substitution of a serine for a glycine affects the first highly
tion screening facilitates genetic counseling and prediction of conserved glycine in a GYG signature sequence of the P-loop of
the chance of recurrence. It also provides prognostic informa- the channel pore, and it abolishes channel function by prevent-
tion, because several studies have shown that cochlear implant ing correct subunit assembly. Impaired KCNQ4 function in the
recipients with GJB2-related deafness do very well.27 inner ear affects Κ+ ion recycling. Normally, mechanosensory
148 | GENETIC SENSORINEURAL HEARING LOSS 2291
FIGURE 148-4. Schematic illustration of cochlear duct cross-section includes an enlarged representation of two important cochlear structures (A and B)
and shows the cochlear expression pattern of genes related to deafness. A, Inset illustrates the stereocilia tip link, mechanotransduction channel, and interac-
tion between anchoring proteins Harmonin-b, MYO1C, CDH23, and PCDH15. B, The gap junction and its constituents. Three-dimensional view of a connexon
hexamer composed of six connexin monomers; each connexin encoded by GJB2 is also known as a connexin 26 molecule, one of which is highlighted. Gap
junctions are composed of two connexons in adjacent cells. Small molecules can pass through the gap junction pore from one cytoplasm to another without
having to cross two cell membranes. 1, Inner hair cells: ACTG1, CDH23, CLDN14, GJA1, GIPC3, ILDR1, KCNQ4, LOXHD1, MYH14, MYO3A, MYO6, MYO7A,
MYO15A, PCDH15, POU4F3, PTPRQ, OTOF, RDX, SERPINB6, STRC, TFCP2L3, TJP2, TMC1, TMHS, TRIOBP, USH1C, and WFS1. 2, Outer hair cells: ACTG1,
CCDC50, CDH23, CLDN14, GIPC3, GJA1, ILDR1, KCNQ4, LOXHD1, MYH9, MYH14, MYO3A, MYO6, MYO7A, MYO15A, OTOF, PCDH15, POU4F3, PTPRQ, PRES,
RDX, STRC, SLC26A5, TFCP2L3, TJP2, TMC1, TMHS, TRIOBP, USH1C, and WFS1. 3, Supporting cells: CLDN14, ESRRB, GJA1, GJB2, GJB6, MYH14, PCDH15,
SLC26A4, TFCP2L3, TMPRSS3, and WFS1. 4, Stereocilia: CDH23, DFNB18, ESPN, PCDH15, STRC, TMIE, and WHRN. 5, Interdental cells: ATP6B1, GJA1,
GJB2, TFCP2L3, and WFS1. 6, Spiral limbus: COCH, COL9A1, CRYM, ESRRB, GJB2, GJB3, and GJB6. 7, Tectorial membrane: COL11A2 OTOA, OTOG, and
TECTA. 8, Cochlear nerve: CCDC50, ESRRB, and GJB3. 9, Spiral ganglion: ESRRB, GIPC3, GJB1, KCNQ4, MPZ, NDP, NDRG1, OTOF, PCDH15, PJVK, PMP22,
SBF2, SLC26A4, TMPRSS3, and WFS1. 10, Reissner's membrane: ESRRB, MYH9, MYH14, POU3F4A, TFCP2L3, and WFS1. 11, Stria vascularis: ATP6B1,
BSND, CCDC50, CLCNKA, CLCNKB, DFNA5, EDN3, ESRRB, GJB2, GJB6, KCNE1, KCNQ1, MITF, MYH14, TFCP2L3, and TMPRSS3. 12, Marginal cells: KCNE1
and KCNQ1. 13, Spiral prominence: ESRRB, MYH14, SLC26A4, and WFS1. 14, Spiral ligament: COCH, COL9A1, CRYM, ESRRB, GJB2, GJB3, GJB6, MYH9,
MYH14, POU3F4, and WFS1. (Modified from Morton CC, Nance WE: Newborn hearing screening: a silent revolution. N Engl J Med 2006;354:2154-64. A, Modified
from Muller U: Harmonin mutations cause mechanotransduction defects in cochlear hair cells. Neuron 2009;62:375-387. B, Modified from Smith RJH: Sensorineural
hearing loss in children. Lancet 2005;365:879-890.)
transduction leads to an increase in cytosolic Κ+ in the outer DFNA8/12 and DFNA13 (Midfrequency Hearing Loss). Identi-
hair cells of the cochlea, the major site of KCNQ4 expression. fication of phenotype-genotype correlations is crucial in
KCNQ4 channels expressed in the base of these cells transport determining the etiology of autosomal-dominant SNHL,34
Κ+ extracellularly, where the ion is taken up by supporting cells and it has implications for prognostic and therapeutic out-
and is cycled back into the scala media.30 The consequence of comes. Some correlations are very robust, such as the low-
abnormal KCNQ4 function is apoptosis of the outer hair cells, frequency audioprofile associated with WFS1-related hearing
and the clinical manifestation of this damage is hearing loss that loss (DFNA6/14/38)35 and the “cookie-bite” (midfrequency)
is progressive and biased to the high frequencies.30 audioprofile associated with TECTA-related hearing loss
2292 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
syndrome, which has been estimated to be 0.3% to 1%.48 SYNDROMIC HEARING
Because an increased risk of SNHL had been reported for these
carriers,48 it was predicted that WFS1 was a common cause of
IMPAIRMENT
low-frequency SNHL. This assumption has been supported by Syndromic forms of hereditary SNHL (Table 148-7) are less
the subsequent discovery of frequent WFS1 mutations in fami- common than nonsyndromic forms. Syndromic hearing impair-
lies with low-frequency SNHL from different populations.49,50 ment refers to deafness that cosegregates with other features to
form a recognizable constellation of findings known as a syn-
X-Linked Nonsyndromic Hearing Impairment drome. Sensorineural deafness has been associated with more
X-linked nonsyndromic hearing impairment accounts for less than 400 syndromes. A discussion of a few of the more common
than 2% of NSHL.51 Five loci and three causative genes have syndromes follows.
been identified (Table 148-5). Reexamination of the original
family used to map DFN1 has revealed other cosegregating fea-
tures, including mental retardation. This type of deafness is AUTOSOMAL-DOMINANT SYNDROMIC
now recognized as a form of X-linked syndromic hearing loss.52 HEARING IMPAIRMENT
Of the remaining DFN loci, DFN3 is most common and is due
to mutations in a transcription factor called POU3F453; it mani- Branchio-oto-renal Syndrome
fests as congenital stapes fixation with radiologic findings that Melnick coined the term branchio-oto-renal (BOR) syndrome in
include widening of the lateral internal auditory canal and dila- 1975 to describe the cosegregation of branchial, otic, and renal
tion of the vestibule.54 Hearing loss is usually mixed, and stape- anomalies in deaf individuals.63 Inheritance is autosomal domi-
dectomy is typically attended by a perilymph gusher.55 The nant, penetrance is nearly 100%, and prevalence is estimated at
hearing impairment associated with the other loci is variable.56,57 1 in 40,000 newborns.64 BOR affects 2% of profoundly deaf
children.64 Otologic findings can involve the external, middle,
Mitochondrial Nonsyndromic Hearing Impairment or inner ear. External ear anomalies include preauricular pits
Mitochondrial nonsyndromic deafness can be caused by various (82%) or tags, auricular malformations (32%), microtia, and
mtDNA mutations, although the 1555 A-to-G mtDNA has been external auditory canal narrowing65,66; middle ear anomalies
characterized best (Table 148-6). As mentioned earlier, this include ossicular malformation (fusion, displacement, underde-
mutation is also associated with aminoglycoside ototoxicity. As velopment), facial nerve dehiscence, absence of the oval window,
a cause of nonsyndromic deafness, the phenotype is similar to and reduction in the size of the middle ear cleft65; and inner
aminoglycoside ototoxicity with a mild, high-frequency loss that ear anomalies include cochlear hypoplasia and dysplasia.67
shows progression.58 The loss is generally of later onset in indi- Enlargement of the cochlear or vestibular aqueducts may be
viduals who have not been exposed to aminoglycosides.3 seen66 as may hypoplasia of the lateral semicircular canal.67
Presbycusis, or age-related hearing loss, also may have a Hearing impairment is the most common feature of BOR
mitochondrial basis.59-62 Because mtDNA mutations accumulate syndrome and is reported in almost 90% of affected individu-
at several times the rate of nuclear DNA mutations, mitochon- als.64 The loss can be conductive (30%) or sensorineural (20%)
drial function ultimately may be impaired, resulting in age- but is most often mixed (50%). It is severe in one third of indi-
related cochlear dysfunction.60 In support of this hypothesis, an viduals and is progressive in one quarter.64 Branchial anomalies
increase in the mtDNA mutation load has been shown in aged occur in the form of laterocervical fistulas, sinuses, and cysts;
cochlea.62 renal anomalies range from agenesis to dysplasia and are found
in 25% of individuals.64 Less common phenotypic findings Neurofibromatosis Type 2
include lacrimal duct aplasia, short palate, and retrognathia.64 Neurofibromatosis type 2 (NF2) is characterized by the devel-
One causative gene is EYA1, the human homologue of the opment of bilateral vestibular schwannomas and other intracra-
Drosophila eyes absent gene.68 The gene contains 16 exons that nial and spinal tumors that include schwannomas, meningiomas,
encode for 559 amino acids.68 EYA1 mutations are found in gliomas, and ependymomas. In addition, patients may have
approximately 25% of patients with a BOR phenotype, and this posterior subcapsular lenticular opacities. Diagnostic criteria
phenotype is hypothesized to reflect a reduction in the amount include 1) bilateral vestibular schwannomas that usually develop
of the EYA1 protein. Mutations in two additional genes, SIX1 by the second decade of life, or 2) a family history of NF2 in
and SIX5, also have been shown more recently to cause BOR a first-degree relative, plus one of the following: unilateral
syndrome.69,70 Both genes act within the genetic network of the vestibular schwannomas before age 30 or any two of meningi-
EYA and PAX genes to regulate organogenesis. oma, glioma, schwannoma, or juvenile posterior subcapsular
148 | GENETIC SENSORINEURAL HEARING LOSS 2295
by cochlear hypoplasia. Most cases of PS result from mutations vestibular dysfunction, and retinal degeneration that begins in
in the SLC26A4 gene that encodes an anion transporter known the third to fourth decade; and type 3 is characterized by pro-
as pendrin, which is expressed in the inner ear (see Fig. 148-4), gressive hearing loss, variable vestibular dysfunction, and vari-
thyroid, and kidney.99 Expression of SLC26A4 has been shown able onset of retinitis pigmentosa. Within each subtype, genetic
throughout the endolymphatic duct and sac, in distinct areas of heterogeneity is found, and numerous subtypes are recognized,
the utricle and saccule, and in the external sulcus region within although the two most common forms are Usher syndrome
the developing cochlea.100 Pendrin is thought to be involved in type 1B (USH1B) and Usher syndrome type 2A (USH2A),
chloride and iodide transport and not sulfate transport.101 which account for 75% to 80% of the Usher syndromes.117
Affected individuals develop goiter in their second decade, USH1B accounts for 75% of Usher syndrome type 1 cases and
although they usually remain euthyroid.99 Thyroid dysfunction is caused by mutations in an unconventional myosin called
can be shown with a perchlorate discharge test, in which radio- MYO7A.118 USH2A is the most common form, and the causative
active iodide and perchlorate are administered. Mutations in gene encodes a 1551–amino acid protein named usherin, a
SLC26A4 prevent rapid movement of iodide from the thyrocyte putative extracellular matrix molecule.119 Numerous other
to the colloid, and perchlorate blocks the Na/I symporter that genes have been implicated in Usher syndrome subtypes, and
moves iodide from the bloodstream into the thyrocyte. The net these are listed in Table 148-7. Cochlear expression patterns of
effect is that iodide in the thyrocyte washes back into the blood- Usher syndrome–related genes are shown in Figure 148-4.
stream in affected individuals, with a release of greater than
10% radioactivity considered diagnostic for PS.97,102 The sensi- Biotinidase Deficiency
tivity of this test is low, which makes genetic testing the pre- Biotinidase deficiency is secondary to an absence of the water-
ferred diagnostic method.97 The hearing impairment is usually soluble B-complex vitamin biotin. Biotin covalently binds to
prelingual, bilateral, and profound, although it can be progres- four carboxylases that are essential for gluconeogenesis, fatty
sive.97 Radiologic studies always show a temporal bone anomaly, acid synthesis, and catabolism of several branched-chain amino
either dilated vestibular aqueducts or Mondini dysplasia.97,103 acids. If biotinidase deficiency is not recognized and corrected
Mutations in SLC26A4 also cause a type of nonsyndromic by daily addition of biotin to the diet, affected individuals
autosomal-recessive deafness called DFNB4.104,105 Whether the develop neurologic features such as seizures, hypertonia, devel-
phenotype is syndromic or nonsyndromic may reflect the opmental delay, ataxia, and visual problems. In at least 75% of
degree of residual function in the abnormal protein. PS and children who become symptomatic, SNHL develops and can be
DFNB4 can be diagnosed by screening this gene for mutations. profound and persistent even after treatment is initiated.120
More recently, mutations in the transcription factor FOXI1 have Cutaneous features are also present and include a skin rash,
also been shown to cause PS in patients heterozygous for a alopecia, and conjunctivitis. With treatment that consists of
mutation in SLC26A4.106 biotin replacement, the neurologic and cutaneous manifesta-
tions resolve; however, the hearing loss and optic atrophy are
Jervell and Lange-Nielsen Syndrome usually irreversible. If a child is brought to medical attention
In 1957, Jervell and Lange-Nielsen described a syndrome char- with episodic or progressive ataxia and progressive sensorineu-
acterized by congenital deafness, prolonged QT interval, and ral deafness, with or without neurologic or cutaneous symp-
syncopal attacks.107 Long QT syndrome itself can be dominantly toms, biotinidase deficiency should be considered. To prevent
or recessively inherited. The dominant disease is called Romano- metabolic coma, diet and treatment should be initiated as soon
Ward syndrome. It is more common and does not include the as possible.120,121 If untreated, 75% of affected infants develop
deafness phenotype.107 The recessive disease is known as Jervell hearing loss that can be profound, and it persists despite the
and Lange-Nielsen syndrome (JLNS). subsequent initiation of treatment.120
JLNS is genetically heterogeneous, and mutations in KVLQT1
and KCNE1 cause this phenotype.108-110 These genes encode for Refsum Disease
subunits of a potassium channel expressed in the heart and Refsum disease is a postlingual, severe, progressive SNHL asso-
inner ear. Hearing impairment is due to changes in endolymph ciated with retinitis pigmentosa, peripheral neuropathy, cere-
homeostasis caused by malfunction of this channel, and it is bellar ataxia, and elevated protein levels in the cerebrospinal
congenital, bilateral, and severe to profound.108-110 Although fluid without an increase in the number of cells.122 It is caused
the prevalence of JLNS among children with congenital deaf- by defective phytanic acid metabolism, and the diagnosis is
ness is only 0.21%,111 it is an important diagnosis to consider established by determining the serum concentration of phy-
because of its cardiac manifestations. The prolonged QT inter- tanic acid. Two genes, PHYH and PEX7, have been implicated
val can lead to ventricular arrhythmias, syncopal episodes, and in most cases of Refsum disease, although a few patients exist
death in childhood.111 Effective treatment with β-adrenergic in whom mutations have not been found.122 Although Refsum
blockers reduces mortality rate from 71% to 6%.111 disease is extremely rare, it is important that it be considered
in the evaluation of a deaf person because it can be easily
Usher Syndromes treated with dietary modification and plasmapheresis.
The Usher syndromes are a genetically and clinically heteroge-
neous group of diseases characterized by SNHL, retinitis pig- X-LINKED SYNDROMES
mentosa, and often vestibular dysfunction.112 The incidence is
estimated at 4.4 per 100,000 population in the United States, Alport Syndrome
and 3% to 6% of congenitally deaf individuals carry this diag- Alport syndrome is a disease of type IV collagen that is mani-
nosis.113 This estimate has recently been revised upward to 1 fested by hematuric nephritis, hearing impairment, and ocular
per 6000 in the United States,114 where it causes 50% of con- changes. The inheritance pattern, although predominantly
comitant deafness and blindness.113 X-linked (~80%), can be autosomal recessive or dominant.123
Three clinical variants of Usher syndrome are recog- Prevalence is estimated at 1 per 5000 in the United States, and
nized.115,116 Type 1 is phenotypically distinguished by the pres- a significant proportion of renal transplant patients have Alport
ence of severe to profound congenital hearing loss, vestibular syndrome.124 Diagnostic criteria include at least three of the
dysfunction, and retinitis pigmentosa that develops in child- following four characteristics: 1) positive family history of
hood; type 2 is distinguished by moderate to severe congenital hematuria with or without chronic renal failure, 2) progressive
hearing loss, with uncertainty related to progression, no high-tone sensorineural deafness, 3) typical eye lesion (anterior
148 | GENETIC SENSORINEURAL HEARING LOSS 2297
reach 6 months of age. A trained pediatric audiologist can adoption of new genomic sequencing technologies has been
measure hearing losses of 20 dB in the better-hearing ear. The especially rapid for genetic hearing loss, because the need has
frequency of testing is usually decided on a case-by-case basis, been great. Interested readers are referred elsewhere for a
although Tomaski and Grundfast148 recommend a more rigid more in-depth review of these genomic technologies and their
schedule in cases of confirmed SNHL in children. They recom- impact on genetic testing for hearing loss.153
mend testing every 3 months in the first year of life, followed Several recent studies have shown the effectiveness of com-
by every 6 months during the preschool years and once a year prehensive, multigene panels for diagnosis of NSHL using
while in school. various different MPS technologies.154-157 This research has
To determine the cause of hearing loss, past recommenda- rapidly been translated into clinical diagnostic tests. In general,
tions included a battery of tests such as thyroid function studies, multigene panels sequence the more than 60 known NSHL
urinalysis, electrocardiogram, ophthalmologic consultation, genes and often include genes that cause Usher and Pendred
and temporal bone imaging. Today, however, genetic testing syndromes. Besides the technology used, differences among
should be ordered instead. Indeed, after a detailed history, these panels typically include the number of genes offered and
physical examination, and audiometric analysis, genetic testing the turnaround time. A listing of clinical laboratories that offer
should be the next test ordered in the evaluation of an indi- these tests is available on the Genetic Testing Registry (http://
vidual with apparent NSHL. This approach limits unnecessary www.ncbi.nlm.nih.gov/gtr).
costly and time-consuming tests and makes focused and Another method for sequencing all known deafness genes
directed testing possible. Apparent syndromic cases can be is whole-exome sequencing, whereby the exons of all 20,000 or so
evaluated with tests relevant to the syndrome on the differen- genes in the human genome are sequenced simultaneously,
tial diagnosis. A notable exception to this is cases of Usher and analysis is focused on known deafness genes. This method
syndrome, which commonly manifest as a nonsyndromic mimic has been effectively used for diagnosis of NSHL.158 These new
prior to progression of retinopathy. For this reason, several diagnostic methods have already improved the diagnostic rate
NSHL screening panels include the genes that cause Usher for NSHL, and it will only improve further as the methods are
syndrome. refined.
Whole-exome and multigene MPS panels are alike in that
Genetic Testing they use similar technology and in both cases uncover many
In the United States, congenital SNHL occurs about three possible causative mutations. This makes interpretation of
times more frequently than Down syndrome, six times more the variants to determine the causative mutation paramount.
frequently than spina bifida, and more than 50 times more However, two key differences exist between multigene panel
frequently than phenylketonuria, making it the most frequently testing and whole-exome sequencing: 1) cost will be highest for
occurring birth defect.149 An estimated 4000 infants are born whole-exome sequencing, and multigene panels are less expen-
each year with severe to profound bilateral hearing loss,150,151 sive, although this difference will be erased or reduced in the
and another 8000 are born with unilateral or mild to moderate next several years; and 2) secondary genomic findings—for
bilateral SNHL.5 At least 50% of congenital hearing loss cases instance, genetic risk factors for other diseases—are more of a
have an underlying genetic cause; this is a compelling reason concern as more genes of the genome are sequenced, which
to provide genetic testing services to all children diagnosed raises ethical concerns that are reduced or mitigated with more
with congenital hearing loss. The implementation of the Early focused genetic tests such as multigene panels.
Hearing Detection and Intervention (EHDI) program in the Based on these advances in genetic testing technologies, the
United States and similar universal hearing screening programs current approach to evaluation of an individual with NSHL
around the world has facilitated increased detection, genetic should proceed as follows: history, physical examination, and
diagnosis, and intervention for these children.152 In addition, audiometry are followed by genetic testing using a multigene
advances in genetic technologies have led to an increase in panel or whole-exome sequencing. In cases of apparent syn-
the availability of genetic diagnostic services for infants with dromic hearing loss, pertinent clinical examination findings
hearing loss.152 and tests will guide genetic screening, typically by single genes,
Genetic testing has undergone rapid change since the com- to arrive at a diagnosis.
pletion of the Human Genome Project, which took 11 years to
sequence the entire genome of a single person—at a cost of
roughly $3 billion and with the coordination of several large
PRENATAL TESTING
sequencing centers. Today, the same sequence can be generated Prenatal diagnosis for some forms of hereditary hearing loss is
in 24 hours for several thousand dollars because of the advent of technically possible by analysis of DNA extracted from fetal
new genomic sequencing technologies called massively parallel cells. Fetal material can be obtained by amniocentesis at 15 to
sequencing (MPS). These advances are especially applicable to a 18 weeks’ gestation or by chorionic villus sampling at 10 to 12
genetic disorder with extreme genetic heterogeneity, such as weeks’ gestation. Gestational age is expressed as menstrual
hearing loss. As could be expected, similarly vast changes in weeks calculated from the first day of the last normal menstrual
scale have also occurred in the clinical genetic testing arena: the period or by ultrasound measurements. The deafness-causing
first genetic tests for deafness that became available in the 1990s allele of a deaf family member must be identified before pre-
focused on detecting a single mutation; in the late 1990s and natal testing can be performed.
early 2000s, sequencing of a single exon or whole gene was Requests for prenatal testing for conditions such as hearing
made available; and in the last 5 years, multigene screening loss are uncommon. Differences in perspective may exist among
panels, and now whole-exome (every exon of the genome) and medical professionals and within families regarding the use of
whole-genome sequencing, have become available on a clinical prenatal testing, particularly if the testing is being considered
basis. for the purpose of pregnancy termination rather than early
As described above, more than 60 different genes with more diagnosis. Although most centers would consider decisions
than a thousand reported mutations are known to cause NSHL. about prenatal testing to be the choice of the parents, careful
The previous status quo, searching for the genetic cause of discussion of these issues is appropriate. Preimplantation
deafness in an individual by assaying for a single causative muta- genetic diagnosis may be available for families in which the
tion or by sequencing a single hearing loss gene, was under- deafness-causing mutation has been identified in a deaf family
standably a low-yield endeavor. It is for this reason that the member.
148 | GENETIC SENSORINEURAL HEARING LOSS 2299
Kelsell DP, Dunlop J, Stevens HP, et al: Connexin 26 mutations in Shibata SB, Raphael Y: Future approaches for inner ear protection and
hereditary non-syndromic sensorineural deafness. Nature 387:80–83, repair. J Commun Disord 43:295–310, 2010.
1997. Smith RJ, Bale JF, Jr, White KR: Sensorineural hearing loss in children.
Kubisch C, Schroeder BC, Friedrich T: KCNQ4, a novel potassium Lancet 365:879–890, 2005.
channel expressed in sensory outer hair cells, is mutated in dominant Strachan T, Read AP: Human molecular genetics, ed 4, New York, 2011,
deafness. Cell 96:437–446, 1999. Garland Science.
Morton CC, Nance WE: Newborn hearing screening: a silent revolu- Tekin M, Arnos KS, Pandya A: Advances in hereditary deafness. Lancet
tion. N Engl J Med 354(20):2151–2164, 2006. 358:1082–1090, 2001.
Morton NE: Genetic epidemiology of hearing impairment. Ann N Y Van Camp G, Smith RJ: Hereditary Hearing Loss Homepage. http://
Acad Sci 630:16–31, 1991. hereditaryhearingloss.org.
Nowak CB: Genetics and hearing loss: A review of Stickler syndrome. Yang T, Vidarsson H, Rodrigo-Blomqvist S, et al: Transcriptional control
J Commun Disord 31:437–453, 1998. of SLC26A4 is involved in Pendred syndrome and nonsyndromic
Pennings RJ, Wagenaar M, van Aarem A, et al: Hearing impairment in enlargement of vestibular aqueduct (DFNB4). Am J Hum Genet 80:
Usher’s syndrome. Adv Otorhinolaryngol 61:184–191, 2002. 1055–1063, 2007.
Read AP, Newton VE: Waardenburg syndrome. J Med Genet 34:656–665,
1997.
148 | GENETIC SENSORINEURAL HEARING LOSS 2300.e1
28. Petersen MB: Non-syndromic autosomal-dominant deafness. Clin
REFERENCES Genet 62(1):1–13, 2002.
1. Smith RJ, Bale JF, Jr, White KR: Sensorineural hearing loss in 29. Liu X, Xu L: Nonsyndromic hearing loss: an analysis of audio-
children. Lancet 365(9462):879–890, 2005. grams. Ann Otol Rhinol Laryngol 103(6):428–433, 1994.
2. Morton CC, Nance WE: Newborn hearing screening–a silent revo- 30. Kubisch C, Schroeder BC, Friedrich T, et al: KCNQ4, a novel
lution. N Engl J Med 354(20):2151–2164, 2006. potassium channel expressed in sensory outer hair cells, is
3. Estivill X, Govea N, Barcelo E, et al: Familial progressive sensori- mutated in dominant deafness. Cell 96(3):437–446, 1999.
neural deafness is mainly due to the mtDNA A1555G mutation 31. Coucke PJ, Van Hauwe P, Kelley PM, et al: Mutations in the
and is enhanced by treatment of aminoglycosides. Am J Hum Genet KCNQ4 gene are responsible for autosomal dominant deafness in
62(1):27–35, 1998. four DFNA2 families. Hum Mol Genet 8(7):1321–1328, 1999.
4. Gorga M, Worthington D: Stimulus calibration in ABR measure- 32. Denoyelle F, Lina-Granade G, Plauchu H, et al: Connexin 26 gene
ments. In Jacobson JT, editor: The Auditory Brainstem Response, linked to a dominant deafness. Nature 393(6683):319–320, 1998.
London, 1985, Taylor & Francis. 33. Kharkovets T, Dedek K, Maier H, et al: Mice with altered KCNQ4
5. Maki-Torkko EM, Lindholm PK, Vayrynen MR, et al: Epidemiol- K+ channels implicate sensory outer hair cells in human progres-
ogy of moderate to profound childhood hearing impairments in sive deafness. EMBO J 25(3):642–652, 2006.
northern Finland. Any changes in ten years? Scand Audiol 27(2): 34. Huygen PPR, Cremers C: Audiometric profiles associated with
95–103, 1998. genetic nonsyndromal hearing impairment: a review and pheno-
6. Fortnum HM, Summerfield AQ, Marshall DH, et al: Prevalence type analysis. In Martini MS, Read D, et al, editors: Genes, Hearing
of permanent childhood hearing impairment in the United and Deafness: From Molecular Biology to Clinical Practice, London, UK,
Kingdom and implications for universal neonatal hearing screen- 2007, Informa HealthCare, pp 185–204.
ing: questionnaire based ascertainment study. Br Med J 323(7312): 35. Cryns K, Pfister M, Pennings RJ, et al: Mutations in the WFS1 gene
536–540, 2001. that cause low-frequency sensorineural hearing loss are small non-
7. Barsky-Firkser L, Sun S: Universal newborn hearing screenings: a inactivating mutations. Hum Genet 110(5):389–394, 2002.
three-year experience. Pediatrics 99(6):E4, 1997. 36. Verhoeven K, Van Laer L, Kirschhofer K, et al: Mutations in the
8. Parving A: The need for universal neonatal hearing screening– human alpha-tectorin gene cause autosomal dominant non-
some aspects of epidemiology and identification. Acta Paediatrica syndromic hearing impairment. Nat Genet 19(1):60–62, 1998.
88(432):69–72, 1999. 37. Kirschhofer K, Kenyon JB, Hoover DM, et al: Autosomal-dominant,
9. Vega Cuadri A, Alvarez Suarez MY, Blasco Huelva A, et al: Oto- prelingual, nonprogressive sensorineural hearing loss: localiza-
acoustic emissions screening as early identification of hearing loss tion of the gene (DFNA8) to chromosome 11q by linkage in an
in newborns [in Spanish]. Acta Otorrinolaringol Esp 52(4):273–278, Austrian family. Cytogenet Cell Genet 82(1-2):126–130, 1998.
2001. 38. Verhoeven K, Van Camp G, Govaerts PJ, et al: A gene for autoso-
10. Lin HC, Shu MT, Chang KC, et al: A universal newborn hearing mal dominant nonsyndromic hearing loss (DFNA12) maps to
screening program in Taiwan. Int J Pediat Otorhinolaryngol 63(3): chromosome 11q22-24. Am J Hum Genet 60(5):1168–1173, 1997.
209–218, 2002. 39. Bork JM, Peters LM, Riazuddin S, et al: Usher syndrome 1D and
11. Van Camp G SR: Hereditary Hearing Loss. Available at http:// nonsyndromic autosomal recessive deafness DFNB12 are caused
hereditaryhearingloss.org. by allelic mutations of the novel cadherin-like gene CDH23. Am J
12. Der Kaloustian VM, Kurban AK: Genetic diseases of the skin, Berlin– Hum Genet 68(1):26–37, 2001.
New York, 1979, Springer. 40. Legan PK, Lukashkina VA, Goodyear RJ, et al: A deafness muta-
13. Strachan T, Read AP: Human molecular genetics, ed 4, New York, tion isolates a second role for the tectorial membrane in hearing.
2011, Garland Science. Nat Neurosci 8(8):1035–1042, 2005.
14. Jorde LB, White RL, Carey JC: Medical genetics, St. Louis, 1995, 41. Mustapha M, Weil D, Chardenoux S, et al: An alpha-tectorin gene
Mosby. defect causes a newly identified autosomal recessive form of sen-
15. Li XC, Friedman RA: Nonsyndromic hereditary hearing loss. Oto- sorineural pre-lingual non-syndromic deafness, DFNB21. Hum
laryngol Clin North Am 35(2):275–285, 2002. Mol Genet 8(3):409–412, 1999.
16. Sundstrom RA, Van Laer L, Van Camp G, et al: Autosomal reces- 42. Meyer NC, Alasti F, Nishimura CJ, et al: Identification of three
sive nonsyndromic hearing loss. Am J Med Genet 89(3):123–129, novel TECTA mutations in Iranian families with autosomal reces-
1999. sive nonsyndromic hearing impairment at the DFNB21 locus. Am
17. Guilford P, Ben Arab S, Blanchard S, et al: A non-syndrome form J Med Genet Part A 143A(14):1623–1629, 2007.
of neurosensory, recessive deafness maps to the pericentromeric 43. Brown MR, Tomek MS, Van Laer L, et al: A novel locus for auto-
region of chromosome 13q. Nat Genet 6(1):24–28, 1994. somal dominant nonsyndromic hearing loss, DFNA13, maps to
18. Kelsell DP, Dunlop J, Stevens HP, et al: Connexin 26 mutations in chromosome 6p. Am J Hum Genet 61(4):924–927, 1997.
hereditary non-syndromic sensorineural deafness. Nature 387 44 McGuirt WT, Prasad SD, Griffith AJ, et al: Mutations in COL11A2
(6628):80–83, 1997. cause non-syndromic hearing loss (DFNA13). Nat Genet 23(4):413–
19. Steel KP: Perspectives: biomedicine. The benefits of recycling. 419, 1999.
Science 285(5432):1363–1364, 1999. 45. Inoue H, Tanizawa Y, Wasson J, et al: A gene encoding a trans-
20. Smith RJ, Robin NH: Genetic testing for deafness–GJB2 and membrane protein is mutated in patients with diabetes mellitus
SLC26A4 as causes of deafness. J Commun Disord 35(4):367–377, and optic atrophy (Wolfram syndrome). Nat Genet 20(2):143–148,
2002. 1998.
21. Kenneson A, Van Naarden Braun K, Boyle C: GJB2 (connexin 26) 46. Khan SY, Ahmed ZM, Shabbir MI, et al: Mutations of the RDX
variants and nonsyndromic sensorineural hearing loss: a HuGE gene cause nonsyndromic hearing loss at the DFNB24 locus. Hum
review. Genet Med 4(4):258–274, 2002. Mutat 28(5):417–423, 2007.
22. Chang EH, Van Camp G, Smith RJ: The role of connexins in 47. Ahmed ZM, Riazuddin S, Bernstein SL, et al: Mutations of the
human disease. Ear Hear 24(4):314–323, 2003. protocadherin gene PCDH15 cause Usher syndrome type 1F. Am
23. Green GE, Scott DA, McDonald JM, et al: Carrier rates in the J Hum Genet 69(1):25–34, 2001.
midwestern United States for GJB2 mutations causing inherited 48. Ohata T, Koizumi A, Kayo T, et al: Evidence of an increased risk
deafness. JAMA 281(23):2211–2216, 1999. of hearing loss in heterozygous carriers in a Wolfram syndrome
24. Morell RJ, Kim HJ, Hood LJ, et al: Mutations in the connexin 26 family. Hum Genet 103(4):470–474, 1998.
gene (GJB2) among Ashkenazi Jews with nonsyndromic recessive 49. Fukuoka H, Kanda Y, Ohta S, et al: Mutations in the WFS1 gene
deafness. N Engl J Med 339(21):1500–1505, 1998. are a frequent cause of autosomal dominant nonsyndromic low-
25. Abe S, Usami S, Shinkawa H, et al: Prevalent connexin 26 gene frequency hearing loss in Japanese. J Hum Genet 52(6):510–515,
(GJB2) mutations in Japanese. J Med Genet 37(1):41–43, 2000. 2007.
26. Choung YH, Moon SK, Park HJ: Functional study of GJB2 in 50. Hansen L, Eiberg H, Barrett T, et al: Mutation analysis of the
hereditary hearing loss. Laryngoscope 112(9):1667–1671, 2002. WFS1 gene in seven Danish Wolfram syndrome families; four new
27. Green GE, Scott DA, McDonald JM, et al: Performance of cochlear mutations identified. Eur J Hum Genet 13(12):1275–1284, 2005.
implant recipients with GJB2-related deafness. Am J Med Genet 51. Morton NE: Genetic epidemiology of hearing impairment. Ann N
109(3):167–170, 2002. Y Acad Sci 630:16–31, 1991.
2300.e2 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
genetic heterogeneity in the Middle Eastern Druze population. 130. Jin H, May M, Tranebjaerg L, et al: A novel X-linked gene, DDP,
Hum Mol Genet 4(9):1637–1642, 1995. shows mutations in families with deafness (DFN-1), dystonia,
105. Li XC, Everett LA, Lalwani AK, et al: A mutation in PDS causes mental deficiency and blindness. Nat Genet 14(2):177–180,
non-syndromic recessive deafness. Nat Genet 18(3):215–217, 1998. 1996.
106. Yang T, Vidarsson H, Rodrigo-Blomqvist S, et al: Transcriptional 131. Koehler CM, Leuenberger D, Merchant S, et al: Human deafness
control of SLC26A4 is involved in Pendred syndrome and non- dystonia syndrome is a mitochondrial disease. Proc Natl Acad Sci U
syndromic enlargement of vestibular aqueduct (DFNB4). Am J S A 96(5):2141–2146, 1999.
Hum Genet 80(6):1055–1063, 2007. 132. Bykhovskaya Y, Yang H, Taylor K, et al: Modifier locus for mito-
107. Komsuoglu B, Goldeli O, Kulan K, et al: The Jervell and Lange- chondrial DNA disease: linkage and linkage disequilibrium
Nielsen syndrome. Int J Cardiol 47(2):189–192, 1994. mapping of a nuclear modifier gene for maternally inherited
108. Neyroud N, Tesson F, Denjoy I, et al: A novel mutation in the deafness. Genet Med 3(3):177–180, 2001.
potassium channel gene KVLQT1 causes the Jervell and Lange- 133. Usami S, Abe S, Akita J, et al: Sensorineural hearing loss associated
Nielsen cardioauditory syndrome. Nat Genet 15(2):186–189, 1997. with the mitochondrial mutations. Adv Otorhinolaryngol 56:203–
109. Tyson J, Tranebjaerg L, Bellman S, et al: IsK and KvLQT1: muta- 211, 2000.
tion in either of the two subunits of the slow component of the 134. Ensink RJ, Huygen PL, Cremers CW: The clinical spectrum of
delayed rectifier potassium channel can cause Jervell and Lange- maternally transmitted hearing loss. Adv Otorhinolaryngol 61:172–
Nielsen syndrome. Hum Mol Genet 6(12):2179–2185, 1997. 183, 2002.
110. Schulze-Bahr E, Wang Q, Wedekind H, et al: KCNE1 mutations 135. Zwirner P, Wilichowski E: Progressive sensorineural hearing loss
cause Jervell and Lange-Nielsen syndrome. Nat Genet 17(3):267– in children with mitochondrial encephalomyopathies. Laryngo-
268, 1997. scope 111(3):515–521, 2001.
111. Ocal B, Imamoglu A, Atalay S, et al: Prevalence of idiopathic long 136. Moraes CT, DiMauro S, Zeviani M, et al: Mitochondrial DNA dele-
QT syndrome in children with congenital deafness. Pediatr Cardiol tions in progressive external ophthalmoplegia and Kearns-Sayre
18(6):401–405, 1997. syndrome. N Engl J Med 320(20):1293–1299, 1989.
112. Keats BJ, Corey DP: The usher syndromes. Am J Med Genet 89(3): 137. Marin-Garcia J, Goldenthal MJ, Sarnat HB: Kearns-Sayre syn-
158–166, 1999. drome with a novel mitochondrial DNA deletion. J Child Neurol
113. Boughman JA, Vernon M, Shaver KA: Usher syndrome: definition 15(8):555–558, 2000.
and estimate of prevalence from two high-risk populations. 138. Katsanos KH, Pappas CJ, Patsouras D, et al: Alarming atrioven-
J Chronic Dis 36(8):595–603, 1983. tricular block and mitral valve prolapse in the Kearns-Sayre syn-
114. Kimberling WJ, Hildebrand MS, Shearer AE, et al: Frequency of drome. Int J Cardiol 83(2):179–181, 2002.
Usher syndrome in two pediatric populations: Implications for 139. Guillausseau PJ, Massin P, Dubois-LaForgue D, et al: Maternally
genetic screening of deaf and hard of hearing children. Genet Med inherited diabetes and deafness: a multicenter study. Ann Intern
12(8):512–516, 2010. Med 134(9 Pt 1):721–728, 2001.
115. Petit C: Usher syndrome: from genetics to pathogenesis. Ann Rev 140. Newkirk JE, Taylor RW, Howell N, et al: Maternally inherited
Genomics Hum Genet 2:271–297, 2001. diabetes and deafness: prevalence in a hospital diabetic popula-
116. Reisser CF, Kimberling WJ, Otterstedde CR: Hearing loss in Usher tion. Diabet Med 14(6):457–460, 1997.
syndrome type II is nonprogressive. Ann Otol Rhinol Laryngol 141. Prezant TR, Agapian JV, Bohlman MC, et al: Mitochondrial ribo-
111(12 Pt 1):1108–1111, 2002. somal RNA mutation associated with both antibiotic-induced and
117. Pennings RJ, Wagenaar M, van Aarem A, et al: Hearing impair- non-syndromic deafness. Nat Genet 4(3):289–294, 1993.
ment in Usher’s syndrome. Adv Otorhinolaryngol 61:184–191, 2002. 142. Yoshida M, Shintani T, Hirao M, et al: Aminoglycoside-induced
118. Weil D, Blanchard S, Kaplan J, et al: Defective myosin VIIA gene hearing loss in a patient with the 961 mutation in mitochondrial
responsible for Usher syndrome type 1B. Nature 374(6517):60–61, DNA. ORL J Otorhinolaryngol Relat Spec 64(3):219–222, 2002.
1995. 143. Fischel-Ghodsian N: Genetic factors in aminoglycoside toxicity.
119. Eudy JD, Weston MD, Yao S, et al: Mutation of a gene encoding Pharmacogenomics 6(1):27–36, 2005.
a protein with extracellular matrix motifs in Usher syndrome type 144. ACMG: Genetics evaluation guidelines for the etiologic diagnosis
IIa. Science 280(5370):1753–1757, 1998. of congenital hearing loss. Genetic Evaluation of Congenital
120. Wolf B, Spencer R, Gleason T: Hearing loss is a common feature Hearing Loss Expert Panel. ACMG statement. Genet Med 4(3):162–
of symptomatic children with profound biotinidase deficiency. 171, 2002.
J Pediatr 140(2):242–246, 2002. 145. Greinwald JH, Jr, Hartnick CJ: The evaluation of children with
121. Heller AJ, Stanley C, Shaia WT, et al: Localization of biotinidase sensorineural hearing loss. Arch Otolaryngol Head Neck Surg
in the brain: implications for its role in hearing loss in biotinidase 128(1):84–87, 2002.
deficiency. Hear Res 173(1-2):62–68, 2002. 146. Tekin M, Arnos KS, Pandya A: Advances in hereditary deafness.
122. van den Brink DM, Brites P, Haasjes J, et al: Identification of PEX7 Lancet 358(9287):1082–1090, 2001.
as the second gene involved in Refsum disease. Am J Hum Genet 147. Green GE, Cunniff C: Genetic evaluation and counseling for con-
72(2):471–477, 2003. genital deafness. Adv Otorhinolaryngol 61:230–240, 2002.
123. Dagher H, Buzza M, Colville D, et al: A comparison of the clinical, 148. Tomaski SM, Grundfast KM: A stepwise approach to the diagnosis
histopathologic, and ultrastructural phenotypes in carriers of and treatment of hereditary hearing loss. Pediatric Clin North Am
X-linked and autosomal recessive Alport’s syndrome. Am J Kidney 46(1):35–48, 1999.
