Morbidity and Mortality Weekly Report
www.cdc.gov/mmwr
Recommendations and Reports June 6, 2008 / Vol. 57 / RR-5
Prevention of Herpes Zoster
Recommendations
of the Advisory Committee on
Immunization Practices (ACIP)
INSIDE: Continuing Education Examination
depardepar
depardepar
depar
tment of health and human sertment of health and human ser
tment of health and human sertment of health and human ser
tment of health and human ser
vicesvices
vicesvices
vices
Centers for Disease Control and PreventionCenters for Disease Control and Prevention
Centers for Disease Control and PreventionCenters for Disease Control and Prevention
Centers for Disease Control and Prevention
MMWR
The MMWR series of publications is published by the Coordinating
Center for Health Information and Service, Centers for Disease
Control and Prevention (CDC), U.S. Department of Health and
Human Services, Atlanta, GA 30333.
Suggested Citation: Centers for Disease Control and Prevention.
[Title]. MMWR Early Release 2008;57[Date]:[inclusive page numbers].
Centers for Disease Control and Prevention
Julie L. Gerberding, MD, MPH
Director
Tanja Popovic, MD, PhD
Chief Science Officer
James W. Stephens, PhD
Associate Director for Science
Steven L. Solomon, MD
Director, Coordinating Center for Health Information and Service
Jay M. Bernhardt, PhD, MPH
Director, National Center for Health Marketing
Katherine L. Daniel, PhD
Deputy Director, National Center for Health Marketing
Editorial and Production Staff
Frederic E. Shaw, MD, JD
Editor, MMWR Series
Teresa F. Rutledge
(Acting) Managing Editor, MMWR Series
David C. Johnson
(Acting) Lead Technical Writer-Editor
David C. Johnson
Project Editor
Peter M. Jenkins
(Acting) Lead Visual Information Specialist
Lynda G. Cupell
Malbea A. LaPete
Visual Information Specialists
Quang M. Doan, MBA
Erica R. Shaver
Information Technology Specialists
Editorial Board
William L. Roper, MD, MPH, Chapel Hill, NC, Chairman
Virginia A. Caine, MD, Indianapolis, IN
David W. Fleming, MD, Seattle, WA
William E. Halperin, MD, DrPH, MPH, Newark, NJ
Margaret A. Hamburg, MD, Washington, DC
King K. Holmes, MD, PhD, Seattle, WA
Deborah Holtzman, PhD, Atlanta, GA
John K. Iglehart, Bethesda, MD
Dennis G. Maki, MD, Madison, WI
Sue Mallonee, MPH, Oklahoma City, OK
Stanley A. Plotkin, MD, Doylestown, PA
Patricia Quinlisk, MD, MPH, Des Moines, IA
Patrick L. Remington, MD, MPH, Madison, WI
Barbara K. Rimer, DrPH, Chapel Hill, NC
John V. Rullan, MD, MPH, San Juan, PR
Anne Schuchat, MD, Atlanta, GA
Dixie E. Snider, MD, MPH, Atlanta, GA
John W. Ward, MD, Atlanta, GA
CONTENTS
Introduction .......................................................................... 1
Methods ...............................................................................2
Background ..........................................................................2
Biology of VZV ................................................................... 2
Clinical Features of Zoster and PHN .................................. 3
Diagnosis ..........................................................................5
Zoster Transmission ...........................................................5
Epidemiology of Zoster and Complications ....................... 6
Treatment and Nonspecific Management of Zoster
and PHN ........................................................................ 11
Prevention of Transmission from Zoster ........................... 11
Zoster Vaccine .................................................................... 11
Vaccine Composition, Dosage, and Administration ......... 11
Storage and Handling ..................................................... 12
Efficacy ............................................................................12
Immunogencity ................................................................ 14
Duration of Efficacy and of Immunity ..............................14
Safety and Adverse Events .............................................. 15
The Economic Burden of Zoster and Cost-Effectiveness of
Vaccination ......................................................................... 15
Summary of Rationale for Zoster Vaccine
Recommendations ............................................................ 17
Recommendations for Use of Zoster Vaccine ...................... 19
Routine Vaccination of Persons Aged
>60 Years ..............19
Simultaneous Administration with Other Adult Vaccines ..19
Groups for Which Vaccine is Not Licensed ....................... 19
Special Groups and Circumstances .................................. 19
Contraindications ............................................................20
Precautions ...................................................................... 21
Program Implementation Issues ......................................21
Future Research and Directions ..........................................22
Additional Information About Zoster and Zoster Vaccine ....23
Acknowledgments ..............................................................23
References .........................................................................23
Disclosure of Relationship
CDC, our planners, and our content experts wish to disclose they have
no financial interests or other relationships with the manufacturers of
commercial products, suppliers of commercial services, or commercial
supporters. This report does not include any discussion of the unlabeled
use of a product or a product under investigational use with the
exception of the discussion of off-label use of zoster vaccine by persons
who report a previous episode of herpes zoster. In addition, guidance is
provided for instances in which zoster vaccine is inadvertently
administered.
1 Vol. 57 / RR-5 MMWR
Prevention of Herpes Zoster
Recommendations of the Advisory Committee
on Immunization Practices (ACIP)
Prepared by
Rafael Harpaz, MD, Ismael R. Ortega-Sanchez, PhD, Jane F. Seward, MBBS,
Division of Viral Diseases, National Center for Immunization and Respiratory Diseases
Summary
These recommendations represent the first statement by the Advisory Committee on Immunization Practices (ACIP) on the
use of a live attenuated vaccine for the prevention of herpes zoster (zoster) (i.e., shingles) and its sequelae, which was licensed
by the U.S. Food and Drug Administration (FDA) on May 25, 2006. This report summarizes the epidemiology of zoster and
its sequelae, describes the zoster vaccine, and provides recommendations for its use among adults aged >60 years in the United
States.
Zoster is a localized, generally painful cutaneous eruption that occurs most frequently among older adults and
immunocompromised persons. It is caused by reactivation of latent varicella zoster virus (VZV) decades after initial VZV
infection is established. Approximately one in three persons will develop zoster during their lifetime, resulting in an estimated
1 million episodes in the United States annually. A common complication of zoster is postherpetic neuralgia (PHN), a
chronic, often debilitating pain condition that can last months or even years. The risk for PHN in patients with zoster is
10%–18%. Another complication of zoster is eye involvement, which occurs in 10%–25% of zoster episodes and can result
in prolonged or permanent pain, facial scarring, and loss of vision. Approximately 3% of patients with zoster are hospital-
ized; many of these episodes involved persons with one or more immunocompromising conditions. Deaths attributable to zoster
are uncommon among persons who are not immunocompromised.
Prompt treatment with the oral antiviral agents acyclovir, valacyclovir, and famciclovir decreases the severity and duration
of acute pain from zoster. Additional pain control can be achieved in certain patients by supplementing antiviral agents with
corticosteroids and with analgesics. Established PHN can be managed in certain patients with analgesics, tricyclic antide-
pressants, and other agents.
Licensed zoster vaccine is a lyophilized preparation of a live, attenuated strain of VZV, the same strain used in the varicella
vaccines. However, its minimum potency is at least 14-times the potency of single-antigen varicella vaccine. In a large
clinical trial, zoster vaccine was partially efficacious at preventing zoster. It also was partially efficacious at reducing the
severity and duration of pain and at preventing PHN among those developing zoster.
Zoster vaccine is recommended for all persons aged >60 years who have no contraindications, including persons who report
a previous episode of zoster or who have chronic medical conditions. The vaccine should be offered at the patient’s first clinical
encounter with his or her health-care provider. It is administered as a single 0.65 mL dose subcutaneously in the deltoid region
of the arm. A booster dose is not licensed for the vaccine. Zoster vaccination is not indicated to treat acute zoster, to prevent
persons with acute zoster from developing PHN, or to treat ongoing PHN. Before administration of zoster vaccine, patients do
not need to be asked about their history of varicella (chickenpox) or to have serologic testing conducted to determine varicella
immunity.
Introduction
The material in this report originated in the National Center for
Immunization and Respiratory Diseases, Anne Schuchat, MD,
Infection with varicella zoster virus (VZV) causes two dis-
Director; and the Division of Viral Diseases, Larry Anderson, MD,
tinct clinical conditions. Primary VZV infection causes
Director.
varicella (i.e., chickenpox), a contagious rash illness that
Corresponding preparer: Rafael Harpaz, MD, National Center for
Immunization and Respiratory Diseases, CDC, 1600 Clifton Rd.,
typically occurs among children. A vaccine for preventing
NE, MS A-47, Atlanta, GA 30333. Telephone: 404-639-6284; Fax:
initial VZV infection has been available in the United States
404-639-8665; E-mail: r[email protected].
since 1995, and the Advisory Committee on Immuniza-
tion Practices (ACIP) recommends routine varicella
2 MMWR June 6, 2008
vaccination for all persons aged >12 months who lack evi-
dence of immunity (1–3). Varicella vaccination has dra-
matically reduced chickenpox cases among children (3).
VZV can reactivate clinically decades after initial infec-
tion to cause herpes zoster (zoster) (i.e., shingles), a local-
ized and generally painful cutaneous eruption that occurs
most frequently among older adults. Approximately 1 mil-
lion new cases of zoster occur in the United States annu-
ally. Approximately one in three persons in the general
population will develop zoster during their lifetime. A com-
mon complication of zoster is postherpetic neuralgia
(PHN), a chronic pain condition that can last months or
even years. In May 2006, a live, attenuated vaccine for pre-
vention of zoster (ZOSTAVAX
®
, manufactured by Merck
& Co., Inc.) was licensed in the United States for use in
persons aged >60 years. This report provides recommen-
dations for use of zoster vaccine for prevention of zoster
and its sequelae.
Methods
In Spring 2005, Merck & Co., Inc. (Whitehouse Sta-
tion, New Jersey) submitted a Biologics License Applica-
tion to the Food and Drug Administration (FDA) for an
investigational live, attenuated vaccine for prevention of
zoster on the basis of a phase 3 clinical trial. These results
were published in June 2005 (4) and presented at the ACIP
meeting later that month. In September 2005, ACIP’s
measles-mumps-rubella and varicella workgroup expanded
its membership to include experts in adult medicine and
in zoster and began review of relevant data on zoster and
the investigational vaccine. Shortly thereafter, this workgroup
reformed as the ACIP shingles workgroup and, during sub-
sequent months, held 19 conference calls to review and
discuss scientific evidence related to herpes zoster and zoster
vaccine, including the epidemiology and natural history of
zoster and its sequelae, and the safety, immunogenicity,
efficacy, financing, storage, and handling of the zoster vac-
cine. The workgroup also reviewed several economic analy-
ses on zoster prevention. Workgroup members participated
in 10 additional conference calls to develop and discuss
recommendation options for preventing zoster. When sci-
entific evidence was lacking, recommendations incorporated
expert opinions of the workgroup members.
Presentations of background materials on zoster and the
vaccine were made during ACIP meetings in October 2005
and the three meetings in 2006. Following vaccine licen-
sure on May 25, 2006, recommendation options were pre-
sented to ACIP in June 2006, and final options were
presented at the October 2006 meeting. During review by
CDC and external partners, modifications were made to
the proposed recommendations to update and clarify word-
ing in the document. As new information on the epidemi-
ology and prevention of zoster becomes available, it will be
reviewed by ACIP and recommendations will be updated
as needed.
Background
Biology of VZV
VZV is an exclusively human pathogen that infects ap-
proximately 98% of the adult population in the United
States (5). The primary infection typically occurs during
childhood and causes varicella. During its viremic phase,
cell-associated VZV gains access to epidermal cells, caus-
ing the typical varicella rash. The virus then enters sensory
nerves in mucocutaneous sites and travels through retro-
grade axonal transport to the sensory dorsal root ganglia
adjacent to the spinal cord where the virus establishes per-
manent latency in neuronal cell bodies (6–7). Latent VZV
is present in approximately 1%–7% of sensory ganglion
neurons, with <10 genomic copies per infected cell (8–10).
Seeding of dorsal root ganglia also might occur during vire-
mia. The magnitude of viremia, the number of skin lesions,
and the burden of VZV that establishes latency during pri-
mary varicella infection might be linked (11). As with other
members of the herpesvirus family, VZV is noninfectious
in its latent form but can reactivate at a later time to form
intact virions in the involved sensory neurons. These viri-
ons then migrate to the skin through axons, spread from
cell to cell, and penetrate the epidermis (12). In its full
clinical expression, zoster causes pain, which is followed by
a vesicular rash distributed across closely overlapping
dermatomes of the involved sensory nerve roots.
The triggers for reactivation of VZV have not been iden-
tified and probably involve multiple factors. However, spe-
cific components of cell-mediated immunity (CMI) have
an important role in controlling the development of zoster
by preventing reactivation within the neuron or the full
clinical expression of reactivated VZV as zoster. The
effectiveness of these protective components of CMI is well
maintained in immunocompetent persons during child-
hood and early adulthood. These CMI components are
believed to be partially or substantially maintained by
periodic immunologic boosting. “Endogenous boosting”
might occur in response to subclinical reactivation of
latent VZV or to development of zoster itself, and “exog-
enous boosting” might occur in response to exposure to
3 Vol. 57 / RR-5 MMWR
VZV circulating in the population as chickenpox (13–19).
Although virtually all adults are infected with VZV (5),
specific immunologic parameters have not been identified
to distinguish who will develop zoster. Anti-VZV antibody
levels per se are not thought to have a role in zoster preven-
tion (20). Parameters that have been monitored and corre-
late with such protection include anamnestic boost in
anti-VZV antibodies in vivo in response to VZV-based vac-
cination, and the presence of and boost in memory CD4 T
cells as measured in vitro by proliferation of peripheral blood
mononuclear cells (PBMC), by frequency of proliferating
PBMC, or by frequency of interferon-γ (IFN-γ ) releasing
PBMC, all in response to VZV-antigens (21,22). These
latter two parameters are generally assessed using a responder
cell frequency (RCF) assay and an IFN-γ enzyme-linked
immunosorbent spot-forming cell (ELISPOT) assay, respec-
tively (22). VZV-specific class I-restricted and unrestricted
cytotoxicities also have been monitored using target cell
lysis (23). Although CMI is necessary for the control of
zoster, other nonimmunologic factors also might be involved
(24).
Clinical Features of Zoster and PHN
The clinical course of acute zoster is variable. It is usually
less severe in children and younger adults. Typically, zoster
begins with a prodrome. Headache, photophobia, and
malaise might occur, with fever being less common.