Dis 38(6):1217–1228, 2001. 149. White KR: The current status of EHDI programs in the United
124. Jais JP, Knebelmann B, Giatras I, et al: X-linked Alport syndrome: States. Ment Retard Dev Disabil Res Rev 9(2):79–88, 2003.
natural history in 195 families and genotype-phenotype correla- 150. Mohr PE, Feldman JJ, Dunbar JL: The societal costs of severe to
tions in males. J Am Soc Nephrol 11(4):649–657, 2000. profound hearing loss in the United States. Policy Anal Brief H Ser
125. Plant KE, Green PM, Vetrie D, et al: Detection of mutations in 2(1):1–4, 2000.
COL4A5 in patients with Alport syndrome. Hum Mutat 13(2):124– 151. Thompson DC, McPhillips H, Davis RL, et al: Universal newborn
132, 1999. hearing screening: summary of evidence. JAMA 286(16):2000–
126. Barker DF, Hostikka SL, Zhou J, et al: Identification of mutations 2010, 2001.
in the COL4A5 collagen gene in Alport syndrome. Science 152. White KR: Early hearing detection and intervention programs:
248(4960):1224–1227, 1990. opportunities for genetic services. Am J Med Genet Part A 130A(1):
127. Gross O, Netzer KO, Lambrecht R, et al: Meta-analysis of genotype- 29–36, 2004.
phenotype correlation in X-linked Alport syndrome: impact on 153. Shearer AE, Hildebrand MS, Sloan CM, et al: Deafness in the
clinical counselling. Nephrol Dial Transplant 17(7):1218–1227, genomics era. Hear Res 282(1-2):1–9, 2011.
2002. 154. Brownstein Z, Friedman LM, Shahin H, et al: Targeted genomic
128. Kashtan CE: Familial hematuric syndromes–Alport syndrome, capture and massively parallel sequencing to identify genes for
thin glomerular basement membrane disease and Fechtner/ hereditary hearing loss in Middle Eastern families. Genome Biol
Epstein syndromes. Contrib Nephrol (136):79–99, 2001. 12(9):R89, 2011.
129. Mohr J, Mageroy K: Sex-linked deafness of a possibly new type. 155. Tang W, Qian D, Ahmad S, et al: A low-cost exon capture method
Acta Genet Stat Med 10:54–62, 1960. suitable for large-scale screening of genetic deafness by the
2300.e4 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
200. Schultz JM, Khan SN, Ahmed ZM, et al: Noncoding mutations of 223. Chaib H, Lina-Granade G, Guilford P, et al: A gene responsible
HGF are associated with nonsyndromic hearing loss, DFNB39. Am for a dominant form of neurosensory non-syndromic deafness
J Hum Genet 85(1):25–39, 2009. maps to the NSRD1 recessive deafness gene interval. Hum Mol
201. Borck G, Ur Rehman A, Lee K, et al: Loss-of-function mutations Genet 3(12):2219–2222, 1994.
of ILDR1 cause autosomal-recessive hearing impairment DFNB42. 224. Grifa A, Wagner CA, D’Ambrosio L, et al: Mutations in GJB6 cause
Am J Hum Genet 88(2):127–137, 2011. nonsyndromic autosomal dominant deafness at DFNA3 locus. Nat
202. Riazuddin S, Ahmed ZM, Fanning AS, et al: Tricellulin is a tight- Genet 23(1):16–18, 1999.
junction protein necessary for hearing. Am J Hum Genet 79(6): 225. Chen AH, Ni L, Fukushima K, et al: Linkage of a gene for domi-
1040–1051, 2006. nant non-syndromic deafness to chromosome 19. Hum Mol Genet
203. Chen W, Kahrizi K, Meyer NC, et al: Mutation of COL11A2 causes 4(6):1073–1076, 1995.
autosomal recessive non-syndromic hearing loss at the DFNB53 226. Donaudy F, Snoeckx R, Pfister M, et al: Nonmuscle myosin heavy-
locus. J Med Genet 42(10):e61, 2005. chain gene MYH14 is expressed in cochlea and mutated in
204. Delmaghani S, del Castillo FJ, Michel V, et al: Mutations in the patients affected by autosomal dominant hearing impairment
gene encoding pejvakin, a newly identified protein of the afferent (DFNA4). Am J Hum Genet 74(4):770–776, 2004.
auditory pathway, cause DFNB59 auditory neuropathy. Nat Genet 227. Zheng J, Miller KK, Yang T, et al: Carcinoembryonic antigen-
38(7):770–778, 2006. related cell adhesion molecule 16 interacts with alpha-tectorin
205. Liu XZ, Ouyang XM, Xia XJ, et al: Prestin, a cochlear motor and is mutated in autosomal dominant hearing loss (DFNA4). Proc
protein, is defective in non-syndromic hearing loss. Hum Mol Genet Natl Acad Sci U S A 108(10):4218–4223, 2011.
12(10):1155–1162, 2003. 228. van Camp G, Coucke P, Balemans W, et al: Localization of a gene
206. Ahmed ZM, Masmoudi S, Kalay E, et al: Mutations of LRTOMT, for non-syndromic hearing loss (DFNA5) to chromosome 7p15.
a fusion gene with alternative reading frames, cause nonsyn- Hum Mol Genet 4(11):2159–2163, 1995.
dromic deafness in humans. Nat Genet 40(11):1335–1340, 2008. 229. Van Laer L, Huizing EH, Verstreken M, et al: Nonsyndromic
207. Du X, Schwander M, Moresco EM, et al: A catechol-O-methyl- hearing impairment is associated with a mutation in DFNA5. Nat
transferase that is essential for auditory function in mice and Genet 20(2):194–197, 1998.
humans. Proc Natl Acad Sci U S A 105(38):14609–14614, 2008. 230. Bespalova IN, Van Camp G, Bom SJ, et al: Mutations in the
208. Shabbir MI, Ahmed ZM, Khan SY, et al: Mutations of human Wolfram syndrome 1 gene (WFS1) are a common cause of low
TMHS cause recessively inherited non-syndromic hearing loss. frequency sensorineural hearing loss. Hum Mol Genet 10(22):2501–
J Med Genet 43(8):634–640, 2006. 2508, 2001.
209. Tlili A, Mannikko M, Charfedine I, et al: A novel autosomal reces- 231. Lesperance MM, Hall JW, 3rd, Bess FH, et al: A gene for autoso-
sive non-syndromic deafness locus, DFNB66, maps to chromo- mal dominant nonsyndromic hereditary hearing impairment
some 6p21.2-22.3 in a large Tunisian consanguineous family. Hum maps to 4p16.3. Hum Mol Genet 4(10):1967–1972, 1995.
Hered 60(3):123–128, 2005. 232. Young TL, Ives E, Lynch E, et al: Non-syndromic progressive
210. Kalay E, Li Y, Uzumcu A, et al: Mutations in the lipoma HMGIC hearing loss DFNA38 is caused by heterozygous missense muta-
fusion partner-like 5 (LHFPL5) gene cause autosomal recessive tion in the Wolfram syndrome gene WFS1. Hum Mol Genet
nonsyndromic hearing loss. Hum Mutat 27(7):633–639, 2006. 10(22):2509–2514, 2001.
211. Waryah AM, Rehman A, Ahmed ZM, et al: DFNB74, a novel auto- 233. Manolis EN, Yandavi N, Nadol JB, Jr, et al: A gene for non-
somal recessive nonsyndromic hearing impairment locus on chro- syndromic autosomal dominant progressive postlingual sensori-
mosome 12q14.2-q15. Clin Genet 76(3):270–275, 2009. neural hearing loss maps to chromosome 14q12-13. Hum Mol
212. Ahmed ZM, Yousaf R, Lee BC, et al: Functional null mutations Genet 5(7):1047–1050, 1996.
of MSRB3 encoding methionine sulfoxide reductase are associ- 234. Robertson NG, Lu L, Heller S, et al: Mutations in a novel cochlear
ated with human deafness DFNB74. Am J Hum Genet 88(1):19–29, gene cause DFNA9, a human nonsyndromic deafness with vestibu-
2011. lar dysfunction. Nat Genet 20(3):299–303, 1998.
213. Grillet N, Schwander M, Hildebrand MS, et al: Mutations in 235. O’Neill ME, Marietta J, Nishimura D, et al: A gene for autosomal
LOXHD1, an evolutionarily conserved stereociliary protein, dominant late-onset progressive non-syndromic hearing loss,
disrupt hair cell function in mice and cause progressive hearing DFNA10, maps to chromosome 6. Hum Mol Genet 5(6):853–856,
loss in humans. Am J Hum Genet 85(3):328–337, 2009. 1996.
214. Rehman AU, Morell RJ, Belyantseva IA, et al: Targeted capture 236. Wayne S, Robertson NG, DeClau F, et al: Mutations in the tran-
and next-generation sequencing identifies C9orf75, encoding scriptional activator EYA4 cause late-onset deafness at the DFNA10
taperin, as the mutated gene in nonsyndromic deafness DFNB79. locus. Hum Mol Genet 10(3):195–200, 2001.
Am J Hum Genet 86(3):378–388, 2010. 237. Tamagawa Y, Kitamura K, Ishida T, et al: A gene for a dominant
215. Li Y, Pohl E, Boulouiz R, et al: Mutations in TPRN cause a progres- form of non-syndromic sensorineural deafness (DFNA11) maps
sive form of autosomal-recessive nonsyndromic hearing loss. Am J within the region containing the DFNB2 recessive deafness gene.
Hum Genet 86(3):479–484, 2010. Hum Mol Genet 5(6):849–852, 1996.
216. Walsh T, Shahin H, Elkan-Miller T, et al: Whole exome sequencing 238. Brown MR, Tomek MS, Van Laer L, et al: A novel locus for auto-
and homozygosity mapping identify mutation in the cell polarity somal dominant nonsyndromic hearing loss, DFNA13, maps to
protein GPSM2 as the cause of nonsyndromic hearing loss chromosome 6p. Am J Hum Genet 61(4):924–927, 1997.
DFNB82. Am J Hum Genet 87(1):90–94, 2010. 239. McGuirt WT, Prasad SD, Griffith AJ, et al: Mutations in COL11A2
217. Schraders M, Oostrik J, Huygen PL, et al: Mutations in PTPRQ cause non-syndromic hearing loss (DFNA13). Nat Genet 23(4):413–
are a cause of autosomal-recessive nonsyndromic hearing impair- 419, 1999.
ment DFNB84 and associated with vestibular dysfunction. Am J 240. Melchionda S, Ahituv N, Bisceglia L, et al: MYO6, the human
Hum Genet 86(4):604–610, 2010. homologue of the gene responsible for deafness in Snell’s waltzer
218. Liu XZ, Xia XJ, Xu LR, et al: Mutations in connexin31 underlie mice, is mutated in autosomal dominant nonsyndromic hearing
recessive as well as dominant non-syndromic hearing loss. Hum loss. Am J Hum Genet 69(3):635–640, 2001.
Mol Genet 9(1):63–67, 2000. 241. Vahava O, Morell R, Lynch ED, et al: Mutation in transcription
219. Sirmaci A, Erbek S, Price J, et al: A truncating mutation in factor POU4F3 associated with inherited progressive hearing loss
SERPINB6 is associated with autosomal-recessive nonsyndromic in humans. Science 279(5358):1950–1954, 1998.
sensorineural hearing loss. Am J Hum Genet 86(5):797–804, 2010. 242. Lalwani AK, Goldstein JA, Kelley MJ, et al: Human nonsyndromic
220. Leon PE, Raventos H, Lynch E, et al: The gene for an inherited hereditary deafness DFNA17 is due to a mutation in nonmuscle
form of deafness maps to chromosome 5q31. Proc Natl Acad Sci U myosin MYH9. Am J Hum Genet 67(5):1121–1128, 2000.
S A 89(11):5181–5184, 1992. 243. van Wijk E, Krieger E, Kemperman MH, et al: A mutation in
221. Lynch ED, Lee MK, Morrow JE, et al: Nonsyndromic deafness the gamma actin 1 (ACTG1) gene causes autosomal domi-
DFNA1 associated with mutation of a human homolog of the nant hearing loss (DFNA20/26). J Med Genet 40(12):879–884,
Drosophila gene diaphanous. Science 278(5341):1315–1318, 1997. 2003.
222. Xia JH, Liu CY, Tang BS, et al: Mutations in the gene encoding 244. Zhu M, Yang T, Wei S, et al: Mutations in the gamma-actin gene
gap junction protein beta-3 associated with autosomal dominant (ACTG1) are associated with dominant progressive deafness
hearing impairment. Nat Genet 20(4):370–373, 1998. (DFNA20/26). Am J Hum Genet 73(5):1082–1091, 2003.
2300.e6 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
245. Greene CC, McMillan PM, Barker SE, et al: DFNA25, a novel locus 267. Trofatter JA, MacCollin MM, Rutter JL, et al: A novel moesin-,
for dominant nonsyndromic hereditary hearing impairment, ezrin-, radixin-like gene is a candidate for the neurofibromatosis
maps to 12q21-24. Am J Hum Genet 68(1):254–260, 2001. 2 tumor suppressor. Cell 72(5):791–800, 1993.
246. Ruel J, Emery S, Nouvian R, et al: Impairment of SLC17A8 encod- 268. Positional cloning of a gene involved in the pathogenesis of
ing vesicular glutamate transporter-3, VGLUT3, underlies nonsyn- Treacher Collins syndrome. The Treacher Collins Syndrome Col-
dromic deafness DFNA25 and inner hair cell dysfunction in null laborative Group. Nat Genet 12(2):130–136, 1996.
mice. Am J Hum Genet 83(2):278–292, 2008. 269. Yang T, Gurrola JG, 2nd, Wu H, et al: Mutations of KCNJ10
247. Peters LM, Anderson DW, Griffith AJ, et al: Mutation of a tran- together with mutations of SLC26A4 cause digenic nonsyndromic
scription factor, TFCP2L3, causes progressive autosomal domi- hearing loss associated with enlarged vestibular aqueduct syn-
nant hearing loss, DFNA28. Hum Mol Genet 11(23):2877–2885, drome. Am J Hum Genet 84(5):651–657, 2009.
2002. 270. Gerber S, Bonneau D, Gilbert B, et al: USH1A: chronicle of a slow
248. Xiao S, Yu C, Chou X, et al: Dentinogenesis imperfecta 1 with or death. Am J Hum Genet 78(2):357–359, 2006.
without progressive hearing loss is associated with distinct muta- 271. Kaplan J, Gerber S, Bonneau D, et al: A gene for Usher syndrome
tions in DSPP. Nat Genet 27(2):201–204, 2001. type I (USH1A) maps to chromosome 14q. Genomics 14(4):979–
249. Modamio-Hoybjor S, Moreno-Pelayo MA, Mencia A, et al: A novel 987, 1992.
locus for autosomal dominant nonsyndromic hearing loss 272. Bitner-Glindzicz M, Lindley KJ, Rutland P, et al: A recessive con-
(DFNA44) maps to chromosome 3q28-29. Hum Genet 112(1):24– tiguous gene deletion causing infantile hyperinsulinism, enter-
28, 2003. opathy and deafness identifies the Usher type 1C gene. Nat Genet
250. Donaudy F, Ferrara A, Esposito L, et al: Multiple mutations of 26(1):56–60, 2000.
MYO1A, a cochlear-expressed gene, in sensorineural hearing loss. 273. Verpy E, Leibovici M, Zwaenepoel I, et al: A defect in harmonin,
Am J Hum Genet 72(6):1571–1577, 2003. a PDZ domain-containing protein expressed in the inner ear
251. D’Adamo P, Pinna M, Capobianco S, et al: A novel autosomal sensory hair cells, underlies Usher syndrome type 1C. Nat Genet
dominant non-syndromic deafness locus (DFNA48) maps to 26(1):51–55, 2000.
12q13-q14 in a large Italian family. Hum Genet 112(3):319–320, 274. Smith RJ, Lee EC, Kimberling WJ, et al: Localization of two genes
2003. for Usher syndrome type I to chromosome 11. Genomics 14(4):995–
252. Modamio-Hoybjor S, Moreno-Pelayo MA, Mencia A, et al: A novel 1002, 1992.
locus for autosomal dominant nonsyndromic hearing loss, 275. Wayne S, Der Kaloustian VM, Schloss M, et al: Localization of the
DFNA50, maps to chromosome 7q32 between the DFNB17 and Usher syndrome type ID gene (Ush1D) to chromosome 10. Hum
DFNB13 deafness loci. J Med Genet 41(2):e14, 2004. Mol Genet 5(10):1689–1692, 1996.
253. Mencia A, Modamio-Hoybjor S, Redshaw N, et al: Mutations in 276. Bolz H, von Brederlow B, Ramirez A, et al: Mutation of CDH23,
the seed region of human miR-96 are responsible for nonsyn- encoding a new member of the cadherin gene family, causes
dromic progressive hearing loss. Nat Genet 41(5):609–613, 2009. Usher syndrome type 1D. Nat Genet 27(1):108–112, 2001.
254. Walsh T, Pierce SB, Lenz DR, et al: Genomic duplication and 277. Chaib H, Kaplan J, Gerber S, et al: A newly identified locus for
overexpression of TJP2/ZO-2 leads to altered expression of apop- Usher syndrome type I, USH1E, maps to chromosome 21q21.
tosis genes in progressive nonsyndromic hearing loss DFNA51. Am Hum Mol Genet 6(1):27–31, 1997.
J Hum Genet 87(1):101–109, 2010. 278. Alagramam KN, Yuan H, Kuehn MH, et al: Mutations in the novel
255. Cheng J, Zhu Y, He S, et al: Functional mutation of SMAC/ protocadherin PCDH15 cause Usher syndrome type 1F. Hum Mol
DIABLO, encoding a mitochondrial proapoptotic protein, causes Genet 10(16):1709–1718, 2001.
human progressive hearing loss DFNA64. Am J Hum Genet 89(1): 279. Mustapha M, Chouery E, Torchard-Pagnez D, et al: A novel locus
56–66, 2011. for Usher syndrome type I, USH1G, maps to chromosome 17q24-
256. Tyson J, Bellman S, Newton V, et al: Mapping of DFN2 to Xq22. 25. Hum Genet 110(4):348–350, 2002.
Hum Mol Genet 5(12):2055–2060, 1996. 280. Weil D, El-Amraoui A, Masmoudi S, et al: Usher syndrome type I
257. Abdelfatah N, Merner N, Houston J, et al: A novel deletion in G (USH1G) is caused by mutations in the gene encoding SANS,
SMPX causes a rare form of X-linked progressive hearing loss in a protein that associates with the USH1C protein, harmonin. Hum
two families due to a founder effect. Hum Mutat 34(1):66–69, Mol Genet 12(5):463–471, 2003.
2013. 281. Ahmed ZM, Riazuddin S, Khan SN, et al: USH1H, a novel locus
258. Zhao H, Li R, Wang Q, et al: Maternally inherited aminoglycoside- for type I Usher syndrome, maps to chromosome 15q22-23. Clin
induced and nonsyndromic deafness is associated with the novel Genet 75(1):86–91, 2009.
C1494T mutation in the mitochondrial 12S rRNA gene in a large 282. Kimberling WJ, Weston MD, Moller C, et al: Localization of Usher
Chinese family. Am J Hum Genet 74(1):139–152, 2004. syndrome type II to chromosome 1q. Genomics 7(2):245–249, 1990.
259. Reid FM, Vernham GA, Jacobs HT: A novel mitochondrial point 283. Hmani M, Ghorbel A, Boulila-Elgaied A, et al: A novel locus for
mutation in a maternal pedigree with sensorineural deafness. Usher syndrome type II, USH2B, maps to chromosome 3 at p23-
Hum Mutat 3(3):243–247, 1994. 24.2. Eur J Hum Genet 7(3):363–367, 1999.
260. Tiranti V, Chariot P, Carella F, et al: Maternally inherited hearing 284. Pieke-Dahl S, Moller CG, Kelley PM, et al: Genetic heterogeneity
loss, ataxia and myoclonus associated with a novel point mutation of Usher syndrome type II: localisation to chromosome 5q. J Med
in mitochondrial tRNASer(UCN) gene. Hum Mol Genet 4(8):1421– Genet 37(4):256–262, 2000.
1427, 1995. 285. Weston MD, Luijendijk MW, Humphrey KD, et al: Mutations in
261. Hutchin TP, Parker MJ, Young ID, et al: A novel mutation in the the VLGR1 gene implicate G-protein signaling in the pathogen-
mitochondrial tRNA(Ser(UCN)) gene in a family with non- esis of Usher syndrome type II. Am J Hum Genet 74(2):357–366,
syndromic sensorineural hearing impairment. J Med Genet 37(9): 2004.
692–694, 2000. 286. Ebermann I, Scholl HP, Charbel Issa P, et al: A novel gene for
262. Friedman RA, Bykhovskaya Y, Sue CM, et al: Maternally inherited Usher syndrome type 2: mutations in the long isoform of whirlin
nonsyndromic hearing loss. Am J Med Genet 84(4):369–372, 1999. are associated with retinitis pigmentosa and sensorineural hearing
263. Kumar S, Deffenbacher K, Marres HA, et al: Genomewide search loss. Hum Genet 121(2):203–211, 2007.
and genetic localization of a second gene associated with autoso- 287. Joensuu T, Hamalainen R, Yuan B, et al: Mutations in a novel gene
mal dominant branchio-oto-renal syndrome: clinical and genetic with transmembrane domains underlie Usher syndrome type 3.
implications. Am J Hum Genet 66(5):1715–1720, 2000. Am J Hum Genet 69(4):673–684, 2001.
264. Ruf RG, Berkman J, Wolf MT, et al: A gene locus for branchio-otic 288. Sankila EM, Pakarinen L, Kaariainen H, et al: Assignment of an
syndrome maps to chromosome 14q21.3-q24.3. J Med Genet 40(7): Usher syndrome type III (USH3) gene to chromosome 3q. Hum
515–519, 2003. Mol Genet 4(1):93–98, 1995.
265. Edery P, Attie T, Amiel J, et al: Mutation of the endothelin-3 gene 289. Ebermann I, Phillips JB, Liebau MC, et al: PDZD7 is a modifier
in the Waardenburg-Hirschsprung disease (Shah-Waardenburg of retinal disease and a contributor to digenic Usher syndrome.
syndrome). Nat Genet 12(4):442–444, 1996. J Clin Invest 120(6):1812–1823, 2010.
266. Baker S, Booth C, Fillman C, et al: A loss of function mutation in 290. Blanton SH, Pandya A, Landa BL, et al: Fine mapping of the
the COL9A2 gene causes autosomal recessive Stickler syndrome. human biotinidase gene and haplotype analysis of five common
Am J Med Genet Part A 155A(7):1668–1672, 2011. mutations. Hum Hered 50(2):102–111, 2000.
148 | GENETIC SENSORINEURAL HEARING LOSS 2300.e7
291. Mihalik SJ, Morrell JC, Kim D, et al: Identification of PAHX, a 297. van den Ouweland JM, Lemkes HH, Ruitenbeek W, et al: Muta-
Refsum disease gene. Nat Genet 17(2):185–189, 1997. tion in mitochondrial tRNA(Leu)(UUR) gene in a large pedigree
292. Mochizuki T, Lemmink HH, Mariyama M, et al: Identification of with maternally transmitted type II diabetes mellitus and deafness.
mutations in the alpha 3(IV) and alpha 4(IV) collagen genes in Nat Genet 1(5):368–371, 1992.
autosomal recessive Alport syndrome. Nat Genet 8(1):77–81, 1994. 298. Shoffner JM, Lott MT, Lezza AM, et al: Myoclonic epilepsy and
293. Roesch K, Curran SP, Tranebjaerg L, et al: Human deafness dys- ragged-red fiber disease (MERRF) is associated with a mitochon-
tonia syndrome is caused by a defect in assembly of the DDP1/ drial DNA tRNA(Lys) mutation. Cell 61(6):931–937, 1990.
TIMM8a-TIMM13 complex. Hum Mol Genet 11(5):477–486, 2002. 299. Zeviani M, Muntoni F, Savarese N, et al: A MERRF/MELAS
294. Berger W, van de Pol D, Warburg M, et al: Mutations in the can- overlap syndrome associated with a new point mutation in the
didate gene for Norrie disease. Hum Mol Genet 1(7):461–465, mitochondrial DNA tRNA(Lys) gene. Eur J Hum Genet 1(1):80–87,
1992. 1993.
295. Chen ZY, Hendriks RW, Jobling MA, et al: Isolation and charac- 300. Kameoka K, Isotani H, Tanaka K, et al: Novel mitochondrial DNA
terization of a candidate gene for Norrie disease. Nat Genet mutation in tRNA(Lys) (8296A–>G) associated with diabetes.
1(3):204–208, 1992. Biochem Biophys Res Commun 245(2):523–527, 1998.
296. Goto Y, Nonaka I, Horai S: A mutation in the tRNA(Leu)(UUR) 301. Ballinger SW, Shoffner JM, Hedaya EV, et al: Maternally transmit-
gene associated with the MELAS subgroup of mitochondrial ted diabetes and deafness associated with a 10.4 kb mitochondrial
encephalomyopathies. Nature 348(6302):651–653, 1990. DNA deletion. Nat Genet 1(1):11–15, 1992.
Otologic Manifestations 149
of Systemic Disease
Saumil N. Merchant† | Joseph B. Nadol Jr
Key Points
■ A wide variety of systemic diseases can affect the temporal bone, including granulomatous and
infectious diseases, neoplasms, disorders of bone, storage and metabolic diseases, and autoimmune
and immunodeficiency disorders.
■ Otologic manifestations of a particular systemic disease are determined by location and extent of
involvement within the temporal bone and the nature of the disease. Any part of the temporal
bone may be affected, and the clinical features may include conductive or sensorineural hearing
loss, vestibular manifestations, otalgia, and facial nerve paralysis.
■ Systemic disease may mimic more common otologic disorders, such as acute or chronic otitis
media, sudden idiopathic deafness, and Bell palsy.
■ Otologic manifestations may constitute a small part of a wider constellation of symptoms and signs,
or they may be the sole and initial presenting feature of a systemic disease.
■ Diagnosis may be challenging, and it requires a high level of suspicion and use of ancillary tests such
as laboratory studies, radiographic evaluation, and biopsy.
■ Management of otologic symptoms must be individualized and often requires coordinated care
across multiple disciplines.
S ystemic diseases that may involve the ear include granuloma- well as the prognosis and treatment, differ greatly. Histopatho-
tous and infectious processes, tumors, bone disorders, storage logically, the primary lesion of LCH is formed by collections of
diseases, collagen vascular and autoimmune diseases, and pathologic Langerhans cells with variable numbers of eosino-
immunodeficiency disorders (Box 149-1). In some of these dis- phils, macrophages, and lymphocytes. Diagnostic characteris-
eases, the initial clinical symptoms may occur in the temporal tics of the pathologic Langerhans cells include nuclei that
bone and may be confused with other diseases limited to the ear. appear deeply indented and elongated on light microscopy,
cytoplasm that is pale and abundant, Birbeck granules on elec-
GRANULOMATOUS AND tron microscopy, expression of CD1 on the cell surface, and
positive immunostaining for S100 protein and for the langerin
INFECTIOUS DISEASES protein.2,3 The etiology and pathogenesis of LCH remain
Chronic otitis media with otorrhea, inflammation, and granula- unknown, although more recent research has begun to shed
tion of the middle ear and mastoid is one of the most common some light on these.2,4,5 Current thinking favors the notion
entities treated by otolaryngologists. Several more generalized that LCH results from immunologic dysfunction that leads to
disease entities—such as Langerhans cell histiocytosis, tubercu- unchecked proliferation of pathologic Langerhans cells.5
losis, Wegener granulomatosis, and mycotic diseases—may Unifocal eosinophilic granuloma, which occurs in children
closely mimic the symptoms of chronic suppurative otitis media, and young adults, shows a male predominance and appears
whereas others, such as Lyme disease and sarcoidosis, may as a solitary osteolytic lesion in the femora, pelvis, scapulae,
mimic idiopathic cranial neuropathy. vertebrae, ribs, mandible, maxilla, or skull, which includes the
temporal bone. The lesion may be asymptomatic, or it may
cause pain, local swelling, or pathologic fracture. No systemic
LANGERHANS CELL HISTIOCYTOSIS manifestations have been reported. The clinical course is
Langerhans cell histiocytosis (LCH), formerly called histiocytosis X, typically benign, the prognosis is excellent, and spontaneous
refers to a group of disorders characterized by a proliferation regression may occur. Local curettage with or without low-dose
of cytologically benign histiocytes. The term histiocytosis X was irradiation (approximately 60 Gy)6 is usually curative, although
proposed by Lichtenstein,1 who considered eosinophilic granu- temporary splinting or casting may be necessary for weight-
loma, Hand-Schüller-Christian disease, and Letterer-Siwe bearing bones. Follow-up examination with a radiographic skel-
disease to be related disorders because of the similarity of their etal survey should be performed to detect lesions at other sites;
pathologic lesions. However, the severity of these diseases, as such lesions are almost always found within 1 year.
Hand-Schüller-Christian disease may be best understood as
a multifocal form of LCH. It usually occurs in children younger
†
Deceased. than 5 years of age, and it is characterized by multifocal
2301
2302 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
marrow transplantation or hematopoietic stem cell transplanta-
Box 149-1. SYSTEMIC DISEASES THAT AFFECT THE EAR
tion has been reported to be successful in a few advanced
Granulomatous and Infectious Diseases refractory cases of LCH.8
Langerhans cell histiocytosis
Tuberculosis Otologic Manifestations
Wegener granulomatosis The mastoid is a common site of involvement in LCH. When
Sarcoidosis small, the lesion is asymptomatic. As it expands, it may manifest
Syphilis in several ways: by erosion of the posterior bony canal wall; by
Lyme disease
erosion through the cortex of the mastoid, zygomatic, or squa-
Mycotic diseases
Cytomegalic inclusion disease mous portions; or by secondary infection.9 The otic capsule and
facial nerve are relatively resistant. Although sensorineural
Neoplastic Diseases
hearing loss (SNHL), vertigo, and facial nerve paralysis can
Multiple myeloma occur, they are infrequent. Similarly, extension beyond the tem-
Leukemia
poral bone to the jugular fossa and skull base is rare.
Lymphoma
Metastatic neoplasms The reported incidence of otologic manifestations in
patients with LCH is 15% to 61%,7 and they may be the first
Diseases of Bone
sign of the disease. The most common symptom is otorrhea,
Paget disease (osteitis deformans) followed by postauricular swelling, hearing loss, and vertigo.10,11
Osteogenesis imperfecta
The most common sign is granulation tissue or aural polyps
Fibrous dysplasia
Osteopetroses in the external auditory canal. The disease may manifest with
Osteitis fibrosa cystica perforation of the tympanic membrane, otitis media, otitis
externa, fistula between the mastoid and the external canal, or
Storage and Metabolic Diseases
nontender postauricular swelling. Occasionally, inner ear symp-
Mucopolysaccharidoses
toms and a positive fistula test are found in the presence of an
Gout
Ochronosis intact tympanic membrane. The disease often mimics chronic
otitis media, and mastoid surgery is frequently performed
Collagen Vascular and Autoimmune Diseases
before the diagnosis is made.9
Multiple sclerosis Diagnosis of LCH is suggested by 1) an inflammatory disor-
Susac disease
der of the middle ear and mastoid that does not respond to
Immunodeficiency Disorders routine antibiotic therapy, 2) bilateral destructive ear disease,
Primary or Congenital 3) an elevated erythrocyte sedimentation rate (ESR) in the
Humoral immunodeficiency disorders absence of acute infection, 4) exuberant granulation tissue
Cellular immunodeficiency disorders after mastoid surgery with a persistently draining cavity, and 5)
Disorders of phagocyte function associated skin and systemic lesions. Radiographs show destruc-
Complement system defects tive lesions in the mastoid and temporal bones (Figs. 149-1 and
Acquired 149-2).9,10,12 The diagnosis is established by biopsy; because the
Acquired immunodeficiency syndrome surface of the granulation tissue often shows infection, necro-
Other sis, and fibrosis, tissue should be acquired from deeper parts of
Genetically determined defects the lesion.13 Microscopic findings include sheets of histiocytes
osteolytic lesions with limited extraskeletal involvement of skin,
lymph nodes, and viscera. Multiple lesions are evident at diag-
nosis or develop within 6 months after a unifocal lesion appears.
Systemic manifestations include fever, anorexia, recurrent
upper respiratory infections, anterior cervical lymphadenopa-
thy, otitis media, and hepatosplenomegaly. The classic triad of
osteolytic skull lesions, exophthalmos as a result of orbital bone
involvement, and diabetes insipidus secondary to pituitary
disease may be seen in 25% of patients.7 Chest radiograph
may show diffuse pulmonary infiltration, particularly in
central and perihilar areas. Hilar lymphadenopathy is rare.
Diagnosis requires biopsy of an accessible lesion. Spontaneous
regression may occur, but the disease is typically chronic, and
low-dose chemotherapy may be required to control systemic
manifestations.
Letterer-Siwe disease is a disseminated form of LCH that occurs
in children younger than 3 years old and manifests with the
diffuse involvement of multiple organs. Manifestations include
fever, seborrheic or eczema-like rash, oral lesions, lymphade-
nopathy, hepatosplenomegaly, multiple bony lesions, diffuse
replacement of marrow with resulting blood dyscrasias, and
pulmonary infiltration with respiratory failure. The disease is
virulent, with a poor prognosis and a high mortality rate. Treat-
ment consists of varying combinations of corticosteroids and
cytotoxic drugs such as methotrexate, mercaptopurine, vincris- FIGURE 149-1. Multiple eosinophilic granulomas in a 32-year-old woman.
tine, vinblastine, chlorambucil, cyclophosphamide, and etopo- Two lytic lesions of the skull show beveled edges (arrows) and nonsclerotic
side. High-dose chemotherapy and radiation followed by bone margins, which are typical of this disease.
149 | OTOLOGIC MANIFESTATIONS OF SYSTEMIC DISEASE 2303
inoculation through a perforation of the tympanic membrane
with a variable number of eosinophils, plasma cells, polymor- is also possible. Middle ear involvement in the absence of active
phonuclear leukocytes, and multinucleated cells (Figs. 149-3 pulmonary disease is rare but may occur.
through 149-5). Areas of hemorrhage and necrosis are common. During the early stages of tuberculous otitis media, the
A definitive diagnosis of LCH is usually made on the basis of tympanic membrane becomes thickened, and the otoscopic
immunohistochemistry and electron microscopic studies.2,4 landmarks become obliterated. Conductive hearing loss results
from middle ear effusion, thickening of the tympanic mem-
brane and middle ear mucosa, and destruction of the ossicles
TUBERCULOSIS (Figs. 149-6 and 149-7). No characteristic pain or tenderness is
The incidence of tuberculous otitis media has decreased dra- evident, but lymphadenopathy in the high jugular chain occurs
matically. During the early part of the twentieth century, 1.3% early. Multiple small perforations of the tympanic membrane
to 18.6% of all cases of chronic otitis media were reportedly may occur, with seropurulent drainage. The perforations
caused by tuberculosis, whereas more recent studies report quickly coalesce to cause loss of the tympanic membrane. Like-
tuberculous otitis media rates of 0.05% to 0.9%.14 The last 20 wise, a myringotomy site in an intact tympanic membrane may
years have witnessed an increase in incidence of tuberculosis, enlarge quickly. The middle ear mucosa appears to be hyper-
however, caused in part by the aggressive nature of tuberculosis emic with polypoid granulation. Osseous involvement results in
in individuals infected with human immunodeficiency virus the sequestration of bone and the destruction of the inner ear,
(HIV) and by an emerging resistance to antituberculosis the facial nerve, or both. Destruction of the mastoid tip may
drugs.15 Mycobacterium tuberculosis is the infective organism in result in an asymptomatic, nontender Bezold abscess (a “cold”
most cases; occasionally, atypical mycobacteria (e.g., M. avium abscess). Rarely, tuberculosis can also manifest as primary
and M. fortuitum) are responsible.16 tuberculous petrositis17 or as SNHL caused by chronic labyrin-
Tuberculosis of the middle ear and mastoid may occur as a thitis and tuberculous meningitis.18
result of hematogeneous or lymphatic spread or by extension
to the middle ear cleft through the eustachian tube. Direct
H
P
ME
M
PA
FIGURE 149-3. Granulation tissue typical of histiocytosis is seen in lytic FIGURE 149-5. Eosinophilic granuloma of the external ear canal with
lesions within the mastoid (M), middle ear (ME), and petrous apex (PA) of a plasma cells (P), eosinophils, and histiocytes (H), which show “folded” nuclei,
4-year old boy with Letterer-Siwe disease of the temporal bone (×4). a morphology peculiar to the histiocytoses (×640).
2304 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
FIGURE 149-6. Tuberculous granulation tissue filled the middle ear of a WEGENER GRANULOMATOSIS
57-year-old man who died of miliary tuberculosis (×12.35). FN, facial nerve; Wegener granulomatosis is a granulomatous inflammatory
I, incus; M, malleus; MA, mastoid; TM, tympanic membrane.
process with necrotizing vasculitis. It primarily affects the upper
and lower respiratory tracts and kidneys, but it can involve
essentially any organ in the body. The disease occurs equally
The diagnosis of tuberculous involvement of the middle ear in men and women, and the mean age of onset is 40 years.
is usually delayed. The characteristic signs and symptoms Common presenting symptoms include headache, sinusitis, rhi-
include 1) multiple perforations of the tympanic membrane norrhea, otitis media, fever, and arthralgia.20,21 Upper airway
that quickly coalesce to form total tympanic membrane loss, and sinus involvement occurs in 75% to 90% of cases. Pulmo-
2) nontender cervical lymphadenopathy, 3) intractable otitis nary manifestations include cough, pleuritic chest pain, hemop-
media with polypoid granulation, and 4) bony sequestration. tysis, and nodular or cavitary infiltrates on chest radiography.
These should alert the physician to this possible diagnosis. Otic Pulmonary manifestations occur in 65% to 85% of cases. Other
capsule involvement with resultant loss of auditory and vestibu- manifestations include glomerulonephritis (60% to 75% of
lar function may be the first symptom and indicates a process cases); eye involvement in the form of conjunctivitis, iritis, scle-
that differs from the more typical chronic otitis media. Defini- ritis, or proptosis (15% to 50% of cases); and dermatologic
tive diagnosis is made by histopathologic examination of tissue findings of necrotic ulcerations, vesicles, or petechiae.
from the middle ear or mastoid, which shows a granulomatous Laboratory findings in WG may include normochromic nor-
process with multinucleated giant cells (Langerhans cells; see mocytic anemia; thrombocytosis; positive rheumatoid factor;
Figs. 149-6 and 149-7) and histologic demonstration of acid-fast and hyperglobulinemia, particularly of IgA. The ESR is almost
organisms of tuberculosis. The accurate identification of the always elevated. The discovery in 1985 of autoantibodies
mycobacterial species and drug-resistant isolates by culture is directed against the cytoplasmic constituents of neutrophils
still the gold standard, although culture is time consuming and (antineutrophil cytoplasmic antibody [ANCA] and cytoplasmic
takes several weeks. The process is facilitated by the use of ANCA [c-ANCA]) in patients with WG was a major advance in
several different types of nucleic acid amplification tests that the diagnosis and understanding of WG.22 A positive ANCA test,
have been approved by the U.S. Food and Drug Administration especially proteinase 3–specific c-ANCA, is very helpful in estab-
for rapid identification of M. tuberculosis.15,19 lishing or supporting a diagnosis of WG. The specificity of posi-
tive c-ANCA testing in WG is greater than 95%; the sensitivity
is variable and depends on the phase and type of WG. In more
than 90% of patients with systemic and active WG, c-ANCA is
positive; whereas in limited WG (e.g., ear or head and neck
only or inactive disease), the sensitivity of c-ANCA is only 65%
to 70%. In addition, the antibody titer parallels the activity of
the disease, although data conflict about the value in using the
titer as a guide for immunosuppressive therapy.
The diagnosis of WG is made histologically by the presence
of necrosis, granulomatous inflammation with multinucleated
TM giant cells, vasculitis, and microabscess formation (Figs. 149-8
and 149-9). Small biopsy specimens, however, such as those
obtained from the ear or upper airway, may lack all of the
various diagnostic features. In such cases, a diagnosis of WG
can be made on the basis of the clinical presentation, ANCA
G testing, and repeat biopsy specimens taken from the same or
related sites.
The etiology and pathogenesis of WG are unknown. Infec-
tious, genetic, and environmental risk factors and combina-
tions thereof have been proposed. The evidence to date
suggests that WG is a complex, immune-mediated disorder in
FIGURE 149-7. A magnified view of the tympanic membrane (TM) seen in which tissue injury results from the interplay of an initiating
Figure 149-6. The TM is intact but greatly thickened by tuberculous granula- inflammatory event and a highly specific immune response.23
tion tissue that contains the typical epithelioid, round, and multinucleated Part of this response consists of the production of ANCA,
giant (G) cells (×100). directed against antigens present within the primary granules
149 | OTOLOGIC MANIFESTATIONS OF SYSTEMIC DISEASE 2305
ET
EAC
TT
ME
CO
IAC
FIGURE 149-10. Bilateral hilar adenopathy and linear parenchymal densi-
FIGURE 149-8. Wegener granulomatosis causing blockage of the eusta- ties in a 56-year-old woman with pulmonary sarcoidosis.
chian tube (ET) and infiltration of the middle ear (ME) in a 41-year-old man
(×10). CO, cochlea; EAC, external auditory canal; IAC, internal auditory
canal; TT, tensor tympani. (Courtesy Leslie Michaels, MD, Institute of Laryngol-
ogy and Otology, London, United Kingdom.) others may have frank granulomatous involvement of the
middle ear and mastoid. The process can extend to involve the
facial nerve and inner ear, and this usually manifests as rapidly
progressive SNHL and loss of vestibular function. Otologic
of neutrophils and monocytes; these antibodies produce tissue disease may be the sole and initial presenting feature in some
damage by interacting with primed neutrophils and endothe- patients with WG.
lial cells.
The prognosis of WG has dramatically improved from a
mortality rate of 82% before the era of immunosuppressive
SARCOIDOSIS
therapy to the current remission rate of more than 75% with Sarcoidosis is a chronic multisystem disorder of unknown etiol-
appropriate medical therapy.24 Induction of remission is usually ogy characterized by the presence of noncaseating granulomas.
achieved with high doses of corticosteroids, cyclophosphamide, It most frequently affects the lungs, although almost all body
or methotrexate given for 3 to 6 months. Maintenance of remis- parts can be affected. The disease shows a female predominance
sion is achieved with lower doses of corticosteroids and less and is 10 times more common in blacks than in whites. The
toxic alternatives to cyclophosphamide, such as azathioprine, onset of disease is usually during the third to fourth decade of
methotrexate, trimethoprim-sulfamethoxazole, or other drug life. Common presenting manifestations include bilateral hilar
combinations. Other therapies that have been used include adenopathy on chest radiography, cough, and granulomatous
leflunomide, mycophenolate mofetil, and the tumor necrosis skin rash. Other manifestations include iridocyclitis, keratocon-
factor inhibitors etanercept and infliximab. junctivitis, peripheral lymphadenopathy, hepatosplenomegaly,
cardiac failure, myalgia, and arthralgia. Neurologic involvement
Otologic Manifestations includes central and peripheral manifestations, and the facial
The middle ear and mastoid are the most common sites within and optic nerves are the most commonly affected cranial nerves.
the temporal bone that are involved in patients with WG,20,25 Either peripheral mononeuritis or polyneuritis may be seen.
and WG may cause obstruction of the eustachian tube with Laboratory findings may include hilar adenopathy on chest
resultant serous otitis media and conductive hearing loss (see radiography (Fig. 149-10), hypercalcemia, and elevated serum
Fig. 149-8). Some patients also have purulent otitis media, and angiotensin-converting enzyme. The histopathologic feature of
GR
G
L
A B
FIGURE 149-9. Wegener granulomatosis in lung biopsy specimen. A, Arteriole shows necrotizing vasculitis with obliteration of the lumen (L) and infiltration
of the vessel wall with polymorphonuclear leukocytes (elastic stain; ×256). B, Vessel wall shows fibrin deposit (F), giant cells (G), and granuloma (GR) (hema-
toxylin and eosin stain; ×256).
2306 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
SYPHILIS
Congenital and acquired syphilis may affect the middle ear in
the late latent and tertiary forms. In the late latent form, the
middle ear and mastoid may be affected by a rarefying osteitis
with leukocytic infiltration of the ossicles and mastoid bone FIGURE 149-13. Area of active bone resorption in a 43-year-old patient
(Figs. 149-12 and 149-13). A similar but larger lesion of tertiary with congenital syphilis. The inflammatory infiltrate includes lymphocytes,
syphilis, the gumma, shows obliterative arteritis and central plasma cells, and multinucleated giant cells (×396).
149 | OTOLOGIC MANIFESTATIONS OF SYSTEMIC DISEASE 2307
The middle ear and mastoid can be involved as a result of
ascending infection along the eustachian tube and tensor
tympani, which is often seen in mucormycosis (Fig. 149-14), Cytomegalovirus
or through superinfection of existing chronic otitis media.45 inclusions
Destruction of the middle ear cleft ensues, often with extension
to the surrounding structures, including thrombosis or rupture Crista lateral
of the internal carotid artery.46 Other routes of infection include semicircular
hematogeneous embolic dissemination, which can result in canal
multiple granulomas throughout the temporal bone, and
through cryptococcal involvement of the CNS, which can cause
invasion and degeneration of the nerve trunks in the internal
auditory canal.47
NEOPLASTIC DISEASES
Although neoplasms of the temporal bone are discussed else-
where (see Chapters 176 and 177), three neoplasms—multiple Cytomegalovirus
myeloma, leukemia, and metastatic tumors—warrant mention inclusions
here, because temporal bone manifestations can occur with
these diseases. 200 µm
B
MULTIPLE MYELOMA FIGURE 149-15. Cytology of the temporal bone of a full-term male infant
Multiple myeloma is a malignancy of plasma cells derived from who died in the first day after birth of disseminated cytomegalic inclusion
disease and necrotizing enterocolitis. Although the right temporal bone was
B lymphocytes, and its major feature is the demonstration of normal, numerous cytomegalic inclusions were apparent within the vestibu-
an abnormal monoclonal protein (M component) in blood, lar labyrinth on the left side, here in the lateral semicircular canal. In this case,
urine, or both. There is a slight male predominance, and the no inclusions were found within the cochlea on either side.
median age of onset is 60 years. Clinical manifestations are the
result of multiple plasma cell tumors and consist of severe bone
pain, pathologic fractures, failure of the bone marrow, renal
EAC failure, hypercalcemia, and recurrent infections.
Laboratory findings include the demonstration of the M
component on serum or urine electrophoresis, normochromic
normocytic anemia, hypercalcemia, and elevated blood urea
nitrogen levels. Typical radiographic findings include punched-
EF
out osteolytic lesions, which are particularly well seen on lateral
skull radiography. Bone marrow aspirates show infiltration by
plasma cells. For decades, the mainstay of therapy has been the
use of alkylating agents, such as melphalan and corticosteroids;
with this regimen, the median survival is approximately 3 years.
TT Important advances have been made more recently that have
substantially altered therapy for patients with myeloma. These
FN advances include the use of hematopoietic cell transplantation;
improved supportive care measures, such as the use of bisphos-
phonates and erythropoietin; and novel agents such as thalido-
mide, lenalidomide, and bortezomib.50,51
FIGURE 149-14. Mucormycosis in a 37-year-old man with diabetes. Inflam- Occasionally, only one plasma cell tumor (without marrow
matory cells have invaded and destroyed the tensor tympani muscle (TT), plasmacytosis) can be found. These lesions can occur in bone
and a hemorrhagic effusion (EF) is apparent in the mesotympanum (×12.35). (solitary bone plasmacytoma) or soft tissue (extramedullary
EAC, external auditory canal; FN, facial nerve. plasmacytoma), including the temporal bone. Both lesions can
149 | OTOLOGIC MANIFESTATIONS OF SYSTEMIC DISEASE 2309
CL
TM
PF
UN
1.0 mm
TM HLI
ME
PA
T
LYMPHOMA
Similar to leukemic infiltrates in the inner ear, both Hodgkin B
and non-Hodgkin lymphomas may result in hearing loss by
either hemorrhagic or malignant infiltrative lesions of the FIGURE 149-21. A, Cytology from a 58-year old man who died of non-
middle and inner ear (Fig. 149-21).63 Hodgkin follicular lymphoma diagnosed at age 52. He was treated with radia-
tion and chemotherapy. At age 58, he developed sudden total hearing loss
in the left ear, a rapidly progressive loss in the right ear, and vertigo. Audi-
METASTATIC NEOPLASMS ometry demonstrated profound loss on the left and a severe to profound
loss on the right. Hemorrhagic and lymphatic infiltration (HLI) was apparent
Secondary malignant tumors usually involve the temporal bone in the perilymphatic scalae of the cochleae. B, HLI of the perilymphatic space
through hematogeneous dissemination. The most common of the lateral semicircular canal.
sites of origin, in order of decreasing frequency, are the breast,
lung, prostate, and skin.64 The lesions are usually destructive
and osteolytic (Fig. 149-22); however, some lesions, such as
those from the prostate or breast, may be osteoblastic. The
petrous apex and internal auditory canal seem to be sites of
predilection for metastases, although any part of the temporal
bone may be involved (Fig. 149-23). The otic capsule seems to
be resistant to neoplastic invasion.65
Although otologic manifestations infrequently are the first
evidence of malignant disease, more often they are preceded
by other systemic symptoms. Involvement of the external canal,
middle ear cleft, or eustachian tube may cause conductive
hearing loss and pain; involvement of the otic capsule may
produce SNHL, vertigo, and facial nerve paralysis. In menin-
geal carcinomatosis, rapidly progressive unilateral or bilateral
SNHL is a common presenting symptom.66 Unilateral SNHL
may mimic a cerebellopontine angle tumor, and bilateral SNHL
may mimic immune-mediated inner ear disease. Diagnosis is
made by cytology of the cerebrospinal fluid.
EAC
ME
MA
PA
FC
IAC
IAC
otic capsule (see Fig. 149-29) and may result in the fixation of
OI
M the stapes footplate.
FIBROUS DYSPLASIA
Fibrous dysplasia is a benign, chronic, slowly progressive bone
disorder of unknown etiology characterized by the replacement
of normal bone with a variable amount of fibrous tissue and
woven bone. It may occur as part of Albright syndrome—
characterized by multiple bony lesions, abnormal pigmenta-
tion, endocrine dysfunction, and precocious puberty in
girls—or it may exist alone in the monostotic or polyostotic
form. The monostotic form is more common and usually occurs
in the skull, ribs, proximal femur, or tibia. In the polyostotic
form, skull lesions are seen in more than 50% of patients.
Clinical manifestations of fibrous dysplasia include bony
deformity, pathologic fracture, and cranial nerve palsies. The
disease starts early in life, usually in childhood; the monostotic
FIGURE 149-29. Osteogenesis imperfecta in a 15-year-old girl. The osteo-
genesis has replaced the enchondral and periosteal layers of the otic capsule. form may become quiescent at puberty, whereas the polyostotic
The neck of the malleus (M) is also involved (×8.5). form can continue to progress. Sarcomatous transformation
can occur, and incidence is estimated at 0.4%.88 Laboratory
findings include an elevated serum alkaline phosphatase level
trauma in childhood that ceased after puberty, a family history in 30% of patients with polyostotic fibrous dysplasia, with
of OI, and blue sclerae are helpful diagnostic clues. usually normal serum calcium and phosphorus levels. The
The conductive loss reflects structural changes in the ossi- typical radiographic findings include a radiolucent area with a
cles. Microfractures of the manubrium,79 fragility of the long well-defined smooth or scalloped edge and a ground-glass
process of the incus, and fracture or resorption of the crura of appearance. Areas of increased radiodensity may also be seen
the stapes have been reported.78,80,81 The stapedial footplate is (Figs. 149-31 and 149-32). The histopathology of fibrous dys-
typically described as thick, soft, and chalklike or granular, plasia consists of the replacement of normal cancellous bone
and it is usually fixed.82 Rehabilitation can be accomplished by by a fibrous stroma arranged in a whorled pattern. A variable
amplification or surgery.83-85 A stapedectomy can give results amount of irregularly arranged spicules of woven bone cause
similar to those seen with otosclerosis, but the procedure is the ground-glass radiographic changes (Fig. 149-33).
extremely delicate. Crimping the prosthesis around the incus Advances have been made toward understanding the cellu-
may cause a pathologic fracture, and a platinum ribbon is pre- lar and molecular basis of fibrous dysplasia. The lesion is com-
ferred to stainless steel wire.78 posed of immature mesenchymal osteoblastic precursor cells.
Histopathology of the temporal bone (Figs. 149-29 and An activating mutation is present in the gene that encodes the
149-30) in cases of OI type 2 has shown deficient ossification α-subunit of stimulatory G protein.89 Elevated levels of cyclic
of the endochondral layer of the otic capsule, which shows
increased amounts of fibrous tissue with numerous blood
vessels. The periosteal layer is often thin and deficient (see Fig.
149-30), and the stapes crura are often thin and incomplete.
The otopathology in cases of OI type 2 is very similar if not
identical to that seen in cases of otosclerosis.86,87 The abnormal
bone involves the periosteal and endochondral layers of the
ED
L
EC
M
FC I
LSCC
A
facial nerve, often with herniation of the nerve into the oval
window niche, has been a consistent finding.98-100 However, no should be performed in any child or young adult with recurrent
observable nerve compression is evident. These children often facial nerve paralysis to determine the possibility of osteopetro-
have recurrent episodes of acute otitis media, serous otitis sis. Total decompression of the facial nerve has been advocated
media, stenosis of the external auditory canal, conductive to ameliorate recurrent palsies.104,105
hearing loss or SNHL, and unilateral or bilateral facial nerve
paralysis.101,102
In the more benign adult form (autosomal-dominant type 2
OSTEITIS FIBROSA CYSTICA
osteopetrosis), the temporal bone is markedly sclerotic with Osteitis fibrosa cystica, also known as von Recklinghausen disease,
obliteration of the mastoid air cells and a narrowing of the is a bone lesion caused by excess parathyroid hormone, and it
eustachian tube and the external and internal auditory canals. is characterized in classic cases by osteoclastic bone resorption,
Exostotic overgrowth of the periosteal bone surrounding the marrow fibrosis, bone cysts, bone pain, and fractures. In most
tympanic cavity can occur (Fig. 149-36), with ankylosis of the cases, it is caused by primary hyperparathyroidism, usually
ossicles and obliteration of the oval and round window niches because of an adenoma. Other manifestations relate to hyper-
(Fig. 149-37). These changes explain the common finding of calcemia and hypercalciuria. Although the temporal bone can
conductive hearing loss. SNHL also occurs, but the inner ears be affected in this disorder,106,107 it is very rare in clinical prac-
appear normal. Narrowing of the eustachian tube predisposes tice. The otic capsule is replaced by abnormal bone composed
a patient to serous otitis media.103 Recurrent acute facial nerve of loosely arranged trabeculae of varying sizes and shapes inter-
paralysis that is similar to Bell palsy, involving one or both sides, spersed with marrow spaces that contain fibrous tissue. SNHL
is a frequent manifestation. The tendency is toward progressive has been attributed to osteitis fibrosa that involves the temporal
residual weakness with each episode. Radiographic studies bone.
CC
RWM RWN
FIGURE 149-35. Higher magnification of the temporal bone shown in FIGURE 149-37. Osseous obliteration of the round window niche (RWN)
Figure 149-34. The calcified cartilage (CC) of endochondral bone appears as of the patient shown in Figure 149-36. Fluid is apparent in the niche adjacent
densely staining, round-to-ovoid profiles (×396). to the round window membrane (RWM; ×18).
2316 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
the development of human gene therapy, which is being actively of cases, severe otitis media in 20%, CMV inclusion-bearing
pursued in many centers. cells in the inner and middle ears in 24%, labyrinthine crypto-
coccosis in 8%, and Kaposi sarcoma deposits in the eighth
Otologic Manifestations cranial nerve in 4%. They concluded that the ear is no less
Otologic disease has been described in all four categories of susceptible to AIDS-associated diseases than any other organ
immunodeficiency disorders. Humoral immune defects result and that it is particularly prone to CMV infection. The spec-
in recurrent and persistent acute and serous otitis media. trum of otologic manifestations and their pathophysiologic
Chronic suppurative otitis media and its attendant complica- mechanisms in AIDS is most likely to expand as more clinical
tions may develop and is often refractory to medical and surgi- and histopathologic data accrue.
cal therapy.132,133 A subgroup consists of children with selective
IgG subclass 2 deficiency, who have been shown to be suscep-
tible to recurrent episodes of otitis media.134 DiGeorge syn- GENETICALLY DETERMINED
drome, T-cell deficiency as a result of thymic hypoplasia, can
manifest varying degrees of anomalies of the external, middle,
DEFECTS
and inner ears with conductive, sensorineural, or mixed hearing Numerous syndromic disorders secondary to genetic defects
losses135; a high incidence of Mondini dysplasia is also seen may have otologic manifestations that consist of hearing loss or
in these ears. Recurrent episodes of acute and chronic otitis vestibular dysfunction or both. Examples include syndromes
media have also been described with neutrophil chemotactic caused by mutations in single genes (autosomal or sex linked),
defects,136 microbicidal disorders (chronic granulomatous mutations in mitochondrial genes, or chromosomal abnormali-
disease),137 and complement system defects.138 ties. Such disorders are beyond the scope of this chapter, and
the reader is referred to Chapter 147 or other sources.147
ACQUIRED IMMUNODEFICIENCY
SYNDROME
For a complete list of references, see expertconsult.com.
Acquired immunodeficiency syndrome (AIDS), which was first
recognized in 1981, is caused by HIV, a lymphotropic virus
that primarily attacks T-helper lymphocytes and renders the SUGGESTED READINGS
patient susceptible to numerous opportunistic infections (see Agrup C, Luxon LM: Immune-mediated inner-ear disorders in neuro-
Chapter 11). otology. Curr Opin Neurol 19:26–32, 2006.
Cunningham MJ, Curtin HD, Jaffe R, et al: Otologic manifestations of
Otologic Manifestations Langerhans’ cell histiocytosis. Arch Otolaryngol Head Neck Surg 115:807,
Otologic manifestations are infrequent in patients with AIDS 1989.
except in children, in whom serous otitis media is common.139 Harris JP, South MA: Immunodeficiency diseases: head and neck mani-
When otologic disease does occur, the microbiology is similar festations. Head Neck Surg 5:114, 1982.
to that of non-AIDS patients, with the addition of unusual Khetarpal U, Schuknecht HF: In search of pathologic correlates of
hearing loss and vertigo in Paget’s disease: a clinical and histopatho-
opportunistic organisms, such as protozoa, fungi, viruses, and logic study of 26 temporal bones. Ann Otol Rhinol Laryngol Suppl
mycobacteria. 145:1, 1990.
Middle ear and mastoid manifestations include acute otitis McCabe BF: Autoimmune sensorineural hearing loss. Ann Otol Rhinol
media, acute mastoiditis, serous otitis media, and bullous myr- Laryngol 88:585, 1979.
ingitis; the severity of these infections varies and depends on McCaffrey TV, McDonald TJ, Facer GW, et al: Otologic manifestations
the immune status of the patient. External ear disease such as of Wegener’s granulomatosis. Otolaryngol Head Neck Surg 88:586,
otitis externa and Kaposi sarcoma can also occur. Tissue diag- 1980.
nosis by biopsy or tympanocentesis is indicated to identify the McGill TJI: Mycotic infection of the temporal bone. Arch Otolaryngol
causative agent before initiating therapy. Head Neck Surg 104:140, 1978.
McKenna MJ, Kristiansen AG, Bartley ML, et al: Association of COL1A1
Pneumocystis jiroveci is an unusual opportunistic protozoan and otosclerosis: evidence for a shared genetic etiology with mild
that is a common cause of middle ear and external ear disease osteogenesis imperfecta. Am J Otol 19:604, 1998.
in patients with AIDS. Subcutaneous masses in the external Megerian CA, Sofferman RA, McKenna MJ, et al: Fibrous dysplasia of
canal or aural polyps that arise from the canal, the tympanic the temporal bone: ten new cases demonstrating the spectrum of
membrane, or the mesotympanum can result in conductive otologic sequelae. Am J Otol 16:408, 1995.
hearing loss, otorrhea, or otalgia. Presumed routes of infection Michaels L, Suocek S, Liang J: The ear in the acquired immunodefi-
include hematogenous spread from another source (e.g., pul- ciency syndrome, 1: temporal bone histopathologic study. Am J Otol
monary), ascending infection through the eustachian tube 15:515, 1994.
from pharyngeal colonization, or airborne transmission of Monsell EM: The mechanism of hearing loss in Paget’s disease of bone.
Laryngoscope 114:598–606, 2004.
the aerosolized organism directly to the external canal.140,141 A Nadol JB, Jr: Positive “fistula sign” with an intact tympanic membrane.
biopsy specimen shows the characteristic organism, and the Arch Otolaryngol Head Neck Surg 100:273, 1974.
infection responds to treatment with oral trimethoprim- Rinaldo A, Brandwein MS, Devaney KO, et al: AIDS-related otological
sulfamethoxazole. Otologic disease that results from P. jiroveci lesions. Acta Otolaryngol 123:672–674, 2003.
may be the initial and only presenting symptom of AIDS.140-142 Ryan AF, Harris JP, Keithley EM: Immune-mediated hearing loss: basic
Inner ear symptoms can occur such as vertigo, tinnitus, and mechanisms and options for therapy. Acta Otolaryngol Suppl 548:38–
SNHL that includes fluctuating and sudden hearing loss.143,144 43, 2002.
SNHL is ascribed to various causes that include otosyphilis, Schuknecht HF: Pathology of the ear, ed 2, Philadelphia, 1993, Lea &
cryptococcal meningitis, tuberculous meningitis, CNS toxoplas- Febiger.
Skolnik PR, Nadol JB, Jr, Baker AS: Tuberculosis of the middle ear:
mosis, and ototoxic medication.142,145 HIV is neurotropic and review of the literature with an instructive case report. Rev Infect Dis
may itself be the primary cause of SNHL, and the facial nerve 8:403, 1986.
can be involved by herpes zoster (Ramsay Hunt syndrome). Zoller M, Wilson WR, Nadol JB, Jr: Treatment of syphilitic hearing loss,
In a study of 49 temporal bones from 25 patients with AIDS, combined penicillin and steroid therapy in 29 patients. Ann Otol
Michaels and colleagues146 found low-grade otitis media in 60% Rhinol Laryngol 88:160, 1979.
149 | OTOLOGIC MANIFESTATIONS OF SYSTEMIC DISEASE 2318.e1
Key Points
■ Sensorineural hearing loss (SNHL) is one of the most common clinical disorders and is associated
with a multitude of etiologies.
■ Because of the wide range of genetic, infectious, vascular, neoplastic, traumatic, toxic, iatrogenic,
degenerative, and immunologic and inflammatory pathologies that can affect the cochlea, a
systematic approach to assessment is crucial to identification of the responsible etiology.
Audiologic, serologic, radiologic, and blood chemistry testing can be used strategically for a
cost-effective approach to diagnosis of SNHL.
■ SNHL results from cochlear hair cell or auditory nerve dysfunction. Psychophysical abnormalities
that result from impaired auditory physiology combine to challenge effective listening.
■ Sudden SNHL is a clinical syndrome with a lengthy differential diagnosis. Prompt assessment and
management of sudden SNHL may offer the opportunity to reverse or ameliorate the hearing loss
using more recently developed therapeutic protocols.
2319
2320 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
portion of the physical examination is more likely to reveal emission. If the stimulus consists of continuous pure tones of
positive findings. two separate frequencies (F1 and F2), the resulting emission is a
continuous tone of frequency (2F1-F2) and is termed a
distortion product otoacoustic emission. OAEs are clearly generated
AUDIOMETRIC TESTING within the cochlea and are believed to be a byproduct of the
Only a brief discussion of audiometric testing is presented here. “cochlear amplifier,” which is dependent on outer hair cell func-
Conventional audiometric testing is discussed more thoroughly tion. They depend highly on the physiologic state of the cochlea
in Chapter 133, and electrophysiologic testing is addressed in and, if present, suggest normal cochlear function. OAEs are
Chapter 134. rarely present with hearing thresholds greater than 30 dB
Audiometric testing serves to verify and quantify the degree hearing loss regardless of etiology. In addition, for an OAE to be
of hearing loss. Air conduction, bone conduction, and speech present, the middle ear mechanism must be functioning nor-
audiometry and tympanometry measurements constitute the mally, because the stimulus and the response must traverse the
minimum test battery in patients with suspected SNHL. Bone- middle ear. The presence of an OAE in an ear with SNHL sug-
conduction and air-conduction pure tone audiometry helps to gests a retrocochlear etiology or possible pseudohypacusis.
determine the type of hearing loss: conductive, sensorineural, Because these etiologies are extremely rare in neonates, OAEs
or mixed. Speech audiometry verifies the pure tone audiomet- also have been useful in neonatal hearing screening programs.
ric results. Speech discrimination testing with assessment of
the performance-intensity function helps to define further the
nature of the SNHL (cochlear vs. retrocochlear) and pro-
VESTIBULAR TESTING
vides essential prognostic information regarding the potential Vestibular testing can be a useful adjunct in the evaluation of
benefits of amplification. Tympanometry with acoustic reflex SNHL in selected patients. Evidence of ipsilateral peripheral
testing verifies the conductive or sensorineural nature of the vestibular hypofunction in a patient with unilateral progressive
hearing loss and provides additional clues regarding etiology. SNHL suggests the presence of a retrocochlear lesion.
Tympanometry can be especially helpful in excluding the pos-
sibility of a conductive component in patients with profound
losses or bilateral losses in the presence of a masking dilemma.
LABORATORY TESTING
Basic audiologic tests also can provide essential diagnostic Laboratory testing rarely proves to be helpful in determining
clues as to whether SNHL is cochlear or retrocochlear in the etiology of SNHL. In most patients, either the fluorescent
origin. By retrocochlear, we mean a lesion proximal to the cochlea, treponemal antibody absorption test or the microhemaggluti-
the most common retrocochlear lesion being vestibular nation test for Treponema pallidum should be obtained, because
schwannoma. The examiner should have a high degree of the prevalence of syphilis is relatively high, it is frequently
suspicion for retrocochlear etiologies when loss is asymmet- asymptomatic, it is important to manage, and it is a potentially
ric, speech discrimination is abnormally reduced or asymmet- treatable cause of SNHL. The Venereal Disease Research Labo-
ric, performance-intensity relationships (“rollover”) on speech ratory test is not helpful in this regard, because it frequently
discrimination testing is abnormal, or abnormalities in acous- becomes negative with inadequate management in the latent
tic reflex are apparent. Other cranial nerve findings, asym- phase of the disease or in neurosyphilis. Routinely obtaining
metric tinnitus, or vestibular complaints—even if mild—should hematologic, metabolic, and endocrine studies does not seem
increase the level of suspicion. to be necessary or cost-effective. Similarly, routine screening for
Auditory brainstem response (ABR) testing is useful in eval- autoimmune disorders does not seem warranted. If these dis-
uating the possibility of a retrocochlear etiology and for estab- orders are clinically significant, they typically are apparent from
lishing thresholds in difficult-to-test patients (young children the history and physical examination. In addition, no clear
or malingerers). In the past, the ABR was believed to be a highly relationship is apparent between the results of any of these tests
sensitive test for the presence of a retrocochlear lesion. It is less and the presence of autoimmune hearing loss.
commonly used in this role today because of reduced sensitivity
in patients with small vestibular schwannomas, tumors readily
detected with the high accuracy offered by magnetic resonance
RADIOGRAPHIC TESTING
imaging (MRI).1-3 New modifications of the ABR may increase Radiographic imaging is warranted in selected patients with
this study’s sensitivity.4 SNHL. MRI with gadolinium enhancement is currently the
Electrocochleography differs from ABR testing in that the gold standard in evaluating potential retrocochlear hearing
reference electrode is placed closer to the cochlea (on or close losses. The role of MRI versus ABR testing in this regard is
to the tympanic membrane or promontory). This allows mea- controversial. It is clear, however, that MRI with gadolinium is
surement of the cochlear microphonic potential, the summat- much more sensitive than ABR for diagnosis of small lesions.1-3
ing potential, and the auditory nerve action potential. Wave I Selective T2-weighted fast spin-echo MRI may be almost as
of the ABR corresponds to the action potential of electroco- sensitive as gadolinium-enhanced standard MRI and is less
chleography. Approximately two thirds of patients with classic expensive.7 Computed tomography (CT) is useful in patients
Meniere disease have an elevated summating potential/action with suspected labyrinthine anomalies, such as large vestibular
potential ratio. It is believed that such a finding suggests the aqueduct syndrome or Mondini dysplasia. CT may also be
presence of endolymphatic hydrops.5,6 Electrocochleography useful in patients with suspected labyrinthine fistula or tempo-
can also be useful in patients in whom wave I of the ABR is ral bone fractures. High-resolution CT with reformatted images
weak or not present, because the location of the reference in the plane of, and perpendicular to, the semicircular canals
electrode inherently enlarges wave I. is the study of choice for showing semicircular canal dehiscence
Otoacoustic emissions (OAEs) consist of acoustic energy syndrome.8
generated by the cochlea and recorded with a microphone in
the external auditory canal. These emissions can be spontane- ETIOLOGY OF SENSORINEURAL
ous in onset or, more commonly, they can be evoked by an
acoustic stimulus delivered to the ear canal. If the acoustic
HEARING LOSS
stimulus consists of a transient sound (a click or tone pip), the SNHL is a common clinical disorder associated with a multi-
resulting emission is termed a transient-evoked otoacoustic tude of etiologies. Because of the wide range of genetic,
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2321
healthy children or sudden deafness in adults may be caused
INFECTIOUS DISORDERS by subclinical mumps infection in individuals without previous
Infectious disease is a leading cause of SNHL in children and immunity.
less so in adults. Infectious etiologies that cause SNHL primarily Cytomegalovirus (CMV) infection is thought by some
in adults are discussed. authors to be a common cause of congenital and progressive
hearing loss in children.24-27 It has also been proposed to be a
Labyrinthitis cause of sudden SNHL in adults.28 Hearing loss associated with
An infectious or inflammatory process within the labyrinth can acquired immunodeficiency syndrome (AIDS) may represent
take two forms pathologically: serous, sometimes referred to as reactivation of latent CMV infections.29
toxic, or suppurative. Serous labyrinthitis is defined as an abnor-
mal process within the labyrinth caused by the degradation of Syphilis
the tissue-fluid environment within the inner ear.19 It may be Congenital or acquired syphilis has been well established as a
caused by bacterial toxins or contamination of perilymph with cause of SNHL. Although hearing loss is not associated with
blood, products of tissue injury, or air at surgery. Bacterial primary acquired syphilis, its incidence has been estimated to
toxins may enter the inner ear during the course of acute or be 80% in patients with symptomatic neurosyphilis, 29% in
chronic OM, presumably through either the oval or the round patients with asymptomatic neurosyphilis, 25% in patients with
window membranes. Because both acute and chronic suppura- late latent syphilis, and 17% in patients with congenital syphi-
tive OM are common, and because SNHL associated with either lis.19 The mechanism of hearing loss in syphilis is either a
condition is rare, these membranes seem to provide an excel- meningolabyrinthitis, as seen in neurosyphilis, or an osteitis of
lent barrier to prevent transmission of bacteria or their toxins the temporal bone with secondary involvement of the laby-
to the inner ear. The principal abnormal finding in patients rinth, as seen in late congenital, late latent, or tertiary syphilis.30
with serous labyrinthitis is endolymphatic hydrops, and the Pathologically, a resorptive osteitis is seen in the temporal bone,
hearing loss and vestibular dysfunction associated with this state and progressive endolymphatic hydrops is noted within the
can be permanent or transient. labyrinth. Clinically, the presentation of syphilitic hearing loss
Commonly, a clinical diagnosis of labyrinthitis is made when often is indistinguishable from Meniere disease, with fluctuat-
patients come to medical attention with sudden onset of SNHL ing hearing loss, tinnitus, aural fullness, and episodic vertigo.
and acute vertigo. The exact etiology in cases such as this is Hennebert sign, a positive fistula test without middle ear disease,
uncertain, but it is probably identical or similar to the etiology and the Tullio phenomenon, which is vertigo or nystagmus on
of sudden SNHL. The evidence tends to support the theory exposure to high-intensity sound, have been strongly associated
that this is most commonly caused by a viral labyrinthitis.20,21 with otosyphilis.19 The generally recommended treatment con-
Suppurative labyrinthitis is caused by bacterial invasion of the sists of an antibiotic protocol adequate for neurosyphilis with
inner ear, and it is manifested by profound hearing loss and the addition of systemic corticosteroids.31
acute vertigo. The route of invasion can be otogenic, from
acute or chronic OM, most commonly caused by a fistula Rocky Mountain Spotted Fever
between the middle ear and the labyrinth. Alternatively, the Rocky Mountain spotted fever is a tick-borne infection caused
route of invasion can be meningogenic, through the cochlear by Rickettsia rickettsii. Headache, fever, myalgias, and an expand-
aqueduct or internal auditory canal. This is the most common ing petechial rash follow the tick bite by approximately 1 week.
etiology of deafness associated with meningitis. The disease results in systemic vasculitis that leads to encepha-
litis, nephritis, and hepatitis. Rapidly progressive SNHL has
Otitis Media been associated with Rocky Mountain spotted fever and may be
SNHL is rarely associated with acute OM, and no study has transient.32,33 Vasculitis that involves the auditory system has
shown a relationship between SNHL and frequency of acute been postulated to be the etiology of the hearing loss. Diagnosis
OM.22 Patients with long-standing chronic OM commonly have is made primarily by clinical presentation and is confirmed by
a mixed hearing loss. Whether the sensorineural component serologic titers. Treatment is with broad-spectrum antibiotics.
of this loss is a result of the infectious process itself or a result
of other factors, such as surgery or chronic use of ototoxic Lyme Disease
topical antibiotics, has been a long-standing controversy. When Lyme disease is a tick-borne spirochetal illness caused by Bor-
controlled for sensorineural losses associated with surgery, no relia burgdorferi. Although the most well-known otolaryngologic
increase in SNHL in patients with chronic OM is apparent.23 manifestation of the disease is facial paralysis, some evidence
suggests that the disease can be a cause of SNHL.34-37 Although
Viral Infections its true significance remains unclear, Lyme disease should be
Herpes zoster oticus is a varicella-zoster virus infection most considered a possible etiology of SNHL in endemic areas.
commonly associated with facial paralysis and a herpetic skin
eruption over the auricle or within the external auditory canal.
Although facial paralysis is the most frequent finding, hearing PHARMACOLOGIC TOXICITY
loss and vertigo can occur singly or in combination.
Measles is now rare in the developed world because of wide- Aminoglycosides
spread vaccination. In the past, measles was a common cause At least 96 different pharmacologic agents have potential oto-
of deafness in children. The hearing loss usually is bilateral and toxic side effects.38,39 Among these, aminoglycoside antibiotics
moderate to profound in degree, and vestibular function can are perhaps the most common offending agents. This group of
be similarly affected. antibiotics includes streptomycin, dihydrostreptomycin, kana-
Similar to measles, mumps is uncommon in the developed mycin, neomycin, amikacin, gentamicin, tobramycin, and netilm-
world because of widespread vaccination. Mumps is a para- icin. Ototoxic drugs are often also nephrotoxic and vice versa
myxovirus infection that causes parotitis. Complications of (aminoglycosides, loop diuretics, potassium bromates, and non-
mumps include orchitis, pancreatitis, SNHL, prostatitis, nephri- steroidal antiinflammatory drugs [NSAIDs]). Alport syndrome,
tis, myocarditis, and meningoencephalitis. SNHL resulting described earlier, is a hereditary disorder that affects the
from mumps is unique, because it is almost always unilateral. kidneys and the inner ear, and associated developmental disor-
Bilateral involvement is rare. Unilateral deafness in otherwise ders result in renal and inner ear abnormalities. The strong
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2323
association between pathology of the renal and auditory systems was available.51 Other ingredients of older ototopical prepara-
has not been well explained. tions also have ototoxic potential (e.g., polymyxin B, propylene
Aminoglycosides target the hair cells and enter the hair cell glycol, acetic acid, and antifungal agents).52,53 It seems prudent
in an energy-dependent process. The end result is death of the to use only agents specifically designed and approved for use in
hair cell. The reader is referred to excellent reviews of the the middle ear for treatment of chronic otitis.
current understanding of the mechanism of aminoglycoside
ototoxicity (see Chapter 154).40-42 The final common pathway Loop Diuretics
of hair cell damage consists of the generation of reactive oxygen The loop diuretics ethacrynic acid, bumetanide, and furosemide
species. Different aminoglycosides have affinities for differing exert their diuretic effect by blocking sodium and water reab-
groups of hair cells, which result in different patterns of oto- sorption in the proximal portion of the loop of Henle. The use
toxicity with different aminoglycosides. Kanamycin, tobramy- of these drugs has been associated with a reversible SNHL and
cin, amikacin, neomycin, and dihydrostreptomycin are more with the potentiation of aminoglycoside-induced hearing loss.
cochleotoxic than vestibulotoxic. Others, such as streptomycin The loss typically is bilateral and symmetric and may be sudden
and gentamicin, are more vestibulotoxic than cochleotoxic. in onset.54,55 These drugs seem to alter metabolism in the stria
In addition, the time course of the toxicity can vary.39 Neo- vascularis, which results in alteration of endolymphatic ion con-
mycin toxicity is typically rapid and profound, whereas a centration and endocochlear potential.56 Risk factors for loop
significant delayed effect has been noted for systemically admin- diuretic–induced ototoxicity include renal failure, rapid infu-
istered streptomycin, dihydrostreptomycin, tobramycin, amika- sion, and concomitant aminoglycoside administration.57
cin, and netilmicin and for gentamicin administered through
the middle ear.43 Antimalarials
The hearing loss may be unilateral or asymmetric and can Quinine has long been known to be associated with the devel-
progress during or after cessation of therapy. Some degree of opment of tinnitus, SNHL, and visual disturbances.58 The drug,
reversibility of the hearing loss sometimes is noted weeks to derived from the bark of the cinchona tree, has a colorful
months after treatment.39 Protective agents, including antioxi- history as an antipyretic. It was dispensed by quacks and in
dants, show promise for preventing or reducing aminoglyco- secret remedies in the seventeenth and eighteenth centuries.
side toxicity. More recently, the use of salicylates has been The syndrome of tinnitus, headache, nausea, and disturbed
proposed.44 A placebo-controlled clinical trial in China has vision is termed cinchonism. Larger doses may cause a more
shown a beneficial effect from the use of aspirin during amino- severe form of the syndrome, which also includes gastrointesti-
glycoside administration.45 nal, CNS, cardiovascular, and dermatologic manifestations.
Well-defined risk factors for aminoglycoside-induced ototox- Quinine is used as an adjunct in the treatment of malaria and
icity have been established and include 1) presence of renal nocturnal leg cramps.59 The ototoxic effect of quinine seems to
disease; 2) longer duration of therapy; 3) increased serum be primarily on hearing and usually is transient. Permanent
levels (either peak or trough levels); 4) advanced age; and 5) hearing loss may occur with large doses or in sensitive patients.
concomitant administration of other ototoxic drugs, particu- Chloroquine and hydroxychloroquine are currently used anti-
larly loop diuretics. Peak and trough serum levels should be malarial drugs structurally related to quinine. They have also
routinely monitored when these drugs are being used, and been associated with ototoxicity and retinopathy. Ototoxicity
particular attention should be paid to avoidance of dosing with these drugs seems to be rare and possibly reversible.60,61
intervals that are too short. With the more recent increased use
of prolonged home-based intravenous (IV) antibiotic therapy, Salicylates
an increase in ototoxic complications has been noted, possibly Aspirin and other salicylates are strongly associated with tinni-
a result of reduced attentiveness to serum levels and dosing tus and reversible SNHL. The hearing loss is dose dependent
intervals. and can be in the moderate-to-severe range. On discontinua-
tion of the drug, hearing returns to normal within 72 hours.58
Ototopical Preparations. Topical preparations that contain Tinnitus consistently occurs at a dose of 6 to 8 g/day of aspirin
neomycin, gentamicin, and tobramycin have long been used and at lower doses in some patients.62,63 Caloric responses also
directly in the ear for treatment of otitis externa and chronic can be reduced by salicylates.64 The site of the ototoxic effect
OM. Placement of aminoglycosides within a healthy middle ear seems to be at the level of basic cochlear mechanics, as evi-
space frequently results in cochlear or vestibular ototoxicity as denced by SNHL, loss of OAEs, reduced cochlear action poten-
shown in experimental animals and in patients. This effect is tials, and alteration of the “tips” of auditory nerve fiber tuning
now used to perform a titrated chemical labyrinthectomy for curves.65 These effects may be a result of alteration in turgidity
patients with Meniere disease.46,47 These same drugs have been and motility of outer hair cells.66
used extensively over the years in countless ears with chronic
OM, with little to no apparent clinically significant effect on Nonsteroidal Antiinflammatory Drugs
hearing or vestibular function.48,49 Reduced permeability of the NSAIDs share many of the therapeutic actions and side effects
inflamed round and oval window membranes, dilution of the of salicylates. Although there are isolated reports of hearing
toxic drugs by purulent fluids, and increased absorption into loss caused by naproxen,67 ketorolac,68 and piroxicam,69 ototox-
the vascular system by the hyperemic mucosa probably account icity as a result of NSAID use is generally rare compared with
for this decreased toxicity in the presence of OM. ototoxicity of salicylates.58,70 Similar to salicylates, animal models
It is clear, however, that use of aminoglycosides in the middle of NSAID ototoxicity show only reversible physiologic changes
ear does cause significant and predictable cochlear and vestibu- without major morphologic changes. Two large epidemiologic
lar toxicity in animals.50 Based on this toxicity, and on the now studies have shown an increased risk of hearing loss related to
widespread effective use of aminoglycosides to create a chemical acetaminophen and ibuprofen use in both men and women.71,72
labyrinthectomy, it is now generally regarded as unwise to use The increased risk appears to be both dose and duration
topical aminoglycoside antibiotics for treatment of OM. In 2004, dependent and is greater in younger subjects.
the American Academy of Otolaryngology–Head and Neck
Surgery convened a consensus panel, which after careful review Analgesics and Analgesic/Narcotic Compounds
of the literature recommended against the use of aminoglyco- The first report of SNHL associated with analgesic abuse was
sides in topical form in the middle ear unless no alternative with propoxyphene in 1978.73 Subsequently, acetaminophen/
2324 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
hydrocodone abuse was found to be associated with SNHL in histologic data suggest that the lesion site in erythromycin tox-
2000.74,75 The hearing loss was severe to profound in the vast icity is the stria vascularis.94
majority of patients. All of the patients were using large amounts
of the drug, but the duration and frequency was quite variable. Cisplatin and Carboplatin
In the majority of the reports, the progression of hearing loss Cisplatin (cis-diamminedichloroplatinum) is a cell cycle–
was usually rapid and usually, but not universally, bilateral. nonspecific cancer chemotherapeutic agent that produces
These patients have generally responded very well to cochlear dose-limiting SNHL and peripheral neuropathy and a dose-
implantation, which suggests a cochlear etiology of the hearing related cumulative renal toxicity, hematologic toxicity, and gas-
loss. Since then, a number of reports have been published of trointestinal toxicity.95 The incidence of hearing loss varies in
SNHL associated with various narcotic-analgesic compounds as adults (25% to 86%) and children (84% to 100%), and chil-
well as street narcotics.76-81 The mechanism of SNHL in these dren seem to be significantly more susceptible to ototoxicity.96
patients is unclear, but it is likely multifactorial. One study The hearing loss initially is worse at high frequencies and is
points to acetominophen as the probable ototoxic agent in bilateral and irreversible. It occasionally is accompanied by tin-
acetaminophen/hydrocodone–associated hearing loss.82 nitus or vertigo. The degree of hearing loss is dose related, but
variability is considerable. Occasionally, severe hearing loss can
Phospodiesterase-5 Inhibitors occur after a single dose.97 If ultra–high-frequency hearing is
The phosphodiesterase-5 (PDE-5) inhibitors sildenafil, varde- tested, 100% of patients show a loss. Many factors influence this
nafil, and tadalafil are widely used agents for the treatment of variability, including mode of drug administration, tumor site,
erectile dysfunction. More recently, they have also shown age, renal function, diet, cranial irradiation, interaction with
some utility in the treatment of pulmonary hypertension. These aminoglycosides and loop diuretics, preexisting hearing loss,
drugs block the degradative action of PDE-5 on nitric oxide– cumulative dose, and total dose per treatment.95,98,99
activated cyclic gaunosine monophosphate in the smooth Carboplatin is a cisplatin analogue with a similar spectrum
muscle cells of the vasculature of the corpus cavernosum. Mild of antineoplastic activity. Carboplatin is less nephrotoxic than
side effects are common and include flushing, headache, nasal cisplatin. Myelosuppression is the dose-limiting toxicity with
congestion, and visual disturbances. In 2007, a case of bilateral, carboplatin. It was initially thought that carboplatin was less
sudden, severe SNHL was reported in a man after daily use of ototoxic than cisplatin, although more recent studies have
sildenafil for 15 days.83 Following this report, a U.S. Food and shown higher rates of ototoxicity than initially appreciated. In
Drug Administration investigation discovered 29 reports of a series of children, high-dose carboplatin therapy was associ-
sudden hearing loss in patients taking PDE-5 inhibitors. ated with a very high rate of ototoxicity.100
Maddox and colleagues84 reviewed these cases and added two Cisplatin and carboplatin ototoxicity seems to be due to the
additional cases. The loss was usually unilateral and partial, and creation of reactive oxygen species within the inner ear that
complete recovery was seen in one third of patients. A second target the hair cells. Cisplatin results in greater outer than
study that reviewed worldwide databases came to approximately inner hair cell damage, whereas carboplatin seems to affect the
the same conclusions.85 Although plausible causative mecha- inner hair cells preferentially.101
nisms have been postulated, whether a causative relationship
exists between PDE-5 inhibitor use and sudden SNHL is impos- Nitrogen Mustards
sible to know with certainty, given the presently available data. Nitrogen mustards are antineoplastic agents that include mech-
lorethamine, chlorambucil, cyclophosphamide, melphalan,
Vancomycin and ifosfamide. Of these drugs, only mechlorethamine has oto-
Vancomycin generally is believed to be ototoxic, but the avail- toxicity as a serious adverse effect, and it has limited usefulness
able data are difficult to evaluate.86 In clinical reports of vanco- today because of its severe toxic profile. Animal and human
mycin ototoxicity, patients almost always also received loop studies with mechlorethamine ototoxicity have revealed severe
diuretics or aminoglycosides. Vancomycin has been associated loss of outer hair cells.70 Other studies have shown shrinkage
with ototoxicity when administered intravenously but not when of the organ of Corti and loss of inner and outer hair cells.102
given orally. Auditory impairment has been reported to be
transient or permanent and is extremely unusual if serum con- Vincristine and Vinblastine
centrations are less than 30 mg/mL. In animals, vancomycin The vinca alkaloids vincristine and, to a lesser extent, vinblas-
ototoxicity does not occur, unless toxic levels are adminis- tine are notable for their potent neurotoxicity. Peripheral neu-
tered.87 Vancomycin is nephrotoxic and is excreted by the ropathy is common, and cranial neuropathies, ataxia, and
kidneys; therefore renal failure can prolong vancomycin half- hearing loss have been reported. Vincristine has been shown
life and can increase the likelihood of ototoxicity. Ototoxicity in animals to cause loss of hair cells and primary auditory
and nephrotoxicity are reputed to be less common with newer, neurons, whereas vinblastine has been shown to result in only
more purified formulations of vancomycin.88 When given orally hair cell loss.95
or in appropriate IV doses, vancomycin ototoxicity seems to
be very rare, but vancomycin may potentiate other ototoxic Eflornithine
drugs.89,90 One case of severe, irreversible hearing loss associ- Eflornithine (difluoromethylornithine) is a potent inhibitor of
ated with intrathecal administration of vancomycin has been ornithine decarboxylase and is very effective in the treatment
reported.91 of trypanosomiasis. It also has proven useful in some patients
with Pneumocystis carinii pneumonia, cryptosporidiosis, leish-
Erythromycin maniasis, and malaria. And although it has shown potential as
Numerous case reports document SNHL associated with eryth- an antineoplastic agent, eflornithine has been reported to
romycin administration.92 In almost all reports, the drug was cause major and dose-related SNHL.103
given intravenously rather than orally. The hearing loss seems
to be uncommon, and in most cases the patient recovers within Deferoxamine
1 to 3 weeks after the drug is stopped. The risk of erythromycin Deferoxamine is an iron-chelating agent used in some patients
ototoxicity seems to be greater in patients with renal or hepatic with chronic iron overload or acute, severe iron intoxication.
insufficiency. Ototoxicity associated with the newer macrolide However, auditory and visual neurotoxicity have been reported
antibiotic azithromycin has also been reported.93 Limited with its use, particularly with larger doses in younger patients.