Abnormal skin sensations and pain of varying severity are
the most common symptoms. These symptoms can pre-
cede the zoster rash by days to weeks (25) and rarely might
be the only clinical manifestation of VZV reactivation
(termed zoster sine herpete) (7). Pain is described as ach-
ing, burning, stabbing, or shock-like. Altered sensitivity to
touch, pain provoked by trivial stimuli, and unbearable
itching are all frequently reported.
Zoster rash is typically unilateral and does not cross the
mid-line, erupting in one or two adjacent dermatomes. The
frequency of zoster occurrence in different dermatomes has
been evaluated in certain studies. In general, thoracic, cer-
vical, and ophthalmic involvement are most common
(Figure 1) (26–28). Small numbers of lesions can occur
outside the primary or adjacent dermatome. The rash is
initially erythematous and maculopapular but progresses
to form coalescing clusters of clear vesicles containing high
concentrations of VZV (Figure 1). The vesicles form over
several days and then evolve through pustular, ulcer, and
crust stages. The rash usually lasts 7–10 days, with com-
plete healing within 2-4 weeks. However, pigmentation
changes and scarring might be permanent. Streptococcal
FIGURE 1.Thoracic distribution of zoster (A), and zoster rash
with coalescing clusters of clear vesicles (B)
or staphylococcal superinfections might complicate zoster
rash (29).
A common and potentially debilitating consequence of
zoster is PHN, a persistent pain after resolution of the rash.
Pathologic observations thought to distinguish PHN from
uncomplicated zoster include axonal and cell body degen-
eration, atrophy of the spinal cord dorsal horn, scarring of
the dorsal root ganglion, and loss of epidermal innervation
of the affected area. Certain experts believe this neuronal
damage might be caused by ongoing viral replication
(30,31). In addition, consensus is lacking regarding crite-
ria needed to distinguish the quality, duration, or underly-
ing pathophysiology of pain occurring with zoster versus
PHN. Therefore, the duration of pain used to define PHN
has been inconsistent, ranging from any duration after reso-
lution of the rash to periods from
>30 days to >6 months
after rash onset.
Regardless of definition, the pain of PHN can last for
weeks or months and occasionally persists for many years.
The nature of PHN pain varies from mild to excruciating
4 MMWR June 6, 2008
in severity, constant, intermittent, or triggered by trivial
stimuli. Approximately half of patients with zoster or PHN
describe their pain as “horrible” or “excruciating”, ranging
in duration from a few minutes to constant on a daily or
almost daily basis (32). The pain can disrupt sleep, mood,
work, and activities of daily living, adversely impacting the
quality of life and leading to social withdrawal and depres-
sion (Table 1) (31–33). Anecdotes of suicide among
patients suffering from PHN have been reported (34; Peter
Richards, MD, personal communication, 2007). Among
persons experiencing zoster, predictors of PHN include the
occurrence and severity of pain both before and after onset
of the rash, the extent of the rash, trigeminal or ophthalmic
distribution (35,36), and the occurrence of viremia (37).
In addition to PHN, zoster is associated with a variety of
other complication. Among persons with zoster, 10%–25%
have eye involvement, called herpes zoster ophthalmicus
(HZO) (38,39) (Figure 2). HZO can occur when reactiva-
tion involves the nasociliary branch of the trigeminal nerve,
sometimes preceded by the presence of zoster vesicles on
the nose (Hutchinson sign). Keratitis occurs in approxi-
mately two thirds of patients with HZO (40), often caus-
ing corneal ulceration. Other complications include
conjunctivitis, uveitis, episcleritis and scleritis, retinitis,
choroiditis, optic neuritis, lid retraction, ptosis, and glau-
coma. Extraocular muscle palsies also occur. Prolonged or
permanent sequelae of HZO include pain, facial scarring,
and loss of vision (41).
An uncommon complication of zoster is Ramsay Hunt
syndrome, a peripheral facial nerve palsy accompanied by
zoster vesicles on the ear, hard palate, or tongue (42). The
pathophysiology of this complication involves reactivation
of VZV in the geniculate ganglion of the facial nerve.
Additional signs and symptoms of Ramsey Hunt syndrome
can include pain, vertigo, hearing loss, sensitivity to sound,
tinnitus, and loss of taste. Many patients do not recover
TABLE 1. Impact of acute herpes zoster and postherpetic
neuralgia on quality of life
Life factor Impact
Physical Chronic fatigue
Anorexia and weight loss
Physical inactivity
Insomnia
Psychological Anxiety
Difficulty concentrating
Depression, suicidal ideation
Social Fewer social gatherings
Changes in social role
Functional Interferes with activities of daily living (e.g., dressing,
bathing, eating, travel, cooking, and shopping)
FIGURE 2. Case of herpes zoster ophthalmicus
Photo/MN Oxman, University of California, San Diego
completely (42). Idiopathic facial palsy (Bell’s palsy) might
be caused by inapparent VZV reactivation (42,43).
Occasionally, zoster can cause motor weakness in
noncranial nerve distributions, called zoster paresis (44,45).
The mechanism has not been determined. The weakness
develops abruptly within 2–3 weeks after onset of the rash
and can involve upper or lower extremities. Diaphragmatic
paralysis also has been described. The prognosis of zoster
paresis is good (46). Zoster also can result in autonomic
dysfunction, causing urinary retention and colon pseudo-
obstruction.
Rarely, patients will experience acute focal neurologic
deficits weeks to months after resolution of the zoster rash,
involving the trigeminal distribution contralateral to the
initial rash. This ischemic stroke syndrome, termed
granulomatous angiitis, is believed to be caused by direct
extension of VZV from the trigeminal ganglion to the
internal carotid artery or its branches, resulting in inflam-
mation (30). Mortality from this syndrome is substantial.
Other rare neurologic complications of zoster include
myelitis, aseptic meningitis, and meningoencephalitis. The
prognosis for these conditions is good, although encepha-
lomyelitis can be life threatening. Guillain-Barré syndrome
also has been reported in association with zoster (47).
In immunocompromised persons, zoster initially might
present typically. However, the rash tends to be more
severe and its duration prolonged (48,49). One specific
risk for persons with some immunosuppressive conditions
5 Vol. 57 / RR-5 MMWR
is dissemination of the zoster rash. True cutaneous dissemi-
nation generally occurs only among immunocompromised
patients, occurring in up to 37% of zoster cases in the
absence of antiviral treatment (49–54). Dissemination usu-
ally begins with a dermatomal rash; however, the rash some-
times begins with no primary dermatomal involvement (54).
Cutaneous dissemination is not life-threatening; however,
it is a marker for VZV viremia that can seed the lungs,
liver, gut, and brain and cause pneumonia, hepatitis,
encephalitis, and disseminated intravascular coagulopathy
in 10%–50% of episodes. Visceral dissemination with no
skin involvement can occur in profoundly
immunocompromised persons. Even with antiviral treat-
ment, the case fatality rate from visceral dissemination is
5%–15%, with most deaths attributable to pneumonia
(49,54,55).
The risk for neurologic zoster complications is generally
increased in immunocompromised persons. These compli-
cations, which can be aggressive and even fatal, include
myelitis, chronic encephalitis, ventriculitis, meningoen-
cephalitis, and cranial palsies (30). However, the risk for
PHN is not appreciably increased among immuno-
compromised persons who develop zoster (30).
Compared with other immunocompromised persons, the
clinical features of zoster are less severe and visceral dis-
semination less common among persons infected with
human immunodeficiency virus (HIV) (56,57). Some
zoster presentations that are unique to persons infected with
HIV include atypical skin eruptions (58,59) and an
aggressive variant of acute retinal necrosis that generally
results in blindness (60). Alveolar bone necrosis and tooth
exfoliation also have been reported (61).
Diagnosis
Zoster diagnosis might not be possible in the absence of
rash (e.g., before rash or in cases of zoster sine herpete).
Patients with localized pain or altered skin sensations might
undergo evaluation for kidney stones, gallstones, or coro-
nary artery disease until the zoster rash appears and the
correct diagnosis is made (62). In its classical manifestation,
the signs and symptoms of zoster are usually distinctive
enough to make an accurate clinical diagnosis once the rash
has appeared (63). Occasionally, zoster might be confused
with impetigo, contact dermatitis, folliculitis, scabies,
insect bites, papular urticaria, candidal infection, dermati-
tis herpetiformis, or drug eruptions. More frequently, zoster
is confused with the rash of herpes simplex virus (HSV),
including eczema herpeticum (4,31,64–66). The accuracy
of diagnosis is lower for children and younger adults in
whom zoster incidence is lower and its symptoms less
often classic.
In some cases, particularly in immunosuppressed per-
sons, the location of rash appearance might be atypical, or
a neurologic complication might occur well after resolu-
tion of the rash. In these instances, laboratory testing might
clarify the diagnosis (67–71). Tzanck smears are inexpen-
sive and can be used at the bedside to detect multinucle-
ated giant cells in lesion specimens, but they do not
distinguish between infections with VZV and HSV. VZV
obtained from lesions can be identified using tissue cul-
ture, but this can take several days and false negative
results occur because viable virus is difficult to recover from
cutaneous lesions. Direct fluorescent antibody (DFA) stain-
ing of VZV-infected cells in a scraping of cells from the
base of the lesion is rapid and sensitive. DFA and other
antigen-detection methods also can be used on biopsy
material, and eosinophilic nuclear inclusions (Cowdry type
A) are observed on histopathology. Polymerase chain reac-
tion (PCR) techniques performed in an experienced labo-
ratory also can be used to detect VZV DNA rapidly and
sensitively in properly-collected lesion material, although
VZV PCR testing is not available in all settings. A modifi-
cation of PCR diagnostic techniques has been used at a few
laboratories to distinguish wild-type VZV from the Oka/
Merck strain used in the licensed varicella and zoster
vaccines.
In immunocompromised persons, even when VZV is
detected by laboratory methods in lesion specimens, dis-
tinguishing chickenpox from disseminated zoster might not
be possible by physical examination (72) or serologically
(73–75). In these instances, a history of VZV exposure, a
history that the rash began with a dermatomal pattern,
and results of VZV antibody testing at or before the time
of rash onset might help guide the diagnosis.
Zoster Transmission
Zoster lesions contain high concentrations of VZV that
can be spread, presumably by the airborne route (76,77),
and cause primary varicella in exposed susceptible persons
(77,78–83). Localized zoster is only contagious after the
rash erupts and until the lesions crust. Zoster is less conta-
gious than varicella (78). In one study of VZV transmis-
sion from zoster, varicella occurred among 15.5% of
susceptible household contacts (78). In contrast, following
household exposure to varicella, a more recent study dem-
onstrated VZV transmission among 71.5% of susceptible
contacts (84). In hospital settings, transmission has been
6 MMWR June 6, 2008
documented between patients or from patients to health-
care personnel, but transmission from health-care person-
nel to patients has not been documented. Persons with
localized zoster are less likely to transmit VZV to suscep-
tible persons in household or occupational settings if their
lesions are covered (85).
Epidemiology of Zoster and
Complications
Risk Factors
Infection with VZV
Wild-type VZV. Because zoster reflects reactivation of
latent VZV, the primary risk factor and a necessary precon-
dition for zoster is previous VZV infection. Approximately
99.5% of the U.S. population aged >40 years has serologic
evidence of previous infection, including all evaluated sub-
groups; therefore, all older adults are at risk for zoster (5),
although many cannot recall a history of varicella (86–90).
Varicella vaccine is effective at preventing initial wild-type
VZV infection in persons not previously infected. Any wild-
type VZV infections prevented cannot reactivate as zoster.
The age at the time of initial VZV infection influences
the age at which zoster occurs. Persons acquiring an intrau-
terine or early childhood infection with VZV are at
increased risk for pediatric zoster (91–93). When VZV
infections occur before age 2 months, the risk for zoster
occurring by the age of 12 years is increased >35-fold com-
pared with the risk for VZV infections occurring after
infancy (92). Other case series suggest that the risk for
pediatric zoster also might be increased in children who
experienced varicella at older ages (94). Conversely, the risk
for zoster might be diminished in persons born in coun-
tries (95) or living in communities (96) where varicella
infection tends to occur at later ages. These observations
suggest that changes in the epidemiology of varicella caused
by varicella vaccination or by other factors can alter the
epidemiology of zoster, particularly pediatric zoster.
Oka/Merck Strain VZV. Among vaccine recipients, the
attenuated Oka/Merck strain of VZV included in varicella
vaccine also can establish a latent infection and clinically
reactivate as zoster (97). Zoster caused by Oka/Merck strain
VZV cannot be distinguished on clinical grounds from
zoster caused by wild-type VZV. The risk for zoster caused
specifically by Oka/Merck strain VZV is unknown because
recipients of varicella vaccine might have already been
infected with wild-type VZV or might have become
infected with wild-type VZV following vaccination (i.e.,
due to vaccine failure) that could also reactivate. Therefore,
the rate of all episodes of zoster among varicella vaccine
recipients define the upper bound for the risk of the subset
of episodes caused by Oka/Merck strain VZV. The risk for
zoster in immunocompromised children was approximately
65% less for those who had received the varicella vaccine
compared with those with previous wild-type varicella in-
fection (98,99). In immunocompetent children, the risk
also appears to be reduced among 1-dose vaccine recipi-
ents compared with children with a history of wild-type
varicella, although longer follow up is needed (99–101).
The risk for zoster in immunocompetent children follow-
ing 2 doses of varicella vaccine has not been studied. Col-
lectively these studies suggest that the risk for Oka/Merck
strain zoster following varicella vaccination is no higher,
and likely considerably lower, than that following wild-
type varicella infection, even though the acquisition of the
Oka/Merck VZV through vaccination generally occurs at a
young age (i.e., varicella vaccination is recommended for
children aged
>12 months [1–3]), which might be a risk
factor for pediatric zoster. As varicella vaccine recipients age,
the risk for and manifestation of Oka/Merck strain zoster
in older persons at greater risk for zoster complications can
be evaluated.
Age
Influence on zoster. Age is the most important risk fac-
tor for development of zoster (Figure 3). Virtually all stud-
ies conducted in numerous settings and with various study
designs have indicated an association between age and
increasing zoster incidence, extending to the oldest cohorts
(4,62,95,102–104). One study indicated that zoster inci-
dence increased with age by a factor of >10, from 0.74 per
1000 person years in children aged <10 years to 10.1 per
1000 person years in persons aged 80–89 years, with much
of the increase beginning at age 50–60 years (13). Approxi-
mately 50% of persons who live to age 85 years will have
experienced zoster (105,106).
The important role of age as a risk factor for zoster is
presumably related to a loss of components of VZV-
specific CMI response because of aging (i.e., immune
senescence) possibly combined with waning immunity that
might occur over time following initial varicella infection.