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2325
SNHL is reversible in some patients when the dosage is 10
reduced.104
0
Percentile
Lipid-Lowering Drugs 10 50 {
Although optic and vestibulocochlear nerve degeneration
TRAUMA produces a temporary SNHL that recovers over the next 24 to
48 hours. This reversible loss is termed a temporary threshold shift
Head Injury (TTS). If the noise is of high enough intensity or is repeated
Blows to the head can cause labyrinthine injury and resultant often enough, a permanent loss of hearing results, which is
SNHL, either directly, through fracture of the labyrinth as a referred to as a permanent threshold shift (PTS). Two distinct types
result of temporal bone fracture, or indirectly, through labyrin- of hearing loss are caused by excessive noise exposure: NIHL
thine concussion. The most common type of temporal bone and acoustic trauma. NIHL is caused by repeated exposures to
fracture, longitudinal fracture, uncommonly extends through sound that is too intense or too long in duration. Each expo-
the labyrinth. The hearing loss associated with longitudinal sure is followed by a TTS, which recovers, but eventually a PTS
fractures typically is similar to that of acoustic trauma (i.e., develops. Acoustic trauma consists of a single exposure to a
limited to the high frequencies and worse at 4 kHz). Similarly, hazardous level of noise that results in a PTS without an inter-
blunt head injury alone, without temporal bone fracture, can current TTS.
result in concussive injury of the labyrinth, which can result in NIHL almost always results in a symmetric, bilateral hearing
SNHL. Transverse fractures almost always traverse the labyrinth loss; it almost never results in a profound loss. Early in the
and result in complete loss of auditory and vestibular function. course of NIHL, the loss usually is limited to the 3, 4, and 6 kHz
Penetrating injuries to the inner ear are rare, but they most range. The greatest loss usually occurs at 4 kHz. As the loss
commonly involve subluxation of the stapes into the vestibule progresses, lower frequencies become involved, but the loss at
with resultant profound SNHL. 3 to 6 kHz is always far worse. The loss progresses most rapidly
during the first 10 to 15 years of exposure and thereafter grows
Noise-Induced Hearing Loss and Acoustic Trauma at a much-reduced rate. Figure 150-3 shows the 50% and 95%
The fact that excessive noise exposure could cause hearing loss confidence limits for 20- and 40-year exposures to 90 dBA
was first recognized in the eighteenth century. In the early sound (decibel with “A” weighting that fully weights 700 to
twentieth century, noise-induced hearing loss (NIHL) was 9000 Hz). Figure 150-4 shows an example of the rate of pro-
termed “boilermaker’s deafness.” Careful descriptions of the gression of NIHL over time. The International Organization
hearing loss sustained in industry would await development of for Standardization has established standards for determining
the audiometer and were first published in the 1930s.106 NIHL and quantifying occupational hearing loss and noise-induced
is now recognized as one of the most common occupationally hearing impairment.108
induced disabilities, and noise exposure is now regulated by the Although other patterns also are seen, the hearing loss from
Occupational Health and Safety Administration (OSHA).107 acoustic trauma is similar to that from NIHL (i.e., worse at high
Noise can be defined loosely as “unwanted sound,” and it can frequencies with a 4-kHz “notch”). The other, most common
be subdivided by intensity, time course (continuous, fluctuating, patterns include flat and downsloping losses. Acoustic trauma
intermittent, impact, impulse), and spectral content (pure tone, frequently is unilateral or asymmetric, and considerable vari-
narrow band, broad band). Impact noise is noise caused by col- ability is found in hearing loss among subjects with identical
lision of two objects and is common in industry. Impulse noise is exposure. Age, gender, race, and coexisting vascular disease
that resulting from a sudden release of energy, such as an explo- have been carefully studied, and when adequately controlled
sion or weapon fire. for other factors, they have not been shown to correlate with
Hearing loss caused by noise is sensorineural in nature. susceptibility to NIHL. An attractive theory suggests that
Rarely, extremely intense impulse exposures can result in tym- patients who are more susceptible to TTS would be more sus-
panic membrane perforations, which adds a conductive com- ceptible to PTS and NIHL, although this has not been shown
ponent to the hearing loss. Most hazardous noise exposure to be the case. At present, there is no known way to predict
2326 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
associated with an audible “pop.” Patients may have a positive
Hennebert sign and positional nystagmus when the involved
15
ear is placed in a dependent position.115 Some authors believe
that perilymphatic fistulae develop spontaneously.116,117
Diagnosis is made by middle ear exploration. Visualization
10
95 of fluid in the region of the oval or round windows is not defini-
tive evidence of a fistula, because serous fluid can ooze from
the middle ear mucosa, or lidocaine from the local anesthetic
can collect in the vicinity.118,119 Treatment consists of packing
90 the area in question with tissue. Because of the lack of a defini-
5
tive diagnostic test for the presence of a fistula, and because
85 even surgical exploration does not reliably diagnose or exclude
the possibility of a fistula, considerable controversy exists with
0
0 5 10 15 20 25 30 35 40
regard to the management of this entity.109,116,120,121 In the 1980s
and early 1990s, it was commonly believed that spontaneous
Duration (years)
perilymphatic fistulae were a common cause of otherwise unex-
FIGURE 150-4. Speech frequency average noise-induced permanent plained hearing loss and vertigo. Many surgical fistula repairs
threshold shift (NIPTS) as a function of duration and level of exposure (Lex) were performed as a result of this belief. It has since become
in dBA time-weighted average (TWA). (From Dobie RA. Medical-legal evalua- clear that spontaneous perilymphatic fistula is rare,122 but no
tion of hearing loss. New York: Van Nostrand Reinhold; 1993.) clear consensus exists regarding diagnosis or management.
Finally, labyrinthine fistula can result from erosion by cho-
lesteatoma, or it may develop spontaneously, as in the superior
semicircular canal dehiscence syndrome. The reader is referred
susceptibility to NIHL—with three exceptions: 1) conductive to Chapters 140 and 165 of this book for detailed descriptions
hearing losses are clearly protective for NIHL in the same way of these entities.
that earplugs or earmuffs would be; 2) the lack of an acoustic
reflex has been shown to predispose patients to NIHL (the Irradiation
protective effect of the acoustic reflex is primarily ≤2 kHz)19; The cochlea seems to be resistant to radiation injury at doses
and 3) patients with an unusually large PTS already should be less than 45 Gy. At doses greater than 45 Gy, a clear, dose-
considered to be more susceptible. TTS and PTS are commonly dependent toxicity manifests as hearing loss.123-125 Radiation
accompanied by tinnitus, and tinnitus after a noise exposure also seems to cause a dose-dependent toxicity to the auditory
should be considered a warning sign. nerve and brainstem.126 The latency period from radiation
A clinician can do little in the management of NIHL or exposure to clinical hearing loss can be 12 months or more.
acoustic trauma. The primary role of otolaryngologists and One report notes that early-onset SNHL after radiation can
audiologists is in prevention and early identification; however, recover to some degree.127 Fractionated irradiation has been
many hazardous noise exposures are not occupational in origin, used to a limited extent in the past to treat vestibular
and many employers are either unable or unwilling to provide schwannoma. Whether it has had an effect on hearing in these
hearing conservation programs. patients is difficult to determine because of the limited data
available.128 More recently, the experience with stereotactic irra-
Barotrauma and Perilymphatic Fistula diation (radiosurgery) for vestibular schwannoma has been
Otitic barotrauma consists of traumatic injury of the tympanic much more extensive; this modality seems to be associated with
membrane and middle ear caused by unequalized pressure substantial risk of SNHL, at least as high as that with microsurgi-
differentials between the middle and external ears. The injury cal removal.129
typically occurs during flying or underwater diving and consists A relative paucity of published literature exists to describe
of pain, hyperemia, possible perforation of the tympanic mem- the experimental effects of radiation on hearing. In a histologic
brane, and edema and ecchymosis of the middle ear mucosa. study in chinchillas sacrificed 2 years after fractionated irradia-
These symptoms can be followed by the development of a tion of the cochlea, Bohne and coworkers130 showed a dose-
hemotympanum or a transudative middle ear effusion, and dependent loss of inner and outer hair cells. Early studies of
conductive hearing loss may result. Any abnormality that results the effects of radiotherapy involving the cochlea on human
in compromised eustachian tube function may predispose to hearing were flawed because of a lack of controls, inadequate
barotrauma. follow-up, and the retrospective nature of the studies. However,
A perilymphatic fistula consists of a pathologic communica- many well-designed studies have now shown that radiotherapy
tion between the perilymphatic space of the inner ear and involving the cochlea causes SNHL in 50% of patients.123-125,131-133
middle ear. Perilymphatic fistulae can be congenital or acquired The hearing loss occurs in a dose-dependent fashion, and it
and can occur at either the round or the oval window.109 seems to increase significantly at doses greater than 45 Gy.
Congenital defects can occur in the stapes footplate in Advanced age, preexisting hearing loss, and adjuvant ototoxic
patients with labyrinthine anomalies such as Mondini dyspla- chemotherapeutic agents are likely to amplify the effects of
sia.110,111 These fistulae can communicate with the subarachnoid radiation. The hearing loss occurs with a latency of 0.5 to
space and result in cerebrospinal fluid leak and possible men- 2 years after treatment and is probably progressive.127 This
ingitis. Typically, these ears have a profound hearing loss. This delayed onset, and the fact that many patients do not survive
phenomenon should be a consideration in the differential for sufficient follow-up, results in underestimation of the fre-
diagnosis of patients with recurrent meningitis.112-114 quency and severity of this complication. Dose-dependent
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2327
injury to the auditory nerve and brainstem also occurs; however, understood. The most common presenting symptoms are head-
the frequency in the absence of other neurologic complications ache and visual blurring, although pulsatile tinnitus, SNHL,
is difficult to determine.126 and vertigo also may be present. It is more commonly seen in
An understanding of the effects of radiation on hearing has obese women.154,155
become much more clinically relevant more recently because The pulsatile tinnitus is usually objective and is eliminated
of the popularity of stereotactic radiotherapy for the treatment by jugular venous compression.156,157 SNHL typically is a fluctu-
of vestibular schwannoma and other similar lesions. Although ating, low-frequency loss that is unilateral or bilateral. Vertigo
many reports suggest that the risk of hearing loss is minimal, and aural fullness also may be present. The most serious mani-
these studies are limited by 1) their retrospective design, 2) lack festation of the disorder is progressive visual loss caused by
of long-term follow-up, and 3) incomplete audiologic charac- optic atrophy. The disease is characterized by remissions and
terization. In addition, it is difficult to compare the studies that relapses, and diagnosis is confirmed by documentation of pap-
are available because of differences in tumor size, radiation illedema on funduscopy or cerebrospinal fluid pressure greater
dose, radiation field, technique of radiation administration, than 200 mm H2O. ABR and electrocochleography abnor-
and follow-up. A wealth of literature is available on the treat- malities can also be seen. Management consists of weight loss,
ment outcomes after stereotactic radiotherapy; however, almost acetazolamide, furosemide, and occasionally lumbar-peritoneal
all of them are limited by one of the above-mentioned short- shunting.157,158
comings. More recent studies report hearing-preservation
rates, defined in different ways and with different follow-up
periods, between 36% and 61%.134-137 The time course of VASCULAR AND HEMATOLOGIC
hearing loss after stereotactic radiotherapy is poorly character- DISORDERS
ized; however, available data would suggest that the loss is pro-
gressive and that early results may not predict later outcomes. Migraine
Migraine is a common disorder, usually restricted to headache
NEUROLOGIC DISORDERS and sometimes the neurologic symptoms of an aura. Numerous
subtypes of migraine are associated with different neurologic
Multiple Sclerosis deficits. Basilar migraine has been associated with numerous
Multiple sclerosis (MS) is a chronic disease characterized auditory and vestibular symptoms and signs that include epi-
pathologically by multiple areas of CNS demyelination, inflam- sodic vertigo, SNHL, tinnitus, aural fullness, distortion, and
mation, and glial scarring. The clinical course is variable and recruitment. Fairly complex and specific diagnostic criteria
ranges from a benign, almost symptom-free disease to a rapidly have been established for basilar migraine.159 In a series of
progressive, disabling disorder. Early in the patient’s course, the 50 patients who met the criteria for basilar migraine, 46% had
disease is characterized by remissions and relapses. The disease bilateral low-frequency SNHL, and an additional 34% had uni-
is said to result in dissemination of neurologic lesions in time lateral low-frequency SNHL.160 The hearing loss often fluctu-
(remissions and relapses) and in space (variability of multiple ated and occasionally was severe. Basilar migraine has been
deficits). The age at onset is typically between 20 and 30 years, implicated as an occasional cause of SNHL. Because of their
and it rarely occurs before age 10 years or after age 60 years. similarity, considerable speculation is found in the literature of
MS is more common in women than in men. The racial and an etiologic association between basilar migraine and Meniere
geographic differences in prevalence are striking, with the disease.
disease being most common in whites and in individuals living Migraine headache can be managed with β-blockers, calcium
at higher latitudes. The cause is unknown but seems to be channel blockers, acetazolamide, NSAIDs, and antiserotonin
related to genetic factors, autoimmune mechanisms, and viral agents. No systematic study to evaluate the use of these drugs
infection. in patients with basilar migraine is currently available. The
SNHL develops in 4% to 10% of patients with MS.138,139 The reader is referred to the excellent reviews of Olsson160 and
hearing loss can be progressive or sudden in onset and can be Harker161 and to Chapter 165 for a more detailed discussion.
bilateral, unilateral, symmetric, or asymmetric.140-143 Frequently,
the loss is sudden and unilateral, and it can recover after days Vertebrobasilar Arterial Occlusion
or weeks.57,144 Audiometrically, speech discrimination can be Several eponyms have been applied to brainstem syndromes,
normal or reduced out of proportion to the increase of pure but all apply to neoplasms with the exception of Wallenberg
tone thresholds. Abnormal patterns of acoustic reflexes can be syndrome (lateral medullary syndrome). Classic brainstem
seen in some patients.141,145,146 Abnormalities of the ABR fre- infarction patterns are seen less often than incomplete or
quently are seen and are a diagnostic criterion for MS. Patterns mixed clinical pictures. To result in SNHL, the occlusion gener-
of abnormality vary and include latency prolongation of wave ally has to involve the anterior inferior cerebellar artery (AICA).
I or the later waves, absence or poor morphology of waveforms, Occlusion of the AICA results in ischemic infarction of the
and waveform abnormalities with increased stimulus presenta- regions of the brainstem supplied by this artery. The occlusion
tion rate.140,143,145,147-149 MRI is often abnormal in MS and typi- typically results from thrombosis or embolism and, rarely, from
cally reveals periventricular white matter plaques on T2-weighted other vascular disorders or surgical occlusion of the vessel. The
images. Plaques can be seen in the cochlear nucleus or inferior area infarcted includes the inferior pons, and many of the find-
colliculus in patients with SNHL.150-153 ings are similar to those of Wallenberg syndrome. In addition,
the AICA usually gives rise to the internal auditory artery, which
Benign Intracranial Hypertension is the principal blood supply to the labyrinth. The findings in
Benign intracranial hypertension, also known as pseudotumor cerebri, patients with acute AICA infarction include acute vertigo with
probably is better termed idiopathic intracranial hypertension, associated nausea and vomiting, facial paralysis, SNHL, tinni-
because it is not always benign in nature. The disorder consists tus, ipsilateral gaze paralysis, ipsilateral loss of pain and tem-
of increased intracranial pressure without evidence of mass perature sensation on the face, contralateral partial loss of pain
lesion, obstructive hydrocephalus, intracranial infection, or and temperature sensation on the trunk and extremities, and
hypertensive encephalopathy. It is associated with a long list ipsilateral Horner syndrome. The vertigo and hearing loss are
of medical disorders but frequently manifests as an isolated caused by ischemic injury of the cochlear and vestibular nuclei
phenomenon. The underlying pathophysiology is poorly in the brainstem and in the labyrinth itself.19 Isolated cerebellar
2328 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
infarction can result in hearing loss, vertigo, facial pain or Polyarteritis Nodosa. Polyarteritis nodosa consists of a nec-
numbness, headache, and ataxia.162 rotizing vasculitis of small-sized and medium-sized arteries. It
can manifest with myriad findings that include weight loss,
Rheologic Disorders and Blood Dyscrasias fatigue, fever, anorexia, arthritis, neuropathy, hypertension,
Waldenström macroglobulinemia is a plasma cell disorder in renal failure, abdominal pain, and SNHL. Diagnosis is made by
which abnormally large amounts of immunoglobulin M (IgM) demonstration of the necrotizing vasculitis on a biopsy speci-
are synthesized and circulate in the plasma. The result is men of involved tissue. SNHL may precede the development
increased blood viscosity and subsequent ischemic lesions. Pro- of systemic symptoms, or it may occur late in the disease.
gressive and sudden hearing losses caused by this disorder have Hearing loss may be unilateral or bilateral and is either rapidly
been reported, and some patients with SNHL have responded or slowly progressive. Facial paralysis also may be seen. Aggres-
to treatment with alkylating agents or plasmapheresis.163 sive doses of steroids and immunosuppressive drugs are given
Cryoglobulinemia results from immune-complex disease, in for management.181,182
which the resulting immune complexes are soluble at body
temperature and precipitate at lower temperatures. The depo- Relapsing Polychondritis. Relapsing polychondritis consists of
sition of these complexes results in glomerulonephritis, vascu- an inflammatory reaction in multiple cartilages. The auricles
litis, and arthritis. The disorder can be associated with usually are the first cartilages to be affected, although arthritis
progressive or sudden SNHL.164 and eye findings are commonly present. The disorder fre-
Sickle cell disease is associated with an increased incidence quently is seen in conjunction with other autoimmune diseases.
of SNHL,165-167 and it has been estimated that SNHL is present The associated hearing loss may be conductive, sensorineu-
in 22% of patients with sickle cell disease.168 The hearing loss ral, or mixed. SNHL can be sudden or progressive in onset
may be progressive or sudden and may be associated with sickle and may be associated with vestibular disturbances. Therapy
cell crises.169-171 Leukemias and lymphomas also have been asso- includes steroids, immunosuppressive drugs, or dapsone.183,184
ciated with SNHL caused by either leukemic infiltrates or hem-
orrhage within the inner ear or by vascular occlusion and Wegener Granulomatosis. Wegener granulomatosis is a syn-
resulting labyrinthine ischemia.19 drome of necrotizing granulomatous vasculitis that principally
Cardiopulmonary bypass has been associated with a slightly involves the lungs, airway, and kidneys. Hearing loss usually is
increased risk of SNHL.172-174 The loss most often is sudden, but conductive because of involvement of the eustachian tube or
one study suggests an increased incidence of bilateral high- middle ear. SNHL may be present if the granulomatous disease
frequency loss postoperatively.175 The etiology would seem to or secondary infection extends into the inner ear.185-187
relate to either an embolic phenomenon or to reduced perfu-
sion of the inner ear. Other Autoimmune Disorders. Other systemic autoimmune
Vascular loops within the cerebellopontine angle (CPA) or disorders less commonly associated with SNHL include sclero-
internal auditory canal have been proposed to be a cause derma,188 temporal arteritis,189 systemic lupus erythemato-
not only of SNHL but also of tinnitus, vertigo, and Meniere sus,190,191 sarcoidosis,192,193 and Vogt-Koyanagi-Harada syndrome.
disease.176-179 Although the concept of vascular compression of
cranial nerves causing intermittent neurologic dysfunction has Primary Autoimmune Inner Ear Disease
been reasonably well accepted in trigeminal neuralgia and McCabe194 first described patients with bilateral SNHL who
hemifacial spasm, it has achieved far less support in auditory were responsive to immunosuppressive drugs. The loss can be
and vestibular dysfunction. Vessel loops are found in contact sudden or progressive in onset and usually involves both ears,
with these nerves routinely during CPA surgery for other either simultaneously or alternately. The hearing loss frequently
reasons, and these patients seem to be suffering no ill effects is associated with vestibular symptoms and can strongly mimic
as a result. The entire CNS is subjected to such pulsation con- Meniere disease. Myriad nonspecific tests of humoral autoim-
tinuously. To date, nothing other than anecdotal evidence has munity may be abnormal. The hallmark of the disease is the
been published in support of this theory. responsiveness of the hearing loss to steroids or cytotoxic drugs.
In some patients, a course of drug treatment can produce a
IMMUNE DISORDERS long-lasting improvement in hearing, and in others, the hearing
improvement depends on continued use of the medications.195
Systemic Autoimmune Disorders In these patients, methotrexate is sometimes used to reduce
Various systemic (non–organ-specific) autoimmune disorders the need for continued high-dose steroids and their resultant
have been associated with SNHL. side effects.196 In still others, SNHL progresses despite aggres-
sive treatment.
Cogan Syndrome. Cogan syndrome is perhaps the prototypic Sera of many of these patients have been shown to contain
autoimmune disorder to affect the inner ear. It consists of an antibody to a 68-kD protein of bovine or guinea pig inner
attacks of acute nonsyphilitic interstial keratitis together with ear extracts. The responsiveness of SNHL to steroid treatment
auditory and vestibular dysfunction. The ocular inflammation correlates with the presence of this antibody.197-200 This 68-kD
may be limited to interstial keratitis but may also include scle- protein has been shown to be a member of the heat shock
ritis and/or uveitis. Cogan syndrome can be associated with protein 70 (HSP70) family.201,202 A significant percentage of
systemic vasculitis, especially of the medium and large vessels, patients with Meniere disease show similar reactivity, which
and aortic involvement is seen in 10% of patients. SNHL may suggests that autoimmunity may play a role in at least a subset
be unilateral or bilateral, and it can be associated with severe of patients with Meniere disease.203,204 Chapter 149 contains a
vertigo, nausea, vomiting, and tinnitus. If treated early enough, full discussion of autoimmune inner ear disease.
SNHL is often responsive to aggressive treatment with steroids
and/or other immunosuppressive medications. If untreated, Acquired Immunodeficiency Syndrome
the hearing loss frequently progresses to a profound loss over SNHL is among the numerous neurologic manifestations of
months. The cochlear inflammatory response can lead to oblit- AIDS. The hearing loss may be a result of an infectious com-
eration and ossification of the cochlear lumen; therefore early plication of AIDS, particularly cryptococcal meningitis or syphi-
cochlear implantation should be considered in cases that do lis, or it may be a primary neurologic manifestation of the
not respond to immunosuppressive agents.180 disease. Human immunodeficiency virus (HIV) should be a
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2329
PARANEOPLASTIC SYNDROMES
Neurologic paraneoplastic syndromes consist of neurologic
abnormalities associated with malignant neoplasms not meta-
static to the nervous system. Rarely, the abnormality may involve
the auditory or vestibular system.213
BONE DISORDERS
Otosclerosis
Otosclerosis primarily causes a conductive hearing loss, but it
is commonly associated with a progressive SNHL, especially
later in the course of the disease. The precise mechanism
remains unclear. CT images of the cochlea in these patients
often reveal a radiolucent area immediately surrounding the
cochlea. Histologically, otosclerotic bone frequently involves
the endosteum, but the degree of endosteal involvement does
not clearly correlate with the degree of SNHL.214 It is doubtful
that isolated cochlear otosclerosis without stapedial involve-
ment (and conductive hearing loss) occurs with any clinically
significant frequency.215 Treatment with sodium fluoride has FIGURE 150-5. Gadolinium-enhanced magnetic resonance image shows a
been reported to retard the progression of hearing loss in medium-sized vestibular schwannoma.
these patients,215-218 although the efficacy of this treatment
remains controversial.219-222 Patients with very advanced otoscle-
rosis can have a bilateral profound mixed hearing loss that can reduced out of proportion to the pure tone thresholds. Acoustic
be audiometrically indistinguishable from a profound SNHL. neuroma can manifest as a sudden loss in 10% of patients,
In these patients, stapedectomy can result in useful hearing although most sudden losses are not a result of acoustic
improvement.30,223,224 neuroma.230 Unilateral or asymmetric tinnitus, with or without
hearing loss, also is a common manifestation of acoustic
Paget Disease neuroma. Patients may have mild or severe vestibular symptoms,
Paget disease (osteitis deformans) is a common but poorly or they may have no symptoms. Bilateral acoustic neuromas are
understood disorder of bone. It is most common in older indi- pathognomonic for neurofibromatosis type 2 (Fig. 150-6).
viduals, with an estimated incidence that ranges from 1% in
individuals 40 to 49 years old to 19% in individuals 80 to 89
years old. Approximately 50% of patients with Paget disease
manifest hearing loss. The loss can be conductive, sensorineu-
ral, or mixed. The stapes footplate is rarely fixed, and surgical
ossicular chain reconstruction is rarely beneficial.225 The treat-
ment of Paget disease consists of calcitonin or etidronate diso-
dium. Some evidence indicates that medical treatment may
stabilize or reverse SNHL.226,227
NEOPLASMS
When patients are initially seen with unilateral or asymmetric
SNHL, particularly when the presentation is not typical for
Meniere disease, neoplasm should be a principal diagnostic
consideration. All patients with asymmetric or progressive
SNHL should be evaluated for a neoplastic etiology. Lesions that
result in SNHL usually are located within the internal auditory
canal or CPA, but tumors located anywhere in the skull base or
temporal bone can result in SNHL if the labyrinth is invaded.
Vestibular schwannoma is the most common neoplasm that
results in SNHL (Fig. 150-5). Best known as acoustic neuroma,
vestibular schwannomas originate from the vestibular nerves
within the CPA or the internal auditory canal. Acoustic neuroma
is common and constitutes 6% of all intracranial neoplasms.228
It has been estimated that 2500 new acoustic neuromas are
diagnosed in the United States annually,229 and acoustic neuro-
mas account for approximately 80% of all CPA neoplasms.228
The most common presenting feature of acoustic neuroma is
progressive unilateral SNHL. Any pattern of hearing loss can FIGURE 150-6. Gadolinium-enhanced magnetic resonance image shows
occur, but most frequently the loss initially involves principally bilateral vestibular schwannomas pathognomonic for neurofibromatosis
the high frequencies. Commonly, speech discrimination is type 2.
2330 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
PSEUDOHYPACUSIS
Pseudohypacusis is simply a factitious or exaggerated hearing
loss and is common, especially in situations in which there is
secondary gain to patients. Pseudohypacusis should be consid-
ered whenever the pattern of loss does not fit the clinical
picture. Discordance between the speech reception threshold
and the pure tone average is a strong indicator of a factitious
loss. Other audiometric studies—such as the Stenger test, ABR,
and OAEs—are helpful in clarifying such situations.
0 0
10 10
20 20
Females Males
30 30
Age (years)
Age (years)
30 30
40 40
50 50 FIGURE 150-8. Median audiograms
40 40
60 60 for patients with little to no noise
exposure as a function of gender, fre-
quency, and age. (From Dobie RA.
50 50
0.5 1 2 3 4 6 0.5 1 2 3 4 6 Medical-legal evaluation of hearing loss.
New York: Van Nostrand Reinhold;
Frequency (kHz) Frequency (kHz) 1993.)
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2331
Schuknecht19 has defined four separate types of presbycusis hypertension. This regimen of sodium restriction and diuretics
on the basis of pathologic findings in human temporal bones. was first proposed by Furstenberg and has since gained wide
In sensory presbycusis, hair cells are progressively lost beginning acceptance.245,246 Despite this wide acceptance, few studies have
at the base of the cochlea. Patients with this abnormal pattern been able to show definitively any beneficial therapeutic effect
tend to have steeply sloping high-frequency hearing losses. with regard to improvement or preservation of hearing.247-249
Neural presbycusis implies a loss of auditory nerve fibers; these The creation of an intralabyrinthine shunt (cochleosaccu-
patients tend to have reduced speech discrimination out of lotomy) has been proposed as a mechanism to control the
proportion to their pure tone thresholds. Atrophy of the stria hydrops and prevent the recurrent membrane ruptures thought
vascularis was seen in strial presbycusis, and these patients have to traumatize the organ of Corti. This procedure has been
relatively flat audiograms. Finally, Schuknecht19 described a shown to be beneficial in controlling vertigo but has been
fourth type of presbycusis termed cochlear conductive or mechani- associated with an unacceptably high incidence of severe
cal presbycusis. No light-microscopic abnormalities are seen in hearing loss.250,251 Surgical decompression of the endolym-
these specimens, and Schuknecht theorized that an age-related phatic sac, with or without shunting of the sac into the mastoid
change in the stiffness of the basilar membrane resulted in the or the subarachnoid space, has been proposed as a way to
hearing loss. These patients have gradually descending (approx- correct the presumed defective sac physiology. Although excel-
imately 25 dB/octave) pure tone thresholds.19 These patterns lent results have been reported, double-blind randomized
are not useful clinically because of the variability of audiometric studies to compare endolymphatic sac with mastoid shunt pro-
shape and severity in individuals with age-related hearing loss, cedures with mastoidectomy alone have failed to show any
and clinically the losses do not fall naturally into these significant difference in maintenance of hearing or control of
patterns.240 vertigo.252-256 The unpredictable, fluctuating nature of Meniere
disease, the lack of an objective diagnostic test, and the high
Meniere Disease and Endolymphatic Hydrops incidence of spontaneous resolution of symptoms make it
Meniere disease is a common disorder that consists of fluctuat- extremely difficult to come to valid statistical conclusions with
ing SNHL, tinnitus, episodic vertigo, and aural fullness. The regard to therapeutic efficacy in Meniere disease.
hearing loss typically begins as a fluctuating low-frequency loss Meniere disease may be considered to be idiopathic endo-
that progresses, gradually or quickly, to a permanent loss that lymphatic hydrops. Although it is the most common cause of
can involve any or all frequencies. The tinnitus is most com- endolymphatic hydrops, many other entities result in similar
monly described as “buzzing” or “roaring,” and it can fluctuate clinical presentations and pathologic findings. The syndrome
in loudness and character. The aural fullness perhaps is the of delayed endolymphatic hydrops consists of the initial devel-
most consistent and stable complaint, but it also typically fluctu- opment of a profound SNHL in one ear, followed by develop-
ates. The vertigo of Meniere disease is the most disabling aspect ment of symptoms of endolymphatic hydrops years later in
of the disorder. either the ipsilateral ear (ipsilateral delayed endolymphatic
The typical presentation consists of episodic, spontaneous hydrops) or the contralateral ear (contralateral delayed endo-
attacks of severe, spinning vertigo that lasts for several hours. lymphatic hydrops).257 Other pathologic processes have been
The attacks frequently are associated with nausea, vomiting, associated with development of endolymphatic hydrops, includ-
and diaphoresis. After the attacks, patients usually are fatigued ing syphilis, temporal bone trauma, serous labyrinthitis, stape-
for 24 hours or more. The attacks of vertigo may be associated dectomy, and autoimmune disease.19 The reader is referred to
with a concomitant or preceding change in hearing loss, tin- Chapter 165 for a more extensive discussion of Meniere disease.
nitus, or aural fullness. Many patients also have various combi-
nations of dysequilibrium or motion-provoked vertigo between
the classic attacks. Subtypes of Meniere disease are described SUDDEN SENSORINEURAL
that have only vestibular symptoms (vestibular Meniere disease)
or auditory symptoms (cochlear Meniere disease).
HEARING LOSS
The hallmark of Meniere disease is variability and unpredict- A subset of patients with SNHL develops hearing loss rapidly,
ability, and this holds true for the hearing loss associated with awakening with it in the morning or developing a progressive
the disease. The hearing loss may fluctuate wildly or may be loss over 12 hours or less. This commonly is a frightening expe-
quite stable over many years. It also may manifest as a sudden, rience for the patient, who might assume that this is a life-
permanent loss. Although most patients progress over the threatening disorder or that it will lead to profound bilateral
years to a moderate to severe loss, many do not, and progres- deafness. Neither scenario typically is the case. The etiology and
sion to a profound loss is rare. The low frequencies are more appropriate evaluation and management of this common syn-
commonly involved than are the middle or high frequencies, drome have been the subject of much debate over the years,
especially in the early stages of the disease, but variability illustrated by the fact that more than 100 etiologies have been
is considerable. The disease is bilateral in perhaps 30% of proposed for this disorder.258
cases.241-244 Patients who have bilateral disease usually develop
bilaterality early in the course of the disease. Patients who
maintain hearing loss in only one ear for the first few years PREVALENCE, NATURAL HISTORY,
rarely develop it in the contralateral ear. This finding supports
the idea that the syndrome we have labeled as Meniere disease
AND PROGNOSIS
is actually the manifestation of several different pathologies. Sudden SNHL is a syndrome and not a diagnosis. Most com-
Although vestibular destructive therapy (chemical or surgical monly, the syndrome has been referred to as sudden deafness or
labyrinthectomy, vestibular nerve section) is effective in control- sudden sensorineural hearing loss, and it has many possible etiolo-
ling the episodic vertigo associated with Meniere disease, no gies. In most patients, the cause is idiopathic. Among patients
therapy to date has been proven to be effective in the treatment who reveal no identifiable etiology, several different pathoge-
of the hearing loss. The most widely accepted medical therapy netic mechanisms seem to be responsible. In reviewing the
for Meniere disease, a sodium-restricted diet and diuretic natural history of the disorder, it should be kept in mind that
administration, is based on the hypothesis that the hydropic the described natural history is most likely the sum of the his-
distension of the membranous labyrinth can be reduced by tories of patients with differing abnormalities. Because of this,
altering body water distribution, as in the management of and because of the difficulty of studying such an entity, either
2332 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
in clinical trials or in animal models, much remains to be hearing loss.260,264 With the possible exception of mumps par-
learned about sudden SNHL. otitis and herpes zoster infections, however, the clinical diagno-
A universal definition of sudden SNHL does not exist, and sis of viral infections is unreliable. Azimi and colleagues265
the rate of progression of SNHL can vary from seconds to days. reported that 53% of mumps meningoencephalitis occurs
For purposes of this discussion, sudden SNHL develops over 12 without parotitis. Other evidence for a viral etiology for sudden
hours or less. This rate of progression can sometimes be unclear SNHL includes studies that document increased viral titers in
in the history, because some patients notice their loss only when such patients,264 pathology consistent with viral infection,266-269
using a telephone. In other patients, sensation of fullness or and viral seroconversion studies.270-273 These seroconversion
tinnitus is the patient’s primary initial complaint. The inci- studies showed a mixture of viruses that included herpes
dence of sudden SNHL has been estimated to range from 5 to simplex, herpes zoster, CMV, influenza, parainfluenza, mumps,
20 per 100,000 persons per year.259 In a typical otologic practice, measles, and adenovirus. The studies failed to show a relation-
this may account for 2% to 3% of unselected outpatient visits. ship between titer results and severity of hearing loss or fre-
Any age group can be affected, but the peak incidence seems quency of recovery.
to be in the sixth decade. The male/female distribution is For some viruses, the evidence of a causative relationship is
essentially equal.260 Bilateral involvement is rare, and simultane- more convincing. The mumps virus has been isolated from the
ous bilateral involvement is very rare.261 perilymph of patients with sudden SNHL,274 and experimental
The most common presentation is a patient noticing a uni- mumps labyrinthitis has been reproduced in hamsters by inocu-
lateral hearing loss on awakening. Others notice a sudden, lation of the subarachnoid space with mumps virus.274,275 Lassa
stable hearing loss or a rapidly progressive loss. Occasionally, fever, an arenavirus infection endemic in West Africa, has been
patients note a fluctuating hearing loss, but most patients have shown to be associated with sudden SNHL in approximately
a stable loss. A sensation of aural fullness in the affected ear is two thirds of the patients.276 The time course, the results of
common and frequently is the only complaint. Tinnitus is audiometric testing, and the patterns of recovery in Lassa fever
present in the ear to a variable degree, and the hearing loss are very similar to that in idiopathic sudden SNHL.277 Measles
sometimes is preceded by the onset of tinnitus. Vertigo or dys- and rubella also are well-documented causes of labyrinthitis,268
equilibrium is present to a variable degree in approximately but these cases rarely manifest in a manner typical of sudden
40% of patients.262 SNHL. Herpes zoster oticus also can cause sudden SNHL,
Four variables seem to affect the prognosis of untreated although it is a clinical entity distinct from idiopathic sudden
idiopathic sudden SNHL: 1) severity of loss, 2) audiogram SNHL. The evidence that herpes zoster may be associated with
shape, 3) presence of vertigo, and 4) age. The more severe the idiopathic sudden SNHL is limited to viral seroconversion
loss, the lower the prognosis for recovery, and profound losses studies. Sudden hearing loss associated with infectious mono-
have an exceptionally poor prognosis. Upsloping and midfre- nucleosis is rare but has been reported.278 For a few viruses,
quency losses recover more frequently than downsloping and strong evidence suggests they may be an occasional cause of
flat losses. The presence of vertigo, particularly with a downslop- idiopathic sudden SNHL. For most other viruses, an association
ing loss, is a poor prognostic indicator, although not all studies with idiopathic sudden SNHL is found, although convincing
concur on this point. Reduced speech discrimination carries a evidence of a causal relationship is lacking.
poor prognosis. Finally, most studies show that children and
adults older than 40 years have a poorer prognosis than Meningitis. Meningitis is a well-recognized and common etiol-
others.259,260,262,263 Most recovery occurs within the first 2 weeks ogy of acquired severe to profound SNHL. It is possible that
after onset; as a corollary, the prognosis for recovery decreases rare cases of idiopathic sudden SNHL may be caused by sub-
the longer the loss persists. Without treatment of any kind, a clinical meningoencephalitis.
significant proportion (30% to 65%) of patients experience
complete or partial recovery.262,263 Syphilis. It has been estimated that the incidence of syphilis in
patients with sudden SNHL is 2% or less. Syphilitic hearing loss
may manifest at any stage of the disease, and it may be associ-
ETIOLOGY OF SUDDEN SENSORINEURAL ated with other manifestations of syphilis, with vestibular symp-
toms, or alone. It may also manifest with unilateral or bilateral
HEARING LOSS sudden SNHL. More typical presentations of syphilitic hearing
The management of sudden SNHL should be focused on loss are discussed in other sections of this chapter. It is impor-
excluding known causes of the syndrome, especially conditions tant to consider the possibility of reactivation of syphilis in
that require treatment. Similar to causes of SNHL in general, patients with HIV infection.209,211
these disorders can be conveniently broken down into infec-
tious, neoplastic, traumatic, ototoxic, immunologic, vascular, Lyme Disease. Lyme disease is a well-established etiology of
developmental, psychogenic, and idiopathic etiologies. Despite acute facial paralysis, and it would not be unreasonable to
a thorough search for an etiology, most cases remain idio- assume that it could also cause SNHL. Hearing loss has not
pathic. Considerable debate continues as to the pathogenesis been strongly associated with Lyme disease, however. The litera-
of the disease in these patients. Principal theories include viral ture contains several descriptions of associations between posi-
infection, vascular occlusion, intracochlear membrane breaks, tive Lyme titers and acute or chronic SNHL, but a causal
and autoimmunity. relationship seems doubtful. In one large study, Lyme titers
were found in 21% of patients with sudden SNHL, and despite
Infectious Disorders antibiotic treatment of all seropositive patients, no difference
Viral Infection. Viral neuritis or cochleitis has long been was reported in outcome between patients with positive titers
thought to be the most common cause of sudden SNHL, and those without.279 Reports of hearing loss in patients with
although much of the evidence for this is circumstantial. SNHL Lyme disease with improved hearing after antibiotic treatment
can complicate clinically evident infections with mumps, are limited to a few case reports.35,280
measles, herpes zoster, and infectious mononucleosis and with
congenital rubella and CMV. Of patients who come to medical Acquired Immunodeficiency Syndrome. At autopsy, 88% of
attention with sudden SNHL, 28% report a viral-like upper HIV-positive patients have evidence of CNS involvement,281 and
respiratory infection within 1 month before the onset of their approximately 10% of patients with AIDS come to medical
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2333
attention because of neurologic symptoms.205 It is not surpris- the presence of such a fistula, and even surgical exploration is
ing that sudden SNHL may be associated with HIV infection; subject to error.295 Except in poststapedectomy patients, it is
it is not a common manifestation of AIDS, but it has been well doubtful that perilymph fistula is a significant cause of SNHL.
documented in the literature.206-209,212,282 In the presence of HIV
infection, sudden SNHL may occur with or without the pres- Intracochlear Membrane Breaks. Intracochlear membrane
ence of opportunistic infection, and it may occur without clini- ruptures and fistulae have been documented pathologically in
cal evidence of AIDS. Sudden SNHL caused by reactivation of patients with endolymphatic hydrops.19 Such breaks have been
latent syphilis may complicate any stage of HIV infection.208,209,211 proposed to be an etiology of SNHL.296 Schuknecht and
As previously mentioned, some cases of sudden SNHL associ- Donovan268 found no evidence of such breaks in a series of
ated with AIDS may be the result of reactivation of latent CMV temporal bones from patients with sudden SNHL; however,
infection. Gussen297-299 found evidence to support the membrane break
theory in a few temporal bones.