Loss of specific immunity allows VZV to complete the pro-
cess of reactivation and spread to the epidermis to produce
the fully expressed clinical illness (12). Although precise
correlates of protection against zoster have not been identi-
fied, certain CMI responses to VZV antigen decline with
age (21,22,107,108).
7 Vol. 57 / RR-5 MMWR
FIGURE 3. Rate* of zoster and postherpetic neuralgia (PHN)
,
by age — United States
1111
1010
99
88
77
66
55
44
33
22
11
00
20 30 40 50 60 70 >80
Zoster
PHN
Age (yrs)
*Per 1,000 person-years.
Defined as >30 days of pain.
Influence on PHN. Among persons experiencing zoster,
the primary risk factor for the development of PHN is age.
Several studies have indicated that the risk for PHN among
persons with zoster increases with age, particularly for per-
sons aged >50 years (13,35,62,109,110) (Figure 3). In one
study, the risk for experiencing at least 2 months of pain
from PHN increased 27.4-fold among patients aged >50
years compared with those aged <50 years (109). Approxi-
mately 80%-85% of PHN occurs in zoster patients aged
>50 years (62).
Sex
Results from a large, randomized, controlled vaccine trial
in the United States (4) indicated that the incidence of
confirmed zoster cases in a cohort of immunocompetent
persons aged >60 years was 11% higher among the women
(11.8 versus 10.7 cases per 1000 person years in women
and men, respectively). A prospective cohort study in the
Netherlands documented 38% more cases among women
than men (odds ratio = 1.38 [95% confidence interval [CI]
= 1.22–1.56) after controlling for age and other zoster risk
factors (111). Other studies (13,102–104,112) using a
variety of methods also demonstrated an age-standardized
excess of zoster among women. However, some researchers
did not find a difference by sex (36,38,105,113–115).
Women with zoster might also be at increased age-specific
risk for developing PHN compared with men (35,62).
Race
Certain studies have suggested racial differences in the
risk for zoster. In North Carolina, reported lifetime zoster
occurrences and reported incidence were lower in blacks
by 65% and 75%, respectively, compared with whites
after controlling for relevant confounders (115,116). A
study in the United Kingdom indicated that zoster risk in
patients was 54% lower among blacks after adjusting for
age, sex, country of birth, or household childhood contacts
(95). The reasons for these racial differences are unknown.
Geographic or Seasonal Variation
Most studies have not documented a seasonal pattern to
zoster incidence (13,38,92,95,105,117). Certain studies
have reported summer seasonality, particularly for exposed
skin sites. This pattern might be related to ultraviolet irra-
diation that peaks during summer months and might serve
as a trigger for zoster (28,118,119). No studies exist
regarding variation in zoster incidence by latitude. Urban/
rural status does not appear to be a risk factor for zoster
(95).
Altered Immunocompetence
Unlike other vaccine-preventable diseases, zoster epide-
miology is not directly related to exposure but to the biol-
ogy underlying the virus-host relation that allows reactivation
of latent VZV. Because CMI plays a key role in controlling
both development of zoster and the features of its clinical
expression, deficiencies in CMI, regardless of their etiol-
ogy, are risk factors for both zoster and its severe manifesta-
tions. Although the magnitude of zoster risk can be
extremely high among immunocompromised persons, the
overall population attributable risk is modest because
immunosuppression is uncommon (62,103,114).
The incidence of zoster is increased substantially in per-
sons with hematologic malignancies and solid tumors (120).
Rates are highest among children with these conditions.
The magnitude of risk depends on both the nature of the
underlying cancer and the type of treatment (121).
Although the incidence of zoster in patients with solid
tumors is <5%, this rate is many-fold higher than that in
unaffected age-matched persons (120). Patients with
Hodgkins disease are at particularly high risk for zoster,
with cumulative risks during the illness and its treatment
as high as 27.3% (51,53,120,122–127).
Zoster is common following hematopoietic stem cell
transplantation (HSCT); rates are 13%–55% during the
first year (54,128,129). Rates are increased following solid
organ transplants (renal, cardiac, liver, and lung) (5%–
17%). Incidence is highest during the months immedi-
ately following the procedure, and the majority of zoster
cases occur within a year of transplantation (130–132).
8 MMWR June 6, 2008
The risk for zoster and its recurrence is elevated in per-
sons infected with HIV. Zoster rates of 29.4–51.5 per 1000
person years have been reported among HIV-infected
adults, reflecting 12- to 17-fold increase compared with
HIV-negative persons (56,133–136). For HIV-infected
children, the risk is even higher (467 per 1000 person years),
especially among children who acquire VZV infection when
they are profoundly immunosuppressed (137). Most studies
have documented increasing zoster risk as CD4+
T-lymphocyte counts decline, but the risk is increased nine-
fold even among HIV-infected women with CD4+
T-lymphocyte counts >750/µL compared with HIV-
negative controls (135). However, the risk might decline at
CD4+ T-cell counts <50 cells/µL (136). Persons infected
with HIV also are at increased risk for recurrences of zoster.
Other Co-morbidities
The risk for zoster appears to be elevated in persons with
inflammatory diseases; however, for most of these condi-
tions, data are insufficient to determine how much of the
risk is attributable to the underlying disease versus its treat-
ment. Zoster has been associated with systemic lupus
erythematosus (SLE), with rates of 15–91 per 1000 per-
son years (138–143). The risk for zoster also is increased
among persons with rheumatoid arthritis (adjusted hazard
ratio = 1.9 [95% CI = 1.8–2.0]), with an incidence of
approximately 10 cases of per 1000 person years reported
(144,145). Patients with Wegener’s granulomatosis have a
reported incidence of 45 zoster cases per 1000 person years
(146), and recurrences in these patients are common. In
one study, Crohns disease and ulcerative colitis were both
associated with a significantly increased risk for zoster
(incident rate ratios = 1.6 and 1.2, respectively). The
increase was, in part, caused by use of immunosuppressive
medications (147). For all these conditions, zoster is gen-
erally not life-threatening, although cutaneous dissemina-
tion is more common, and deaths have been reported in
such patients (138,141,142).
Certain studies have evaluated the risk for zoster in per-
sons with other noninflammatory co-morbid conditions,
although findings have not been consistent. Two studies
have documented an association between zoster and diabe-
tes mellitus (148,149). However, this association was not
indicated in two other studies (150,151). Another study
documented an increased risk for zoster in persons who
subsequently had multiple sclerosis diagnosed (152).
Exposure to VZV/External Boosting
VZV can be transmitted from zoster lesions to cause pri-
mary varicella in susceptible persons. Although some
experts have suggested that zoster can be caused directly
by exposure to VZV from varicella or from other cases of
zoster (72,153,154), in general, zoster is not associated with
epidemics of varicella. In addition, zoster does not have a
seasonal pattern to suggest it is spread directly from vari-
cella (13,28,38,92,105,117). Theoretically, reactivation of
latent VZV might be triggered by exposure to exogenous
VZV (123,153); however, no evidence suggests that such
episodes occur more frequently than would be expected to
occur by chance.
Conversely, exposure to varicella might reduce the risk
for zoster (13). Protection might be partially maintained
by exposure to varicella circulating in the population and
the resulting exogenous boosting of VZV-specific immu-
nity (15,117,155). An analysis of surveillance data from
the United Kingdom indicated an inverse relation between
annual varicella incidence in children aged <5 years and
zoster incidence in adults aged 15–44 years (117). A case-
control study in the United Kingdom (15) documented a
graded reduction in zoster risk as a function of number of
varicella contacts over a 10-year period. Multivariate analy-
sis suggested a 74% reduction in risk for zoster among per-
sons with three to four varicella exposures compared with
those with no exposures, with a significant trend suggest-
ing some reduction with fewer than three exposures. Social
contacts with children (as a proxy for varicella exposure)
and occupational contact with sick children were protec-
tive (15). A cohort analysis based on data from a sentinel
physician network in the United Kingdom (155) suggested
that adults living with children had both increased vari-
cella exposure and a 25% decrease in zoster incidence. The
analysis estimated that this boosting effect lasted an aver-
age of 20 years (95% CI = 7–41 years). However, persons
living or interacting with children might have different
underlying health compared with persons without expo-
sure to children, which might be a confounder in these
studies. Other evidence that varicella exposure might pro-
tect against zoster includes possible effects household
exposure to varicella had against subsequent development
of zoster among children with leukemia (156). Finally, the
efficacy of the zoster vaccine (4) supports the concept that
exposure to exogenous VZV can reduce risk for zoster, pre-
sumably by boosting specific immunity against VZV.
Contrary evidence also exists that varicella exposure does
not reduce the risk for zoster. Women are at greater risk for
zoster (13,102,103,110,111) despite the fact that women
probably have more exposure to young children who expe-
rience varicella. A Japanese study indicated that the risk for
zoster in children was not diminished by repeated varicella
exposures (92).
9 Vol. 57 / RR-5 MMWR
Although a sufficient number of varicella exposures could
reduce the risk for zoster in select populations, it is unclear
whether such levels of exposure play an epidemiologically
important role in reducing the risk for zoster among the
general population of older adults who are at the highest
risk for the disease, and, if so, how long such effects would
last in the elderly.
Other Risk Factors
As with orofacial and genital flairs of HSV, zoster has been
anecdotally linked to stress. However, only two rigorous
evaluations of the role of psychological stress on zoster have
been conducted. A case-control study documented a sig-
nificant association with number of stressful life events
within 6 months of reported zoster (p = 0.012) (157). A
prospective cohort study indicated a nonsignificant asso-
ciation (p = 0.078) between zoster risk and negative life
events.
Trauma or surgery could lead to reactivation of VZV in
the affected dorsal root ganglion and increase the risk for
zoster rash in that dermatome. Such a development would
seem to be specific and easily ascertained, and certain
reports and case series describe such events (158–161). One
case-control study collected information about recent trauma
and/or surgery in patients who developed zoster and in
matched controls. The frequency of trauma in nonzoster
sites was similar between the two groups, but zoster
patients were significantly more likely than controls to
report trauma at the site of their zoster during the month
before zoster onset (adjusted OR = 12.1 [95% CI = 1.5–
97.6]; p = 0.002) (24). The basis by which these stimuli
provoke zoster is unclear, but they suggest that
nonimmunologic factors can play important roles in the
pathophysiology of zoster.
Finally, one study indicated that dietary micronutrient
intake was protective against zoster. Body mass index did
not appear to be associated with zoster risk (162). Genetic
predisposition for zoster also has been reported (163).
Population Rates of Zoster and PHN
Zoster
Zoster is not a reportable condition in the United States;
therefore, incidence has been inferred from a variety of stud-
ies. Observed rates have varied substantially on the basis of
methods for case ascertainment, access to health care, and
case definitions. The age distribution in the population
being studied also is an important consideration when com-
paring these studies because zoster can vary dramatically
across study sites. Conclusions cannot be drawn from cross-
study comparisons without adjusting for age or comparing
age-specific rates directly. Differences in the prevalence of
immunosuppression or in racial makeup also can influence
population-wide zoster incidence. In addition, the incidence
of zoster appears to have been increasing over recent decades,
even after adjusting for other factors, although this increase
has not been observed consistently.
Despite these limitations, certain analyses of zoster inci-
dence in the United States have been conducted. The inci-
dence in all studies ranged from 3.2–4.2 per 1000
population per year (age-adjusted to the 2000 U.S. popu-
lation) (62,103,104,114,164,165), translating into an
estimated 1 million cases annually. In all studies, a sub-
stantial increase in zoster incidence occurred with age and
extended to the oldest strata; for all persons aged
>60 years,
the annual incidence was approximately 10 per 1000 per-
sons (62,103,104,114,164,165), similar to the annual
incidence of 11.1 per 1000 observed during the zoster vac-
cine trial (4). On the basis of these data, an estimated 32%
of persons in the United States will experience zoster dur-
ing their lifetime (CDC unpublished data, 2007).
Certain studies provide evidence of increasing age-
specific zoster incidence in the United States
(38,62,165,166), although other studies have shown no
such trend (104). The observed increases cannot be solely
attributed to changes in the epidemiology of varicella,
because documentation of increases predated licensure of
varicella vaccine in the United States in 1995 (38) and
because age-specific increases over time also are being
reported in certain international settings, including in the
absence of varicella vaccination programs (105,167,168).
Because the basis for this increase remains unclear, predict-
ing whether the age-specific risk for zoster will continue to
increase in the future is difficult.
Recurrent Zoster
Effectively evaluating the risks for recurrent zoster
(i.e., second or subsequent episodes) in immunocompetent
persons requires large populations, long-term follow up,
adequate duration, and laboratory confirmation. Although
data are limited, certain studies suggest a recurrence rate
that is comparable to the rate of initial episodes (13,38,114).
A community-based study of clinician-diagnosed zoster was
conducted in Olmsted County, Minnesota. The observa-
tional period lasted 6 years. Of 1,669 persons that experi-
enced an episode of zoster during that period, 24 experienced
a second episode, suggesting a high incidence of zoster
recurrence and providing no evidence that an episode of
zoster protects against recurrence (62). Similar observations
were noted in an older survey-based study (169). In the
Shingles Prevention Study, two of approximately 20,000
10 MMWR June 6, 2008
vaccine placebo recipients had two episodes of zoster within
3 years of the initial episode. These cases provide the first
laboratory-confirmed evidence that zoster can recur in
immunocompetent persons, even soon after the initial
episode (4).
Zoster Hospitalizations and Deaths
Hospitalizations. Conclusions about hospitalization for
zoster should be interpreted carefully if they are derived
from administrative data. Hospital administrative data
often do not distinguish zoster episodes that were reasons
for hospitalizations from those episodes that were inciden-
tal to the hospitalization or that occurred during prolonged
hospital stay. PHN at the time of an unrelated hospitaliza-
tion also might be coded as zoster. In addition, underlying
immunosuppressive conditions might not be available or
might not be collected from administrative data. These fac-
tors preclude determination of the portion of hospitaliza-
tions that could be prevented by a live-attenuated vaccine
that is contraindicated for immunosuppressed persons.
Given these limitations, crude annual rates of zoster hos-
pitalization have ranged from 2.1 per 100,000 population
in a Northern California managed care population (170)
to 4.4 per 100,000 population in England (171). A crude
rate of 16.1 per 100,000 population was identified in an
analysis of Connecticut-wide hospitalization data that
included all zoster episodes, not just primary discharge
diagnosis (172).