Neoplasms
Acoustic Neuroma. It is common for sudden SNHL to be the Pharmacologic Toxicity
initial manifestation of a vestibular schwannoma (acoustic Toxicity from any of the drugs discussed in the previous section
neuroma). According to Moffat and associates,230 10.2% of on ototoxic causes of SNHL may result in the relatively sudden
acoustic neuromas initially manifest with sudden SNHL. The onset of hearing loss. In addition to these drugs, others have
prevalence of acoustic neuroma among patients with sudden been associated with sudden SNHL. Interferon has been associ-
SNHL is less clear, although estimates range from 0.8%261 to ated with SNHL that has been reversible in most patients.300,301
3%.283,284 No clear criteria suggest that sudden SNHL may be a The insecticides malathion and methoxychlor have been associ-
result of an acoustic neuroma. The presence of tinnitus in the ated with bilateral SNHL.302
ipsilateral ear before sudden SNHL is suggestive, but it is not
present in most cases.284 In addition, midfrequency and high- Immunologic Disorders
frequency hearing loss are more commonly associated with The finding that many patients with SNHL seem to benefit
acoustic neuroma than are low-frequency losses, and electro- from glucocorticoid therapy and the finding of cross-reacting
nystagmography abnormalities are more common with acoustic circulating antibodies in many patients with sudden and rapidly
neuroma.284 progressive SNHL suggest that at least a subset of SNHL cases
Responsiveness of the hearing loss to treatment with steroids are caused by inner ear autoimmunity.200 In addition, many
is not a reliable indicator that a retrocochlear lesion can be well-known autoimmune diseases have been associated with
ruled out. Many cases have been reported of steroid-responsive SNHL, including Cogan syndrome,303,304 systemic lupus erythe-
SNHL and SNHL with spontaneous recovery that have been matosus,190 temporal arteritis, and polyarteritis nodosa.
found to be caused by acoustic neuroma.284,285 The clinician
should have a high level of suspicion for acoustic neuroma in Vascular Disorders
any patient with SNHL. Most investigators recommend that Sudden hearing loss can occur with occlusion of the cochlear
ABR or gadolinium-enhanced MRI be obtained in patients with blood supply. Because of the abruptness of onset of SNHL and
sudden SNHL,286 although no relationship has been found the fact that the cochlea depends on a single terminal branch
between tumor size and SNHL.284 Because of this and the of the posterior cerebral circulation, vascular occlusion has
numerous more recent reports of false-negative ABR tests in been thought by some authors to be an attractive hypothetic
patients with small acoustic neuromas,3,287-289 it seems warranted etiology for idiopathic sudden hearing losses. Other aspects
to evaluate all patients with sudden SNHL with gadolinium- that argue against a circulatory etiology include high incidence
enhanced MRI. of spontaneous recovery, significant incidence in young
patients, lack of an apparent increased incidence in diabetics,
Other Neoplasms. Neoplasms of the CPA or internal auditory the fact that the loss frequently is limited to just a few frequen-
canal other than acoustic neuromas have also been associated cies, and the fact that most patients do not have vertigo. Similar
with SNHL. These include meningioma,290 cholesteatoma, to viral etiologies, a few cases of SNHL are clearly a result of
hemangioma,291 arachnoid cyst, and metastatic neoplasms. In vascular occlusion, but most cases remain idiopathic. Temporal
addition, skull base neoplasms that erode into the inner ear bone studies have not found evidence of vascular occlusion in
can rarely manifest with SNHL. cases of idiopathic SNHL.19 Series of patients with idiopathic
SNHL have been evaluated for hemostatic abnormalities, but
Trauma and Membrane Ruptures no significant association has been found.305 The role of vascu-
Head Injury. Sensorineural hearing loss of any degree can lar occlusion versus viral infection in these idiopathic cases has
occur after closed or open head injury. The mechanism of been the subject of extensive debate over the years. At this
injury in such patients has been shown pathologically to vary point, it seems doubtful that a significant proportion of cases
from mild loss of outer or inner hair cells or cochlear mem- of idiopathic sudden SNHL have a vascular etiology.
brane breaks to fracture across the labyrinth or intralabyrin- Migraine,160,306,307 hemoglobin sickle cell disease,166,168-171 and
thine hemorrhage.19 Many of these injuries are pathologically macroglobulinemia163,164 have been documented to be associ-
indistinguishable from injuries of acoustic trauma.292 Some ated with sudden SNHL. Rare cases of thromboangiitis obliter-
patients experience a variable degree of recovery from head ans (Buerger disease) have been associated with sudden
injury–induced hearing loss, a process probably equivalent to SNHL.308 Small cerebellar infarctions may mimic labyrinthine
the temporary threshold shift seen with acoustic trauma. lesions, including sudden onset of hearing loss.309 Cardiopul-
monary bypass173,174 and noncardiac surgery172 have been associ-
Perilymphatic Fistula. Round or oval window fistulae can occur ated with an increased risk of sudden SNHL. Sudden SNHL
congenitally, after stapedectomy, or after barotrauma. SNHL is has also been reported after spinal manipulation with probable
well described after events that cause barotrauma.293,294 Some injury to the vertebrobasilar arterial system.310
investigators theorize that these fistulae can occur after heavy It has long been believed that patients with diabetes have a
lifting or straining or even spontaneously. Patients with such higher incidence of idiopathic sudden SNHL than people
fistulae can have sudden or fluctuating SNHL and varying without diabetes. This belief has been based on the higher
degrees of vestibular symptoms. No test is reliable for detecting incidence of other acute cranial neuropathies and on the
2334 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
diffuse vascular abnormalities found in patients with diabetes. at tapering doses. They found that all patients (n = 14) with
Histologic studies of human temporal bones from patients with midfrequency losses (loss at 4 kHz better than 8 kHz) had a
diabetes mellitus have not consistently found any abnormal complete recovery regardless of treatment. They noted that
alterations.19,311 In a careful study of the relationship of idio- among patients with losses greater than 90 dB HL at all fre-
pathic sudden SNHL to diabetes, Wilson28 found that diabetic quencies, no difference in recovery was reported between the
patients with idiopathic sudden SNHL were less likely to recover steroid-treated versus placebo-treated groups. Among the
hearing at higher frequencies. No significant difference was remaining patients (nonprofound losses with hearing at 4 kHz
reported in audiologic patterns between diabetic and nondia- better than at 8 kHz), a significant increase in recovery was
betic patients with idiopathic sudden SNHL. An attempt to reported in the steroid-treated group. Of patients in the steroid-
compare the incidence of diabetes in patients with idiopathic treated group, 78% experienced complete or partial recovery
sudden SNHL with a control population was inconclusive. compared with 38% in the placebo-treated group.
In a similar study, Moskowitz and colleagues315 confirmed a
Developmental Abnormalities significantly improved recovery rate in a steroid-treated group
Large vestibular aqueduct syndrome is associated with SNHL compared with a nontreated control group. Direct steroid treat-
and frequently occurs in a stepwise fashion associated with ment to the inner ear via middle ear instillation or round
minor head trauma. It seems plausible that other, as yet unde- window microcatheter has seen increasing use on an empiric
fined developmental abnormalities may predispose individuals basis. This treatment has the potential advantage of very high
to sudden SNHL, either spontaneously or after minor head steroid concentrations within the inner ear without the associ-
trauma. ated systemic side effects. Anecdotal reports indicate that this
treatment may be more effective than orally administered ste-
Idiopathic Disorders roids and that local complications are infrequent.316 Refer to
Meniere Disease. Some patients seen with typical sudden Chapter 155 for a more complete discussion.
SNHL ultimately may develop a history more suggestive of An alternative management regimen proposed by some
endolymphatic hydrops or even frank Meniere disease; this investigators involves attempts to improve blood flow or oxy-
probably constitutes only 5% of all patients with sudden SNHL. genation to the inner ear. Vasodilators have been used exten-
In a review of 1270 patients with Meniere disease, Hallberg312 sively in the treatment of sudden SNHL. Any proposed
found that only 4.4% had initially been seen with sudden vasodilator would have to cross the blood-brain barrier and
SNHL. A subset of these patients very likely had an autoim- have an effect on intracranial circulation. IV histamine infu-
mune etiology of their hearing loss. sion, oral papaverine, and oral nicotinic acid have been used
most frequently. Fisch and Nagahara others317,318 have shown
Multiple Sclerosis. MS is a demyelinating disorder of the CNS that breathing a gas mixture with increased partial pressures of
manifested by differing neurologic lesions separated by space oxygen and carbon dioxide (carbogen) results in increased
and time. Sudden SNHL is a rare initial manifestation of perilymph oxygen tension in cats and humans.
MS.140,152,313 Among patients with MS, auditory abnormalities Other agents proposed to improve cochlear blood flow
are common.142,145 include low-molecular-weight dextran, mannitol, pentoxifyl-
line, and heparin. In addition, the iodinated radiographic
Sarcoidosis. CNS manifestations are rare (incidence of 1% to contrast agent diatrizoate was serendipitously noted during a
5%), although among patients with neurosarcoidosis, 20% vertebral angiogram to improve hearing in patients with sudden
have eighth cranial nerve findings. This eighth cranial nerve SNHL. Morimitsu and coworkers319 showed in the same report
involvement may manifest as sudden SNHL, although it only that in a small series, 54% of patients treated with diatrizoate
rarely is an isolated finding.193 had a complete recovery compared with 19% of a control
group treated with vasodilators. It was later shown that triiodin-
Psychogenic Disorders ated benzoic acid derivatives such as diatrizoate have a specific
Pseudohypacusis frequently manifests as a sudden loss. In most effect on the stria vascularis, protecting the endocochlear
patients, malingering is readily apparent after initial audiologic potential from furosemide-induced depression.320
studies. Clinical studies that used the previously mentioned agents
have shown poor to mixed results. The varying definition of
recovery used by different authors complicates the interpretation
TREATMENT of these results. No controlled study has shown a beneficial
Treatment of sudden SNHL should be based on its etiology. effect from papaverine, nicotinic acid, or pentoxifylline. Don-
If an etiology is apparent, the appropriate treatment should aldson321 found no improvement with aggressive heparin
follow: antibiotics for infectious causes, withdrawal of the therapy in a series of patients. In a prospective, randomized
offending drug for ototoxicity, and so on. Most cases are idio- double-blind study, Probst and colleagues322 found no differ-
pathic, and treatment decisions should be made on the basis ence in outcome between placebo and treatment with low-
of empiric guidelines. Because of the poor understanding of molecular-weight dextran, pentoxifylline, or both. In a
idiopathic sudden SNHL, controversy surrounds its treatment. randomized prospective trial that compared papaverine/
Because of the dictum, “first, do no harm,” new or unconven- dextran infusion treatment with inhaled carbogen, the average
tional treatment protocols should be well reasoned and care- hearing improvement in the carbogen group was better,
fully applied. It seems prudent to avoid the use of potentially although no significant difference in hearing recovery was seen
harmful treatment protocols outside of controlled clinical between the two groups after 5 days of treatment.317 After 1 year
trials, because serious complications and deaths have been of treatment, a statistically significant improvement in hearing
reported after treatment of idiopathic sudden SNHL.314 was reported in the carbogen-treated group.323
Steroids in moderate doses have become the most widely Redleaf and coworkers324 reviewed their 10-year experience
accepted treatment option for idiopathic sudden SNHL. Wilson with diatrizoate and dextran and noted that 74% of their 36
and associates263 performed a double-blind randomized trial of patients showed audiometric improvement with treatment.
steroids versus placebo for idiopathic sudden SNHL. Their Only 36% improved to 50% of their premorbid hearing levels,
active drug group was treated with either a 10- or 12-day course which was very close to the 32% recovery noted in the placebo
of orally administered dexamethasone or methylprednisolone group of the Wilson study.263 Another, even more empiric
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2335
29. Meynard JL, el Amrani M, Meyohas MC, et al: Two cases of cyto-
REFERENCES megalovirus infection revealed by hearing loss in HIV-infected
1. El-Kashlan HK, Eisenmann D, Kileny PR: Auditory brain stem patients. Biomed Pharmacother 51:461, 1997.
response in small acoustic neuromas. Ear Hear 21:257, 2000. 30. Goodhill V: Syphilis of the ear: a histopathologic study. Ann Otol
2. Schmidt RJ, Sataloff RT, Newman J, et al: The sensitivity of audi- Rhinol Laryngol 48:676, 1939.
tory brainstem response testing for the diagnosis of acoustic neu- 31. Darmstadt GL, Harris JP: Luetic hearing loss: clinical presenta-
romas. Arch Otolaryngol Head Neck Surg 127:19, 2001. tion, diagnosis, and treatment. Am J Otolaryngol 10:410, 1989.
3. Wilson DF, Hodgson RS, Gustafson MF, et al: The sensitivity of 32. Dolan S, Everett ED, Renner L: Hearing loss in Rocky Mountain
auditory brainstem response testing in small acoustic neuromas. spotted fever. Ann Intern Med 104:285, 1986.
Laryngoscope 102:961, 1992. 33. Steinfeld HJ, Silverstein J, Weisburger W, et al: Deafness associated
4. Don M, Kwong B, Tanaka C, et al: The stacked ABR: a sensitive with Rocky Mountain spotted fever. Md Med J 37:287, 1988.
and specific screening tool for detecting small acoustic tumors. 34. Hanner P, Rosenhall U, Edstrom S, et al: Hearing impairment in
Audiol Neurootol 10:274, 2005. patients with antibody production against Borrelia burgdorferi
5. Arts HA, Kileny PR, Telian SA: Diagnostic testing for endolym- antigen. Lancet 1:13, 1989.
phatic hydrops. Otolaryngol Clin North Am 30:987, 1997. 35. Hyden D, Roberg M, Odkvist L: Borreliosis as a cause of sudden
6. Ruth RA, Lambert PR, Ferraro JA: Electrocochleography: methods deafness and vestibular neuritis in Sweden. Acta Otolaryngol Suppl
and clinical applications. Am J Otol 9(Suppl):1, 1988. 520:320, 1995.
7. Shelton C, Harnsberger HR, Allen R, et al: Fast spin echo mag- 36. Lesser TH, Dort JC, Simmen DP: Ear, nose and throat manifesta-
netic resonance imaging: clinical application in screening for tions of Lyme disease. J Laryngol Otol 104:301, 1990.
acoustic neuroma. Otolaryngol Head Neck Surg 114:71, 1996. 37. Moscatello AL, Worden DL, Nadelman RB, et al: Otolaryngologic
8. Belden CJ, Weg N, Minor LB, et al: CT evaluation of bone dehis- aspects of Lyme disease. Laryngoscope 101:592, 1991.
cence of the superior semicircular canal as a cause of sound- and/ 38. Huang MY, Schacht J: Drug-induced ototoxicity: pathogenesis and
or pressure-induced vertigo. Radiology 226:337, 2003. prevention. Med Toxicol Adverse Drug Exp 4:452, 1989.
9. Toriello HV, Reardon W, Gorlin RJ: Hereditary Hearing Loss and Its 39. Matz GJ: Clinical perspectives on ototoxic drugs. Ann Otol Rhinol
Syndromes. ed 2, Oxford, UK, 2004, Oxford University Press. Laryngol Suppl 148:39, 1990.
10. Davis RR, Kozel P, Erway LC: Genetic influences in individual 40. Forge A, Schacht J: Aminoglycoside antibiotics. Audiol Neurootol
susceptibility to noise: a review. Noise Health 5:19, 2003. 5:3, 2000.
11. Davis RR, Newlander JK, Ling X, et al: Genetic basis for suscepti- 41. Rybak LP, Ramkumar V: Ototoxicity. Kidney Int 72:931, 2007.
bility to noise-induced hearing loss in mice. Hear Res 155:82, 2001. 42. Schacht J: Biochemical basis of aminoglycoside ototoxicity. Otolar-
12. Garringer HJ, Pankratz ND, Nichols WC, et al: Hearing impair- yngol Clin North Am 26:845, 1993.
ment susceptibility in elderly men and the DFNA18 locus. Arch 43. Magnusson M, Padoan S: Delayed onset of ototoxic effects of
Otolaryngol Head Neck Surg 132:506, 2006. gentamicin in treatment of Meniere’s disease: rationale for
13. Johnson KR, Zheng QY, Erway LC: A major gene affecting age- extremely low dose therapy. Acta Otolaryngol 111:671, 1991.
related hearing loss is common to at least ten inbred strains of 44. Chen Y, Huang WG, Zha DJ, et al: Aspirin attenuates gentamicin
mice. Genomics 70:171, 2000. ototoxicity: from the laboratory to the clinic. Hear Res 226:178,
14. Kashtan CE: Alport syndrome: an inherited disorder of renal, 2007.
ocular, and cochlear basement membranes. Medicine (Baltimore) 45. Sha SH, Qiu JH, Schacht J: Aspirin to prevent gentamicin-induced
78:338, 1999. hearing loss. N Engl J Med 354:1856, 2006.
15. Jackler RK, Luxford WM, House WF: Congenital malformations 46. Chia SH, Gamst AC, Anderson JP, et al: Intratympanic gentamicin
of the inner ear: a classification based on embryogenesis. Laryn- therapy for Meniere’s disease: a meta-analysis. Otol Neurotol 25:544,
goscope 97:2, 1987. 2004.
16. Griffith AJ, Arts A, Downs C, et al: Familial large vestibular aque- 47. Nedzelski JM, Schessel DA, Bryce GE, et al: Chemical labyrinthec-
duct syndrome. Laryngoscope 106:960, 1996. tomy: local application of gentamicin for the treatment of unilat-
17. Stinckens C, Standaert L, Casselman JW, et al: The presence of eral Meniere’s disease. Am J Otol 13:18, 1992.
a widened vestibular aqueduct and progressive sensorineural 48. Linder TE, Zwicky S, Brandle P: Ototoxicity of ear drops: a clinical
hearing loss in the branchio-oto-renal syndrome: a family study. perspective. Am J Otol 16:653, 1995.
Int J Pediatr Otorhinolaryngol 59:163, 2001. 49. Rohn GN, Meyerhoff WL, Wright CG: Ototoxicity of topical
18. Kitamura K, Takahashi K, Noguchi Y, et al: Mutations of the agents. Otolaryngol Clin North Am 26:747, 1993.
Pendred syndrome gene (PDS) in patients with large vestibular 50. Matz G, Rybak L, Roland PS, et al: Ototoxicity of ototopical anti-
aqueduct. Acta Otolaryngol 120:137, 2000. biotic drops in humans. Otolaryngol Head Neck Surg 130:S79, 2004.
19. Schuknecht HF: Pathology of the Ear, ed 2, Philadelphia, 1993, Lea 51. Roland PS, Stewart MG, Hannley M, et al: Consensus panel on
& Febiger. role of potentially ototoxic antibiotics for topical middle ear use:
20. Stokroos RJ, Albers FW: The etiology of idiopathic sudden senso- introduction, methodology, and recommendations. Otolaryngol
rineural hearing loss: a review of the literature. Acta Otorhinolar- Head Neck Surg 130:S51, 2004.
yngol Belg 50:69, 1996. 52. Marsh RR, Tom LW: Ototoxicity of antimycotics. Otolaryngol Head
21. Stokroos RJ, Albers FW, Schirm J: The etiology of idiopathic Neck Surg 100:134, 1989.
sudden sensorineural hearing loss: experimental herpes simplex 53. Morizono T: Toxicity of ototopical drugs: animal modeling. Ann
virus infection of the inner ear. Am J Otol 19:447, 1998. Otol Rhinol Laryngol Suppl 148:42, 1990.
22. Rahko T, Karma P, Sipila M: Sensorineural hearing loss and acute 54. Koegel L, Jr: Ototoxicity: a contemporary review of aminoglyco-
otitis media in children. Acta Otolaryngol 108:107, 1989. sides, loop diuretics, acetylsalicylic acid, quinine, erythromycin,
23. Noordzij JP, Dodson EE, Ruth RA, et al: Chronic otitis media and and cisplatinum. Am J Otol 6:190, 1985.
sensorineural hearing loss: is there a clinically significant relation? 55. Matz GJ: Aminoglycoside ototoxicity. Am J Otolaryngol 7:117, 1986.
Am J Otol 16:420, 1995. 56. Rybak LP: Pathophysiology of furosemide ototoxicity. J Otolaryngol
24. Barbi M, Binda S, Caroppo S, et al: A wider role for congenital 11:127, 1982.
cytomegalovirus infection in sensorineural hearing loss. Pediatr 57. Franklin DJ, Coker NJ, Jenkins HA: Sudden sensorineural hearing
Infect Dis J 22:39, 2003. loss as a presentation of multiple sclerosis. Arch Otolaryngol Head
25. Fowler KB, Boppana SB: Congenital cytomegalovirus (CMV) Neck Surg 115:41, 1989.
infection and hearing deficit. J Clin Virol 35:226, 2006. 58. Jung TT, Rhee CK, Lee CS, et al: Ototoxicity of salicylate, nonste-
26. Pass RF: Congenital cytomegalovirus infection and hearing loss. roidal antiinflammatory drugs, and quinine. Otolaryngol Clin North
Herpes 12:50, 2005. Am 26:791, 1993.
27. Woolf NK, Koehrn FJ, Harris JP, et al: Congenital cytomegalovirus 59. Goodman LS, Gilman A, Brunton LL, et al: Goodman and Gilman’s
labyrinthitis and sensorineural hearing loss in guinea pigs. J Infect the Pharmacological Basis of Therapeutics, 11th ed, New York, 2006,
Dis 160:929, 1989. McGraw-Hill.
28. Wilson WR: The relationship of the herpesvirus family to sudden 60. Hadi U, Nuwayhid N, Hasbini AS: Chloroquine ototoxicity: an
hearing loss: a prospective clinical study and literature review. idiosyncratic phenomenon. Otolaryngol Head Neck Surg 114:491,
Laryngoscope 96:870, 1986. 1996.
2335.e2 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
122. Friedland DR, Wackym PA: A critical appraisal of spontaneous 149. Verma NP, Lynn GE: Auditory evoked responses in multiple scle-
perilymphatic fistulas of the inner ear. Am J Otol 20:261, 1999. rosis: wave I abnormality. Arch Otolaryngol 111:22, 1985.
123. Grau C, Møller K, Overgaard M, et al: Sensori-neural hearing loss 150. Armington WG, Harnsberger HR, Smoker WR, et al: Normal and
in patients treated with irradiation for nasopharyngeal carcinoma. diseased acoustic pathway: evaluation with MR imaging. Radiology
Int J Radiat Oncol Biol Phys 21:723, 1991. 167:509, 1988.
124. Honoré HB, Bentzen SM, Møller K, et al: Sensori-neural hearing 151. Cure JK, Cromwell LD, Case JL, et al: Auditory dysfunction caused
loss after radiotherapy for nasopharyngeal carcinoma: individual- by multiple sclerosis: detection with MR imaging. AJNR Am J Neu-
ized risk estimation. Radiother Oncol 65:9, 2002. roradiol 11:817, 1990.
125. Pan CC, Eisbruch A, Lee JS, et al: Prospective study of inner ear 152. Drulovic B, Ribaric-Jankes K, Kostic VS, et al: Sudden hearing loss
radiation dose and hearing loss in head-and-neck cancer patients. as the initial monosymptom of multiple sclerosis. Neurology 43:
Int J Radiat Oncol Biol Phys 61:1393, 2005. 2703, 1993.
126. Grau C, Møller K, Overgaard M, et al: Auditory brain stem 153. Gebarski SS, Gabrielsen TO, Gilman S, et al: The initial diagnosis
responses in patients after radiation therapy for nasopharyngeal of multiple sclerosis: clinical impact of magnetic resonance
carcinoma. Cancer 70:2396, 1992. imaging. Ann Neurol 17:469, 1985.
127. Ho WK, Wei WI, Kwong DL, et al: Long-term sensorineural 154. Sismanis A: Otologic manifestations of benign intracranial hyper-
hearing deficit following radiotherapy in patients suffering from tension syndrome: diagnosis and management. Laryngoscope 97:1,
nasopharyngeal carcinoma: a prospective study. Head Neck 21:547, 1987.
1999. 155. Sismanis A, Hughes GB, Abedi E, et al: Otologic symptoms and
128. Maire JJP, Floquet AA, Darrouzet VV, et al: Fractionated radiation findings of the pseudotumor cerebri syndrome: a preliminary
therapy in the treatment of stage III and IV cerebello-pontine report. Otolaryngol Head Neck Surg 93:398, 1985.
angle neurinomas: preliminary results in 20 cases. Int J Radiat 156. Sismanis A, Butts FM, Hughes GB: Objective tinnitus in benign
Oncol Biol Phys 23:147, 1992. intracranial hypertension: an update. Laryngoscope 100:33, 1990.
129. Arts HA, Telian SA, El-Kashlan H, et al: Hearing preservation and 157. Sismanis A, Smoker WR: Pulsatile tinnitus: recent advances in
facial nerve outcomes in vestibular schwannoma surgery: results diagnosis. Laryngoscope 104:681, 1994.
using the middle cranial fossa approach. Otol Neurotol 27:234, 158. Sugerman HJ, Felton WL, 3rd, Salvant JB, Jr, et al: Effects of surgi-
2006. cally induced weight loss on idiopathic intracranial hypertension
130. Bohne BBA, Marks JJE, Glasgow GGP: Delayed effects of ionizing in morbid obesity. Neurology 45:1655, 1995.
radiation on the ear. Laryngoscope 95:818, 1985. 159. Classification and diagnostic criteria for headache disorders,
131. Grau C, Overgaard J: Postirradiation sensorineural hearing loss: cranial neuralgias and facial pain. Headache Classification Com-
a common but ignored late radiation complication. Int J Radiat mittee of the International Headache Society. Cephalalgia 8
Oncol Biol Phys 36:515, 1996. (Suppl 7):1, 1988.
132. Herrmann FF, Dörr WW, Müller RR, et al: A prospective study on 160. Olsson JE: Neurotologic findings in basilar migraine. Laryngoscope
radiation-induced changes in hearing function. Int J Radiat Oncol 101:1, 1991.
Biol Phys 65:1338, 2006. 161. Harker LA: Migraine. In Jackler RK, Brackmann DE, editors:
133. Kwong DL, Wei WI, Sham JS, et al: Sensorineural hearing loss in Neurotology, Philadelphia, 2003, Elsevier.
patients treated for nasopharyngeal carcinoma: a prospective 162. Rubenstein RL, Norman DM, Schindler RA, et al: Cerebellar
study of the effect of radiation and cisplatin treatment. Int J Radiat infarction—a presentation of vertigo. Laryngoscope 90:505,
Oncol Biol Phys 36:281, 1996. 1980.
134. Iwai Y, Yamanaka K, Shiotani M, et al: Radiosurgery for acoustic 163. Wells M, Michaels L, Wells DG: Otolaryngological disturbances in
neuromas: results of low-dose treatment. Neurosurgery 53:282, Waldenstrom’s macroglobulinaemia. Clin Otolaryngol Allied Sci
2003. 2:327, 1977.
135. Massager N, Nissim O, Delbrouck C, et al: Irradiation of cochlear 164. Nomura Y, Tsuchida M, Mori S, et al: Deafness in cryoglobuline-
structures during vestibular schwannoma radiosurgery and associ- mia. Ann Otol Rhinol Laryngol 91:250, 1982.
ated hearing outcome. J Neurosurg 107:733, 2007. 165. Ajulo SO, Osiname AI, Myatt HM: Sensorineural hearing loss in
136. Paek SH, Chung H-T, Jeong SS, et al: Hearing preservation after sickle cell anaemia—a United Kingdom study. J Laryngol Otol 107:
gamma knife stereotactic radiosurgery of vestibular schwannoma. 790, 1993.
Cancer 104:580, 2005. 166. Friedman EM, Herer GR, Luban NL, et al: Sickle cell anemia and
137. Prasad DD, Steiner MM, Steiner LL: Gamma surgery for vestibular hearing. Ann Otol Rhinol Laryngol 89:342, 1980.
schwannoma. J Neurosurg 92:745, 2000. 167. Odetoyinbo O, Adekile A: Sensorineural hearing loss in children
138. Grenman R: Involvement of the audiovestibular system in multi- with sickle cell anemia. Ann Otol Rhinol Laryngol 96:258, 1987.
ple sclerosis: an otoneurologic and audiologic study. Acta Otolar- 168. Elwany S, Kamel T: Sensorineural hearing loss in sickle cell crisis.
yngol Suppl 420:1, 1985. Laryngoscope 98:386, 1988.
139. Noffsinger D, Olsen WO, Carhart R, et al: Auditory and vestibular 169. Hotaling AJ, Hillstrom RP, Bazell C: Sickle cell crisis and sensori-
aberrations in multiple sclerosis. Acta Otolaryngol Suppl 303:1, 1972. neural hearing loss: case report and discussion. Int J Pediatr Oto-
140. Daugherty WT, Lederman RJ, Nodar RH, et al: Hearing loss in rhinolaryngol 17:207, 1989.
multiple sclerosis. Arch Neurol 40:33, 1983. 170. O’Keeffe LJ, Maw AR: Sudden total deafness in sickle cell disease.
141. Jerger J, Oliver TA, Rivera V, et al: Abnormalities of the acoustic J Laryngol Otol 105:653, 1991.
reflex in multiple sclerosis. Am J Otolaryngol 7:163, 1986. 171. Tavin ME, Rubin JS, Camacho FJ: Sudden sensorineural hearing
142. Jerger JF, Oliver TA, Chmiel RA, et al: Patterns of auditory abnor- loss in haemoglobin SC disease. J Laryngol Otol 107:831, 1993.
mality in multiple sclerosis. Audiology 25:193, 1986. 172. Millen SJ, Toohill RJ, Lehman RH: Sudden sensorineural hearing
143. Schweitzer VG, Shepard N: Sudden hearing loss: an uncommon loss: operative complication in non-otologic surgery. Laryngoscope
manifestation of multiple sclerosis. Otolaryngol Head Neck Surg 92:613, 1982.
100:327, 1989. 173. Plasse HM, Mittleman M, Frost JO: Unilateral sudden hearing loss
144. Drulovic B, Ribaric-Jankes K, Kostic V, et al: Multiple sclerosis as after open heart surgery: a detailed study of seven cases. Laryngo-
the cause of sudden “pontine” deafness. Audiology 33:195, 1994. scope 91:101, 1981.
145. Keith RW, Garza-Holquin Y, Smolak L, et al: Acoustic reflex 174. Plasse HM, Spencer FC, Mittleman M, et al: Unilateral sudden loss
dynamics and auditory brain stem responses in multiple sclerosis. of hearing: an unusual complication of cardiac operation. J Thorac
Am J Otol 8:406, 1987. Cardiovasc Surg 79:822, 1980.
146. Wiegand DA, Poch NE: The acoustic reflex in patients with asymp- 175. Shapiro MJ, Purn JM, Raskin C: A study of the effects of cardio-
tomatic multiple sclerosis. Am J Otolaryngol 9:210, 1988. pulmonary bypass surgery on auditory function. Laryngoscope
147. Hopf HC, Maurer K: Wave I of early auditory evoked potentials 91:2046, 1981.
in multiple sclerosis. Electroencephalogr Clin Neurophysiol 56:31, 176. Benecke JE, Jr, Hitselberger WE: Vertigo caused by basilar artery
1983. compression of the eighth nerve. Laryngoscope 98:807, 1988.
148. Musiek FE, Gollegly KM, Kibbe KS, et al: Electrophysiologic and 177. Jannetta PJ: Neurovascular cross-compression in patients with
behavioral auditory findings in multiple sclerosis. Am J Otol 10:343, hyperactive dysfunction symptoms of the eighth cranial nerve.
1989. Surg Forum 26:467, 1975.
2335.e4 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
178. Jannetta PJ: Neurovascular compression in cranial nerve and sys- 207. Hart CW, Cokely CG, Schupbach J, et al: Neurotologic findings
temic disease. Ann Surg 192:518, 1980. of a patient with acquired immune deficiency syndrome. Ear Hear
179. McCabe BF, Harker LA: Vascular loop as a cause of vertigo. Ann 10:68, 1989.
Otol Rhinol Laryngol 92:542, 1983. 208. Kwartler JA, Linthicum FH, Jahn AF, et al: Sudden hearing loss
180. McDonald TJ, Vollertsen RS, Younge BR: Cogan’s syndrome: due to AIDS-related cryptococcal meningitis—a temporal bone
audiovestibular involvement and prognosis in 18 patients. Laryn- study. Otolaryngol Head Neck Surg 104:265, 1991.
goscope 95:650, 1985. 209. Little JP, Gardner G, Acker JD, et al: Otosyphilis in a patient with
181. Vathenen AS, Skinner DW, Shale DJ: Treatment response with human immunodeficiency virus: internal auditory canal gumma.
bilateral mixed deafness and facial palsy in polyarteritis nodosa. Otolaryngol Head Neck Surg 112:488, 1995.
Am J Med 84:1081, 1988. 210. Madriz JJ, Herrera G: Human immunodeficiency virus and
182. Wolf M, Kronenberg J, Engelberg S, et al: Rapidly progressive acquired immune deficiency syndrome AIDS-related hearing dis-
hearing loss as a symptom of polyarteritis nodosa. Am J Otolaryngol orders. J Am Acad Audiol 6:358, 1995.
8:105, 1987. 211. Smith ME, Canalis RF: Otologic manifestations of AIDS: the oto-
183. Cody DT, Sones DA: Relapsing polychondritis: audiovestibular syphilis connection. Laryngoscope 99:365, 1989.
manifestations. Laryngoscope 81:1208, 1971. 212. Timon CI, Walsh MA: Sudden sensorineural hearing loss as a
184. Damiani JM, Levine HL: Relapsing polychondritis—report of ten presentation of HIV infection. J Laryngol Otol 103:1071, 1989.
cases. Laryngoscope 89:929, 1979. 213. Gulya AJ: Neurologic paraneoplastic syndromes with neurotologic
185. Bakthavachalam S, Driver MS, Cox C, et al: Hearing loss in Wegen- manifestations. Laryngoscope 103:754, 1993.
er’s granulomatosis. Otol Neurotol 25:833, 2004. 214. Schuknecht HF, Barber W: Histologic variants in otosclerosis.
186. Fenton JE, O’Sullivan TJ: The otological manifestations of Wegen- Laryngoscope 95:1307, 1985.
er’s granulomatosis. J Laryngol Otol 108:144, 1994. 215. Bretlau P, Salomon G, Johnsen NJ: Otospongiosis and sodium
187. Takagi D, Nakamaru Y, Maguchi S, et al: Otologic manifestations fluoride: a clinical double-blind, placebo-controlled study on
of Wegener’s granulomatosis. Laryngoscope 112:1684, 2002. sodium fluoride treatment in otospongiosis. Am J Otol 10:20, 1989.
188. Abou-Taleb A, Linthicum FH, Jr: Scleroderma and hearing loss 216. Bretlau P, Causse J, Causse JB, et al: Otospongiosis and sodium
(histopathology of a case). J Laryngol Otol 101:656, 1987. fluoride: a blind experimental and clinical evaluation of the effect
189. Kramer MR, Nesher G, Sonnenblick M: Steroid-responsive of sodium fluoride treatment in patients with otospongiosis. Ann
hearing loss in temporal arteritis. J Laryngol Otol 102:524, 1988. Otol Rhinol Laryngol 94:103, 1985.
190. Bowman CA, Linthicum FH, Jr, Nelson RA, et al: Sensorineural 217. Causse JR, Causse JB: Early detection of otosclerosis by impedance-
hearing loss associated with systemic lupus erythematosus. Otolar- audiometry screening. Scand Audiol Suppl 17:47, 1983.
yngol Head Neck Surg 94:197, 1986. 218. Causse JR, Causse JB: Clinical studies on fluoride in otospongiosis.
191. MacFadyen DJ, Schneider RJ, Chisholm IA: A syndrome of brain, Am J Otol 6:51, 1985.
inner ear and retinal microangiopathy. Can J Neurol Sci 14:315, 219. Forquer BD, Linthicum FH, Bennett C: Sodium fluoride: effec-
1987. tiveness of treatment for cochlear otosclerosis. Am J Otol 7:121,
192. Jahrsdoerfer RA, Thompson EG, Johns MM, et al: Sarcoidosis 1986.
and fluctuating hearing loss. Ann Otol Rhinol Laryngol 90:161, 220. Kornblut AD: Sodium fluoride therapy for otosclerosis. Arch Oto-
1981. laryngol Head Neck Surg 117:450, 1991.
193. Souliere CR, Jr, Kava CR, Barrs DM, et al: Sudden hearing loss as 221. Shambaugh GE, Jr: Fluoride therapy for otosclerosis. Arch Otolar-
the sole manifestation of neurosarcoidosis. Otolaryngol Head Neck yngol Head Neck Surg 116:1217, 1990.
Surg 105:376, 1991. 222. Snow JB, Jr: Current status of fluoride therapy for otosclerosis. Am
194. McCabe BF: Autoimmune sensorineural hearing loss. Ann Otol J Otol 6:56, 1985.
Rhinol Laryngol 88:585, 1979. 223. Lippy WH, Battista RA, Schuring AG, et al: Far-advanced otoscle-
195. Hughes GB, Kinney SE, Barna BP, et al: Practical versus theoretical rosis. Am J Otol 15:225, 1994.
management of autoimmune inner ear disease. Laryngoscope 94: 224. Wiet RJ, Morgenstein SA, Zwolan TA, et al: Far-advanced otoscle-
758, 1984. rosis: cochlear implantation vs stapedectomy. Arch Otolaryngol
196. Sismanis A, Thompson T, Willis HE: Methotrexate therapy for Head Neck Surg 113:299, 1987.
autoimmune hearing loss: a preliminary report. Laryngoscope 104: 225. Harner SG, Rose DE, Facer GW: Paget’s disease and hearing loss.
932, 1994. Otolaryngology 86, 1978.
197. Harris JP: Autoimmunity of the inner ear. Am J Otol 10:193, 1989. 226. el Sammaa M, Linthicum FH, House HP, et al: Calcitonin as treat-
198. Harris JP, Sharp PA: Inner ear autoantibodies in patients with ment for hearing loss in Paget’s disease. Am J Otol 7:241, 1986.
rapidly progressive sensorineural hearing loss. Laryngoscope 100: 227. Lando M, Hoover LA, Finerman G: Stabilization of hearing loss
516, 1990. in Paget’s disease with calcitonin and etidronate. Arch Otolaryngol
199. Moscicki RA: Immune-mediated inner ear disorders. Baillieres Clin Head Neck Surg 114:891, 1988.
Neurol 3:547, 1994. 228. Zulch K: Epidemiology of brain tumors: general statistical and
200. Moscicki RA, San Martin JE, Quintero CH, et al: Serum antibody biological data. In Brain Tumors: Their Biology and Pathology, ed 3,
to inner ear proteins in patients with progressive hearing loss: Berlin, 1986, Springer-Verlag.
correlation with disease activity and response to corticosteroid 229. Jackler RK: Acoustic neuroma. In Jackler RK, Brackmann D,
treatment. JAMA 272:611, 1994. editors: Neurotology, ed 2, Philadelphia, 2005, Elsevier-Mosby.
201. Billings PB, Keithley EM, Harris JP: Evidence linking the 68 kilo- 230. Moffat DA, Baguley DM, von Blumenthal H, et al: Sudden deaf-
dalton antigen identified in progressive sensorineural hearing loss ness in vestibular schwannoma. J Laryngol Otol 108:116, 1994.
patient sera with heat shock protein 70. Ann Otol Rhinol Laryngol 231. Harner SG: Hearing in adult-onset diabetes mellitus. Otolaryngol
104:181, 1995. Head Neck Surg 89:322, 1981.
202. Bloch DB, San Martin JE, Rauch SD, et al: Serum antibodies to 232. Anand VT, Mann SB, Dash RJ, et al: Auditory investigations in
heat shock protein 70 in sensorineural hearing loss. Arch Otolar- hypothyroidism. Acta Otolaryngol 108:83, 1989.
yngol Head Neck Surg 121:1167, 1995. 233. Debruyne F, Vanderschueren-Lodeweyckx M, Bastijns P: Hearing
203. Gottschlich S, Billings PB, Keithley EM, et al: Assessment of in congenital hypothyroidism. Audiology 22:404, 1983.
serum antibodies in patients with rapidly progressive sensorineu- 234. O’Malley BW, Jr, Li D, Turner DS: Hearing loss and cochlear
ral hearing loss and Meniere’s disease. Laryngoscope 105:1347, abnormalities in the congenital hypothyroid (hyt/hyt) mouse.
1995. Hear Res 88:181, 1995.
204. Rauch SD, San Martin JE, Moscicki RA, et al: Serum antibodies 235. Vanderschueren-Lodeweyckx M, Debruyne F, Dooms L, et al: Sen-
against heat shock protein 70 in Meniere’s disease. Am J Otol 16: sorineural hearing loss in sporadic congenital hypothyroidism.
648, 1995. Arch Dis Child 58:419, 1983.
205. Carne CA: ABC of AIDS. Neurological manifestations. BMJ (Clin 236. Garty BZ, Daliot D, Kauli R, et al: Hearing impairment in idio-
Res Ed) 294:1399, 1987. pathic hypoparathyroidism and pseudohypoparathyroidism. Isr J
206. Grimaldi LM, Luzi L, Martino GV, et al: Bilateral eighth cranial Med Sci 30:587, 1994.
nerve neuropathy in human immunodeficiency virus infection. 237. Ikeda K, Kobayashi T, Kusakari J, et al: Sensorineural hearing loss
J Neurol 240:363, 1993. associated with hypoparathyroidism. Laryngoscope 97:1075, 1987.
150 | SENSORINEURAL HEARING LOSS IN ADULTS 2335.e5
298. Gussen R: Sudden hearing loss associated with cochlear mem- 314. Zaytoun GM, Schuknecht HF, Farmer HS: Fatality following the
brane rupture: two human temporal bone reports. Arch Otolaryn- use of low molecular weight dextran in the treatment of sudden
gol 107:598, 1981. deafness. Adv Otorhinolaryngol 31:240, 1983.
299. Gussen R: Sudden deafness associated with bilateral Reissner’s 315. Moskowitz D, Lee KJ, Smith HW: Steroid use in idiopathic sudden
membrane ruptures. Am J Otolaryngol 4:27, 1983. sensorineural hearing loss. Laryngoscope 94:664, 1984.
300. Kanda Y, Shigeno K, Kinoshita N, et al: Sudden hearing loss associ- 316. Lefebvre PP, Staecker H: Steroid perfusion of the inner ear for
ated with interferon. Lancet 343:1134, 1994. sudden sensorineural hearing loss after failure of conventional
301. Kanda Y, Shigeno K, Matsuo H, et al: Interferon-induced sudden therapy: a pilot study. Acta Otolaryngol 122:698, 2002.
hearing loss. Audiology 34:98, 1995. 317. Fisch U: Management of sudden deafness. Otolaryngol Head Neck
302. Harell M, Shea JJ, Emmett JR: Bilateral sudden deafness following Surg 91:3, 1983.
combined insecticide poisoning. Laryngoscope 88:1348, 1978. 318. Nagahara K, Fisch U, Yagi N: Perilymph oxygenation in sudden
303. Cote DN, Molony TB, Waxman J, et al: Cogan’s syndrome mani- and progressive sensorineural hearing loss. Acta Otolaryngol 96:57,
festing as sudden bilateral deafness: diagnosis and management. 1983.
South Med J 86:1056, 1993. 319. Morimitsu T, Nakashima T, Matsumoto I, et al: Dysfunction of stria
304. Haynes BF, Pikus A, Kaiser-Kupfer M, et al: Successful treatment vascularis as a new theory of sudden deafness. Adv Otorhinolaryngol
of sudden hearing loss in Cogan’s syndrome with corticosteroids. 22:57, 1977.
Arthritis Rheum 24:501, 1981. 320. Hirashima N: Blocking effect of radio-contrast media on cochlear
305. Einer H, Tengborn L, Axelsson A, et al: Sudden sensorineural depression. Ann Otol Rhinol Laryngol 87:32, 1978.
hearing loss and hemostatic mechanisms. Arch Otolaryngol Head 321. Donaldson JA: Heparin therapy for sudden sensorineural hearing
Neck Surg 120:536, 1994. loss. Arch Otolaryngol 105:351, 1979.
306. Lipkin AF, Jenkins HA, Coker NJ: Migraine and sudden sensori- 322. Probst R, Tschopp K, Ludin E, et al: A randomized, double-blind,
neural hearing loss. Arch Otolaryngol Head Neck Surg 113:325, 1987. placebo-controlled study of dextran/pentoxifylline medication in
307. Viirre ES, Baloh RW: Migraine as a cause of sudden hearing loss. acute acoustic trauma and sudden hearing loss. Acta Otolaryngol
Headache 36:24, 1996. 112:435, 1992.
308. Kirikae I, Nomura Y, Shitara T, et al: Sudden deafness due to 323. Fisch U, Nagahara K, Pollak A: Sudden hearing loss: circulatory.
Buerger’s disease. Arch Otolaryngol 75:502, 1962. Am J Otol 5:488, 1984.
309. Huang CY, Yu YL: Small cerebellar strokes may mimic labyrinthine 324. Redleaf MI, Bauer CA, Gantz BJ, et al: Diatrizoate and dextran
lesions. J Neurol Neurosurg Psychiatry 48:263, 1985. treatment of sudden sensorineural hearing loss. Am J Otol 16:295,
310. Brownson RJ, Zollinger WK, Madeira T, et al: Sudden sensorineu- 1995.
ral hearing loss following manipulation of the cervical spine. 325. Wilkins SA, Jr, Mattox DE, Lyles A: Evaluation of a “shotgun”
Laryngoscope 96:166, 1986. regimen for sudden hearing loss. Otolaryngol Head Neck Surg 97:
311. Jorgensen MB: The inner ear in diabetes mellitus: histological 474, 1987.
studies. Arch Otolaryngol 74:373, 1961. 326. Hughes GB, Freedman MA, Haberkamp TJ, et al: Sudden senso-
312. Hallberg OE: Sudden deafness of obscure origin. Laryngoscope rineural hearing loss. Otolaryngol Clin North Am 29:393, 1996.
66:1237, 1956. 327. Tucci DL, Farmer JC, Jr, Kitch RD, et al: Treatment of sudden
313. Shea JJ, 3rd, Brackmann DE: Multiple sclerosis manifesting as sensorineural hearing loss with systemic steroids and valacyclovir.
sudden hearing loss. Otolaryngol Head Neck Surg 97:335, 1987. Otol Neurotol 23:301, 2002.
151 Tinnitus and Hyperacusis
Carol A. Bauer
Key Points
■ Tinnitus is a common condition that affects 30% of people older than age 55; the 5-year incidence
is 5%.
■ Disturbing tinnitus that impairs daily life activity affects 1% to 5% of those who have tinnitus.
■ Auditory deprivation from hearing loss induces central neural changes that result in tinnitus.
Mechanisms responsible for tinnitus involve peripheral triggers and central plasticity.
■ Sound therapy is an effective form of tinnitus treatment that benefits most patients when combined
with education and adjunct treatments directed toward factors that exacerbate tinnitus.
■ Tinnitus that can be modulated with somatic manipulation may respond to therapy that targets
temporomandibular joint and cervical spine disorders.
■ Medications are helpful for specific forms of tinnitus.
■ Associated comorbidities of anxiety and depression warrant referral for professional evaluation and
treatment with counseling and behavioral therapy.
■ Clinicians knowledgeable in auditory function and head and neck physiology are well equipped to
provide most tinnitus patients with effective treatment, which includes education, rehabilitation of
hearing loss, and identification and treatment of exacerbating factors.
2336
151 | TINNITUS AND HYPERACUSIS 2337
with mild tinnitus in the initial survey had no tinnitus on 5-year to prevent the onset or progression of NIHL and may possibly
follow-up, and only 20% reported progression to moderate limit the incidence of tinnitus. Intense sound exposure triggers
or severe tinnitus. Forty-five percent of participants with tinni- a reduction of blood flow and a cascade of metabolic events in
tus rated as “significant” at baseline reported no tinnitus at the cochlea, with formation of reactive oxygen and nitrogen
follow-up (43%) or reported improvement to mild tinnitus species that damage cellular lipids, proteins, and DNA and
(57%). This rate of spontaneous improvement in tinnitus has culminate in increased cell death.11 Interventions that target
significant implications for designing clinical trials to assess the these molecular mechanisms of NIHL include antioxidant
efficacy of tinnitus interventions. therapy such as with vitamin E, salicylate, and N-acetylcysteine.12,13
Ginkgo biloba extract contains multiple compounds with vaso-
tropic, potential neuroprotective, and antioxidant effects.
SUBJECTIVE TINNITUS Although uncontrolled trials and anecdotal reports have sug-
Subjective tinnitus is the most common form that affects adults, gested the efficacy of ginkgo, results from a meta-analysis and
and it is the focus of this chapter. Subjective tinnitus is most randomized controlled trials do not support its use.14,15
commonly related to sensorineural hearing loss (SNHL) from Presbycusis is SNHL related to aging. Most cases of presby-
acoustic trauma and presbycusis, and it is less commonly the cusis cannot be strictly and solely attributed to aging but rather
result of conductive hearing loss, endolymphatic hydrops, and involve some combination of cochlear injury from additional
cerebellopontine angle neoplasms. Subjective tinnitus can be sources, such as cumulative noise injury, metabolic or vascular
subtyped based on etiology, the pattern of associated hearing dysfunction, and genetic predisposition. For example, elderly
loss, psychoacoustic features, exacerbating factors, psychologic patients with diabetes have significantly higher pure tone
comorbidities, and the presence of somatic modulators. Tin- thresholds, lower otoacoustic emission amplitude, and lower
nitus subtype classification schemes can be useful to identify speech recognition in noise16 than age-matched individuals
forms of tinnitus that are responsive to specific targeted treat- without diabetes.17 Interactions between age and other factors
ment programs. Box 151-2 lists some useful features for subtyp- that affect the cochlea and auditory pathway challenge the iden-
ing tinnitus. tification of a single mechanism for presbycusis-associated tin-
nitus and development of an effective targeted intervention.
Hearing Loss Subtypes
The two most common types of hearing loss associated with Somatic Tinnitus Subtype
tinnitus are noise-induced hearing loss (NIHL) and presbycu- Somatic tinnitus is a unique form of tinnitus in which the loud-
sis. NIHL is a significant and growing health problem. Although ness, laterality, or tonality of the tinnitus can by modulated by
reduction of exposure to occupational noise has been effec- somatic modulation. This form of tinnitus was originally
tive in the last several decades, a notable increase has been observed in a small group of patients after surgical removal of
reported in the incidence of NIHL from recreational and leisure large vestibular schwannomas.18,19 Postoperatively, these patients
activity in children and adolescents and from military combat– had the ability to modulate their chronic tinnitus by exagger-
related noise exposure in young adults.6-8 Acute transient tin- ated eye movements, leg motion, or cutaneous stimulation of
nitus is nearly universal immediately after unprotected exposure the hands or face.20 The presumed mechanism of action for
to damaging acoustic stimuli, such as gunfire and amplified this unusual form of tinnitus is neural sprouting or aberrant
music. A Web-based survey of 9693 adolescents determined that reinnervation after auditory deafferentation. Subsequent to
61% of respondents experienced hearing loss and temporary these observations, a more general form of somatosensory
tinnitus after attending concerts.9 The prevalence of chronic modulation of tinnitus has been recognized, in which tinnitus
tinnitus associated with NIHL is 50% to 70%.10 Chronic tinnitus can be modulated by maneuvers or stimulation of the head and
induced by acoustic trauma occurs at a younger age than tin- neck region. It has been reported that forceful isometric con-
nitus associated with other types of hearing loss. Consequently, traction of the head and neck muscles can modify the loudness
acoustic trauma–induced tinnitus is experienced for a longer and pitch of tinnitus in 65% to 80% of patients with idiopathic
portion of the life span than other forms of tinnitus. tinnitus.21,22 Furthermore, tinnitus can be induced by strong
NIHL and associated tinnitus are preventable. In addition to contractions of muscles in the jaw, head, or neck in 15% to 58%
obvious proactive methods, such as wearing ear-protective hard- of subjects without a history of tinnitus.22,23
ware, intervention in the periexposure period may prove useful The association between tinnitus and somatic pathology of
the head and neck is underscored by the reported higher inci-
dence of tinnitus in individuals with temporomandibular joint
(TMJ) dysfunction and normal audiometric thresholds com-
pared with controls.24 One third of patients with symptoms of
Box 151-2. SUBJECTIVE TINNITUS SUBTYPES
TMJ dysfunction reported modulation of tinnitus with jaw
Pattern of hearing loss movement or pressure applied to the TMJ.25 When tinnitus
Noise-induced hearing loss occurs in association with disorders of the head and neck—
Presbycusis such as TMJ dysfunction, unilateral facial pain, otalgia, and
Unilateral occipital or temporal headache—successful tinnitus alleviation
High-frequency hearing loss may be possible using interventions that target the somatic
Outer hair cell dysfunction
Somatic tinnitus
dysfunction.
Temporomandibular joint dysfunction Levine and colleagues26 systematically reviewed the efficacy
Cervical dysfunction of treatments that target somatosensory systems. They defined
Gaze evoked somatic tinnitus syndrome as tinnitus that is 1) perceived in the
Cutaneous evoked ear, 2) ipsilateral to the somatic trigger, and 3) not associated
General somatosensory modulated with any new hearing complaints. Tinnitus that is strongly lat-
Typewriter tinnitus eralized to one ear in the presence of symmetric hearing,
Exacerbated by sleep or rest including symmetric hearing loss, would theoretically have a
Musical/complex somatic etiologic component by the definition of Levine and
Associated affective disorder
Intrusive (versus habituated)
colleagues. The review by these authors presents evidence that
somatic tinnitus is often responsive to acupuncture, electric
2338 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
Until more recently, most pharmacologic interventions for neurotransmitters may be important in the distress associated
tinnitus were empirically determined. Anecdotal experience with tinnitus.
and fortuitous observations of tinnitus relief were the primary Treatment with sertraline, a SSRI, improved tinnitus loud-
sources of innovation. Over the past 50 years, pharmacologic ness (P = .014) and severity (P = .024) in a group of tinnitus
treatment of tinnitus has become more rational. Anesthetics patients with an associated depressive or anxiety disorder.103
(lidocaine, tocainide, mexiletine), anticonvulsants (carbamaze- Despite this positive evidence, it is unclear whether SSRI treat-
pine, gabapentin), and tranquilizers (diazepam, clonazepam, ment improved tinnitus directly or indirectly through allevia-
oxazepam) have been investigated as tinnitus treatments. They tion of mood disorder. A placebo-controlled trial of the SSRI
have in common the general property of facilitating neural paroxetine in a group of subjects without coexisting mood or
inhibition. Antidepressants such as trimipramine, nortriptyline, anxiety disorder did not produce improvement in any tinnitus
amitriptyline, and selective serotonin reuptake inhibitors measure over placebo.104
(SSRIs) have been tested for their ability to ameliorate the
comorbid mood disturbance associated with tinnitus. Consider-
ing only well-controlled trials, mixed results have been obtained
CLINICAL EVALUATION OF TINNITUS
for all drugs tested to date. Clinical treatment of a patient with tinnitus should begin with
The hypothesis that tinnitus emerges from increased central a general medical evaluation followed by a complete head
neural activity after loss of inhibition can be used to guide and neck examination. Objectives of the evaluation include a
pharmacologic intervention and is supported by work with descriptive characterization of the tinnitus (psychoacoustic
animal models,92-94 and it provides a rational basis for numerous properties, impact on daily life, reactive components); determi-
pharmacologic interventions that include lidocaine, carbam- nation of etiology; and identification of factors that exacerbate,
azepine, alprazolam, and gabapentin. Despite the increase in ameliorate, or trigger the tinnitus. At a minimum, the results
targeted tinnitus drug trials, use of a single agent to treat a of the examination educate the patient about the tinnitus, and
heterogeneous large sample of subjects has been for the most patient education is a powerful therapeutic component of the
part unsuccessful. Several reasons, none mutually exclusive, clinical process that should not be undervalued or underesti-
may account for this lack of success. Most tinnitus clinical trials mated. A thorough examination also facilitates development of
randomly select and assign participants to treatment groups. a directed individual treatment plan.
However, evidence is emerging that tinnitus is a heterogeneous Tinnitus can be described in terms of its psychoacoustic
disorder with variable pathologic features.34,95,96 Testing drugs properties and in terms of the affective or reactive responses to
with a single mechanism of action is unlikely to succeed when the tinnitus. The affective or reactive components include the
using randomly determined heterogeneous sample groups. For comorbid problems of depression, sleep disruption, difficulty
the same reason, trials that use small dose ranges may also fail. with concentration, sadness, anxiety, and fear. The reactive
Studies that enroll subjects with a single tinnitus etiology components of tinnitus are highly individual and are significant
may be more likely to identify successful treatments. A study factors in tinnitus disability. Careful evaluation of both compo-
that examined the effect of gabapentin on tinnitus in two spe- nents is important for the clinician to appreciate fully the
cific patient subpopulations obtained positive results.97 The impact of the tinnitus on the individual and to formulate a
study was designed to test the hypothesis that acoustic trauma directed treatment plan.
leads to a loss of inhibition in the auditory pathway mediated The qualitative features of tinnitus can be assessed using
by the inhibitory neurotransmitter γ-aminobutyric acid (GABA). standardized questionnaires and psychophysical measure-
The hypothesis was advanced after an animal experiment that ments. Relevant information includes localization (left, right,
showed gabapentin, a GABA analogue, to be effective in reduc- in the head, outside of the head), constancy (episodic, fluctuat-
ing the loudness of tinnitus in rats.93 This study used a very wide ing, constant, pulsatile), pitch, loudness, and sound quality
dose range and tested one group of patients with objective and (tonal, hissing, buzzing, clicking, ringing). This information
historic evidence of traumatic sound exposure and a second can be useful in determining etiology: pulsatile tinnitus can
group without such evidence. Gabapentin was found to reduce imply a vascular source, whereas fluctuating tinnitus may be
tinnitus annoyance significantly in the trauma group at a daily linked to specific triggers such as foods, illnesses, stress, or
dose level of 1800 to 2400 mg. In contrast, a subsequent clinical acoustic trauma. Episodic tinnitus may relate to unstable
trial to study the effect of only a single dose in a randomly hearing thresholds that derive from cochlear dysfunctions,
selected group of patients did not obtain a significant effect.98 such as hydrops or perilymph fistulae. Clicking or tapping tin-
The therapeutic effect was likely missed because only a single nitus may occur with mechanical disorders that affect either the
dose level was used, and the treatment group included various middle ear muscles (stapedial or tensor tympani spasm) or the
tinnitus etiologies. Drugs with a specific mechanism of action auditory nerve (vascular loop or demyelination). Identification
are unlikely to be effective in all subjects characterized by such of tinnitus with specific characteristics such as these is very
heterogeneity. Stratified study designs with segregation of tin- useful and can lead to a directed course of therapy.
nitus types, etiologies, and hearing characteristics should be
more successful in identifying effective drug therapies. Standardized Outcome Measures
The most extensively used drugs for tinnitus treatment are Numerous standardized questionnaires are available for mea-
antidepressants. Attempting to treat tinnitus with antidepres- suring tinnitus severity and perceived disability. Standardized
sants is sensible for two reasons: first, the association between assessment is useful for documenting clinical outcomes and
severe tinnitus and mood disorders is well recognized; second, reporting the results of clinical trials. Standardized measures
the pharmacologic mechanism of action of many antidepres- are also crucial for determining the subjective impact of tin-
sants involves receptors and neurotransmitters located in the nitus. Finally, standardized questionnaires are useful for stratify-
auditory pathway.99 Although GABA deficiency seems to con- ing patients according to the severity and impact of their
tribute to tinnitus pathology,97,100-102 the role of other neu- tinnitus, which facilitates identification of specific problems
rotransmitter systems in triggering or maintaining tinnitus is and serves to triage patient care from minimal counseling to
currently unknown. Serotonin is known to function as a modu- intensive rehabilitation.
lator of sensory systems, learning, and memory. Along with The THI is a widely used self-assessment tool.105 This 25-item
acetylcholine, serotonin can affect behavioral conditioning questionnaire has good construct validity, strong internal con-
and associated plastic changes in auditory cortex. Both sistency, and good test-retest reliability. The THI yields a total
151 | TINNITUS AND HYPERACUSIS 2343
score and three subscale scores; the subscales encompass func- migraine,95 Lyme disease,96 benzodiazepine withdrawal,97 or as
tional limitations in mental (e.g., difficulty concentrating), part of a syndrome.98 Patients with tinnitus and hyperacusis
social, occupational, and physical domains (e.g., difficulty frequently have loudness discomfort levels that are lower
sleeping); emotional responses to tinnitus (e.g., anger, depres- than those of individuals with similar levels of SNHL, again
sion, anxiety); and catastrophic reactions to tinnitus (e.g., des- indicating that these phenomena are distinctly different from
peration, loss of control, failure to cope). In addition to its recruitment.99
good internal consistency and test-retest reliability, the THI has Andersson and associates100 assessed the prevalence of
high convergent validity with the 27-item Tinnitus Handicap hyperacusis in the general population in two surveys of the
Questionnaire and the 52-item Tinnitus Questionnaire.106 The adult Swedish population. The point prevalence of hyperacusis
95% confidence interval for the THI is 20 points, which sug- was 5.9% (postal survey) and 7.7% (Internet survey). Partici-
gests that a difference in scores of 20 points or greater repre- pants who reported hearing impairment were excluded from
sents a statistically and clinically significant change. the prevalence calculations, which minimized the inclusion of
patients with cochlear damage and reduced dynamic range in
Tinnitus Comorbidities the sample. Other surveys of hyperacusis report a higher preva-
Severe, debilitating tinnitus is frequently associated with depres- lence in the general population (22%).101 The prevalence of
sion, anxiety, and other mood disorders.79,107 Comorbid emo- hyperacusis within the tinnitus population is unknown,
tional disturbance is not unique to tinnitus and has been shown although estimates range from 40% to 80%, but no systematic
to accompany many chronic illnesses.83,108 It is well recognized survey has been conducted for accurate estimation.102,103 In the
that coexisting mood disorders hamper improvement and general population and the tinnitus population, the varied
interfere with treatment of conditions such as chronic pain and prevalence estimates likely reflect the adopted operational defi-
tinnitus. The identification and treatment of comorbid condi- nition of hyperacusis.
tions in tinnitus patients is an important aspect of the clinical Objective tests of loudness discomfort or sound intolerance
evaluation. In an outpatient setting, several tools are available in patients with hyperacusis have been adapted from proce-
to screen for mood disorders, such as the Beck Depression dures developed for assessing recruitment.104,112 Several varia-
Inventory and the Hamilton Anxiety Scale. An important thera- tions exist on the technique for measuring loudness discomfort
peutic adjunct is teaching coping skills through behavior modi- levels, but no consensus or standardization has been estab-
fication and cognitive therapy.84 lished. In addition, issues of intersubject and intrasubject vari-
ability,113 test-retest reliability,114 operator dependency, and
Tinnitus and Insomnia poor face validity for test stimuli all limit the utility of adapting
Patients commonly report that tinnitus interferes with their measures of loudness intolerance to patients with hyperacusis.
ability to fall asleep, and sleep disruption is a significant comor- Very few validated, well-established self-report question-
bid condition in adults and children.85,86 A robust correlation naires specifically assess hyperacusis. Dauman and Bouscau-
has been shown between the reported loudness and severity of Faure103 developed a scale for assessing hyperacusis in patients
tinnitus and degree of sleep disruption.87,109 A positive feedback with tinnitus called the Multiple-Activity Scale for Hyperacusis,
loop may exist in which tinnitus leads to sleep deprivation, and which uses a structured interview for rating the annoyance
the sleep deprivation exacerbates somatic complaints, tinnitus induced by sound exposure from different physical and social
among them. Depression and anxiety exacerbated by sleep loss activities. They reported hyperacusis in 197 of 249 clinical
may significantly interact with and compound the cycle of tin- patients (79%) screened with the instrument and reported
nitus and poor quality sleep. Coping ability may also deteriorate substantial to severe annoyance from hyperacusis present in
as a result of sleep deprivation or lack of restorative sleep. Sleep 42%. No correlation was found between the severity of hyper-
difficulties include insufficient sleep, poor quality sleep, and acusis, as indicated by the Multiple-Activity Scale for Hyperacu-
nonrestorative sleep. Altered sleep architecture reflected in a sis, and audiometric threshold shift, which again indicates
significant decrease in time spent in stage 3 and 4 REM sleep that hyperacusis is a phenomenon distinct from loudness
was demonstrated in subjects with chronic tinnitus compared recruitment. The Hyperacusis Questionnaire is a 14-item instru-
with matched controls.110 Psychologic and behavioral manage- ment, in which hyperacusis is assessed using a four-alternative
ment of sleep disturbance is an effective treatment modality for Likert scale.115
chronic insomnia and may provide benefit for patients with The impact of hypersensitivity to sound can range from
tinnitus and concomitant sleep disturbance.111 general avoidance of social situations, such as concerts and
Pharmacologic sleep aids such as melatonin can reduce tin- restaurants, to specific sound aversions: vacuum cleaners,
nitus severity, particularly in patients with pronounced sleep traffic, clinking dishes, children playing, and so on. In extreme
difficulties.91,92 Bedside sound generators have also been shown cases, patients with severe hypersensitivity become housebound
to improve sleep quality significantly and to lessen tinnitus in an effort to control acoustic exposure, and they wear ear
distress. The bedside sounds most frequently chosen are often plugs and ear muffs for extended periods. In all cases of sound
selected for their perceived positive emotional effect.41 hypersensitivity, it is important to address the physiologic com-
ponent of loudness discomfort and reduced dynamic range and
also the psychologic component of fear, anxiety, social with-
HYPERACUSIS drawal, and maladaptation that accompanies the condition.
Hyperacusis has been defined as noise intolerance, annoyance The cognitive influence on sound sensitivity is illustrated by the
caused by ordinary sounds, and abnormal discomfort for supra- high correlation of loudness tolerance with anxiety.116 A survey
threshold sound.93 These definitions distinguish hyperacusis, of 62 Swedish patients with hyperacusis showed that nearly half
considered by many to be a central phenomenon, from recruit- had a concomitant psychiatric disorder with a predominance
ment, the rapid growth of perceived loudness with increasing of anxiety-related personality traits.117
stimulus level observed in association with cochlear hearing loss The efficacy of sound therapy for treating hyperacusis has
and outer cell dysfunction. Hyperacusis frequently occurs in had mixed results. Dauman and Bouscau-Faure103 reported that
association with tinnitus, but it can be present without tinnitus TRT was more effective in improving hyperacusis (63%) than
or any associated hearing loss. Hyperacusis can occur after the tinnitus (47%) in 32 patients evaluated at three time points
loss of the stapedial reflex in association with acute facial after therapy. They reported that hyperacusis remains a problem
paralysis94 and in association with general conditions such as for a significant proportion of patients treated with TRT. Gold
2344 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
26. Levine RA, Nam ED, Oron Y, et al: Evidence for a tinnitus sub-
REFERENCES group responsive to somatosensory based treatment modalities.
1. Nondahl DM, Cruickshanks KJ, Wiley TL, et al: Prevalence and Prog Brain Res 166:195–207, 2007.
5-year incidence of tinnitus among older adults: the epidemiology 27. Mardini MK: Ear-clicking “tinnitus” responding to carbamaze-
of hearing loss study. J Am Acad Audiol 13(6):323–331, 2002. pine. N Engl J Med 317(24):1542, 1987.
2. Cooper JC, Jr: Health and Nutrition Examination Survey of 28. Levine RA: Typewriter tinnitus: a carbamazepine-responsive syn-
1971-75: Part II. Tinnitus, subjective hearing loss, and well-being. drome related to auditory nerve vascular compression. ORL J Oto-
J Am Acad Audiol 5(1):37–43, 1994. rhinolaryngol Relat Spec 68(1):43–46; discussion 46–47, 2006.
3. Sismanis A: Tinnitus : advances in evaluation and management. Oto- 29. Brantberg K: Paroxysmal staccato tinnitus: a carbamazepine
laryngologic clinics of North America, v. 36, no. 2, Philadelphia, 2003, responsive hyperactivity dysfunction symptom of the eighth
W.B. Saunders Company, pp xii, 235–407. cranial nerve. J Neurol Neurosurg Psychiatry 81(4):451–455, 2010.
4. Evered D, Lawrenson G: Tinnitus. Ciba Foundation symposium; 85, 30. Ramachandran VS, Hirstein W: The perception of phantom
Summit, NJ, 1981, Ciba Pharmaceutical Co. Medical Education limbs. The D. O. Hebb lecture. Brain 121(Pt 9):1603–1630, 1998.
Administration. viii, 325. 31. Simmel ML: Phantom experiences following amputation in child-
5. Sindhusake D, Golding M, Newall P, et al: Risk factors for tinnitus hood. J Neurol Neurosurg Psychiatry 25:69–78, 1962.
in a population of older adults: the blue mountains hearing study. 32. Moller AR: The role of neural plasticity in tinnitus. Prog Brain Res
Ear Hear 24(6):501–507, 2003. 166:37–45, 2007.
6. Barney R, Bohnker BK: Hearing thresholds for U.S. Marines: 33. Merzenich MM, Nelson RJ, Stryker MP, et al: Somatosensory corti-
comparison of aviation, combat arms, and other personnel. Aviat cal map changes following digit amputation in adult monkeys.
Space Environ Med 77(1):53–56, 2006. J Comp Neurol 224(4):591–605, 1984.
7. Helfer TM, Jordan NN, Lee RB: Postdeployment hearing loss in 34. Yang TT, Gallen CC, Ramachandran VS, et al: Noninvasive detec-
U.S. Army soldiers seen at audiology clinics from April 1, 2003, tion of cerebral plasticity in adult human somatosensory cortex.
through March 31, 2004. Am J Audiol 14(2):161–168, 2005. Neuroreport 5(6):701–704, 1994.
8. Niskar AS, Kieszak SM, Holmes AE, et al: Estimated prevalence of 35. Diesch E, Struve M, Rupp A, et al: Enhancement of steady-state
noise-induced hearing threshold shifts among children 6 to 19 auditory evoked magnetic fields in tinnitus. Eur J Neurosci 19(4):
years of age: the Third National Health and Nutrition Examina- 1093–1104, 2004.
tion Survey, 1988-1994, United States. Pediatrics 108(1):40–43, 36. Arnold W, Bartenstein P, Oestreicher E, et al: Focal metabolic
2001. activation in the predominant left auditory cortex in patients suf-
9. Chung JH, Des Roches CM, Meunier J, et al: Evaluation of noise- fering from tinnitus: a PET study with [18F]deoxyglucose. ORL J
induced hearing loss in young people using a web-based survey Otorhinolaryngol Relat Spec 58(4):195–199, 1996.
technique. Pediatrics 115(4):861–867, 2005. 37. Giraud AL, Chéry-Croze S, Fischer G, et al: A selective imaging of
10. Axelsson A, Sandh A: Tinnitus in noise-induced hearing loss. Br J tinnitus. Neuroreport 10(1):1–5, 1999.
Audiol 19(4):271–276, 1985. 38. Mirz F, Pedersen B, Ishizu K, et al: Positron emission tomography
11. Le Prell CG, Hughes LF, Miller JM: Free radical scavengers vita- of cortical centers of tinnitus. Hear Res 134(1–2):133–144, 1999.
mins A, C, and E plus magnesium reduce noise trauma. Free Radic 39. Norena AJ, Eggermont JJ: Enriched acoustic environment after
Biol Med 42(9):1454–1463, 2007. noise trauma reduces hearing loss and prevents cortical map reor-
12. Kopke R, Beilefeld E, Liu J, et al: Prevention of impulse noise- ganization. J Neurosci 25(3):699–705, 2005.
induced hearing loss with antioxidants. Acta Otolaryngol 125(3): 40. Goodhill V: The management of tinnitus. Trans Am Laryngol
235–243, 2005. Rhinol Otol Soc 54th Meeting:220–231, 1950.
13. Ewert DL, Liu J, Li W, et al: Antioxidant treatment reduces blast- 41. Handscomb L: Use of bedside sound generators by patients with
induced cochlear damage and hearing loss. Hear Res 285(1–2):29– tinnitus-related sleeping difficulty: which sounds are preferred
39, 2012. and why? Acta Otolaryngol Suppl 556:59–63, 2006.
14. Drew S, Davies E: Effectiveness of Ginkgo biloba in treating tin- 42. Bentler RA, Tyler RS: Tinnitus management. ASHA 29(5):27–32,
nitus: double blind, placebo controlled trial. BMJ 322(7278):73, 1987.
2001. 43. McNeill C, Távora-Vieira D, Alnafjan F, et al: Tinnitus pitch,
15. Rejali D, Sivakumar A, Balaji N: Ginkgo biloba does not benefit masking, and the effectiveness of hearing aids for tinnitus therapy.
patients with tinnitus: a randomized placebo-controlled double- Int J Audiol 51(12):914–919, 2012.
blind trial and meta-analysis of randomized trials. Clin Otolaryngol 44. Searchfield GD, Kaur M, Martin WH: Hearing aids as an adjunct
Allied Sci 29(3):226–231, 2004. to counseling: tinnitus patients who choose amplification do
16. Bainbridge KE, Cowie CC, Rust KF, et al: Mitigating case mix better than those that don’t. Int J Audiol 49(8):574–579, 2010.
factors by choice of glycemic control performance measure 45. Surr RK, Montgomery AA, Mueller HG: Effect of amplification on
threshold. Diabetes Care 31(9):1754–1760, 2008. tinnitus among new hearing aid users. Ear Hear 6(2):71–75, 1985.
17. Frisina ST, Mapes F, Kim S, et al: Characterization of hearing loss 46. Folmer RL, Carroll JR: Long-term effectiveness of ear-level devices
in aged type II diabetics. Hear Res 211(1–2):103–113, 2006. for tinnitus. Otolaryngol Head Neck Surg 134(1):132–137, 2006.
18. Wall M, Rosenberg M, Richardson D: Gaze-evoked tinnitus. Neurol- 47. Trotter MI, Donaldson I: Hearing aids and tinnitus therapy:
ogy 37(6):1034–1036, 1987. a 25-year experience. J Laryngol Otol 122(10):1052–1056, 2008.
19. Whittaker CK: Tinnitus and eye movement. Am J Otol 4(2):188, 48. Vernon J, Schleuning A: Tinnitus: a new management. Laryngo-
1982. scope 88(3):413–419, 1978.
20. Cacace AT, Cousins JP, Parnes SM, et al: Cutaneous-evoked tin- 49. Terry AM, Jones DM: Preference for potential tinnitus maskers:
nitus. I. Phenomenology, psychophysics and functional imaging. results from annoyance ratings. Br J Audiol 20(4):277–297, 1986.
Audiol Neurootol 4(5):247–257, 1999. 50. Vernon JA, Meikle MB: Tinnitus masking:unresolved problems.
21. Abel MD, Levine RA: Muscle contractions and auditory percep- Ciba Found Symp 85:239–262, 1981.
tion in tinnitus patients and nonclinical subjects. Cranio 22(3):181– 51. Reavis KM, Rothholtz VS, Tang Q, et al: Temporary suppression
191, 2004. of tinnitus by modulated sounds. J Assoc Res Otolaryngol 13(4):561–
22. Sanchez TG, Buera GC, Lorenzi MC, et al: The influence of vol- 571, 2012.
untary muscle contractions upon the onset and modulation of 52. Henry JA, Schechter MA, Zaugg TL, et al: Clinical trial to compare
tinnitus. Audiol Neurootol 7(6):370–375, 2002. tinnitus masking and tinnitus retraining therapy. Acta Otolaryngol
23. Levine RA, Abel M, Cheng H: CNS somatosensory-auditory inter- Suppl 556:64–69, 2006.
actions elicit or modulate tinnitus. Exp Brain Res 153(4):643–648, 53. Jastreboff PJ, Hazell JW: A neurophysiological approach to tinni-
2003. tus: clinical implications. Br J Audiol 27(1):7–17, 1993.
24. Chole RA, Parker WS: Tinnitus and vertigo in patients with tem- 54. Bartnik G, Fabijanska A, Rogowski M: Effects of tinnitus retraining
poromandibular disorder. Arch Otolaryngol Head Neck Surg 118(8): therapy (TRT) for patients with tinnitus and subjective hearing
817–821, 1992. loss versus tinnitus only. Scand Audiol Suppl 52:206–208, 2001.
25. Rubinstein B, Axelsson A, Carlsson GE: Prevalence of signs and 55. Berry JA, Gold SL, Frederick EA, et al: Patient-based outcomes in
symptoms of craniomandibular disorders in tinnitus patients. patients with primary tinnitus undergoing tinnitus retraining
J Craniomandib Disord 4(3):186–192, 1990. therapy. Arch Otolaryngol Head Neck Surg 128(10):1153–1157, 2002.
2344.e2 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
111. Espie CA, Inglis SJ, Tessier S, et al: The clinical effectiveness of 115. Khalfa S, Dubal S, Veuillet E, et al: Psychometric normalization of
cognitive behaviour therapy for chronic insomnia: implementa- a hyperacusis questionnaire. ORL J Otorhinolaryngol Relat Spec
tion and evaluation of a sleep clinic in general medical practice. 64(6):436–442, 2002.
Behav Res Ther 39(1):45–60, 2001. 116. Stephens SD: The treatment of tinnitus—a historical perspective.
112. Sherlock LP, Formby C: Estimates of loudness, loudness discom- J Laryngol Otol 98(10):963–972, 1984.
fort, and the auditory dynamic range: normative estimates, com- 117. Jüris L, Andersson G, Larsen HC, et al: Psychiatric comorbidity
parison of procedures, and test-retest reliability. J Am Acad Audiol and personality traits in patients with hyperacusis. Int J Audiol
16(2):85–100, 2005. 52(4):230–235, 2013.
113. Stephens SD, Blegvad B, Krogh HJ: The value of some supra- 118. Gold SL, Formby C, Gray WC: Celebrating a decade of evaluation
threshold auditory measures. Scand Audiol 6(4):213–221, 1977. and treatment: the University of Maryland Tinnitus & Hyperacusis
114. Bornstein SP, Musiek FE: Loudness discomfort level and reliability Center. Am J Audiol 9(2):69–74, 2000.
as a function of instructional set. Scand Audiol 22(2):125–131, 1993.
Noise-Induced Hearing Loss 152
Brenda L. Lonsbury-Martin | Glen K. Martin
Key Points
■ Noise-induced hearing loss (NIHL) is second only to age-related hearing loss as the most prevalent
form of hearing loss.
■ NIHL that results from relatively brief noise exposures can be reversible, as happens with exposure
that occurs at an evening spent in a loud entertainment venue.
■ Permanent NIHL is caused by either an acoustic trauma from a brief exposure to a very intense
blast of sound or chronic long-term exposure to loud sounds, such as those associated with a noisy
occupation.
■ An accelerating incidence of high-frequency hearing loss in younger individuals points to early,
chronic noise exposure, possibly from personal entertainment devices.
■ NIHL is a complex condition influenced by environmental and genetic factors.
■ NIHL is associated not only with cochlear injury but also with upstream damage to the auditory
pathway.
■ Genetic association studies have identified genetic factors primarily related to oxidative stress that
influence an individual’s susceptibility to NIHL.
■ Current research on the administration of certain antioxidants or dietary supplements before or
after noise exposure shows promise for developing a pharmacologic treatment for NIHL in the
near future.
■ NIHL is a preventable condition, and the otolaryngologist plays a critical role in educating patients
about protecting their ears from the adverse effects of noise overexposure.