In a community-based study in Olmsted County, Min-
nesota, approximately 3% of patients with zoster were hos-
pitalized for the illness (62). Although values differ
substantially, all studies indicate that zoster hospitaliza-
tion rates increase with age (170,172–174). In the Con-
necticut study, zoster hospitalization rates were
approximately 75-fold greater among persons aged >85 years
than in persons aged <30 years (172). Although precise
denominators are not available, risks for hospitalization also
increased among persons with altered immunocompetence;
approximately 30% of all persons hospitalized with zoster
episodes had one or more immunocompromising condi-
tions, primarily malignancies (82%) and HIV infection
(8%) (62,172). Central nervous system and ophthalmo-
logic complications accounted for most of the reported com-
plications among hospitalized zoster cases (172–174),
although bacterial superinfection was common in one
series (175). Another study indicated that 0.5% of patients
with confirmed zoster were hospitalized before their zoster
rash developed for prodromal pain syndromes including
suspected myocardial infarction, severe new-onset headache,
back pain, and abdominal pain resulting in appendectomy
(62).
Deaths. On the basis of clinical experience and in the
absence of zoster-related deaths in cohort studies, certain
experts believe that zoster mortality appears to be uncom-
mon, particularly among healthy persons (176). Vital
records might not distinguish deaths attributed to zoster
from incidental deaths occurring merely in the presence of
zoster, and they might not capture information on the im-
munologic status among those deaths. An Australian study
using administrative data indicated that 1% of patients hos-
pitalized with a primary zoster diagnosis died; the number
of deaths directly attributable to zoster was not validated
(174). Certain analyses have indicated that almost all zoster
deaths occur in the elderly, with a rate >10-fold higher
among persons aged >65 years (171,173,174). Immuno-
suppression also appears to be a risk factor for zoster mor-
tality. In one study, 52% of patients hospitalized with zoster
who died had one or more immunocompromising condi-
tion (e.g., malignancies, leukemia, and HIV). In that study,
the risk for death in persons with immunocompromising
conditions was 8.7%; the risk in persons without these
conditions was 3.7% (172).
PHN
Drawing conclusions from studies on the risk for PHN is
difficult because definitions for PHN vary and results are
influenced by many factors, including the source and age
of the study population. Among zoster patients treated with
a placebo in clinical trials of antiviral drugs, approximately
one third still had pain after 3 months and approximately
one fourth had pain at 6 months (177,178). However, these
trials might include a population of patients with more
severe zoster pain, thereby introducing a detection bias that
could inflate estimated risks for PHN. In a phase 3 clinical
trial of zoster vaccine (4), zoster occurred among 642
placebo recipients; the risk for pain persisting at least 30,
60, 90, 120 or 180 days among these person was 30.3%,
17.6%, 12.5%, 8.4%, and 5.1%, respectively. Results from
the trial might not reflect risks for progression to PHN in
community settings because ascertainment, diagnosis, and
antiviral treatment of zoster were standardized and thor-
ough. However, in a community-based study in Olmsted
County, Minnesota, in which almost all medical events were
captured, the risk for PHN in patients with zoster was 18%,
13%, and 10% when defining PHN as at least 30, 60, and
90 days of pain, respectively (62).
11 Vol. 57 / RR-5 MMWR
Treatment and Nonspecific Management
of Zoster and PHN
The treatment of acute zoster, the prevention of PHN
development among patients with acute zoster, and the
treatment of patients with current PHN are complex clini-
cal problems with ongoing uncertainties and active research
(31). Acyclovir, famciclovir, and valacyclovir are approved
by the FDA for treatment of zoster in immunocompetent
patients. All three are nucleoside analogs that inhibit rep-
lication of human herpes viruses, including VZV. Clinical
trials have indicated that these agents, taken orally, reduce
the duration of viral shedding and lesion formation,
reduce the time to rash healing, and decrease the severity
and duration of acute pain from zoster and the risk for pro-
gression to PHN. Because all three antiviral agents are safe
and well tolerated, many experts recommend that treat-
ment should be considered for all eligible patients with
zoster, and specifically recommend treatment for persons
aged >50 years who have moderate or severe pain, moder-
ate or severe rash, or involvement of nontruncal dermatomes
(31). In clinical trials, treatment has been initiated within
72 hours of rash onset, a biologically arbitrary time point
that often is not feasible in clinical practice. The benefits of
later treatment have not been studied (31). If treatment
cannot be initiated within 72 hours of rash onset, experts
recommend that it should be initiated as soon as possible,
particularly in the presence of new vesicle formation or of
complications.
Two clinical trials have assessed the role of corticoster-
oids in combination with acyclovir for treatment of zoster
and prevention of subsequent PHN (179,180). Patients at
risk for steroid-related toxicities (e.g., those with diabetes
mellitus or gastritis) were excluded from the trials. A 3-week
tapering course of corticosteroids diminished acute zoster
pain and decreased the time to cutaneous healing,
cessation of analgesic therapy, and return of uninterrupted
sleep and normal daily activities. However, no evidence
indicated that use of corticosteroids prevented development
of PHN. Theoretically, corticosteroids should be equally
effective in combination with valacyclovir or famciclovir;
however, combinations of these agents have not been stud-
ied in clinical trials. No evidence indicates that topical
antiviral therapy or corticosteroids without systemic
antiviral therapy have a role in treatment of zoster.
A variety of approaches have been used with varying
degrees of success for control of acute zoster pain, includ-
ing acetaminophen, nonsteroidal anti-inflammatory agents,
tricyclic antidepressants, opiates, anticonvulsants, capsaisin,
and topical anesthetics (31). In more severe instances of
pain, referral to a pain specialist, or even hospitalization
and administration of epidural analgesics, is often consid-
ered. Many of these same modalities are used with varying
degrees of success for control of chronic PHN pain
(26,181,182). Elderly persons, who already have reduced
physiologic reserve and typically take multiple medications
for pre-existing chronic conditions, might be unable to
tolerate psychotropic and other medications for manage-
ment of their acute zoster or chronic PHN pain (31,33).
Patients with uncomplicated zoster should be advised to
keep the rash clean and dry, to avoid topical antibiotics,
and, if possible, to keep the rash covered. They should alert
their physician if the rash worsens or they have fever, which
could indicate bacterial superinfection (31).
Prevention of Transmission from Zoster
Some health-care institutions might exclude personnel
with zoster from work until their lesions dry and crust (85).
Persons with localized zoster should avoid contact with
susceptible persons at high risk for severe varicella in
household and occupational settings until lesions are crusted.
Such persons include pregnant women, all premature
infants born to susceptible mothers, infants born at
<28 weeks’ gestation or who weigh <1000 g regardless of
maternal immune status, and immunocompromised per-
sons of all ages (85). Persons with opportunities for contact
with such high risk-persons in household or occupational
settings should be informed about how to recognize the
signs and symptoms of zoster. If a person susceptible to
varicella infection has close exposure to a persons with zoster,
postexposure prophylaxis with varicella vaccine or
VARIZIG
should be considered (3,85,183).
Zoster Vaccine
Vaccine Composition, Dosage, and
Administration
The zoster vaccine licensed in the United States
(ZOSTAVAX
®
, Merck & Co., Inc.) is a lyophilized prepa-
ration of the Oka/Merck strain of live, attenuated VZV,
the same strain used in the varicella vaccines (VARIVAX
®
,
PROQUAD
®
). The Oka strain was isolated in Japan (184)
in the early 1970s from vesicular fluid from a healthy child
who had varicella; the strain was attenuated through
sequential propagation in cultures of human embryonic
lung cells, embryonic guinea-pig cells, and human diploid
cells (WI-38). Further passage of the virus was performed
at Merck Research Laboratories in human diploid cell
12 MMWR June 6, 2008
cultures (MRC-5). The cells, virus seeds, virus bulks, and
bovine serum used in the manufacturing are all tested to
provide assurance that the final product is free of adventi-
tious agents.
Zoster vaccine, when reconstituted as directed in the
package label using the supplied diluent, is a sterile prepa-
ration for subcutaneous administration. Each 0.65-mL dose
contains a minimum of 19,400 PFU (4.29 log
10
) of Oka/
Merck strain of VZV when reconstituted and stored at room
temperature for up to 30 minutes. Zoster vaccine is similar
to VARIVAX
®
. However, its minimum potency is at least
14-times the potency of VARIVAX
®
, which contains a mini-
mum of 1,350 (approximately 3.13 log
10
) PFU.
PROQUAD
®
contains 3.993 log
10
PFU, similar in
potency to ZOSTAVAX
®
. Each dose of zoster vaccine also
contains additional VZV antigenic component from non-
viable Oka/Merck VZV. Additional vaccine components in
each dose include 31.16 mg of sucrose, 15.58 mg of
hydrolyzed porcine gelatin, 3.99 mg of sodium chloride,
0.62 mg of monosodium L-glutamate, 0.57 mg of so-
dium phosphate dibasic, 0.10 mg of potassium phosphate
monobasic, 0.10 mg of potassium chloride; residual com-
ponents of MRC-5 cells including DNA and protein; and
trace quantities of neomycin and bovine calf serum. The
product contains no thimerosal or other preservatives.
Zoster vaccine should be administered as a single
0.65-mL dose subcutaneously in the deltoid region of the
upper arm; a booster dose is not licensed for the vaccine.
The vaccine should not be injected intravascularly or
intramuscularly and should only be reconstituted and
injected using a sterile syringe free of preservatives, anti-
septics, and detergents, which can inactivate the vaccine
virus.
Storage and Handling
To maintain potency, lyophilized zoster vaccine must be
stored frozen at an average temperature of <5°F (<-15°C)
until it is reconstituted for injection. Any freezer that has a
separate sealed freezer door and reliably maintains an
average temperature of <5°F (<-15°C) is acceptable for stor-
ing zoster vaccine. Providers should check the adequacy of
their freezer by verifying its temperature before obtaining
vaccine. In general, the freezer compartments of dormitory
style units are incapable of reliably maintaining tempera-
tures cold enough to store zoster vaccine and are unaccept-
able for storage. For certain refrigerator/freezer models, it is
necessary to reduce the temperature to the coldest setting
to maintain zoster vaccine at the correct temperature. How-
ever, this might reduce the temperature in the refrigerator
compartment and result in freezing of any vaccines or other
pharmaceutical products being refrigerated. As a result, both
the refrigerator and freezer temperatures should be moni-
tored and the temperature recorded at least twice a day.
Any out-of-range temperature readings require immediate
and documented corrective action. When a freezer is tem-
porarily unavailable (e.g., during transport or equipment
failure), zoster vaccine should be stored in a suitable con-
tainer (i.e., the original shipping container or a compa-
rable container with a properly fitting lid) with an adequate
quantity of dry ice (i.e., a minimum of six pound per box)
so that dry ice would persist in the container if
unreconstituted vaccine must be transported back to the
freezer. Dry ice placed in a suitable container will maintain
a temperature of <5°F (<-15°C). The diluent, which does
not contain preservative or other antiviral substances that
could inactivate the vaccine virus, should be stored sepa-
rately, either at room temperature or in the refrigerator.
The vaccine should be reconstituted according to the
directions in the package label and only with the diluent
supplied. Before reconstitution, zoster vaccine should be
protected from light. Once reconstituted, the vaccine should
be used immediately to minimize loss of potency. The vac-
cine must be discarded if not used within 30 minutes after
reconstitution. Information regarding stability under con-
ditions other than those recommended is available from
the manufacturer at 800-637-2590.
Efficacy
The efficacy of zoster vaccine was evaluated in a phase 3
vaccine trial termed the Shingles Prevention Study, a double-
blind randomized, placebo-controlled trial involving
38,546 healthy adults aged >60 years who had a history of
varicella or at least 30 years of residence in the continental
United States (as a marker of previous infection). Persons
excluded from the trial included those with a history of
zoster, with allergies to components of the vaccine, with
immunocompromising conditions, or with conditions that
might have interfered with study evaluations (e.g., cogni-
tive impairment, <5 year life expectancy, dermatologic dis-
orders, chronic pain, hearing loss, or lack of mobility). The
study population ranged in age from 59–99 years (median:
69.4 years), and comprised 41.0% females, 95.4% white,
2.1% blacks, 1.3% Hispanics, and 1.2% other or unknown
race/ethnicity. On enrollment, approximately 90% of the
participants had at least one underlying chronic medical
condition.
Persons were randomized to receive a single subcutane-
ous dose of zoster vaccine or placebo; the mean duration of
13 Vol. 57 / RR-5 MMWR
follow up was 3.1 years. Active case ascertainment was con-
ducted through monthly telephone contact supplemented
by a close-out interview. Zoster cases were confirmed by
PCR testing (93%), viral culture (1%), or evaluation by a
panel of five physicians with expertise in zoster diagnosis
(6%). Patients with confirmed zoster were followed for at
least 182 days to assess the outcome of the condition,
including presence and severity of pain. Approximately 95%
of persons were followed to completion of the study. Out-
comes evaluated included incidence of zoster, incidence of
PHN (defined as pain level of three or more [on a numeri-
cal rating scale of 0-10] persisting at least 90 days after
rash onset), and burden of illness (BOI), measured using a
mean value of severity-by-duration index for each treatment
group, thus incorporating the incidence, severity, and
duration of pain and discomfort from zoster). A total of
957 confirmed cases of zoster occurred among study par-
ticipants: 315 among vaccine recipients and 642 among
placebo recipients. The proportion of vaccine and placebo
recipients that received antiviral treatment within 72 hours
of rash onset, as clinically indicated, was 64.1% and 65.9%,
respectively.
The vaccine reduced the risk for developing zoster by
51.3% (95% CI = 44.2–57.6; p<0.001 (Table 2) (4). The
vaccine was 66.5% (95% CI = 47.5–79.2; p<0.001) effi-
cacious for preventing PHN. When the definition of PHN
was changed from 30 days of pain to 182 days of pain
following rash onset, vaccine efficacy increased from 58.9%
to 72.9% (Table 3). Zoster vaccine had an independent
effect of reducing PHN among patients who developed
zoster (39% [95% CI = 7%–59%]) (Table 2). The mean
severity-by-duration of zoster was reduced by 57%
(p = 0.016) in vaccine recipients who developed PHN.
Zoster vaccine reduced BOI by 61.1% (95% CI = 51.1–
69.1; p<0.001) (Table 2). The vaccine reduced the degree
of interference in activities of daily living (ADLI) caused
by zoster, in part because of the reduction in zoster itself,
but also because of a decrease in ADLI among those vac-
cine recipients who did develop zoster (185). No evidence
indicated that vaccine recipients experiencing zoster were
protected from other sequelae such as scarring, bacterial
superinfection, palsies, or ocular or visceral complications
(186).