O ne of the most common causes of permanent hearing MEASUREMENT OF NOISE
impairment is exposure to excessive sounds. Hundreds of mil- The term noise is commonly used to designate an undesirable
lions of individuals worldwide have noise-induced hearing loss sound. In the scientific and clinical fields that deal with hearing,
(NIHL), which results in a reduced quality of life because of this term has come to mean any excessively loud sound that has
social isolation and possible relentless tinnitus in addition to the potential to harm hearing. The temporal patterns of envi-
impairment in communication with family members, cowork- ronmental noise are typically described as continuous, fluctuat-
ers, and friends. In the United States alone, approximately 30 ing, intermittent, or impulsive.3 Continuous noise, or steady-state
million workers are exposed to hazardous job-related noises. noise, remains relatively constant, whereas fluctuating noise
The costs are immense: almost $250 million is spent per year increases and decreases in level over time, and intermittent noise
in terms of compensation and early retirement payments for is interrupted for varying time periods. Impulsive noise, or
work-related NIHL.1 Moreover, disability compensation associ- impact noise caused by explosive or metal-on-metal mechanical
ated with occupational hearing loss linked to military service events, have rapidly changing pressure characteristics that
represents an even greater cost to our society. In a recent report consist of intense, brief (i.e., milliseconds) wave fronts, fol-
for fiscal year 2009, the U.S. Government Accountability Office lowed by much smaller reverberations and echoes that occur
reported to Congress that some of the most common impair- over many seconds. The amount of noise, usually referred to
ments for veterans who receive disability benefits were hearing as the sound-pressure level (SPL), is conventionally measured by
related and that annual payments for such conditions exceeded a sound-level meter in decibel (dB) units using a frequency-
$1.1 billion.2 weighting formula called the A-scale. The dBA-scale metric of
This chapter presents and discusses recent perspectives on sound level essentially mimics the threshold-sensitivity curve for
the effects of excessive noise on hearing that address the scien- the human ear, so the low-frequency and high-frequency com-
tific and practical aspects of NIHL. Although NIHL has been ponents are given less emphasis as auditory hazards. Standard
studied experimentally for more than a century, only in the last sound-level meters have electronic networks designed to
few decades have some major breakthroughs occurred in our measure noise magnitude automatically in dBA, whereas to
basic understanding of the ear’s reaction to damaging sounds, measure impulse or impact noise, a more intricate peak-reading
along with a better understanding of the environmental and sound-level meter is needed that is capable of accurately mea-
genetic factors that contribute to NIHL. This steady progres- suring sounds with essentially instantaneous onset times.
sion in the knowledge base about NIHL promises to improve The personal noise dosimeter is typically used to measure
significantly the detection and treatment of this disorder over noise exposure in the workplace. This instrument provides a
the coming years. readout of the noise dose or the percent exposure experienced
2345
2346 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
by a single worker, typically over a specific shift. The logging than is known about NIHL. Consequently, it is well established
dosimeter integrates a function of sound pressure over time that a single exposure to a severe sound that causes violent
and calculates the daily (8-hour) dose with respect to the changes in air pressure can produce direct mechanical damage
current permissible noise level for a continuous noise of less to the delicate tissues of the peripheral auditory apparatus,
than 90 dBA lasting 8 hours. More recently, personal noise which includes components of the middle ear (tympanic mem-
dosimeters have been offered to the consumer as a portable, brane, ossicles) and inner ear (organ of Corti). In contrast,
compact, and affordable device that can be used to protect regular exposure to less intense but still noisy sounds involves
hearing. The instrument measures and displays noise dose con- the insidious destruction of cochlear components that eventu-
tinuously for 16 hours, and the dosimeter provides an early ally and unavoidably leads to an elevation in hearing thresholds,
warning that the user is approaching overexposure and should along with other common symptoms of hearing impairment.
use hearing protection. A particular noise—such as from power Acoustic trauma was previously a relatively rare event typi-
tools, music concerts, or sporting events—can also be measured cally associated with accidental explosions in industrial settings.
for 2 minutes, and then the estimated dose per hour is calcu- However, military servicemen and servicewomen caught in
lated and displayed to determine whether permissible exposure roadside home-made bomb or improvised explosive device
levels would be exceeded. By putting valuable health informa- blasts in recent armed conflicts are returning home in epi-
tion into the hands of consumers, such easy-to-use, inexpensive demic numbers with tinnitus and profound, permanent hearing
(<$100) dosimeters empower individuals to take appropriate losses.5 Among the blast-related comorbidities is mild traumatic
steps to prevent NIHL. brain injury associated with central auditory processing com-
plaints.6 Overall, then, acoustic trauma is a hearing problem
that is increasing, at least in combat troops. Because many of
NATURE OF THE HEARING LOSS these postdeployment cases are being treated in the private
Depending on the level of the sound exposure, either revers- sector, otolaryngologists may see acoustic trauma in increasing
ible or permanent damage can occur to the peripheral auditory numbers.
end organ. The reversible loss, typically referred to as a tempo- Irreversible NIHL is a specific pathologic state that exhibits
rary threshold shift (TTS), results from exposures to moderately a recognized set of symptoms and objective findings.7 NIHL
intense sounds, such as might be encountered at a live music includes 1) a permanent sensorineural hearing loss with
event or by using noisy power tools. Hearing problems associ- damage principally to cochlear hair cells, primarily to OHCs;
ated with TTS include elevated thresholds, particularly for the 2) a history of long-term exposure to dangerous noise levels
higher midfrequency region that includes the 3- to 6-kHz fre- (i.e., >85 dBA for 8 hours/day) sufficient to cause the degree
quencies. The TTS condition is often accompanied by many and pattern of hearing loss described by audiologic findings;
other common symptoms of hearing impairment that include 3) a gradual loss of hearing over the first 5 to 10 years of expo-
tinnitus, loudness recruitment, muffled sounds, and diplacusis. sure; 4) hearing loss that involves initially the higher frequen-
Depending on the duration of the exposure, recovery from cies, from 3 to 8 kHz, before including frequencies of 2 kHz or
TTS can occur over periods that range from minutes to hours less; 5) speech-recognition scores consistent with the audiomet-
or days. ric loss; and 6) hearing loss that stabilizes after the noise expo-
After exposure, if TTS does not recover before the ear is sure is terminated.
reexposed to excessive sound, a permanent change in hearing A patient with NIHL commonly consults a physician because
can occur that is referred to as a permanent threshold shift (PTS). of difficulties in hearing and understanding ordinary speech,
In PTS, the elevation in hearing thresholds is irreversible, especially in the presence of background noise. Many variations
because lasting structural damage occurs to the critical ele- can be found in the detailed configuration of the audiogram
ments of the cochlea. The precise relationship between the of a noise-damaged ear, depending on the temporal and spec-
TTS and PTS stages of hearing loss caused by noise exposure tral distribution of the noise stimulus and on the stage of
is unknown. Although it seems logical to assume that repeated hearing loss. The pattern of hearing loss most commonly associ-
episodes of TTS would eventually lead to PTS, experimental ated with the earlier stages of NIHL is illustrated in Figure
evidence suggests that the fundamental processes that underlie 152-1, A. The beginning region of impairment involves the
the development of reversible versus permanent NIHL are sensitive midfrequency range, primarily 3 to 6 kHz, and the
unrelated. Nordmann and colleagues4 used a survival fixation corresponding hearing loss is classically described as the “4-kHz
approach to show that the histopathologic manifestations of notch.” This pattern of maximal hearing loss, with little or no
TTS and PTS noise damage to the chinchilla cochlea are dis- loss below 2 kHz, typically occurs regardless of the noise-
tinct. Specifically, TTS was correlated with a buckling of the exposure environment. The audiogram results in Figure 152-1,
supporting pillar cell bodies in the frequency region of the A, also show the sensorineural aspect of NIHL in that thresh-
maximal exposure effect. The morphologic abnormality consis- olds for bone-conducted stimuli are essentially identical to the
tently correlated with PTS was a focal loss of hair cells and a thresholds for air conduction. The profile of noise-induced
complete degeneration of the corresponding population of threshold hearing is usually symmetric for both ears, particu-
nerve fiber endings. Because PTS eventually develops from larly for individuals who have been working in noisy industrial
repeated exposures to stimuli that initially produce only TTS, settings in which “surround” sounds are present.
it is likely that the latter condition is also associated with subtle Commonly, other forms of noxious sound, such as the
changes to the sensitive outer hair cell (OHC) system that go gunfire associated with sport shooting, cause an asymmetric
undetected by conventional light microscopy. pattern of hearing loss similar to the one illustrated in Figure
Traditionally, PTS caused by acoustic overstimulation has 152-1, B. In this case, the ear pointed toward the source of noise
been separated into two distinct classes. One type, called acoustic (gun barrel)—which is the right ear of the left-handed shooter
trauma, is caused by a single, brief exposure to a very intense depicted in Figure 152-1, B—would have worse hearing than
sound (e.g., an explosive blast), and it results in a sudden, the ear directed away from the source (in this example, the left
usually painful loss of hearing. The other type of hearing loss or protected ear) by 15 to 30 dB or more and particularly at
is commonly referred to as noise-induced hearing loss (NIHL) and higher frequencies because of the absence of the protective
results from chronic exposure to less intense levels of sound. A head shadow effect.
great deal more is known about the anatomic processes that The development of a hearing loss caused by habitual expo-
underlie the symptoms of and recovery from acoustic trauma sure to moderately intense levels of noise typically consists of
152 | NOISE-INDUCED HEARING LOSS 2347
–10 –10
RE
0 0
LE
10 10
FIGURE 152-1. Audiometric patterns of <
two stages. Initially, the middle to high frequencies exhibit the loss worsened at the higher frequencies and spread to the lower
resulting hearing loss. As the length of time of exposure to loud frequencies. In addition, for average exposure times of less
noise increases, hearing loss becomes greater and begins to than 10 years, hearing levels for the press and hammer opera-
affect adjacent higher and lower frequencies. In a classic cross- tors who were exposed to mean levels of 108 and 99 dB SPL,
sectional study of occupational NIHL, Taylor and colleagues8 respectively, deteriorated similarly. For long-term exposures of
showed the gradual loss of hearing sensitivity in workers caused 10 years or more, the results of Taylor and colleagues8 indicated
by habitual exposure to the intense sounds of drop-forging that hearing losses that resulted from the hammer-induced
tools commonly used in foundries. Figure 152-2 illustrates the impact noise were greater than those that resulted from the
progressive effects of exposure to the wideband noise depicted more continuous noise of the press equipment. Finally, a char-
in Figure 152-2, A, of two types of forging tools, presses and acteristic feature of NIHL clearly documented in Figure 152-2,
hammers, on the magnitude of hearing loss as the length of B and C, is that hearing levels are rarely increased beyond about
exposure increased. For the operators of the press and hammer 70 to 90 dB of hearing loss on average even after more than
equipment (see Fig. 152-2, B and C, respectively), the approxi- 30 years of continuous noise exposure.
mate 10- to 20-dB threshold shifts, typically observed at the
higher frequencies during the first 1 to 2 years of exposure,
grew to be a 20-dB or greater loss, from 3 to 6 kHz, after a 3-year
COCHLEAR DAMAGE
exposure. After an 8-year exposure, a 40-dB or greater thresh- The primary site of anatomic damage is at the level of the
old shift was apparent. mechanosensory receptors of the auditory system’s end organ.
Detailed comparisons of the NIHL growth curves of Figure Loud sound damages the inner hair cells (IHCs) and OHCs of
152-2, B and C, reveal that with continuing exposure, hearing the organ of Corti, and the OHCs in particular are most affected
120 10 10
Sound pressure level (dB)
0 0
110 –10 –10
–20 –20
100 –30 –30
–40 –40
90 –50 –50
–60 –60
80 –70 –70
Hammer –80 –80
Press
70 –90 –90
.063 .125 .25 .5 1 2 4 8 0 1 2 3 4 5 6 0 1 2 3 4 5 6
A One-third octave band center B Frequency (kHz) C Frequency (kHz)
frequency (kHz)
Control 1–2 yr 2–3 yr 4–7 yr
in the initial stages. In instances that involve very intense acous- of IHCs and OHCs), with its dense network of nerve fibers, to
tic stimulation, supporting-cell elements also can be directly the complete absence of hair cells and their corresponding
affected. Depending on the physical attributes of the exposure nerve fibers (the much lighter adjacent area) can be noted.
stimulus—such as time-varying characteristics or the intensity, Figure 152-3, B, graphically reconstructs the histopathologic
frequency, spectral content, duration, or schedule—noise can features of this cochlea as a cytocochleogram by depicting the
cause damage to hair cells that ranges from total destruction number of remaining hair cells, in the form of percentages,
to effects evident only in the ultrastructure of specialized sub- averaged over 1-mm sections. A typical finding in individuals
cellular regions (e.g., the fusing or bending of the individual exposed to the occupational noise exemplified in this case is
cilia that make up the stereociliary bundle). Whenever degen- the almost symmetric pattern of degeneration observed for the
erative processes or structural modifications to the cochlea two ears. The inset at the top right of the cytocochleogram
reach a significant level, an associated reduction in hearing shows the patient’s audiogram obtained about 1 year before his
capability can be detected. death, which shows the severity of the anatomic damage in
Johnsson and Hawkins9 were among the first investigators to functional terms by revealing an abrupt hearing loss for test
describe the typical patterns of cochlear injury for humans with frequencies below 2 kHz.
chronic exposure to different types of loud sound. The photo- Examination of human temporal bone specimens by other
micrograph of the cochlear tissue in Figure 152-3, A, depicts laboratories10 has yielded documentation of the progressive
some common histopathologic consequences of NIHL for a stages of noise damage as depicted by epidemiologic data such
patient with a lengthy history of industrial noise exposure. The as those of Figure 152-2. First, in the predictable sequence of
authors reported that the 50-year-old patient had worked inter- events, a small region of hair cell and nerve fiber degeneration
mittently over a 5- to 6-year period in an automotive body- appears bilaterally at a cochlear region that corresponds to the
stamping plant and that he had a long history of using 4-kHz notch. Typically, these discrete lesions gradually grow in
recreational firearms. The sharp transition in the basal end a basilar direction (i.e., toward the high-frequency extent of the
(following the uncoiling to the right) from the normal-looking cochlea) to involve a greater portion of the organ of Corti.
organ of Corti (a darkish stripe that corresponds to the region Finally, as exposure to noise continues over years, the remain-
ing sensory and neural elements in the basal end of the cochlea
are destroyed, which results in an abrupt loss of mid- to high-
frequency hearing, such as that depicted by the clinical audio-
gram inset shown in Figure 152-3, B.
RESEARCH ON NOISE-INDUCED
HEARING LOSS
Scientific interest in the damaging effects of excessive sound
on hearing has a long history for many reasons. First, the exper-
imental strategy of exposing animals to noise and examining
their ears for the sites of the resulting acoustic injury was used
in the past as the independent variable in establishing some of
our basic knowledge about hearing. Particularly, with the use
of intense tones as the damaging agent, frequency information
that relates physical distance along the basilar membrane to the
A “best” frequency of the injured region provided an initial basis
for understanding the tonotopicity of the cochlea and the
100 central projections of such frequency-related information.11
% Hair cells remaining
kHz
0.25 0.5 1 2 4 8
90
0 Additionally, noise-damage strategies have been used to con-
80
70
20
tribute to our understanding of the function of IHCs and OHCs
dB
40
60 60
80
by permitting differences in their central terminations in the
50 100
ventral and dorsal cochlear nuclei to be distinguished.12
40
30 Although useful as an analytic strategy, the major impetus
20 behind the more contemporary interest in the effects of noise
10
Nerve fibers
on hearing originates from a desire to understand the funda-
0
mental processes by which exposure to loud sound leads to
Apex 30 25 20 15 10 5 Base acoustic injury. The reward of achieving an appreciation of the
B Length of basilar membrane from basal end (mm) basic processes that underlie NIHL lies in the ability to prevent,
FIGURE 152-3. A, Low-power photomicrograph of a soft surface prepara-
or at least predict, an individual’s susceptibility to PTS or
tion of organ of Corti from the left cochlea of a 50-year-old man exposed perhaps even to initiate regeneration of damaged or lost critical
extensively to occupational noise shows a pattern of abrupt degeneration of cellular components, which would eventually lead to the recov-
the basal region. A small patch of organ of Corti (arrow) remains near the ery of hearing.
basal end. B, Modified cytocochleograms for two ears, along with an inset The research literature on the effects of noise on hearing
audiogram measured 1 year earlier, shows nerve fiber degeneration and a and on the anatomic elements of the ear is voluminous. Early
sharp pattern of hair cell degeneration expressed as percentage remaining experiments performed more than 70 years ago were straight-
per millimeter of length of basilar membrane measured from the basal end. forward anatomic studies based on the strategy of exposing
Note relative symmetry of corresponding abrupt, high-frequency loss of various animal models to intense noises, followed by a general
cochlear elements. Separate curves represent inner hair cells (solid lines) and
outer hair cells (dashed lines) averaged over three rows of outer hair cells
description of the resulting histopathology at the cellular level.
for left (X) and right (O) ears. Yellow horizontal line along the abscissa indicates More modern noise studies in animal models have attempted
presence of nerve fibers in osseous spiral lamina. (Modified from Johnsson LG, to establish a structure/function relationship between noise-
Hawkins JE. Degeneration patterns in human ears exposed to noise. Ann Otol induced morphologic damage and the inability to detect acous-
Rhinol Laryngol 1976;85:725-739.) tic signals.
152 | NOISE-INDUCED HEARING LOSS 2349
In this extensive literature, a great disparity in the experi- loss of hair cells through oncosis or apoptosis sometimes occurs
mental findings to relate the effects of missing hair cells to the in noise-injured ears. However, a newly defined third death
corresponding hearing sensitivity is frequently apparent. Such pathway—associated with the lack of a basolateral plasma mem-
contrasting findings and the confusion they have caused are brane, cellular debris arranged in the shape of an intact OHC,
related to numerous confounding variables, which include and a nucleus deficient in nucleoplasm—is more commonly
poor analysis of the problem (e.g., different durations or recov- observed in noise-damaged OHCs.22
ery intervals, exposure frequencies, or bandwidths); lack of
understanding of the limitations of applied functional methods, NEW KNOWLEDGE ABOUT
such as auditory evoked potential or psychoacoustic hearing
tests, and anatomic techniques; and use of animals of unrelated CELLULAR AND MOLECULAR
ages with unknown histories, which could include animals with MECHANISMS OF NOISE-INDUCED
prior exposure to intense sound and/or ototoxic medications.
Also, such disparate studies typically exposed animal subjects
HEARING LOSS
to a single noise at levels much greater than 100 dB SPL in an It has long been known that NIHL is associated with cochlear
attempt to mimic, within a relatively brief study interval, damage damage that initially involves the loss of sensory OHCs. Never-
patterns that develop in humans from intermittent exposure to theless, recently it has been shown that moderate sound over-
much less intense noises over many years. Consequently, stimulation can also produce a more proximal injury that
although early noise studies showed that the longer an animal involves a rapid and irreversible loss of cochlear nerve termi-
was exposed to extreme levels of sound, the greater the result- nals on IHCs, which is followed by a subsequent gradual degen-
ing cochlear injury, they contributed little to our knowledge eration of spiral ganglion cells in the presence of a full recovery
concerning how NIHL develops in humans who work for long of cochlear thresholds and no permanent loss of IHCs and
periods in noisy work settings. OHCs.23,24 Although these new histopathologic findings about
In contrast, research conducted over the past few decades the noise-damaged inner ear are ground breaking, the research
has made use of more realistic experimental protocols that frontiers that currently promise to provide fresh insights into
incorporate intermittent exposure stimuli of intensities and the fundamental basis of NIHL, and eventually into the devel-
durations designed to approximate the effects of a working opment of a cure for this disorder, consist of hair cell regenera-
lifetime of exposure to occupational noise. Additionally, in tion and/or repair, “training” protocols that target the
general, more recent studies have been developed sequentially cochlear-efferent system to make hair cells more resistant, the
within a program of research so that a thorough understanding use of protective agents and strategies before and after noise
of a particular effect is achieved. exposure, and understanding the genetic basis of sus ceptibility
and resistance to the adverse effects of sound overexposure.
0 0
DPOAE level (dB SPL)
–10 –10
–20 –20
–30 –30
–40 –40
–50 –50
5 6 7 8 910 20 30 40 50 5 6 7 8 910 20 30 40 50
A Frequency (kHz) B Frequency (kHz)
FIGURE 152-4. A and B, Mean difference distortion product graph—that is, preexposure distortion product otoacoustic emission (DPOAE) levels minus
postexposure DPOAE levels—for 65-dB sound-pressure level (SPL) primary tones that compare effects of an extreme 8-hour exposure with a 10-kHz octave
band of noise (shaded region from approximately 7 to 14 kHz) at 105 dB SPL for 2-month-old CBA mice (A) compared with 2-month-old MOLF mice (B).
Bold horizontal dotted line at 0 indicates no change between preexposure baseline DPOAE levels and postexposure counterparts measured at 2 days (red
circles), 1 week (open yellow circles), and 2 weeks (×) after overexposure. Note the only minor recovery of DPOAEs for CBA mice between 2 days and 2
weeks after exposure, especially for frequencies below 30 kHz. In contrast, DPOAEs for MOLF mice essentially returned to baseline values. Although not
shown here, immediately after exposure, both strains exhibited essentially no DPOAEs greater than approximately 10 kHz. Noise-floor differences (dashed
lines without symbols in lower portion of plots) represent preexposure DPOAEs subtracted from postexposure noise floors to indicate maximum possible
DPOAE loss. Vertical bars represent ±1 standard deviation for 2 days after exposure. CBA, n = 10, 19 ears; MOLF, n = 8, 15 ears.
2352 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
octave-band noise centered at 10 kHz) on control CBA and determine effective health criteria for controlling such noises.
MOLF mice at 2 months of age.61 Although CBA mice showed Most notably, a great disadvantage in gaining a more complete
only an expected minor recovery of DPOAEs to baseline levels understanding of NIHL in humans is that no more recent
at 2 days and 1 and 2 weeks postexposure, MOLF DPOAEs studies have been done of the hearing status of contemporary
essentially returned to their preexposure levels by 1 to 2 weeks workers, at least not in the countries of North America and
after exposure. In combination, findings in inbred mouse Europe. Reliance on data collected 30 or more years ago, such
models of NIHL provide the basis for applying suitable molecu- as those illustrated in Figure 152-2,8 has likely resulted in under-
lar techniques that permit the mapping of an NIHL gene to estimates of the amount of hearing loss that is due to occupa-
specific chromosomal loci (e.g., identifying differentially tional noise, especially for individuals with intermittent or
expressed genes using DNA microarrays or suppressive subtrac- impulsive noise exposures.
tive hybridization). Successful identification of this gene, and
perhaps its related modifier genes, would have great implica-
tions for developing a diagnostic indicator of the susceptibility EARLY DETECTION OF
of a particular human ear to the adverse effects of sound
overexposure.
NOISE-INDUCED HEARING LOSS
Genetic association studies on oxidative stress genes have One aspect of NIHL that has not received a great deal of
identified the first heritable factors that likely influence an research attention is the development of more sensitive mea-
individual’s susceptibility to NIHL. Konings and coworkers62 sures capable of detecting small, acoustically induced injuries
investigated whether variations in the form of single nucleotide to the organ of Corti (i.e., the beginning stages of NIHL) so
polymorphisms (SNPs) of the catalase gene (CAT), one of the that individuals vulnerable to the long-term damage caused by
genes involved in oxidative stress, influenced noise susceptibil- continuous exposure can be identified. In recent years, evi-
ity. By comparing audiometric data and DNA samples from the dence has been accumulating that the routine audiometric
10% most susceptible and 10% most resistant Swedish and testing of behavioral thresholds to pure tones at octave intervals
Polish noise-exposed workers, significant interactions were does not meet this need, because by the time such a loss is
observed between noise-exposure levels and several SNPs in identified using methods that test behavioral hearing sensitivity,
both populations. These findings indicate that CAT is poten- permanent cochlear damage has already occurred. Many
tially a noise susceptibility gene. Further research by these threshold and suprathreshold psychoacoustic tests to detect
investigators identified hundreds of common point mutations subtle deteriorations in hearing acuity, including psychophysi-
in the genome as susceptibility alleles for genes known to play cal tuning curves and frequency discrimination tasks, either
functional and/or morphologic roles in the cochlea. To date, have not proved to be of general usefulness or have proved to
the most promising results were obtained for genes involved in be too cumbersome methodologically to implement in a clini-
potassium recycling,63 heat shock protein 70,64 protocadherin cal or workplace setting because of the limitations associated
15, and myosin 14.65 With further development of high- with the necessarily brief evaluation periods.
throughput genotyping methods and expansion of SNP data- The diagnostic technique that incorporates OAEs is ideal
bases, the identification of NIHL susceptibility genes promises for assessing the normality of cochlear processing in ears sus-
to lead to genetic tests that identify at-risk individuals and pected of being overstimulated by loud sound because of their
permit personalized gene therapy if necessary. well-recognized sensitivity to the OHC class of receptor cell,
Although the findings of the more recent studies in particu- which is primarily involved in the initial stages of NIHL. The
lar have resulted in advancing our scientific knowledge of the functional status of OHCs in established instances of NIHL has
sound-damage process, many major empiric issues remain to been well described in many more recent studies of the practi-
be satisfied. These issues include development of low-cost tech- cal applicability of evoked OAE testing in the audiology clinic.66
nical methods for controlling noise at the source, physical pro- Numerous reports in the literature have shown that in groups
tection of individuals from excessive exposure, identification of of noise-exposed humans who have been followed serially,
individuals who are in the early stages of NIHL, prediction of reductions in OAE levels are more sensitive than pure tone
the degree of risk from potentially hazardous noises, and deter- audiometric thresholds in detecting the early states of perma-
mination of whether particular individuals or ears already nent noise-induced cochlear damage.67-69 In these studies,
damaged by noise are more susceptible to injury. decreases in emission magnitudes were measured in the
Ethical issues prevent the deliberate exposure of human absence of any change in the corresponding audiometric
subjects to extreme noises as a means of studying NIHL threshold frequencies.
experimentally. However, the complexity of measurements Figure 152-5 illustrates the ability of the two major types of
demanded by the alternate cross-sectional study design that evoked OAEs—the DPOAE, in its graphic, DP-gram form that
describes the hearing of humans exposed occupationally is depicts emission level as a function of the f2 test frequency
considerable. These difficulties include differences inherent in (lower left in Fig. 152-5), and the TEOAE, elicited by clicks, in
the population (e.g., race, gender, presence of ear disease); its spectral form (right plots in Fig. 152-5)—to describe the
problems involved in controlling concomitant exposure to non- configuration of a developing NIHL. The 43-year-old woman
occupational noise or past exposure history; and technical in this example had participated in recreational rifle shooting
problems with the descriptive techniques themselves, which for 3 years before OAE testing, and she claimed to have worn
range from variability in audiometric measures to difficulties in protective headphone devices continuously during this period.
performing valid measurements of the noise environment This right-shouldered shooter came to the otology clinic with
itself. Because of the complicated experimental designs complaints of hearing loss, tinnitus, muffled hearing, and dif-
demanded by all of these controls, few faultless clinical, epide- ficulty in understanding speech in background noise. It is clear
miologic, or experimental studies can be found of the effects from the test results that the magnitude and frequency extents
of excessive noise on hearing in humans. of the TEOAEs reflected the pattern of normal hearing illus-
The importance of performing longitudinal field studies trated by the clinical audiogram at the top left of Figure 152-5.
of communities or particular population segments—such as The DP-gram at lower left clearly shows abnormal activity:
elderly individuals, children, and chronically ill individuals— the right ear (yellow circles) shows below-average response
who have been habitually exposed to loud environmental levels, and the left ear (red circles) exhibits a significant
noises produced by road traffic or aircraft is obvious to reduction in emitted responses for frequencies above 3 kHz.
152 | NOISE-INDUCED HEARING LOSS 2353
43 F TEOAE-R
–10 20
0 15 Repro by frequency
TEOAE-L
20 20
Repro by frequency
15 15
1 k 2 k 3 k 4 k 5 k (Hz)
DPOAE level (dB SPL)
FIGURE 152-5. Early detection of noise-induced hearing loss in a 43-year-old rifle shooter who complained of muffled hearing, tinnitus, and difficulty hearing
speech in background noise. Note normal pure tone audiogram bilaterally (top left). In corresponding distortion product (DP) graphs (lower left), greater
functional loss seen in the left ear (red circles) compared with the right ear (yellow circles), particularly for frequencies above 3 kHz, which was due to protec-
tive head shadow effect for this right-handed woman. Emission levels for 2f1-f2 distortion product otoacoustic emissions (DPOAEs) are plotted in the bottom
left in response to 75-dB sound pressure level primary tones as a function of the f2 eliciting primary tone. Spectral plots shown on the right are for the transient
(click) evoked otoacoustic emission (TEOAEs). Emitted response (open area) for the better functioning right (R) ear was more prominently distributed above
the related noise floor (shaded area) than were emissions for the left (L) ear for frequencies above 3 kHz. In addition, higher reproducibility (repro) values
for the right ear (top right of each plot) reflected its more robust activity levels. In the DPOAE plot, variability of emission level (± 1 standard deviation) in
normal-hearing ears is represented by the bold dashed lines in the top portion. Similar variability of the related noise floor is indicated by pair of dashed lines
along the bottom of the plot. ANSI, American National Standards Institute.
True to classic principles, the left ear, which was exposed more surviving OHCs in an impaired ear based on tests of OAEs, and
to the muzzle end of the gun, went unprotected by the head particularly the DP-gram, can assist the clinician in achieving
shadow effect. This example of the ability of evoked OAEs to an optimal match between the patterns of hearing-aid amplifi-
detect cochlear pathology caused by noise exposure attests to cation and an NIHL.
the potential usefulness of OAE procedures in identifying Also, especially important to hearing conservation is the
the primary site of pathology associated with hearing com- correct identification of individuals who exhibit pseudohypacu-
plaints secondary to a sensorineural loss and in monitoring the sis and seek monetary compensation for a purported work-
development of potential hearing impairments in hearing- related hearing impairment. An example of this application is
conservation programs. illustrated in Figure 152-8 for a 42-year-old man who was
Other evidence that supports the usefulness of evoked OAEs referred to the audiology clinic for assessment by a local work-
in examining patients with noise damage is illustrated in Figures er’s compensation committee. In this case, the patient claimed
152-6 and 152-7. In Figure 152-6, the results of OAE testing in to have poor hearing caused by on-the-job exposure to intense
a 21-year-old man who had just completed 3 years of military noise to the extent that he could not hear frequencies greater
service in the infantry show the precision of DPOAEs in particu- than about 4 kHz. According to the OAE findings, it is likely
lar in tracking the asymmetric pattern of hearing loss for this that the patient, a wood-lathe operator, had an asymmetric
left-handed shooter. Additionally, the OAE findings presented noise-related hearing loss that was worse in the ear that was
in Figure 152-7 show the ability of evoked emissions to reflect more exposed to the tool; in this case, the right ear. However,
accurately the magnitude and frequency extent of the more based on the type of hearing-loss data presented in Figure
severe but essentially symmetric hearing loss experienced by a 152-1, A, for an industrial worker, it is unlikely that lathe-related
49-year-old man who had been working for more than 20 years noises could cause such a profound deafness for frequencies
in a fish cannery. above 4 kHz. Additionally, the normal levels of DPOAEs and
Both examples attest to several other benefits of OAE testing TEOAEs for frequencies below 2 kHz do not support the
in NIHL patients, including hearing aid fitting and identifying patient’s claim that the elevated audiometric hearing levels
nonorganic hearing loss. A significant advantage of the digital over the low-frequency to midfrequency regions were due to
hearing aid over the older analog models is its ability to provide noise overexposure. Findings such as these support the use of
frequency-specific amplification based on the configuration of objective tests of evoked OAEs in determining the authenticity
a particular patient’s hearing loss. Knowledge of the pattern of of compensation claims for work-related hearing problems.
21 M TEOAE-R
–10 20
0 15 Repro by frequency
DPOAE TEOAE-L
20 20
15 15 Repro by frequency
1 k 2 k 3 k 4 k 5 k (Hz)
10
DPOAE level (dB SPL)
FIGURE 152-6. Early stages of noise-induced hearing loss in a 21-year-old veteran who had just completed a 3-year tour of duty. For this left-handed infan-
tryman, greater hearing loss is evident from approximately 3 to 4 kHz for the right ear (yellow circles) depicted in the audiogram (top left). In the corresponding
distortion production (DP) graph (bottom left), abnormally low levels of distortion product otoacoustic emission (DPOAE) activity are apparent for frequencies
greater than 1.5 kHz, which were poorer for the right ear. Transient (click) evoked otoacoustic emission (TEOAE) spectra on the right also show poorer
emitted responses from 1 to 2 kHz and show lower reproducibility (repro) values for the right (R) ear. Above 2 kHz, no emitted response is seen for either
ear. In the DPOAE plot, variability of emission level (± 1 standard deviation) in normal-hearing ears is represented by the bold dashed lines in the top portion.
Similar variability of the related noise floor is indicated by pair of dashed lines along the bottom of the plot. ANSI, American National Standards Institute.
49 M TEOAE-R
–10 20
0 15 Repro by frequency
Hearing level (dB) (ANSI 69)
10 1 k 2 k 3 k 4 k 5 k (Hz)
TEOAE level (dB SPL)
20 10 99 93 0 0 0 (%)
30 5
40 0
50
–5
60 Right ear
70 Left ear –10
80 –15
90 –20
100
110 –25
120 –30
250 500 1K 2K 4K 8K 0 1 2 3 4 5 6
Frequency (kHz) Frequency (kHz)
TEOAE-L
20 20
15 15 Repro by frequency
1 k 2 k 3 k 4 k 5 k (Hz)
10
DPOAE level (dB SPL)
10 96 88 0 0 0 (%)
5 5
0
0
–5
–5
–10
–10
–15
–20 –15
–25 –20
–30 –25
–35 –30
0.5 0.7 1 2 3 4 5 6 8 10 0 1 2 3 4 5 6
f2 frequency (kHz) Frequency (kHz)
FIGURE 152-7. More advanced stage of noise-induced hearing loss in a 49-year-old man who had worked for more than 20 years in a fish cannery. Audio-
grams at top left exhibit symmetric hearing loss for frequencies above 2 kHz. Distortion product otoacoustic emission (DPOAE) plots indicate a symmetric
pattern of dysfunction in a similar manner. Transient evoked otoacoustic emission (TEOAE) spectra on the right also show a lack of emitted responses for
frequencies above 2 kHz. In the DPOAE plot, variability of emission level (± 1 standard deviation) in normal-hearing ears is represented by the bold dashed
lines in the top portion. Similar variability of the related noise floor is indicated by pair of dashed lines along the bottom of the plot. ANSI, American National
Standards Institute; repro, reproducibility.
152 | NOISE-INDUCED HEARING LOSS 2355
42 M TEOAE-R
–10 20
0 15 Repro by frequency
TEOAE-L
20 20
Repro by frequency
15 15
1 k 2 k 3 k 4 k 5 k (Hz)
DPOAE level (dB SPL)
FIGURE 152-8. A possible case of pseudohypacusis in a 42-year-old factory worker who operated a wood lathe. Audiograms at top left show a relatively
flat hearing loss up to approximately 4 kHz. For frequencies above 4 kHz, the patient claimed that test tones were inaudible. Distortion product otoacoustic
emission (DPOAE) graphs show normal-appearing function up to approximately 3 to 4 kHz, when reductions in distortion product otoacoustic emission
(DPOAE) levels are evident; this occurs initially for the right ear (yellow circles), which was more exposed to the lathe. Transient evoked otoacoustic emission
(TEOAE) spectral plots on the right also show poorer click-evoked OAEs for the right ear. Together, normal levels of evoked emissions for frequencies below
3 kHz and an asymmetric pattern of reduction in OAEs suggest that both exposure to noise and aging contributed to decreased levels of activity for frequen-
cies above 3 kHz. In the DPOAE plot, variability of emission level (± 1 standard deviation) in normal-hearing ears is represented by the bold dashed lines in
the top portion. Similar variability of the related noise floor is indicated by pair of dashed lines along the bottom of the plot. ANSI, American National Standards
Institute; repro, reproducibility.
reported. In a series of experimental studies in the rat model,
INTERACTIVE EFFECTS Fechter and colleagues78 discovered that the simultaneous
It is well established that noise in combination with certain exposure to noise and the environmental pollutants carbon
chemical agents produces stronger reactions than each stimu- monoxide or hydrogen cyanide produced more permanent
lus applied singly. The four major categories of ototoxic drugs hearing loss at the high frequencies than the sum of the losses
are 1) aminoglycoside antibiotics, 2) platinum derivative anti- produced by each agent administered alone. Various other
tumor agents, 3) loop diuretics, and 4) salicylates. The latter chemicals present in the environment as commercial products
two drug classes cause reversible effects, whereas aminoglyco- or chemical intermediaries and contaminants—such as the
sides and platinum derivatives cause permanent damage to the organic solvents toluene and hexane, the pollutants methyl
inner ear and to hearing. Many laboratories have established mercury and lead acetate, and the organic compounds trimeth-
in animal models that kanamycin, neomycin, or amikacin yltin chloride and styrene, used in the manufacture of plastics
applied in combination with different types of noise produces and polyurethane foam and rubber—have been identified as
a marked potentiating interaction.70,71 Other studies of the tem- potent ototoxic agents that can interact synergistically with
poral aspects of interactive effects indicate that the degree of excessive noise. The ototoxicity of environmental agents such
potentiating interaction is the same, whether the drug is given as metals, solvents, and asphyxiants and their interaction with
concurrently with the noise exposure or several months later.72 noise are issues that have received a great deal of attention in
Other evidence from humans73 and from controlled laboratory the literature.79 Although many of these environmental toxi-
research in a rat model74 indicates that additional loss may cants have been associated with direct injury to inner ear struc-
occur when subjects are treated with aspirin and exposed to tures, possible additional anatomic damage to more central
noise concomitantly, although other findings infer that the auditory pathways is also very likely.
combination of salicylate plus noise does not produce greater
effects than noise alone.75 Finally, experimental evidence in OTHER ADVERSE EFFECTS
several animal models showed that the heavy-metal antineoplas-
tic agent cis-diamminedichloroplatinum, commonly known as
CAUSED BY NOISE
cisplatin, significantly increased the amount of hearing and Damage to the vestibular system is a potential problem with
sensory cell losses from exposure to noise.76,77 noise, because balance receptors are coupled physically with
In recent years, the interactive effects of noise with chemical the auditory receptors; that is, they share the membranous
agents common to industry and the environment have been labyrinth. In addition to the anatomic proximity of the
2356 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
vestibular labyrinth to the acoustic-energy delivery system, the helped attenuate the noise sufficiently to prevent TTS in most
great similarity in cochlear and vestibular hair cell ultrastruc- patients undergoing MRI evaluation. The use of the higher
ture, and the common arterial blood supply of the cochlea and gradient fields that are applied at faster slew rates, which are
vestibular end organs via the same end artery support the pos- used in current functional MRI, demands increased hearing-
sibility of vestibular damage associated with NIHL. Theoreti- protection systems to minimize the risk of noise trauma.91
cally, the limiting membrane, the membranous partition that Other nonauditory problems concern the general bother-
separates the utricle and semicircular canals from the rest of some or fatiguing effects of noise, which may lead to nonspe-
the vestibule, protects most vestibular sensory cells from the cific health disorders because of interference with the restorative
adverse effects of intensive stapes vibration. processes associated with rest and sleep. In conditions of
Nevertheless, some studies of industrial workers and military chronic exposure, noise is considered to act as a biologic
personnel have reported a high occurrence of vestibular symp- stressor that can lead to prolonged activation of the autonomic
toms and signs and complaints of dysequilibrium.80 Such simple nervous system and the pituitary-adrenal complex, which
correlations between noise exposure and vestibular distur- results in general health impairment.92 Noise has also been
bances are highly controversial because of the typical low inci- related to disorders that involve gastrointestinal motility, such
dence of clinical vestibular symptoms associated with gradually as peptic ulcers,93 and, as noted earlier, to circulatory problems
developing NIHL.81 A study of military personnel with mild to such as hypertension.94 Certain types of noise can be annoying
severe NIHL using sophisticated vestibular testing in the form and can lead to emotional unrest.95 Finally, noise can have a
of a computerized rotatory-chair system along with routine elec- deleterious effect on task performance, especially if speech
tronystagmographic measures of caloric responses showed understanding is involved.96 Generally, data relevant to the non-
more direct correlations between hearing loss and vestibular auditory effects of noise tend to be inconclusive, because the
upset.82 In combination, the results showed a symmetric, cen- variables are difficult to identify and isolate for objective study.
trally compensated decrease in the response of the vestibular
end organ associated with a symmetric hearing loss. A more
recent study83 that combined caloric testing with vestibular-
LEGAL ISSUES
evoked myogenic potentials in patients with chronic NIHL One practical issue that the lawmakers of industrialized societ-
showed absent or delayed electrophysiologic potentials, which ies have had to consider concerns the balancing of two conflict-
indicates that the vestibular system, especially the sacculocollic ing goals: How can laborers be protected against the hazards
reflex pathway, was damaged in addition to the cochlear portion of the workplace without placing an enormous financial burden
of the inner ear. on society, either by satisfying compensatory obligations or by
Together these findings imply that a subclinical, well- preventing such work-related effects through costly engineer-
compensated malfunction of the vestibular system is associated ing modifications of the industrial process? Over the years,
with NIHL. Beyond the medicolegal implications is the daunt- concerns about protecting the public and the work force from
ing possibility that a currently asymptomatic vestibulopathy environmental insults such as noise have been written into a
produced by noise exposure might progress to a disturbing combination of law and governmental regulations. Federal,
vertigo under certain environmental conditions.84 state, and community legislative and regulatory actions are con-
Considerable interest has been shown recently in the inter- tinually being reviewed and altered. Although the purpose of
action of vibration and noise, which are common cofactors in this review is not to detail such legal controls, regarding their
the workplace. Although most research shows that vibration historic development or current status, a brief discussion is
alone does not affect hearing, the results of epidemiologic appropriate. For a lucid analysis of governmental statutes
studies in humans and controlled laboratory studies in animal and regulations pertaining to noise control, see the review by
models indicate a synergistic interaction of concomitant vibra- Dobie.7
tion (either whole body or hand-arm vibration) and noise that The major difficulty encountered in composing regulatory
results in an increase in the degree of NIHL.85 control has been in defining in practical terms what constitutes
Exposure to infrafrequency and ultrafrequency sounds a hazardous noise. One popular approach toward expressing
outside of the human hearing range has also been studied. the danger of a particular noise by a simple number is embod-
Infrasonic or vibratory stimuli are defined as sounds in the range ied in the equal-energy principle,97 which assumes that perma-
of 0.1 to 20 Hz, and there are no known instances in which nent damage to hearing is related to total sound energy, a
infrasound alone clearly caused permanent damage to the product of the noise level in dBA and the duration of exposure.
human cochlea. However, studies in the chinchilla model One tenet of this postulate is that an equal amount of noise
showed that the presence of intense sounds in the audible energy causes an equal amount of hearing loss.
frequency range along with simultaneous infrasound increases Burns and Robinson,98 who investigated the hearing loss of
cochlear damage.86 thousands of industrial workers, concluded that the equal-
In contrast, earlier reports have shown detrimental effects energy principle could be applied to determine daily exposure
of microwaves on hearing.87 The thermal properties of ultra- doses, because hearing loss caused by work-related noise expo-
high-frequency sound waves in the gigahertz range, rather than sure seemed to be a simple function of noise energy. Atherley
mechanical energy, seem to have produced these adverse and Martin99 extended the concept to impulse noise by applica-
experimental effects. Overall, it is probably safe to conclude tion of the equivalent continuous sound level (Leq) principle.
that the auditory system’s built-in middle ear filter limits the The Leq is defined as the A-weighted level of a continuous,
extreme frequencies of sound that are hazardous to the steady sound that produces, in a specified interval, an exposure
inner ear.88 that has the same total acoustic energy as that of an actual time-
Magnetic resonance imaging (MRI) systems have acoustic varying sound over the identical interval.100 In other words, if
noise associated with their clinical scanning actions. For spe- one sound contains twice as much energy as a second sound
cific protocols that incorporate faster and noisier sequences, but lasts half as long, both sounds would be characterized by
peaks of 120 to 130 dBA have been measured, particularly the same equivalent sound level. The Leq concept holds that
during high-speed echoplanar imaging.89 Early on, Brummett these two exposures produce the same damage to the ear.
and associates90 showed that sounds generated by MRI are suf- Although notions such as the equal-energy principle have
ficiently intense to cause some TTS in a significant number of proved useful in the practical definition of noise-control vari-
patients. However, their results also showed that earplugs ables, their validity has been more difficult to establish. Over
152 | NOISE-INDUCED HEARING LOSS 2357
the years, experimental evidence has been conflicting in that sure to overly intense sounds, particularly with respect to chil-
it has tended to be either for or against the equal-energy rule. dren and adolescents.