In general, with increasing age at vaccination, the vac-
cine retained efficacy against severity of zoster better than
against zoster itself. Thus, efficacy for the prevention of
zoster was highest among persons aged 60–69 years and
declined with increasing age (Table 2). Declines in efficacy
of preventing zoster were observed with each 5-year increase
in age throughout the age range of participants (187).
However, no significant differences were observed among
persons aged 60–69 years versus those aged
>70 years in
vaccine efficacy at reducing BOI or PHN, probably
because the independent effect of reducing PHN among
patients who developed zoster was greatest among persons
aged 70–79 years (Table 2). For persons aged >80 years,
efficacy against zoster was 18% (Table 2), but efficacy against
PHN (39%) was better retained (186). No significant dif-
ferences by sex were observed in the efficacy of the vaccine
at reducing BOI, PHN, or zoster (4). No evidence indi-
cated that the vaccine was less efficacious for prevention of
zoster (vaccine efficacy: 51.6%; 95% CI = 41.4–60.1),
PHN (vaccine efficacy: 60.9%; 95% CI = 31.3–78.7), or
for reduction in BOI (vaccine efficacy: 60.1%; 95% CI =
46.1–70.4) among subjects with functional limitations
(188).
TABLE 2. Efficacy of ZOSTAVAX
®
compared with a placebo, by age group — Shingles Prevention Study*
ZOSTAVAX
®
Placebo Efficacy
% HZ % HZ
cases cases PHN
§
Burden of
Age group No. HZ PHN with No. HZ PHN with HZ PHN
§
with HZ illness (BOI)
(yrs)
subjects cases cases
§
PHN
§
subjects cases cases
§
PHN
§
% (95% CI**) % (95% CI) % (95% CI) % (95% CI)
60–69 10,370 122 8 6.6 10,356 334 23 6.9 64 (56–71) 65.7 (20.4–86.7) 5 (-107–56) 65.5 (51.5–75.5)
70–79 7,621 156 12 7.7 7,559 261 45 17.2 41 (28–52) 66.8 (43.3–81.3) 55 (18–76) 55.4 (39.9–66.9)
>80 1,263 37 7 18.9 1,332 47 12 25.5 18 (-29–48)
26 (-69–68)
Total 19,254 315 27 8.6 19,247 642 80 12.5 51 (44–58) 66.5 (47.5–79.2) 39
††
(7–59) 61.1 (51.1–69.1)
* The analysis was performed on the Modified Intent-To-Treat (MITT) population that included all subjects randomized in the study who were followed for at least 30 days
postvaccination and did not develop an evaluable case of herpes zoster (HZ) within the first 30 days postvaccination.
Age strata at randomization were aged 60–69 years and >70 years.
§
Postherpetic neuralgia (PHN) was defined as HZ-associated pain rated as three or more, on a scale ranging from 0 (no pain) to 10 (pain as bad as you can imagine), persisting
or appearing more than 90 days after onset of HZ rash using Zoster Brief Pain Inventory.
VE for PHN and BOI calculated for the age groups 60–69 and >70 years.
** Confidence interval.
††
Age-adjusted estimate based on the age strata (age 60–69 years and >70 years) at randomization.
Sources: Oxman MN, Levin MJ, Johnson GR, et al. Zoster Prevention Study Group. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med
2005;352:2271-84; ZOSTAVAX
®
. Package Insert. Merck & Co., Inc, Whitehouse Station, NJ 08889.
14 MMWR June 6, 2008
TABLE 3. Efficacy of zoster vaccine on the incidence of postherpetic neuralgia (PHN) among persons aged >60 years, by
duration of pain* — Shingles Prevention Study
Vaccine group Placebo group Efficacy
Persistence of No. confirmed Incidence No. confirmed Incidence
PHN among
all subjects
cases of HZ
with PHN
per 1,000
person/yr
§
cases of HZ
with PHN
per 1,000
person/yr
§
% (95% CI**)
30 days 81 1.39 196 3.39 58.9 (46.6–68.7)
60 days 45 0.77 113 1.96 60.4 (43.6–72.6)
90 days 27 0.46 80 1.38 66.5 (47.5–79.2)
††
120 days 17 0.29 54 0.93 68.7 (45.2–83.0)
182 days 9 0.16 33 0.57 72.9 (42.1–88.6)
* For the secondary end point, PHN was defined as the pain and discomfort associated with herpes zoster rated as three or more, on a scale ranging
from 0 (no pain) to 10 (pain as bad as you can imagine), persisting or appearing more than 90 days after the onset of herpes zoster rash. Efficacy
analyses were performed with the use of a follow-up interval that excluded the first 30 days after vaccination and the modified intention-to-treat
population, which excluded persons who withdrew or in whom a confirmed case of herpes zoster developed, within the first 30 days after vaccination.
Of the three persons who developed more than 1 confirmed case of herpes zoster, only the first case was included.
PHN was defined as the pain and discomfort associated with herpes zoster that was rated as three or more persisting or appearing more than 30, 60,
90, 120, and 182 days after the onset of herpes zoster rash.
§
For the total population and the subgroups stratified according to sex, the incidence of PHN in each treatment group (vaccine or placebo) was the
weighted average of the observed incidence of PHN stratified according to age group, with weights proportional to the total number of person-years of
follow-up in each age group.
Vaccine efficacy for the incidence of PHN and 95% confidence interval (CI).
** Confidence interval.
††
Vaccine efficacy for the incidence of PHN for all persons was the protocol-specified secondary end point.
Source: Oxman MN, Levin MJ, Johnson GR, et al. Zoster Prevention Study Group. A vaccine to prevent herpes zoster and postherpetic neuralgia in older
adults. N Engl J Med 2005;352:2271–84.
Twelve clinical lots of zoster vaccine were used in the
Shingles Prevention Study, nine of which were heat treated
to accelerate aging of the vaccine. Potency upon shipment
to study sites ranged from 21,000–62,000 PFUs/dose, but
potency and accelerated aging did not significantly influ-
ence vaccine efficacy with regard to zoster, PHN, or BOI.
Immunogencity
A substudy of the Shingles Prevention Study was con-
ducted among 1,395 persons to assess VZV-specific
immunity at baseline and 6 weeks following administra-
tion of zoster vaccine or placebo. The longer-term duration
of immunogenicity also was assessed. Anamnestic antibody
response was evaluated using gpELISA to measure increases
in VZV antibody levels after vaccination. RCF and IFN-γ
ELISPOT were used to measure the number of memory
T-cells. With all three assays, VZV-specific immunity mea-
sured 6 weeks after vaccination increased following receipt
of vaccine but not placebo. In both vaccine and placebo
recipients, immune responses were inversely related to the
risk for developing zoster; this association with protection
was greatest for anamnestic antibody response following
vaccination for which gpELISA Geometric Mean Titers
(GMTs) increased 1.7-fold (95% CI = 1.6–1.8). However,
for all three assays, no threshold level of immunity that
predicted complete protection from zoster was observed.
No clear dose response for increases in GMTs was
observed; similar increases were achieved in vaccine
recipients throughout the dosage range used in the Shingles
Prevention Study (189). Peak CMI responses were present
1–3 weeks following vaccination (187,190,191), as would
be expected for anamnestic responses that would occur in
persons with previous VZV infection. The impact of age
on CMI response to vaccination also was evaluated. RCF
and IFN-γ ELISPOT responses were greater in persons aged
60–69 years than in persons aged >70 years (p<0.01) (192).
The increase in GMTs as a measure of anamnestic anti-
body response in persons aged 50–59 years was compa-
rable to that in persons aged >60 years (193). In a
prelicensure study, subjects aged 55–70 years acquired
VZV-specific class I-restricted and unrestricted cytotoxic-
ity following vaccination with even low levels (4,000 PFUs)
of either live or heat-inactivated Oka/Merck strain of VZV
(23).
Duration of Efficacy and of Immunity
Vaccine efficacy for zoster prevention declined during the
first year following vaccination, but remained stable through
the remaining 3 years of follow up (Figure 4). Vaccine effi-
cacy for PHN prevention had a similar pattern, with an
initial decline and subsequent stabilization. After conclu-
sion of the Shingles Prevention Study, approximately 7,500
vaccine recipients will be followed to extend observation to
10 years. Because placebo recipients were offered zoster
vaccine at the conclusion of the Shingles Prevention Study,
zoster rates in these 7,500 persons will be compared with
15 Vol. 57 / RR-5 MMWR
FIGURE 4. Duration of zoster vaccine efficacy for preventing
zoster and postherpetic neuralgia (PHN)
HZ PHN
100
50
0
Vaccine efficacy
95% CI
-50
-100
0 1 2 3 4 0 1 2 3 4
Time since the start of follow-up (yrs)
historic controls. Increases in RCF and IFN-γ ELISPOT
responses persisted for 3–6 years following vaccination
(192,194).
Safety and Adverse Events
Serious Adverse Events
Adverse events were monitored in the Shingles Prevention
Study population, with more comprehensive ascertainment
in a safety substudy comprising 6,616 persons (3,345 vac-
cine recipients and 3,271 placebo recipients) (Table 4). In
the Shingles Prevention Study population, the number and
types of serious adverse events (4) during the 42 days after
receipt of vaccine or placebo were similar (1.4%). How-
ever, rates of serious adverse events in the safety substudy
were higher in vaccine recipients (1.9%) than in placebo
recipients (1.3%), with a relative risk of 1.5 (95% CI =
1.0–2.3). Nonetheless, no temporal or clinical patterns of
adverse events were observed in vaccine recipients to sug-
gest a causal relation (4,186). The incidence of death and
hospitalizations was similar in the two treatment groups
throughout the observation time (4,186).
Mild Local and Systemic Reactions
In the Shingles Prevention Study safety substudy, self-
reported injection site adverse events (e.g., erythema, pain,
swelling, warmth, and pruritis) were more common among
vaccine recipients (48.3%) than placebo recipients (16.6%)
(p<0.05) (Table 4) (4); the risk for these events was higher
in vaccine recipients aged 60–69 years (58.3%) than in
persons aged >70 years (41.3%) (189). Most injection site
adverse events were mild and resolved within 4 days (187).
Less-serious systemic adverse events, including headaches,
were more common in vaccine recipients (6.3%) than in
placebo recipients (4.9%) (p<0.05) (Table 4) (4). The risk
for fevers after vaccination did not differ between vaccine
recipients and controls.
The safety and tolerability of zoster vaccine was evalu-
ated in a separate study among persons aged 50–59 years,
including 62 persons who received the standard potency
(approximately 58,000 PFUs) and 123 persons who
received high potency (approximately 207,000 PFUs)
(195). Although the numbers of persons was small, both
vaccines were safe and well tolerated; however, injection
site reactions were more common (69.4% and 82.9%,
respectively) than those observed in person aged >60 years
in the Shingles Prevention Study (48.3%).
Vaccine Virus Rash and Transmission
Varicella-like rashes, including injection site varicella-like
lesions, generalized varicella-like rashes, and zoster-like
rashes, were evaluated in the Shingles Prevention Study
during the first 42 days of observation (Table 4). Twenty
vaccine recipients and seven placebo recipients had lesions
at the injection site (p<0.05) (4); the lesions were tested
for VZV by PCR in one of these persons in each group,
and results were negative in both. Among the vaccine
recipients, lesions occurred a median of 3–4 days after
vaccination and lasted a median of 5 days.
Generalized varicella-like rashes occurred at similar rates
in the two groups (Table 4). Zoster-like rashes were less
common in vaccine versus placebo recipients during this
42-day period (p<0.05). Oka/Merck strain VZV was not
detected in any of 10 lesion specimens from vaccine recipi-
ents available for PCR testing. In early studies conducted
as part of the manufacturer’s clinical program for develop-
ment of zoster vaccine, samples from rashes in two vacci-
nated persons were confirmed to be Oka/Merck-strain VZV
(186). Both experienced noninjection-site varicella-like
rashes; one had 21 lesions on day 17 lasting 8 days and the
other developed five lesions on day 8 that lasted 16 days.
No varicella-like rashes were documented during any clini-
cal zoster vaccine trials of laboratory-confirmed zoster
attributed to Oka/Merck strain VZV. In addition, no evi-
dence existed of transmission of vaccine virus from vaccine
recipients to contacts.
The Economic Burden of Zoster and
Cost-Effectiveness of Vaccination
The economic burden of zoster in the elderly is substan-
tial and includes direct costs attributed to health-care use
and indirect costs attributed to losses in productivity from
temporary or more permanent disability. In addition,
16 MMWR June 6, 2008
TABLE 4. Adverse events among all persons and among persons in an adverse-events substudy*
Vaccine group Placebo group Difference in risk
Event No. (%) No. (%) % (95% CI
)
No. in group 19,270 19,276
Day of vaccination to end of study
Death 793 (4.1) 795 (4.1) 0.01 (-1.2–1.2)
§
Death by age group (yrs)
60–69 218 (2.1) 246 (2.4) -0.80 (-2.0–0.4)
§
> 7 0 575 (6.5) 549 (6.2) 0.95 (-1.2–3.1)
§
Vaccine-related serious adverse event
2 (<0.1) 3 (<0.1) NC
Day of vaccination to day 42
Death 14 (0.1) 16 (0.1) -0.01 (-0.1–0.1)
>1 serious adverse events 255 (1.4) 254 (1.4) 0.01 (-0.2–0.3)
Varicella-like rash at injection site 20 (0.1) 7 (0.04) 0.07 (0.02–0.13)**
Varicella-like rash not at injection site 18 (0.1) 14 (0.1) 0.02 (-0.04–0.09)
Herpes zoster-like rash 17 (0.1) 36 (0.2) -0.10 (-0.18– -0.03)**
Rash unrelated to herpes zoster 595 (3.2) 620 (3.3) -0.13 (-0.49–0.23)
Confirmed case of herpes zoster 7 (<0.1) 24 (0.1) -0.09 (-0.16– -0.03)**
Persons in the adverse event substudy 3345 3271
Day of vaccination to end of study
Person hospitalized 1,137 (34.0) 1,115 (34.1) 0.1 (-8.8–9.0)
§
Hospitalization related to herpes zoster 5 (0.2) 6 (0.2) -0.1 (-0.7–0.5)
§
Day of vaccination to day 42
>1 serious adverse events 64 (1.9) 41 (1.3) 0.7 (0.1–1.3)**
>1 adverse events 1,929 (58.1) 1,117 (34.3) 23.7 (21.3–26.0)**
>1 systemic adverse events 820 (24.7) 768 (23.6) 1.0 (-1.0–3.1)
>1 vaccine-related systemic adverse events
209 (6.3) 160 (4.9) 1.4 (0.3–2.5)**
Documented temperature >38.3
o
C (>100.9
o
F) 27 (0.8) 27 (0.9) 0 (-0.5–0.4)
Self-reports of feeling abnormal temperature
††
231 (7.2) 190 (6.0) 1.2 (0.0–2.4)
>1 adverse event at injection site
§§
1,604 (48.3) 539 (16.6) 31.7 (28.3–32.6)**
Erythema 1,188 (35.8) 227 (7.0) 28.8 (26.9–30.6)**
Pain or tenderness 1,147 (34.5) 278 (8.5) 26.0 (24.1–27.9)**
Swelling 871 (26.2) 147 (4.5) 21.7 (20.1–23.4)**
Pruritus 237 (7.1) 33 (1.0) 6.1 (5.2–7.1)**
Warmth 57 (1.7) 11 (0.3) 1.4 (0.9–1.9)**
Hematoma 53 (1.6) 46 (1.4) 0.2 (-0.4–0.8)
R a s h 10 (0.3) 3 (0.1) 0.2 (0.0–0.5)
* The rates of death and of hospitalization are percentages of persons in each treatment group. Otherwise, percentages are rates weighted in proportion
to the number of persons with safety follow-up in each age group. NC denotes not calculated. Three persons who had withdrawn from the study because
of worsening health and subsequently died were included in the safety analysis.
Confidence interval.
§
The difference in risk (vaccine group versus placebo group) and the 95% confidence intervals (CIs) for deaths and hospitalizations are based on the
rates per 1000 persons-years of follow-up to account for differential follow-up among the study participants as a result of staggered enrollment.
Otherwise, the differences in risk and 95% CIs are based on an asymptotic method for the difference of two binomial proportions where the proportions
are weighted according to the number of persons with safety follow-up in each age group. Negative values for the difference in risk result when the rate
in the placebo group is larger than that in the vaccine group.
Events classified as possibly related to vaccination were assessed by a blinded investigator at each site.
** P<0.05 for the comparison with the placebo group.
††
A temperature of >38.3
o
C (>100.9
o
F) was not documented.
§§
None of the adverse events related to the injection site were considered to be serious adverse events.
Source: Oxman MN, Levin MJ, Johnson GR, et al. Zoster Prevention Study Group. A vaccine to prevent herpes zoster and postherpetic neuralgia in older
adults. N Engl J Med 2005;352:2271–4.
much of the economic burden of zoster is borne by indi- emergency department visits, and 1–5 for the number of
vidual patients as reduced quality of life because of pain medications prescribed. Approximately 1%–4% of zoster
and suffering. Certain studies provide a range of estimates episodes result in hospitalization, with a mean duration of
for health-care use among persons aged >60 years for treat- 4.8 days. (196–199). Health-care use for zoster and PHN
ment of zoster and PHN. The estimates vary widely increases substantially with the age of patients (196–198).
because of differing assumptions regarding the risk for PHN Costs associated with acute zoster have been evaluated.
and of complications resulting from zoster. Estimated health- Among patients with acute episodes of zoster, average
care use per case of zoster ranges from 1.3–3.1 for the num- expenditures ranged from $112–$287 per episode of out-
ber of outpatient visits, 0.005–0.12 for the number of patient care, $73–$180 per antiviral treatment, and
Recommendations and Reports
June 6, 2008 / Vol. 57 / RR-5
Morbidity and Mortality Weekly Report
www.cdc.gov/mmwr
Continuing Education Activity Sponsored by CDC
Prevention of Herpes Zoster Recommendations of the Advisory Committee
on Immunization Practices (ACIP)
EXPIRATION — June 6, 2010
You must complete and return the response form electronically or by mail by contact hours Certified Health Education Specialist (CHES) credit. If you
Xxxxxxxx XX, 200X, to receive continuing education credit. If you answer all return the form electronically, you will receive educational credit
of the questions, you will receive an award letter for 2.75 hours Continuing immediately. If you mail the form, you will receive educational credit in
Medical Education (CME) credit; 0.25 Continuing Education Units (CEUs); approximately 30 days. No fees are charged for participating in this continuing
2.75 contact hours Continuing Nursing Education (CNE) credit; or 3.0 education activity.
INSTRUCTIONS
By Internet
1. Read this MMWR (Vol. 57, RR-5), which contains the correct answers to
the questions beginning on the next page.
2. Go to the MMWR Continuing Education Internet site at http://
www.cdc.gov/mmwr/cme/conted.html.
3. Select which exam you want to take and select whether you want to register
for CME, CEU, CNE, or CHES credit.
4. Fill out and submit the registration form.
5. Select exam questions. To receive continuing education credit, you must
answer all of the questions. Questions with more than one correct answer
will instruct you to “Indicate all that apply.”
6. Submit your answers no later than June 6, 2010.
7. Immediately print your Certificate of Completion for your records.
By Mail or Fax
1. Read this MMWR (Vol. 57, RR-5), which contains the correct answers to
the questions beginning on the next page.
2. Complete all registration information on the response form, including
your name, mailing address, phone number, and e-mail address.
3. Indicate whether you are registering for CME, CEU, CNE, or CHES
credit.
4. Select your answers to the questions, and mark the corresponding letters
on the response form. To receive continuing education credit, you must
answer all of the questions. Questions with more than one correct answer
will instruct you to “Indicate all that apply.”
5. Sign and date the response form or a photocopy of the form and send no
later than June 6, 2010, to
Fax: 404-498-2388
Mail: MMWR CE Credit
CCHIS, Centers for Disease Control and Prevention
1600 Clifton Rd, N.E., MS E-90
Atlanta, GA 30333
6. Your Certificate of Completion will be mailed to you within 30 days.
ACCREDITATION
Continuing Medical Education (CME). CDC is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical
education for physicians. CDC designates this educational activity for a maximum of 2.75 hours in category 1 credit toward the AMA Physician’s Recognition
Award. Each physician should claim only those hours of credit that he/she actually spent in the educational activity.
Continuing Education Unit (CEU). CDC has been reviewed and approved as an Authorized Provider by the International Association for Continuing
Education and Training (IACET), 1620 I Street, N.W., Suite 615, Washington, DC 20006. CDC has awarded 0.25 CEUs to participants who successfully
complete this program.
Continuing Nursing Education (CNE). CDC is accredited as a provider of continuing nursing education by the American Nurses Credentialing Center’s
Commission on Accreditation. CDC will award 2.75 contact hour(s) in CNE credit.
Certified Health Education Specialist (CHES). CDC is a designated provider of continuing education contact hours (CECH) in health education by the
National Commission for Health Education Credentialing, Inc. This program is a designated event for CHESs to receive 3.0 category I contact hour(s) in health
education. The CDC provider number is GA0082.
depardepar
depardepar
depar
tment of health and human sertment of health and human ser
tment of health and human sertment of health and human ser
tment of health and human ser
vicesvices
vicesvices
vices
Centers for Disease Control and PreventionCenters for Disease Control and Prevention
Centers for Disease Control and PreventionCenters for Disease Control and Prevention
Centers for Disease Control and Prevention
CE-2 MMWR June 6, 2008
Goals and Objectives
This report provides recommendations on use of the licensed live-attenuated vaccine for prevention of herpes zoster (zoster) and its most frequent complication,
post-herpetic neuralgia (PHN). The recommendations were developed by CDC’s Advisory Committee on Immunization Practices. The goal of this report is to
provide recommendations on the zoster vaccine for clinicians, public health officials, and others interested in preventing zoster and PHN in the United States. Upon
completion of this educational activity, the reader should be able to 1) describe the epidemiology of zoster in the United States, 2) identify recommendations for
zoster vaccination in the United States, and 3) and describe the characteristics of zoster vaccine.
To receive continuing education credit, please answer all of the following questions.
1. Which of the following statements about herpes zoster is true?
A. It most frequently manifests in a sacral nerve dermatomal distribution.
B. It most frequently manifests in a thoracic, cervical, or ophthalmic
dermatomal distribution.
C. It most frequently presents as a disseminated rash.
2. The diagnosis of herpes zoster…
A. Can usually be made on clinical grounds alone.
B. Is made on the basis of testing antibody to varicella zoster virus.
C. Neither A nor B.
3. Which of the following statements is true regarding the epidemiology
of herpes zoster?
A. Approximately one in 10 persons acquire zoster during their lifetime.
B. After adjusting for changes in age and other factors in the population,
the rate for zoster in the United States has been stable in recent
decades.
C. Recent data suggest approximately 1 million cases of zoster occur
annually in the United States.
D. Exposure to varicella has been demonstrated to have major influence
on the rate for zoster in the population.
E. All of the above.
4. Which of the following groups are at increased risk for herpes zoster?
A. Persons at advanced age.
B. Patients with atopic dermatitis.
C. Blacks.
D. Persons that have been vaccinated with varicella vaccine.
E. All of the above.
5. The most significant burden from zoster in the U.S. population is
caused by…
A. A high number of hospitalizations.
B. A high number of deaths.
C. Blindness caused by ophthalmic zoster.
D. Impact on quality of life attributed to pain and suffering.
6. Which of the following is true regarding zoster vaccine?
A. It must be refrigerated before reconstitution.
B. It is made from a different strain of varicella zoster virus than used in
varicella vaccine.
C. It should not be administered within 28 days of inactivated influenza
vaccine.
D. It contains thimerosol.
E. None of the above.
7. Which of the following is true regarding the efficacy of zoster vaccine
in the Shingles Prevention Study?
A. The vaccine reduced overall incidence of zoster by approximtaely
51%.
B. The vaccine was most effective at preventing the post-herpatic
neuralgia (PHN) of the longest duration.
C. The vaccine was not very effective at preventing zoster in persons aged
>80 years but was much more effective at preventing PHN in these
persons.
D. Efficacy of the vaccine at preventing zoster was fairly stable over a
period of 6 years.
E. A, B, and C.
8. The Advisory Committee for Immunization Practices (ACIP)
recommends zoster vaccine for which of the following?
A. Adults aged >50 years about to undergo kidney transplantation.
B. Adults aged >60 years, with 1 booster dose 10 years later.
C. Adults aged >60 years irrespective of prior history of varicella or zoster.
D. Adults aged >60 who are experiencing zoster to prevent PHN.
E. B, C, and D.
9. Zoster vaccine is contraindicated by ACIP for which of the following
persons?
A. Persons with insulin-dependent diabetes mellitus.
B. Persons infected with human immunodeficiency virus before
progression to acquired immunodeficiency syndrome.
C. Persons that received chemotherapy for leukemia during the
preceding 3 months.
D. A, B, and C.
E. B and C.
10. In the Shingles Prevention Study, which of the following adverse
reactions occurred with significantly greater frequency in the zoster
vaccine recipients as compared with controls?
A. A generalized varicella-like rash.
B. Injection site reactions.
C. Fever.
D. All of the above.
11. What best describes your professional activities?
A. Physician.
B. Nurse.
C. Health educator.
D. Office staff.
E. Other.
12. I plan to use these recommendations as the basis for … (Indicate all
that apply.)
A. health education materials.
B. insurance reimbursement policies.
C. local practice guidelines.
D. public policy.
E. other.
MMWR Response Form for Continuing Education Credit
June 6, 2008/Vol. 57/No. RR-5
Prevention of Herpes Zoster Recommendations of the Advisory
Committee on Immunization Practices (ACIP)
13. Overall, the length of the journal report was …
A. much too long.
B. a little too long.
C. just right.
D. a little too short.
E. much too short.
14. After reading this report, I am confident I can describe the
epidemiology of zoster in the United States.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
15. After reading this report, I am confident I can identify
recommendations for zoster vaccination in the United States.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
16. After reading this report, I am confident I can describe the
characteristics of zoster vaccine.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
Vol. 57 / No. RR-5 Recommendations and Reports CE-3
Detach or photocopy.
To receive continuing education credit, you must
provide your contact information (1. please print or type);
indicate your choice of CME, CME for nonphysicians, CEU, CNE, or CHES credit;2.
answer3. all of the test questions;
sign and date this form or a photocopy;4.
submit your answer form by5. June 6, 2010.
Failure to complete these items can result in a delay or rejection of your application for
continuing education credit.
Check One
Last Name (print or type) First Name
CME Credit
CME for
nonphysicians
Street Address or P.O. Box
Credit
CEU Credit
Apartment or Suite
CNE Credit
CHES Credit
City State ZIP Code
CPE Credit
Phone Number Fax Number
E-Mail Address
Fill in the appropriate blocks to indicate your answers. Remember, you must answer all
of the questions to receive continuing education credit!
1. [ ] A [ ] B [ ] C
14. [ ] A [ ] B [ ] C [ ] D [ ] E
2. [ ] A [ ] B [ ] C
15. [ ] A [ ] B [ ] C [ ] D [ ] E
3. [ ] A [ ] B [ ] C [ ] D [ ] E
16. [ ] A [ ] B [ ] C [ ] D [ ] E
4. [ ] A [ ] B [ ] C [ ] D [ ] E
17. [ ] A [ ] B [ ] C [ ] D [ ] E
5. [ ] A [ ] B [ ] C [ ] D
18. [ ] A [ ] B [ ] C [ ] D [ ] E
6. [ ] A [ ] B [ ] C [ ] D [ ] E
19. [ ] A [ ] B [ ] C [ ] D [ ] E
7. [ ] A [ ] B [ ] C [ ] D [ ] E
20. [ ] A [ ] B [ ] C [ ] D [ ] E
8. [ ] A [ ] B [ ] C [ ] D [ ] E
21. [ ] A [ ] B [ ] C [ ] D [ ] E
9. [ ] A [ ] B [ ] C [ ] D [ ] E
22. [ ] A [ ] B [ ] C [ ] D [ ] E
10. [ ] A [ ] B [ ] C [ ] D
23. [ ] A [ ] B [ ] C [ ] D [ ] E
11. [ ] A [ ] B [ ] C [ ] D [ ] E
24. [ ] A [ ] B [ ] C [ ] D [ ] E
12. [ ] A [ ] B [ ] C [ ] D [ ] E
25. [ ] A [ ] B
13. [ ] A [ ] B [ ] C [ ] D [ ] E
26. [ ] A [ ] B [ ] C [ ] D [ ] E [ ] F
Signature Date I Completed Exam
17. The learning outcomes (objectives) were relevant to the goals of this
report.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
18. The instructional strategies used in this report (text, tables, and figures)
helped me learn the material.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
19. The content was appropriate given the stated objectives of the report.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
20. The content expert(s) demonstrated expertise in the subject matter.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
(Continued on pg CE-4)
CE-4 MMWR June 6, 2008
21. Overall, the quality of the journal report was excellent.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
22. The recommendations will improve the quality of my practice.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
23. The availability of continuing education credit influenced my decision
to read this report.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
24. The MMWR format was conductive to learning this content.
A. Strongly agree.
B. Agree.
C. Undecided.
D. Disagree.
E. Strongly disagree.
25. Do you feel this course was commercially biased? (Indicate yes or no; if
yes, please explain in the space provided below.)