Generally, it seems that the principle cannot be generalized Although a known prevention exists, for fiscal and technical
indiscriminantly to stimuli throughout the range of noise reasons, it is unlikely that the general loud sound levels of our
parameters, because exposure to impulse or intermittent noises environment will be reduced. As previously noted, whereas
may lead to more or less degeneration in the organ of Corti clinical trials on the safety and efficacy of certain antioxidant
than would be expected according to the equal-energy princi- compounds to prevent or reverse NIHL are currently in prog-
ple. However, some aspect of the equal-energy concept has ress, to date no proven treatment or cure for noise damage
been adopted by most industrialized nations, including the exists. Consequently, detecting the early stages of NIHL is
United States, as a means of measuring the potential hazard of important to prevent further injury to the crucial sensory-cell
a particular noise exposure. Present regulations use a 5-dB receptors of the organ of Corti.
exchange rate, along with a 90-dBA time-weighted average, to The role of the otolaryngologist is first to identify the cause
define the permissible exposure limit over an 8-hour workday. and extent of a reported hearing loss through systematic
Consequently, exposure to 90 dBA for 8 hours is equivalent in medical and audiometric evaluation. As part of the course of
sound-energy terms to an exposure of 95 dBA for half that otologic management, the patient should be educated about
time. For more complete particulars, see Table G-16 at www the hazards of noise and about preventive measures to preserve
.osha.gov, which details the permissible noise exposures devel- remaining hearing. The physician should make basic decisions
oped by the U.S. Occupational Safety and Health Administra- about the appropriate aural rehabilitative course of action.
tion (OSHA).101 Finally, the otolaryngologist should be sensitive to any emo-
Currently, industries that employ laborers who work where tional problems the patient may have in accepting a hearing
noise levels are greater than 85 dBA—except for farming impairment, which may be dealt with by counseling or special
and construction workers, who work in hard-to-control provisions.
environments—implement a hearing-conservation program Although NIHL is not medically or surgically treatable, it is
that consists of several components as dictated by OSHA.101 almost entirely preventable. Its prevention requires education,
First, preemployment audiometric assessment and annual engineering, and administrative controls and the proper use of
audiometric monitoring are required so that noise-induced hearing protection. One of the otolaryngologist’s most impor-
cochlear impairment can be detected before it becomes too tant contributions is in teaching the patient to guard against
severe. When a hearing loss is identified, the worker must be further losses.106 When recommending the use of personal
notified of the disorder and counseled about the use of per- hearing protectors or reinforcing compliance with ongoing
sonal hearing protectors. The second portion of the conserva- hearing-protection measures, the physician should be aware
tion program requires workers in areas of high noise levels (≥ that the commonly used personal protectors in the form of
85 dBA) to wear ear protectors and to participate in a noise inserted earplugs or earmuffs vary considerably in effectiveness
education program that informs the employee of the hazardous and produce an attenuation that is highly frequency depen-
effects and the correct fitting of personal protectors. An impor- dent.107 When sealed correctly into the ear canal, earplugs
tant part of hearing conservation is the otologic referral, which reduce the noise that reaches the middle ear by 15 to 30 dB,
is essential when in-plant audiometry has detected a substantial and they work best for the mid to higher frequency range (i.e.,
hearing loss. 2 to 5 kHz). Earmuffs are more effective protectors, especially
for frequencies between 500 Hz and 1 kHz, in which noise is
attenuated by 30 to 40 dB.108 In areas with extremely high noise
ROLE OF THE levels, earplugs do not afford sufficient protection, and indi-
viduals should be advised to wear both earplugs and earmuffs.
OTOLARYNGOLOGIST Also, sound energy associated with high noise levels may reach
Patients with NIHL constitute a notable portion of an otolar- the inner ear by passing through vibrating bone and tissues
yngologist’s patient population, particularly for otolaryngolo- adjacent to the ear. Bone-conduction and tissue-conduction
gists who are in private practice. With the increase in the thresholds set a practical limit to the possible attenuation pro-
population older than 65 years, the psychologic, economic, and vided by hearing-protection devices.
social impact of NIHL is still growing. Modern stereo systems A final point concerning the use of hearing protectors
in the form of personal music players or personal listening should be impressed on the patient. When data are considered
devices (PLDs) such as the MP3 and iPod systems have suffi- that relate the maximal protection in dBA to the percentage of
cient noise levels and listening durations to put consumers at time that the devices are worn, it is clear that hearing protectors
risk for developing NIHL.102 Because the number of young should be worn all the time, because if they are removed for
people subject to social noise exposure has tripled since the even a few minutes, their effective cumulative attenuation capa-
early 1980s,103 the widespread use of these devices among chil- bility is severely reduced. Removing hearing protection for only
dren and adolescents is a growing worry. Such concern is sup- 15 minutes of an 8-hour shift can cut protection efficacy in half.
ported by the finding uncovered in the audiometric database Similarly, a poorly fitting hearing protector does not prevent
of a large-scale national cross-sectional survey of an accelerating hearing loss.1
prevalence of sensorineural hearing loss in young adults 20 to In many states, compensation is available for occupational
29 years old.104 This growing prevalence of high-frequency NIHL, and physicians are sometimes required to serve as expert
hearing loss in young adults may relate to an increase in PLD witnesses as to the probable cause of a claimant’s hearing loss.
use. However, the outcome of at least one relevant experimen- Such medicolegal testimony requires that the patient undergo
tal study that used similarly aged subjects, who were systemati- careful otologic and audiometric evaluation to exclude hearing
cally exposed to music from an MP3 player at their preferred losses caused by unrelated ear disorders such as cerumen
listening levels, does not agree with this explanation.105 It is impaction, middle ear effusion, aging, genetic deficits, otoscle-
clear that to demonstrate whether exposure to PLDs as teenag- rosis, and an array of other ear diseases (e.g., Meniere disease,
ers influences hearing loss in old age, long-term longitudinal acoustic neuroma). To permit the reversible effects (i.e., TTS)
cohort studies are needed.103 In any case, otolaryngologists to recover, most states require a recovery time that ranges from
are expected to continue to be prominently involved with the 14 “quiet” hours to 24 weeks away from the occupational noise
problem of NIHL and leisure-time activities that involve expo- source before audiometric assessment. By ruling out organic
2358 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
disease, giving careful attention to the history of job-related and also know how much more we need to learn about basic ear
recreation-related noise exposure, and documenting the extent processes before such damage can be corrected. As our under-
and degree of involvement of both ears, an informed decision standing of the fundamental mechanisms involved in the
concerning any causal relationship between the noted disability process of NIHL progresses, developing a medical intervention
and the work environment can be made. to minimize permanent injury seems achievable. In the mean-
Hearing impairment is the medical term used to refer to the time, we know what needs to be done to prevent or check the
hearing level at which individuals begin to experience difficul- damage process. In light of the economic impracticability of
ties in everyday life. Hearing impairment manifests in practical noise control at the engineering or administrative level, espe-
terms, such as in a difficulty understanding speech. By the time cially in industry, it seems that education of the public about
an individual becomes aware of decreased speech intelligibility, the potential hazards of excessive sound and the beneficial use
considerable damage to the organ of Corti likely has occurred, of protective devices will be the major weapons against NIHL.
because speech reception is not altered greatly until a hearing Consequently, the educational role of otolaryngologists is para-
loss is more than 40 dB. The amount of loss at frequencies most mount for the conservation of hearing threatened by habitual
important to speech—at 2, 3, and 4 kHz—is used by OSHA101 as noise exposure.
a basis for calculating amounts of compensation, because NIHL
occurs initially at frequencies of 2 kHz or greater. The measure
of hearing impairment is called the hearing handicap, which is
ACKNOWLEDGMENTS
always based on the functional state of both ears. An official The authors acknowledge the long-time support of the National
guide devised by the American Academy of Otolaryngology– Institutes of Health (DC00613, DC03114) and the Veterans
Head and Neck Surgery109 provides a detailed explanation Administration (VA/RRD C7107R, C6212L) for their programs
and formula for evaluating and calculating an NIHL handicap. of research and Barden B. Stagner for conducting data analysis
Generally, the disability benefits of the U.S. Social Security and assisting with assembling the illustrations used here.
Administration110 are less generous than those for either mili-
tary veterans or typical worker’s compensation programs. For a complete list of references, see expertconsult.com.
Because of an increasingly noisy environment and the prob-
ability that many individuals will unwittingly sustain hearing
losses because of exposure to loud sounds at home, on the job, SUGGESTED READINGS
during recreational activity, and through misuse of PLDs, oto- Bao J, Hungerford M, Luxmore R, et al: Prophylactic and therapeutic
laryngologists and other health care professionals who deal functions of drug combinations against noise-induced hearing loss.
with hearing problems should educate the public about hearing Hear Res 304:33–40, 2013.
conservation. Such education is needed, especially for children Furman AC, Kujawa SG, Liberman MC: Noise-induce cochlear neu-
and adolescents, who, as noted previously, are regularly exposed ropathy is selective for fibers with low spontaneous rates. J Neuro-
physiol 110:577–586, 2013.
to the amplified music associated with personal music players, Gallun FJ, Diedesch AC, Kubli LR, et al: Performance on tests of central
dance clubs, and live concerts. A common image that can be auditory processing by individuals exposed to high-intensity blasts.
used to motivate the public and workers in industry to be aware J Rehabil Res Dev 49:1005–1025, 2012.
of the insidious damage of noise exposure is the grass meta- Gallun FJ, Lewis MS, Folmer RL, et al: Implications of blast exposure
phor that represents the sensory cells of the inner ear. Typically, for central auditory function: a review. J Rehabil Res Dev 49:1059–1074,
blades of grass straighten up after they have been bent by being 2012.
walked on. However, if the same patch is walked on day after Guthrie OW, Xu H: Noise exposure potentiates the subcellular distribu-
day, the grass eventually dies and leaves a bare spot; this is tion of nucleotide excision repair proteins within spiral ganglion
similar to what happens to the ear’s hair cells when assaulted neurons. Hear Res 294:21–30, 2012.
Konings A, Van Laer L, Van Camp G: Genetic studies on noise-induced
continually by high-intensity sounds. hearing loss: a review. Ear Hear 30:151–159, 2009.
The physician can be particularly effective in teaching indi- Le Prell CG, Dell S, Hensley B, et al: Digital music exposure reliably
viduals how to recognize the danger signals of potential NIHL, induces temporary threshold shift in normal-hearing human sub-
including the need to shout to be heard and the development jects. Ear Hear 33:e44–e58, 2012.
of the sensation of pain, muffled sound, and tinnitus.111,112 The Maison SF, Usubuchi H, Liberman MC: Efferent feedback minimizes
influence of otolaryngologists as hearing experts in the areas cochlear neuropathy from moderate noise exposure. J Neurosci 33:
of education and motivation can be an important force in pre- 5542–5552, 2013.
venting NIHL. National Institutes of Health: It’s a Noisy Planet: Protect Their Hearing.
Accessed September 3, 2013 at www.noisyplanet.nidcd.nih.gov.
Sliwinska-Kowalska M, Pawelczyk M: Contribution of genetic factors to
SUMMARY noise-induced hearing loss: a human studies review. Mutat Res 752:
61–65, 2013.
We know more now than ever about the workings of the human Verbeek JH, Kateman E, Morata TC, et al: Interventions to prevent
ear, including how it responds initially to excessive sound, and occupational noise-induced hearing loss. Cochrane Database Syst Rev
how it eventually fails to reverse the progressive damage. We 10:CD006396, 2012.
152 | NOISE-INDUCED HEARING LOSS 2358.e1
29. Tucci DL, Rubel EW: Physiologic status of regenerated hair cells
REFERENCES in the avian inner ear following aminoglycoside ototoxicity. Oto-
1. National Institute of Occupational Safety and Health. Available at laryngol Head Neck Surg 103:443–450, 1990.
www.cdc.gov/niosh/docs/2001-103. Accessed September 3, 2013. 30. Cotanche DA, Lee KH, Stone JS, et al: Hair cell regeneration in
2. United States Government Accountability Office, 2011. GAO the bird cochlea following noise damage or ototoxic drug damage:
Report No. 11-114. Hearing loss prevention: improvements to a review. Anat Embryol (Berl) 198:1–18, 1994.
DOD hearing conservation programs could lead to better 31. Zheng JL, Gao WQ: Overexpression of Math1 induces robust
outcomes. production of extra hair cells in postnatal rat inner ears. Nat
3. Occupational Safety and Health Administration, Department of Neurosci 3:480–586, 2000.
Labor: Occupational noise exposure: hearing conservation 32. Bermingham-McDonogh O, Rubel EW: Hair cell regeneration:
amendment. Fed Reg 46:4078–4179, 1981. winging our way towards a sound future. Curr Opin Neurobiol 13:
4. Nordmann AS, Bohne BA, Harding GW: Histopathological differ- 119–126, 2003.
ences between temporary and permanent threshold shift. Hear Res 33. Kawamoto K, Ishimoto S, Minoda R, et al: Math1 gene transfer
139:13–30, 2000. generates new cochlear hair cells in mature guinea pigs in vivo.
5. Helfer TM: Noise-induced hearing injuries, active component, J Neurosci 23:4395–4440, 2003.
U.S. Armed Forces, 2007-2010. MSMR 18:7–10, 2011. 34. Yang SM, Chen W, Guo WW, et al: Regeneration of stereocilia of
6. Helfer TM, Jordan NN, Lee RB, et al: Noise-induced hearing hair cells by forced Atoh1 expression in the adult mammalian
injury and comorbidities among postdeployment U.S. Army sol- cochlea. PLoS One 7:e46355, 2012.
diers: April 2003-2009. Am J Audiol 20:33–41, 2011. 35. Canlon B, Borg E, Flock A: Protection against noise trauma by
7. Dobie RA: Medical-Legal Evaluation of Hearing Loss. ed 2, Toronto, preexposure to a low level acoustic stimulus. Hear Res 34:197–200,
2001, Cengage Delmar Learning. 1988.
8. Taylor W, Lempert B, Pelmear P, et al: Noise levels and hearing 36. Franklin DJ, Lonsbury-Martin BL, Stagner BB, et al: Altered
thresholds in the drop forging industry. J Acoust Soc Am 76:807– susceptibility of 2f1-f2 acoustic-distortion products to the effects of
819, 1984. repeated noise exposure in rabbits. Hear Res 53:185–208, 1991.
9. Johnsson LG, Hawkins JE: Degeneration patterns in human ears 37. Miyakita T, Hellström PA, Frimanson E, et al: Effect of low level
exposed to noise. Ann Otol Rhinol Laryngol 85:725–739, 1976. acoustic stimulation on temporary threshold shift in young
10. McGill TJI, Schuknecht HF: Human cochlear changes in noise- humans. Hear Res 60:149–155, 1992.
induced hearing loss. Laryngoscope 86:1293–1302, 1976. 38. Kemp DT: Stimulated acoustic emissions from within the human
11. Ou HC, Harding GW, Bohne BA: An anatomically based auditory system. J Acoust Soc Am 64:1386–1391, 1978.
frequency-place map for the mouse cochlea. Hear Res 145:123– 39. Brownell WE: Outer hair cell electromotility and otoacoustic emis-
129, 2000. sions. Ear Hear 11:82–92, 1990.
12. Morest DK, Bohne BA: Noise-induced degeneration in the brain 40. Collet L, Kemp DT, Veuillet E, et al: Effect of contralateral audi-
and representation of inner and outer hair cells. Hear Res 9:145– tory stimuli on active cochlear micro-mechanical properties in
151, 1983. human subjects. Hear Res 43:251–261, 1990.
13. Caiazzo AJ, Tonndorf J: Ear canal resonance and temporary 41. Kim DO, Dorn PA, Neely ST, et al: Adaptation of distortion
threshold shift. Otolaryngology 86:820, 1978. product otoacoustic emission in human. J Assoc Res Otolaryngol 2:
14. Schuknecht HF: Pathology of the Ear, Cambridge, MA, 1974, 31–40, 2001.
Harvard University Press. 42. Maison SF, Liberman MC: Predicting vulnerability to acoustic
15. Wang Y, Hirose K, Liberman MC: Dynamics of noise-induced cel- injury with a noninvasive assay of olivocochlear reflex strength.
lular injury and repair in the mouse cochlea. J Assoc Res Otolaryngol J Neurosci 20:4701–4707, 2000.
3:248–268, 2002. 43. Luebke AE, Stagner BB, Martin GK, et al: Adaptation of distortion
16. Clark JA, Pickles JO: The effects of moderate and low levels of product otoacoustic emissions predicts susceptibility to acoustic
acoustic overstimulation on stereocilia and their tip links in the over-exposure in alert rabbits. J Acoust Soc Am 135:1941–1949, 2014.
guinea pig. Hear Res 99:119–128, 1996. 44. Desai A, Reed D, Cheyne A, et al: Absence of otoacoustic emis-
17. Liberman MC: Chronic ultrastructural changes in acoustic sions in subjects with normal audiometric thresholds implies
trauma: serial-section reconstruction of stereocilia and cuticular exposure to noise. Noise Health 1:58–65, 1999.
plates. Hear Res 26:65–88, 1987. 45. Canlon B, Agerman K, Dauman R, et al: Pharmacological strate-
18. Liberman MC, Dodds LW: Acute ultrastructural changes in acous- gies for preventing cochlear damage induced by noise trauma.
tic trauma: serial-section reconstruction of stereocilia and cuticu- Noise Health 1:13–23, 1998.
lar plates. Hear Res 26:45–64, 1987. 46. Kopke RD, Coleman JK, Liu J, et al: Candidate’s thesis: enhancing
19. Rzadzinska AK, Schneider ME, Davies C, et al: An actin molecular intrinsic cochlear stress defenses to reduce noise-induced hearing
treadmill and myosins maintain stereocilia functional architecture loss. Laryngoscope 112:1515–1532, 2002.
and self-renewal. J Cell Biol 164:887–897, 2004. 47. Kopke RD, Jackson RL, Coleman JK, et al: NAC for noise: from
20. Manor U, Kachar B: Dynamic length regulation of sensory stereo- the bench top to the clinic. Hear Res 226:114–125, 2007.
cilia. Semin Cell Dev Biol 19:502–510, 2008. 48. Bielefeld EC, Kopke RD, Jackson RL, et al: Noise protection with
21. Ahmad M, Bohne BA, Harding GW: An in vivo tracer study of N-acetyl-l-cysteine (NAC) using a variety of exposures, NAC doses,
noise-induced damage to the reticular lamina. Hear Res 175:82– and routes of administration. Acta Otolaryngol 127:914–919, 2007.
100, 2003. 49. Kramer S, Dreisbach L, Lockwood J, et al: Efficacy of the antioxi-
22. Bohne BA, Harding GW, Lee SC: Death pathways in noise- dant N-acetylcysteine (NAC) in protecting ears exposed to loud
damaged outer hair cells. Hear Res 223:61–70, 2007. music. J Am Acad Audiol 17:265–278, 2006.
23. Kujawa SG, Liberman MC: Adding insult to injury: cochlear nerve 50. Campbell K, Claussen A, Meech R, et al: D-methione (D-met)
degeneration after ‘temporary’ noise-induced hearing loss. J Neu- significantly rescues noise-induced hearing loss: timing studies.
rosci 29:14077–14085, 2009. Hear Res 282:138–144, 2011.
24. Lin HW, Furman AC, Kujawa SG, et al: Primary neural degenera- 51. D-Methione to Reduce Noise-Induced Hearing Loss (NIHL),
tion in the Guinea pig cochlea after reversible noise-induced NCT01345474. Available at ClinicalTrials.gov/ct2/show/
threshold shift. J Assoc Res Otolaryngol 12:605–616, 2011. NCT01345474. Accessed May 26, 2014.
25. Corwin JT, Cotanche DA: Regeneration of sensory hair cells after 52. Le Prell CG, Gagnon PM, Bennett DC, et al: Nutrient-enhanced
acoustic trauma. Science 240:1772–1774, 1988. diet reduces noise-induced damage to the inner ear and hearing
26. Ryals BM, Rubel EW: Hair cell regeneration after acoustic trauma loss. Transl Res 158:38–53, 2011.
in adult Coturnix quail. Science 240:1774–1776, 1988. 53. Le Prell CG, Dolan DF, Bennett DC, et al: Nutrient plasma levels
27. Cruz RM, Lambert PR, Rubel EW: Light microscopic evidence of achieved during treatment that reduces noise-induced hearing
hair cell regeneration after gentamicin toxicity in chick cochlea. loss. Transl Res 158:54–70, 2011.
Arch Otolaryngol Head Neck Surg 13:1058–1062, 1987. 54. Ward WD: General auditory effects of noise. Otolaryngol Clin North
28. McFadden EA, Saunders JC: Recovery of auditory function follow- Am 12:473–491, 1979.
ing intense sound exposure in the neonatal chick. Hear Res 41: 55. Barrenas ML, Lindgren F: The influence of eye colour on suscep-
205–215, 1989. tibility to TTS in humans. Br J Audiol 25:303–307, 1991.
2358.e2 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
107. Neitzel R, Somers S, Seixas N: Variability of real-world hearing 110. Social Security Administration: The Social Security Quick Calcula-
protector attenuation measurements. Ann Occup Hyg 50:679–691, tor page. Available at www.ssa.gov/OACT/quickcalc. Accessed
2006. September 2, 2013.
108. Giardino DA, Durkt G, Jr: Evaluation of muff-type hearing protec- 111. Folmer RL: The importance of hearing conservation instruction.
tors as used in a working environment. Am Ind Hyg Assoc J 57:264– J Sch Nurs 19:140–148, 2003.
271, 1996. 112. Peters RJ: The role of hearing protectors in leisure noise. Noise
109. American Academy of Otolaryngology–Head and Neck Surgery. Health 5:47–55, 2003.
Available at entnet.org. Accessed September 3, 2013.
Infections of the Labyrinth 153
John C. Goddard | William H. Slattery III
Key Points
■ Cytomegalovirus is the most common cause of nongenetic hearing loss in the United States.
■ The use of antiviral medications in the treatment of children with congenital cytomegalovirus
infection may help minimize deterioration in hearing.
■ Congenital rubella remains an important cause of sensorineural hearing loss (SNHL) worldwide.
■ Prevention of congenital rubella through routine vaccination is the best method of avoiding SNHL
and other sequelae.
■ Early identification and treatment of children with congenital syphilis will often help avoid late
complications associated with labyrinthine involvement.
■ Measles and mumps infections remain important causes of childhood hearing loss in countries that
lack routine vaccination programs.
■ Suppurative labyrinthitis may develop as a sequela of bacterial and nonbacterial meningitis as well
as from infectious conditions that affect the middle ear, mastoid, and temporal bone.
■ Routine vaccination against common bacterial pathogens, widespread use of antibiotic therapy, and
early identification of infection have all led to the reduction in the number of meningogenic and
otogenic cases of suppurative labyrinthitis.
■ Idiopathic disorders such as sudden SNHL, vestibular neuritis, and labyrinthitis have not
conclusively been linked to an infectious etiologic agent.
I nfections of the labyrinth may arise from direct invasion by a noninvasive manner that avoids loss of cochlear and/or ves-
bacteria, fungi, parasites, and viruses as well as from toxic sub- tibular function.9 Although sampling of perilymph may be
stances associated with an adjacent infectious process. In the achieved during cochlear implant surgery and from labyrin-
preantibiotic era, suppurative (bacterial) labyrinthitis was a fre- thine tissue collected during surgeries that sacrifice the inner
quent sequela of acute pyogenic otitis media and often increased ear (i.e., labyrinthectomy), patients who undergo these proce-
the likelihood of associated meningitis and subsequent death.1 dures do not always have a history of labyrinthine infection.10
In developed countries, routine use of antibiotic therapy has Moreover, the time interval between a suspected labyrinthine
significantly reduced the rate of otitis media–related complica- infection and subsequent surgical intervention may be signifi-
tions, and suppurative labyrinthitis occurs only rarely.2 Syphi- cantly delayed, which makes meaningful analysis difficult. Alter-
litic (luetic) involvement of the labyrinth has been well natively, postmortem temporal bone histopathologic analysis
documented histopathologically and was a frequently observed
clinical picture during the 1900s.3 As the rates of syphilis have
fallen significantly within the United States, so too have cases
Box 153-1. ELEMENTS NEEDED FOR CONFIRMATION
of luetic-induced sensorineural hearing loss (SNHL) and ves-
OF AN INNER EAR INFECTION
tibular dysfunction.4 Measles, mumps, and rubella (MMR)
infections have been a significant source of morbidity and mor- 1. Clinical association of infectious agent with a specific cochlear or
tality worldwide, with associated hearing loss in certain cases.5-7 vestibular syndrome
The development of MMR vaccines in the 1960s, however, led a. Epidemiologic studies of infectious agent and syndrome
to a dramatic decline in the incidence of these viral diseases, b. Clinical studies of syndrome and isolation of infectious agent at
other sites or serologic antibody titer rise
with a concomitant decrease in the number of cases with
2. Clear evidence of infectious agent presence within inner ear tissue
hearing loss.8 Whereas the United States and other developed a. Isolation of infectious agent vRNA or mRNA, excluding mRNA
countries have seen a decline in the overall number of labyrin- latency-associated transcripts, in inner ear tissue
thine infections associated with the above-named conditions, b. Histologic demonstration in inner ear tissue by electron
the incidence of these conditions in developing countries microscopy of infectious agent or by light microscopy finding of
remains significant. Consequently, it is imperative that otolar- inclusion bodies or characteristic cell morphology plus isolation of
yngologists remain knowledgeable about all of the potential infectious agent at other body sites
causes of labyrinthine infections. 3. In experimental animals, demonstration that suspect infectious agent
Identification and isolation of a specific causative agent in can cause similar auditory or vestibular signs and inner ear pathology
suspected cases of labyrinthine infection remains elusive (Box Modified from Davis LE. Neurovirology of deafness, labyrinthitis and Bell’s
153-1). Much of the difficulty in determining causation stems palsy. In Johnson RT, ed: Neurovirology. Minneapolis: American Academy of
from an inability to obtain tissue samples from the inner ear in Neurology; 2002.
2359
2360 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
remains an invaluable tool for examining the architecture of chamber filled with sodium-rich perilymph. The membranous
the labyrinth, particularly in patients with known clinical histo- labyrinth is a completely separate system suspended within this
ries. However, the physical and chemical processing of tempo- perilymph-filled chamber and is composed of potassium-rich
ral bone specimens limits the ability to use certain analytic endolymph. The membranous utricular and saccular ducts
techniques, such as the polymerase chain reaction (PCR), to connect their respective end organs with the endolymphatic
identify viral or other infectious agents.9,11 It is hoped that con- duct, which travels within the bony vestibular aqueduct and
tinued refinement of microbiologic, viral, and genetic tech- terminates as the endolymphatic sac along the posterior face
niques along with the development of novel approaches to of the petrous bone.
safely sample the inner ear will allow for more consistent and The vasculature of the inner ear will not be reviewed here,
reliable identification of the sources of labyrinthine infections but its importance as a potential route of (hematogenous)
in the future. spread of infection to the inner ear must be remembered.
Specific anatomic aspects of the labyrinth provide the remain-
ing routes for spread of infection to the inner ear; these include
BACKGROUND ANATOMY the cochlear aqueduct, internal auditory canal (IAC), and oval
and round windows. The cochlear aqueduct is a bony channel that
AND PHYSIOLOGY contains subarachnoid-like tissue known as the periotic duct.12
The inner ear has been dubbed the labyrinth, owing to the Although it is believed that the cochlear aqueduct is not respon-
complexity of its anatomic design. Although it is beyond the sible for the transmission of high pressure levels of cerebrospi-
scope of this chapter to review the anatomy and physiology of nal fluid (CSF) to the inner ear seen in perilymphatic gushers,
the labyrinth in its entirety, a basic understanding is a prereq- it may serve as a conduit for the spread of infection to and from
uisite to a discussion of infections of the labyrinth. Functionally the intracranial cavity.13-15 The cochlear aqueduct traverses the
characterized by six different sensory end organs—the cochlear inferior petrous bone from the subarachnoid space of the pos-
organ of Corti, the three ampullae of the semicircular canals, terior fossa to the basal aspect of the cochlea and opens into
and the utricular and saccular maculae—the labyrinth is more the medial aspect of the scala tympani. Some histologic studies
broadly characterized as a bony outer shell and a membranous of the cochlear aqueduct suggest higher patency in children,
internal compartment (Fig. 153-1). The shell of the labyrinth whereas others demonstrate patency along the entire length of
comprises otic capsule bone, which defines the shape of the the aqueduct in 34% of specimens.13-15 The cribrose area at the
semicircular canals, vestibule, and cochlea and encloses a fundus of the IAC serves as the point of entry of the cochlear
Ampulla
Utricle
Endolymphatic
sac
Dura
Ampulla
Stapes in
Endolymphatic oval window
duct
Saccule Vestibular
cecum
Round window
Helicotrema
Cochlear duct
nerve fibers into the modiolus. In cases of congenital deficiency likely to have a child with a significant CMV infection than
of the cribrose area, CSF pressure may be transmitted more women infected during pregnancy.21,23 However, reactivation of
completely to the perilymph.16 This location may also serve as a latent CMV infection (i.e., secondary CMV infection) remains
a route for spread of infection between the intracranial cavity a potential means of maternal-fetal transmission.
and the inner ear. The final potential anatomic routes of spread The current gold standard for the diagnosis of congenital
are from the oval and round windows, which provide the most CMV infection is isolation of the virus from urine, saliva, blood,
direct interface between the middle ear and the labyrinth. or other body fluids within the first 2 to 3 weeks of life.21,23 PCR
Although the stapes footplate serves as a bony barrier at the analysis of CMV DNA has now become the preferred technique
level of the oval window, the round window membrane is three for viral isolation and has replaced viral culture methods of
to four cell layers thick and may offer less resistance to the detection.24,25 Although interest has been shown in using the
spread of various molecules.17 dried blood samples routinely obtained for genetic screening
purposes at birth as a means of identifying CMV DNA, the
sensitivity of this method has been poor when compared with
PERINATAL LABYRINTHINE PCR testing of saliva.10,25-27 Peripheral blood antibody detection
is also not useful for diagnosing congenital CMV. Finally, testing
INFECTIONS performed more than 3 weeks after birth is not considered
A number of infections may affect a pregnancy and jeopardize adequate for a diagnosis of congenital CMV because of the high
the health of the developing fetus or newborn baby. These frequency of asymptomatic CMV infection among infants aged
perinatal infections are typically viral and may impact a variety 3 weeks and older.
of organ systems, including the inner ear. The most common The majority of children born with congenital CMV are
perinatal viral infections that affect the labyrinth are cytomega- asymptomatic, and 80% never develop any significant symp-
lovirus (CMV) and rubella, although herpes simplex virus toms or disabilities.21 Occurring rarely, the most severe form of
(HSV) and other viruses may affect the inner ear as well. Non- congenital CMV, termed cytomegalic inclusion disease (CID),
viral perinatal infections may affect the labyrinth and include affects multiple organ systems and is associated with significant,
toxoplasmosis (parasitic) and syphilis (bacterial), among permanent disabilities.21,23 In patients with this severe pheno-
others. type, SNHL, microcephaly, and learning difficulties are seen in
nearly 50% of cases.28 Hepatosplenomegaly, jaundice, blue-
berry muffin rash, and computed tomography (CT) evidence
PERINATAL VIRAL LABYRINTHINE of intracerebral calcifications are also typical of CID.23 Whereas
the implications for those with CID are fairly clear, the prepon-
INFECTIONS derance of newborns with congenital CMV infection are asymp-
Viruses have been implicated in the development of labyrin- tomatic and have an unpredictable course. Early longitudinal
thine infections for decades. However, as mentioned above, the studies that examined the percentage of asymptomatic cases of
ability to identify and isolate a causative organism in cases of congenital CMV with SNHL reported rates that ranged from
labyrinthine infection remains challenging. Although strong 7% to 15%.29-33 Among these patients, SNHL was often bilateral
clinical evidence supports a causative relationship between and ranged from mild to profound, although unilateral, pro-
many viral infections and the development of cochleovestibular gressive, and delayed-onset losses were also noted.29-33 Addition-
symptoms, CMV is the only perinatal viral infection that affects ally, newborn screening was normal in up to 50% of cases.30
the labyrinth that has actually been isolated from perilymph or More recent studies that examined the rates and characteristics
detected within inner ear tissue.18-20 Other congenital viruses— of hearing loss among asymptomatic patients with congenital
such as rubella, HSV, and human immunodeficiency virus CMV infection found rates of SNHL to be as high as 23%.26,34-36
(HIV)—have not yet been identified within similar inner ear Examination of peripheral blood viral load levels suggests the
samples. risk of SNHL may actually be lower in patients with reduced
viral loads.37 However, specific factors that influence the pattern
Congenital CMV and severity of SNHL in asymptomatic cases of congenital CMV
According to the Centers for Disease Control and Prevention remain unclear. The data concerning vestibular abnormalities
(CDC), CMV is the most common congenital viral infection among patients with congenital CMV are more limited, but
and the most common cause of nongenetic hearing loss in the reports do suggest that patients who are symptomatic at birth
United States.21 Approximately 1 in 150 children is born with can develop vestibular deficits over time.38
congenital CMV infection, which translates into 30,000 chil- The clinical evidence that validates congenital CMV-related
dren per year in the United States alone. Clinical symptoms labyrinthine infections has been strengthened by the ability of
and permanent disabilities will occur in up to 15% to 20% of researchers and clinicians to isolate CMV viral particles from
cases of congenital CMV, including the possible development inner ear tissue.18-20,39-42 In the 1970s, Davis and colleagues9,42,43
of SNHL.21-23 The nature of the hearing loss in these children cultured CMV from perilymph at autopsy from an infant with
is quite variable, and it may be delayed, progressive, bilateral, CID and subsequently identified CMV antigen in cells of the
or even fluctuating.23 Because of the potentially delayed pre- membranous labyrinth and intracellular inclusions within the
sentation of CMV-related hearing loss, newborn hearing screen- inner ear (Fig. 153-2). Prior descriptions of temporal bone
ing may be normal.23-25 histopathologic findings observed in infants dying of symptom-
Congenital CMV infections occur primarily via transmission atic congenital CMV suggest significant changes within the
in utero, although they may occur at the time of delivery or entire endolymphatic system but little histologic damage to the
postnatally. Whereas mothers are the source of transmission to auditory and vestibular nerves, along with the spiral and ves-
their offspring, pregnant women often become infected tibular ganglia.41-45 Hydrops of the cochlea and saccule and
through sexual contact with a partner or spouse or through collapse of the Reissner membrane have also been observed.9
contact with the saliva or urine of an infected child.21 Maternal Teissier and colleagues39 more recently examined six CMV-
CMV infection can occur at any time before or during preg- infected fetuses that ranged in age from 19 to 35 weeks post-
nancy and is typically asymptomatic, although mononucleosis- conception. Cytomegalic cells with inclusion bodies were noted
like symptoms may develop.21 Women who are infected 6 within the marginal cells of the stria vascularis and nonsensory
months or more prior to becoming pregnant are much less epithelium of the vestibular apparatus.39 Extensive involvement
2362 PART VII | OTOLOGY, NEUROTOLOGY, AND SKULL BASE SURGERY
and histopathologic studies. Rates of hearing loss after menin-
gitis may approach 35%, and severe to profound hearing loss
and labyrinthine ossification occurs in a number of cases.92-97
The spread of bacterial infection from the subarachnoid space
to the labyrinth most likely occurs via the cochlear aqueduct or
the cribrose area of the fundus of the IAC.12,91,92,98-101 However,
vascular routes, abnormal traumatic or congenital bony dehis-
cences between the labyrinth and subarachnoid space, and the
endolymphatic sac may also be involved in transmitting these
infections.92
Diagnosis of meningogenic labyrinthitis relies on clinical
history and supportive audiometric data. Clinical findings in
adults may include lethargy, fever, neck stiffness, photophobia,
and headache. In infants and young children, the initial symp-
toms may be more variable. The onset of hearing loss and/or
vestibular deficits may not be readily identifiable, particularly
in infants with meningitis. However, audiometric documenta-
tion of SNHL in patients with a recent history of meningitis is
suggestive of labyrinthine involvement. Further support for
Figure 153-3. Endolymphatic hydrops as a result of syphilis. Arrows depict meningogenic causes of SNHL are provided in cases where
abnormal position of Reissner membrane (hematoxylin & eosin stain, ×20). normal hearing was documented prior to the meningitic event.
(Courtesy Dr. Fred Linthicum, Eccles Temporal Bone Laboratory, House Research Ototoxicity from IV antibiotic therapy used to treat meningitis
Institute, Los Angeles.) must also be considered as a possible etiology of SNHL, espe-
cially in locales where gentamycin and other aminoglycoside
rodents or birds, will have contaminated feces. Consequently, antibiotics are commonly used. Standard pure tone audiometry
people may transmit the cysts to themselves or others via hand with word-recognition testing should be performed as soon as
to oral contact after the handling of litter box materials. Con- a suspicion of meningogenic labyrinthine involvement is enter-
genital toxoplasmosis is relatively uncommon and occurs in an tained. However, such testing is not always practical, and alter-
estimated 1 in 10,000 newborns.85 The majority of newborns natively, otoacoustic emission and auditory brainstem response
with congenital toxoplasmosis are asymptomatic, although testing, which do not require voluntary participation, may be
involvement of the CNS, visual, and other systems is possible.86 used.90 Whereas normal auditory brainstem response results
In a recent systematic review, the prevalence of toxoplasmosis- early in the course of meningitis may be encouraging, follow-up
associated hearing loss in infected newborns was found to behavioral audiometry is imperative.102 CT imaging of the post-
range from 0% to 26%.87 Determination of antibodies (IgM meningitic patient may also support a diagnosis of meningo-
and IgG) against T. gondii in the hearing-impaired newborn genic labyrinthine involvement when ossification of labyrinthine
should be performed in suspected cases to aid in diagnosis. structures is noted. A lack of middle ear or mastoid infection
Treatment consists of 12 months of antiparasitic medication, at the time of clinical meningitis is also important in distin-
and evidence suggests a reduction in SNHL rates among guishing meningogenic from otogenic labyrinthitis. Timing of
patients with high compliance and early treatment initiation.87 the occurrence of labyrinthine ossification following meningitis
No known reports of temporal bone histopathology in patients is variable, although animal models suggest that ossification
with congenital toxoplasmosis exist in the literature. following S. pneumoniae meningitis occurs as early as 3 days to
3 weeks after inoculation.103,104 The incidence of labyrinthine
ossification following meningitis is also not known, although
ACQUIRED LABYRINTHINE INFECTIONS studies of children with SNHL following meningitis demon-
Acquired labyrinthine infections may be a result of a variety of strate some degree of ossification in up to 70% of cases.105,106
bacterial, viral, or parasitic pathogens. Some of the more Although it does appear that S. pneumoniae meningitis is associ-
common causes of infection are discussed here. ated with a higher incidence of SNHL, there does not appear
to be a significant difference in the development of labyrin-
Meningogenic Labyrinthine Infections thine ossification among S. pneumoniae, N. meningitidis, and
Bacterial Meningogenic Labyrinthine Infections. Meningitis HIB.107
may result from bacterial, viral, fungal, parasitic, or toxic inva- Temporal bone findings in cases of meningogenic labyrin-
sion of the subarachnoid space. In the United States alone, thitis have been fairly well documented.12,92,98-101 Schuknecht12
4100 cases of bacterial meningitis, with nearly 500 associated distinguishes between terminal meningogenic labyrinthitis and
deaths, occur annually.88 Streptococcus pneumoniae, Neisseria men- a more standard form of meningogenic labyrinthitis. In terminal
ingitidis, and Haemophilus influenzae type B (HIB) are the most meningogenic labyrinthitis, suppuration of the labyrinth is thought
common causes of meningitis in children; whereas Escherichia to be a terminal event in a patient who is moribund and in
coli, Group B Streptococcus, and Listeria monocytogenes are often whom cochleovestibular symptoms are not present. In nonter-
implicated in newborns. Adolescents and adults are usually minal meningogenic labyrinthitis, studies of temporal bones
infected by N. meningitides and S. pneumoniae.89 Introduction of reveal fibrosis and ossification within the labyrinth, particularly
the HIB vaccine in the United States pediatric population in within the basal portions of the cochlea.12,98-101 In the most
the 1990s led to a significant reduction in the number of cases recent review of a series of temporal bone specimens with sup-
of HIB meningitis.90 purative labyrinthitis secondary to meningitis, inflammatory
Proper recognition of the link between labyrinthine infec- cells were commonly found within both the vestibular and
tions and meningitis was first recognized in the late 1800s.91 In cochlear perilymphatic spaces.92 Sensory and neural structures
the preantibiotic era, meningitis was suspected to account for were often intact, although loss of spiral ganglion neurons was
as many as 20% of cases of SNHL in children.91 Since that time, seen in 10% to 15% of cases.92
our understanding of meningogenic labyrinthine infections Adherence to childhood and adult vaccination schedules is
has increased greatly through a combination of epidemiologic an important means of meningitis prevention, particularly in
153 | INFECTIONS OF THE LABYRINTH 2365
A B
Figure 153-4. Cryptococcal labyrinthine involvement. A, Arrow depicts eighth nerve infiltration by Cryptococcus in the fundus of the internal auditory canal
(hematoxylin & eosin [H&E] stain, ×100). B, High-power view demonstrates cryptococcal spores (H&E stain, ×400). (Courtesy Dr. Fred Linthicum, Eccles
Temporal Bone Laboratory, House Research Institute, Los Angeles.)
ACQUIRED SYPHILITIC
LABYRINTHINE INFECTIONS
As with congenital syphilis, acquired syphilis results from the
spirochete Treponema pallidum. Primary, secondary, and tertiary Figure 153-6. Syphilitic osteitis. Infiltration of the endolymphatic duct (large
forms of syphilis describe the different clinical features of this arrow) with lymphocytic cells and destruction of adjacent bone (small arrow)
multisystem disease.12,77 Primary syphilis is defined by a chancre are shown (hematoxylin & eosin stain, ×100). (Courtesy Dr. Fred Linthicum,
at the site of inoculation, and it lacks inner ear symptomatology Eccles Temporal Bone Laboratory, House Research Institute, Los Angeles.)