A. Yes.
B. No.
26. How did you learn about the continuing education activity?
A. Internet.
B. Advertisement (e.g., fact sheet, MMWR cover, newsletter, or journal).
C. Coworker/supervisor.
D. Conference presentation.
E. MMWR subscription.
F. Other.
Correct answers for questions 1–10.
B; 2. A; 3. C; 4.A; 5. D; 6. E; 7. E; 8. C; 9. C; 10. B. 1.
DEPDEP
DEPDEP
DEP
ARTMENT OF HEALARTMENT OF HEAL
ARTMENT OF HEALARTMENT OF HEAL
ARTMENT OF HEAL
TH AND HUMAN SERTH AND HUMAN SER
TH AND HUMAN SERTH AND HUMAN SER
TH AND HUMAN SER
VICESVICES
VICESVICES
VICES
Centers for Disease Control and Prevention (CDC)
Atlanta, Georgia 30333
Official BusinessOfficial Business
Official BusinessOfficial Business
Official Business
Penalty for Private Use $300
NO POSTAGE
NECESSARY
IF MAILED
IN THE
UNITED STATES
BUSINESS REPLY MAIL
FIRST CLASS MAIL PERMIT NO. 99110 ATLANTA, GA 30333
Postage Will Be Paid by Department of Health and Human Services
MMWR CE CREDIT
COORDINATING CENTER FOR HEALTH INFORMATION
AND SERVICE — MAILSTOP E-90
CENTERS FOR DISEASE CONTROL AND PREVENTION
1600 CLIFTON RD., N.E.
ATLANTA, GEORGIA 30333
17 Vol. 57 / RR-5 MMWR
$3,221–$7,206 per hospitalization (2006 dollars). Addi-
tional costs associated with managing non-PHN complica-
tions (e.g., ocular, neurologic, and cutaneous) ranged from
$1,158–$11,255 per complication, and from $566–
$1,914 per episode of PHN. Among the subset of patients
with PHN persisting from 30 days to 12 months, annual-
ized health-care costs, including costs of the acute episode,
ranged from $2,159 to $5,387 (200,201). Although indi-
rect costs from death can occur with zoster, these costs
result mostly from losses in work time caused by tempo-
rary or more permanent disability. Patients with zoster
(including those progressing to PHN) lose an average of
>129 hours of work per episode (197,198), including losses
of 12 or more hours of work time and 69 hours of leisure
time during the first 30 days (196). Data on the national
economic impacts of zoster and its complications on
quality of life have not been reported.
Five studies have estimated the cost-effectiveness of a
1-dose routine vaccination program of immunocompetent
persons aged >60 years (196,197,199,202,203) (Table 5).
One of these studies has not been published (196). All five
studies used a Markov cohort model (105), followed a cost-
utility analytic approach that included a societal perspec-
tive (204,205), and used quality-adjusted life-year (QALY)
scores to assess the incremental impact of the vaccine
program on quality of life. Costs and health benefits were
measured in 2005–2006 U.S. dollars, and a 3% discount
rate was used to adjust health outcomes and costs. Model
assumptions varied regarding duration of vaccine protec-
tion, the efficacy of the vaccine for preventing PHN among
vaccine recipients who developed zoster, costs associated
with vaccine adverse events, and costs attributed to losses
in work productivity. None of the five models incorporated
costs for losses in leisure time. Assuming a routine vaccina-
tion program with 100% coverage, the estimated QALYs
gained ranged from 0.0016 (0.6 days) to 0.0087 (3 days).
At a vaccine cost of $150 per dose, the societal costs of
routinely vaccinating immunocompetent persons aged >60
years range from $27,000 to $112,000 per QALY gained.
In the sensitivity analyses, variables with the strongest
influence on outcomes include vaccine costs, duration of
vaccine efficacy, risks for PHN as a complication, and costs
and QALY scores for zoster and its complications.
Although costs per QALY gained are most appropriately
used to prioritize among competing programs for purposes
of resource allocation, policymakers often decide whether
or not to support programs by comparing their cost per
QALY against a standard threshold. A threshold suggested
by the World Health Organization is three times the gross
domestic product per capita, which would be $94,431 for
the United States (206). Alternatively, policymakers often
decide about supporting programs by comparing their cost
per QALY with the values for other widely accepted inter-
ventions. Compilations of such cost effectiveness data have
been published and maintained in on-line registries
(207,208). The estimated cost per QALY for zoster vacci-
nation covers a wide range that appears acceptable in com-
parison to either standard thresholds or to other established
interventions, but it is at the intermediate-to-high end of
that range.
Summary of Rationale for Zoster
Vaccine Recommendations
The availability of a safe and effective vaccine for zoster
offers an opportunity to decrease the burden of this disease
and its complications among persons with high levels of
risk. In the United States, the vaccine is licensed for use
among persons aged >60 years, and routine vaccination of
this population is recommended for several reasons. First,
zoster causes substantial morbidity in the United States,
with approximately 1,000,000 new cases occurring annu-
ally (62). Many of these cases cause debilitating pain, and
when PHN develops, the pain can last for months or even
years. Other complications include involvement of the eye
that can threaten sight, bacterial superinfections, and dis-
figuring facial scarring. Second, although effective antiviral
medications for treatment of zoster are available, adminis-
tration must be initiated within 72 hours of rash onset for
maximum benefit. Many patients might not obtain such
rapid diagnosis and treatment, and even when they do, the
treatment is only partially effective at alleviating the symp-
toms and shortening their duration. Third, available treat-
ments for PHN often do not completely alleviate the pain
and might be poorly tolerated by the older patients (31,33).
Finally, available evidence suggests the cost-effectiveness of
zoster vaccine is within the range of some other public
health interventions.
In a large, placebo-controlled clinical trial, the zoster vac-
cine reduced BOI attributed to zoster by 61.1 % and the
incidence of PHN by 66.5 %. The vaccine reduced the
overall incidence of zoster by 51.3 % and substantially
reduced its associated pain (4). Although the vaccine was
more efficacious in persons aged 60–69 years, substantial
efficacy against zoster was observed in persons aged
>70 years, and PHN was prevented in older age groups.
Prevention of zoster and its sequelae is particularly impor-
tant among the oldest persons because they experience the
highest incidence of zoster and PHN, they might be least
18 MMWR June 6, 2008
TABLE 5. Economic burden and projected cost-effectiveness of a herpes zoster vaccination program, by selected studies* —
United States
Study No. 1
Study No. 2 Study No. 3 Study No. 4 Study No. 5
§
Costs
Acute zoster
Outpatient care $246–$312
$214 (SD** = $52.7) $287 ($221–$422) $112 ($56–$224) $289 ($178–$527)
Antiviral medicines $73–$135 $155 NA
††
$180 ($135–$238) NA
Hospitalization $6,815 (SD = $11,608) $8,521 $7,206 ($6,529–$8,401) $6,884 $3,221 ($130–$5,200)
Out-of-pocket
§§
$7–$20 NA NA NA N/A
Complications (nonpostherpetic
neuralgia [PHN])
¶¶
Ocular $1,158 (SD = $2,231) $1,827 (SD= $2,844) $11,255 ($9,255–$13,255) NA NA
Neurologic $1,642 (SD = $3,483) $2,623 (SD= $3,965) NA NA NA
Cutaneous $1,851 (SD = $1,838) $3,739 (SD= $5,684) NA NA NA
PHN***
Outpatient care and prescriptions $903–$1071 $1,914 (SD= $2,667) $566 ($483–$673) $996 NA
Out-of-pocket $29–$118 NA NA NA NA
Productivity losses per case
†††
$1,062 $2,437 $782 (0–$1,000) $744 ($185–$1,934) NA
Cost-Effectiveness
Outcomes prevented per million person-years
Zoster 58,000 66,000 NA NA NA
Complications (non-PHN) 7,000 8,500 NA NA NA
PHN 32,000 20,000 NA NA NA
Premature deaths 5 37 NA NA NA
Resource-use averted per million person-years
Outpatient visits 285,000 274,000 NA NA NA
Emergency department visits 28,000 8,700 NA NA NA
Hospitalization days 6,000 8,900 NA NA NA
Prescription fills 176,000 354,000 NA NA NA
QALYs gained per person-year
§§§
0.0087 0.0057 0.0021 0.0016 NA
Vaccine cost per dose
¶¶¶
$150 ($50–$200) $150 $145 ($50–$150) $200 ($50–$500) $170 ($123–$245)
Cost per QALY****
,§§§
$55,000 $27,000 $112,000 $100,000 $50,000
* Study No. 1: Ortega-Sanchez IR. Projected cost-effectiveness of vaccinating US elderly to prevent shingles. Oral presentation to the Advisory Committee for Immunization
Practices (ACIP) Meeting, June 2006; Study No. 2: Pellissier JM, Brisson M, Levin MJ. Evaluation of the cost-effectiveness In the United States of a vaccine to prevent herpes
zoster and postherpetic neuralgia in older adults. Vaccine 2007;25:8326–37; Study No. 3: Rothberg MB, Virapongse A, Smith KJ. Cost-effectiveness of a vaccine to prevent
herpes zoster and postherpetic neuralgia in older adults. Clin Infect Dis 2007;44:1280–8; Study No. 4: Hornberger J, Robertus K. Cost-effectiveness of a vaccine to prevent
herpes zoster and postherpetic neuralgia in older adults. Ann Intern Med 2006;145:317–25; Study No. 5: Ho A, Coplan PM, Lee A, et al. Cost-effectiveness of vaccination against
herpes zoster in the elderly population. Canadian J Anesthesia (Suppl 1): 26394. Studies used a Markov cohort model. Except for Study 5, which considered vaccinating
immunocompetent persons aged >65 years, studies No. 1–4 evaluated a routine vaccination program of immunocompentent persons aged >60 years. Baseline for comparison
in all studies was “no vaccination. Study No. 5 measured medical costs of zoster in 2002 U.S. dollars and Studies No. 1–4 measured age-specific costs in 2005–2006 U.S. dollars.
For rates of zoster incidence and medical costs, Study No. 1 used data mainly from MedStat. For rates of zoster incidence and complications, the study used:Yawn BP, Saddier
S, Wollan P, Sauver JS, Kurland M, Sy L. A population-based study of the incidence and complication rates of herpes zoster before zoster vaccine introduction. Mayo Clin
Proc 2007;82:1341–9. For quality-of-life scores with zoster, out-of-pocket, and productivity costs, the study used:Lieu TA, Ortega-Sanchez IR, Ray GT, et al. Community and
patient values for preventing zoster. Pharmacoeconomics 2008;26:235–49.
§
For incidence of zoster, Study No. 5 used data mainly from: Oxman MN, Levin MJ, Johnson GR, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older
adults. N Engl J Med 2005;352:2271–84; CDC. Prevention of varicella: updated recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR
2007;56(No. RR-4). Medical costs from 1995 were updated to 2002 U.S. dollars.
Ranges in medical costs described age-specific costs for zoster or PHN cases among persons aged >60 years. High and low medical costs also were used for sensitivity analysis.
** SD = Standard deviation. In addition to reported mean costs, SD values were used for sensitivity analysis.
††
NA = not available.
§§
Out-of-pocket costs are monetary expenses associated with zoster or PHN as reported by patients and include medical and nonmedical payments, over-the-counter medications,
travel expenses, and other monetary expenses.
¶¶
Study Nos. 1–3 included costs of frequent non-PHN complications observed <30 days after zoster reactivation (i.e., ocular, neurologic, and cutaneous complications).
*** Study Nos. 1 and 2 used costs attributable to PHN cases lasting >90 days, and study No. 4 used cost attributable to PHN cases lasting 182 days ($166 per month). Although
Study No. 3 assumed that 43%–56% of PHN cases lasted >12 months, attributable costs were not explicitly defined.
†††
Productivity losses (i.e., indirect costs) in cases of zoster and PHN were related to monetary value of time of work missed or income lost. Losses from leisure time or unpaid
activities were not included. Although zoster can cause death, no productivity costs associated with premature deaths were included.
§§§
Source of quality-of-life values to estimate quality-adjusted live years (QALYs) differed across studies. Source of quality-of-life values to estimate quality-adjusted life years
(QALYs) differed across studies. For all five studies, quality-of-life values are based on published data with valuation by representative samples of community members. In
addition, study No. 1 also used published quality-of-life values using valuation by zoster patients, yielding values of QALYs gained per person-year of 0.019 and cost per QALY
of $23,000.
¶¶¶
Range in vaccine costs were used for sensitivity analyses. Study No. 4 reported an aggregated cost for the vaccine and vaccine administration equal to $200. Except for Study
No. 5, vaccine administration costs also were included (i.e., $3.50 in Study No. 3, $18 in Study No. 2, and $15–$30 in Study No. 1).
**** Estimates of cost per QALY saved assumed that PHN cases were avoided only through the prevention of zoster cases.When PHN cases were prevented in persons with zoster
for whom the vaccine failed to prevent zoster (i.e., burden of illness), a societal cost of $35,000 per QALY saved was estimated by Study No. 1.
able to seek medical attention for zoster and PHN and to to treat PHN, they might have the least reserve to tolerate
request treatment of ongoing pain, they might be least able zoster and its complications, and they are most likely to
to tolerate the medications and procedures commonly used suffer social and psychological consequences from PHN.
19 Vol. 57 / RR-5 MMWR
Recommendations for Use of
Zoster Vaccine
Routine Vaccination of Persons Aged
>60 Years
ACIP recommends routine vaccination of all persons aged
>60 years with 1 dose of zoster vaccine. Persons who report
a previous episode of zoster and persons with chronic medical
conditions (e.g., chronic renal failure, diabetes mellitus,
rheumatoid arthritis, and chronic pulmonary disease) can
be vaccinated unless those conditions are contraindications
or precautions. Zoster vaccination is not indicated to treat
acute zoster, to prevent persons with acute zoster from
developing PHN, or to treat ongoing PHN. Before routine
administration of zoster vaccine, it is not necessary to ask
patients about their history of varicella (chickenpox) or to
conduct serologic testing for varicella immunity.
Simultaneous Administration with Other
Adult Vaccines
Immunogenicity of zoster vaccine and trivalent inacti-
vated influenza vaccine is not compromised when the two
vaccines are administered simultaneously (186). However,
no data exist on administration of zoster vaccine with other
vaccines routinely recommended for persons aged >60 years,
which are all inactivated. In general, the simultaneous
administration of most widely used live, attenuated and
inactivated vaccines has not resulted in impaired immune
response or an increased rate of adverse events (209). There-
fore, zoster vaccine can be administered with other
indicated vaccines during the same visit (e.g., Td, Tdap,
and pneumococcal polysaccharide vaccines). Each vaccine
must be administered using a separate syringe at a differ-
ent anatomic site. If simultaneous administration is not
possible, zoster vaccine can be administered at any time
before or after an inactivated vaccine, but at least 4 weeks
before or after another live, attenuated vaccine (209).
Groups for Which Vaccine is Not
Licensed
Vaccination of Persons Aged <60 Years
The vaccine is not licensed for persons aged <60 years,
and no recommendation exists for routine vaccination of
persons aged <60 years. In the clinical trial, the zoster vac-
cine was evaluated among persons aged >60 years. The vac-
cine was most effective and well tolerated in the youngest
persons (Table 1) (4). Although the vaccine would
probably be safe and effective in persons aged <60 years,
data are insufficient to recommend vaccination of these
persons at this time.
Vaccination of Persons Who Have Received
Varicella Vaccine
Zoster vaccination is not recommended for persons of
any age who have received varicella vaccine. However, health-
care providers do not need to inquire about varicella vacci-
nation history before administering zoster vaccine because
virtually all persons currently or soon to be in the recom-
mended age group have not received varicella vaccine. In
the United States, varicella vaccination began in 1995. Since
that time, few adults aged >40 years would have been sus-
ceptible to varicella and thus eligible to receive varicella
vaccine (5). The number of persons eligible for zoster vac-
cination who have received varicella vaccine is extremely
small and will remain so for at least a decade.
Special Groups and Circumstances
Persons with a Reported History of Zoster
Persons with a reported history of zoster can be vacci-
nated. Repeated zoster has been confirmed in immuno-
competent persons soon after a previous episode (4).
Although the precise risk for and severity of zoster as a func-
tion of time following an earlier episode are unknown, some
studies suggest it may be comparable to the risk in persons
without a history of zoster (62,169). Furthermore, no labo-
ratory evaluations exist to test for the previous occurrence
of zoster, and any reported diagnosis or history might be
erroneous (4,64,65). Although the safety and efficacy of
zoster vaccine have not been assessed in persons with a his-
tory of zoster, different safety concerns are not expected in
this group.
Persons Anticipating Immunosuppression
The risk for zoster and its severe morbidity and mortal-
ity is much greater among persons who are immunosup-
pressed. Review of vaccination status for zoster and other
vaccines should be a key component of the medical assess-
ment for immunocompetent patients aged >60 years who
might be anticipating initiation of immunosuppressive treat-
ments or who have diseases that might lead to immunode-
ficiency. Such patients without a history of zoster vaccination
should receive 1 dose of zoster vaccine at the first possible
clinical encounter while their immunity is intact. Zoster
vaccine should be administered at least 14 days before ini-
tiation of immunosuppressive therapy, although some
20 MMWR June 6, 2008
experts advise waiting 1 month after zoster vaccination to
begin immunosuppressive therapy if delay is possible (210).
Persons Receiving Antiviral Medications
Licensed antiviral medications active against members of
the herpesvirus family include acyclovir, famciclovir, and
valacyclovir. These agents might interfere with replication
of the live, VZV-based zoster vaccine. All three agents have
relatively short serum half-lives and are quickly cleared from
the body. Persons taking chronic acyclovir, famciclovir, or
valacyclovir should discontinue these medications at least
24 hours before administration of zoster vaccine, if pos-
sible (209). These medications should not be used for at
least 14 days after vaccination, by which time the immu-
nologic effect should be established (209).
Persons Receiving Blood Products
Zoster vaccine can be administered to persons at any time
before, concurrent with, or after receiving blood or other
antibody-containing blood product because persons with
a history of varicella indefinitely maintain high levels of
antibody to VZV, and the levels are comparable to those
found in donated blood and antibody-containing blood
products (e.g., whole blood, packed red blood cells, and
plasma immune globulin, hyperimmune globulin, and
intravenous immune globulin) (192,211).
Nursing Mothers
Most live vaccines, including varicella vaccine, are not
secreted in breast milk (209,212). Therefore, breast feed-
ing is not a contraindication for zoster vaccination. However,
this situation will be extremely rare in the target age group
for this vaccine.
Contraindications
Allergy to Vaccine Components
Zoster vaccine is contraindicated for persons who have a
history of anaphylactic reaction to any component of the
vaccine, including gelatin and neomycin. Neomycin allergy
is usually manifested as a contact dermatitis, which repre-
sents a delayed-type immune response. A history of con-
tact dermatitis to neomycin is not a contraindication for
receiving zoster vaccine (209).
Immunocompromised Persons
Zoster vaccine should not be administered to persons with
primary or acquired immunodeficiency including:
Persons with leukemia, lymphomas, or other malignant
neoplasms affecting the bone marrow or lymphatic
system. However, patients whose leukemia is in remis-
sion and who have not received chemotherapy (e.g.,
alkylating drugs or antimetabolites) or radiation for at
least 3 months can receive zoster vaccine (209).
Persons with AIDS or other clinical manifestations of
HIV, including persons with CD4+ T-lymphocyte val-
ues <200 per mm
3
or <15% of total lymphocytes.
Persons on immunosuppressive therapy, including
high-dose corticosteroids (>20 mg/day of prednisone
or equivalent) lasting two or more weeks. Zoster vacci-
nation should be deferred for at least 1 month after
discontinuation of such therapy (209). Short-term cor-
ticosteroid therapy (<14 days); low-to-moderate dose
(<20 mg/day of prednisone or equivalent); topical (e.g.,
nasal, skin, inhaled); intra-articular, bursal, or tendon
injections; or long-term alternate-day treatment with
low to moderate doses of short-acting systemic corti-
costeroids are not considered to be sufficiently immu-
nosuppressive to cause concerns for vaccine safety.
Persons receiving this dose or schedule can receive zoster
vaccine. Therapy with low-doses of methotrexate (<0.4
mg/Kg/week), azathioprine (<3.0 mg/Kg/day), or 6-
mercaptopurine (<1.5 mg/Kg/day) for treatment of
rheumatoid arthritis, psoriasis, polymyositis, sarcoido-
sis, inflammatory bowel disease, and other conditions
are also not considered sufficiently immunosuppres-
sive to create vaccine safety concerns and are not
contraindications for administration of zoster vaccine.
• Persons with clinical or laboratory evidence of other
unspecified cellular immunodeficiency. However, per-
sons with impaired humoral immunity (e.g.,
hypogammaglobulinemia or dysgammaglobulinemia)
can receive zoster vaccine.
Persons undergoing hematopoietic stem cell transplan-
tation (HSCT). The experience of HSCT recipients with
VZV-containing vaccines (e.g., zoster vaccine) is lim-
ited. Physicians should assess the immune status of
the recipient on a case-by-case basis to determine the
relevant risks. If a decision is made to vaccinate with
zoster vaccine, the vaccine should be administered at
least 24 months after transplantation (209).
Persons receiving recombinant human immune media-
tors and immune modulators, especially the antitumor
necrosis factor agents adalimumab, infliximab, and
etanercept. The safety and efficacy of zoster vaccine
administered concurrently with these agents is
unknown. If it is not possible to administer zoster vac-
cine to patients before initiation of therapy, physicians
should assess the immune status of the recipient on a
case-by-case basis to determine the relevant risks and
21 Vol. 57 / RR-5 MMWR
benefits. Otherwise, vaccination with zoster vaccine
should be deferred for at least 1 month after discon-
tinuation of such therapy.
Pregnancy
Zoster vaccine is not recommended for use in pregnant
women, although these women are unlikely to be in the
vaccine target age group. The effects of the live, attenuated
VZV-based zoster vaccine on the fetus are unknown.
Women should avoid becoming pregnant for 4 weeks fol-
lowing zoster vaccination. Having a pregnant household
member is not a contraindication to zoster vaccination. If a
pregnant woman is vaccinated or becomes pregnant within
1 month of vaccination, she should be counseled about
potential effects on the fetus. Wild-type VZV poses a small
risk to the fetus (3), and the fetal risk from the attenuated
zoster vaccine is probably even lower. Furthermore, virtu-
ally all persons receiving the vaccine will have preexisting
VZV immunity, which is expected to limit viral replica-
tion and presumably further reduce fetal risk. In most cir-
cumstances, the decision to terminate a pregnancy should
not be based on whether zoster vaccine was administered
during pregnancy. Merck & Co., Inc., in collaboration with
CDC, has established a pregnancy registry to monitor the
maternal-fetal outcomes of pregnant women who are inad-
vertently administered live-attenuated VZV-based vaccines
within 1 month of pregnancy (telephone: 800-986-8999).
Patients and health-care providers should report any expo-
sure to zoster vaccine during pregnancy to this registry.
Precautions
Moderate to Severe Illness
Zoster vaccination of persons who have severe acute ill-
ness should be postponed until recovery. The decision to
delay vaccination depends on the severity of symptoms and
the etiology of the disease. Zoster vaccine can be adminis-
tered to persons who have mild acute illnesses with or
without fever (209).
Program Implementation Issues
Following Good Adult Vaccination Practices
Zoster vaccine should be offered to patients aged >60
years at the first available clinical encounter with their pro-
vider. The average adult in this age group has 5–8 clinical
encounters with their provider annually (213). Strategies
to promote zoster vaccination include linking delivery of
zoster vaccine to delivery of other indicated adult vaccines
(e.g., influenza) and preventive-health interventions (214–
217), standing orders so that patients will automatically
be offered indicated vaccines rather than requiring case-
by-case physicians’ orders (218), and practice-based audits
and/or physician-reminder systems (218). Residents of
nursing homes and other long-term–care facilities who are
at least aged 60 years and without contraindications should
be included in routine zoster vaccination activities. When
administering zoster vaccine, health-care providers should
review the patients vaccination status for all indicated adult
vaccines (219,220).
ACIP recommends that health-care providers keep per-
manent documentation of all administered vaccines, includ-
ing zoster vaccine, in the vaccine recipients permanent
medical record (209). The type of the vaccine, manufac-
turer, anatomic site, route of delivery, date of administra-
tion, lot number, and name of the administering facility
should be recorded. To help avoid the administration of
unnecessary doses, every patient should be given a record
of the vaccination.
Administration Errors
The zoster vaccine, ZOSTAVAX
®
, is a live, attenuated
vaccine containing Oka/Merck strain VZV. The vaccine is
similar to the varicella vaccine, VARIVAX
®
, except the mini-
mum PFU-content of the ZOSTAVAX
®
is at least 14-fold
higher than the minimum PFU-content of VARIVAX
®
.
Opportunities for administration errors are possible.
For providers who serve both children and adults, physi-
cal separation of products, careful visual inspection and
reading of labels, and preparation of vaccine for patient use
only at time of vaccination can help prevent errors. If a
provider mistakenly administers high-potency zoster vac-
cine to a child indicated for varicella vaccine, the level of
protection against varicella would probably be at least the
same as for conventional doses of varicella vaccine. This
erroneous dose should count as a single valid dose of vari-
cella vaccine. If the erroneous dose was administered in lieu
of the first dose of varicella vaccine, a second dose of vari-
cella vaccine is required. Administration errors involving
zoster vaccine should be reported to VAERS whether or
not an adverse event occurs.
Early clinical trials for prevention of varicella were con-
ducted in susceptible children using a formulation of live-
attenuated Oka/Merck strain VZV at doses of 17,430 PFU,
approaching the range of PFU in zoster vaccine (>19,400
PFU). This high-dose formulation was well tolerated and
efficacious (221). The more recently licensed live, attenu-
ated Oka-strain VZV vaccine (PROQUAD
®
) prepared in
22 MMWR June 6, 2008
combination with measles, mumps, and rubella vaccine
(MMRV) is formulated with a broad range of titers that
extend to over 60,000 PFU (222,223).
Varicella vaccine (VARIVAX
®
) is not indicated for pre-
vention of zoster. MMRV vaccine (PROQUAD
®
) is not
licensed for use in persons aged >13 years. If a provider
mistakenly administers varicella vaccine to persons indi-
cated for zoster vaccine, no specific safety concerns exists,
but the dose should not be considered valid and the
patient should be administered a dose of zoster vaccine dur-
ing that same visit. If the error is not immediately detected,
a dose of zoster vaccine should be administered as soon as
possible but not within 28 days of the varicella vaccine
dose to prevent potential interference of 2 doses of live
attenuated virus.
Risk for Transmission of Oka/Merck Strain
after Receiving Zoster Vaccine
Persons having close household or occupational contact
with persons at risk for severe varicella need not take any
precautions after receiving zoster vaccine except in rare
instances in which a varicella-like rash develops, when stan-
dard contact precautions are adequate. Although transmis-
sion of Oka/Merck strain VZV has been documented
following varicella vaccination, such transmission is rare and
has only been documented when the vaccine recipient first
developed a varicella-like rash. Rates of varicella-like rash
appear to be less common following zoster vaccination than
following varicella vaccination (4), and transmission of the
Oka/Merck strain VZV from recipients of zoster vaccine
has not been detected. The risk for transmitting the
attenuated vaccine virus to susceptible persons should be
weighed against the risk for developing wild-type zoster
that could be transmitted to a susceptible person. If a sus-
ceptible, immunocompromised person is inadvertently
exposed to a person who has a vaccine-related rash,
VARIZIG
need not be administered because disease as-
sociated with this type of transmission is expected to be
mild. Acyclovir, valacyclovir, and famciclovir are active
against live-attenuated Oka/Merck strain VZV and can be
used in the unlikely situations in which a severe illness
develops in the susceptible contact.
Reporting of Adverse Events after
Vaccination
As with any newly licensed vaccine, surveillance for rare
adverse events associated with administration of zoster vac-
cine is important for assessing its safety in widespread use.
Vaccine safety surveillance in the age group for which zoster
vaccine is recommended (aged >60 years) will present chal-
lenges because of the high prevalence of chronic conditions,
the frequent use of multiple medications, and the common
occurrence of medical events. Coincident adverse events can
be anticipated following zoster vaccination, but many of
these could be caused by the vaccine as well. All clinically
significant adverse events should be reported to VAERS even
if causal relation to vaccination is not certain. VAERS
reporting forms and information are available electronically
at http://www.vaers.hhs.gov or by telephone (800-822-
7967). Web-based reporting is also available, and provid-
ers are encouraged to report electronically at https://
secure.vaers.org/VaersDataEntryintro.htm.
Future Research and Directions
Key questions remain regarding optimal implementation
of zoster vaccination and preventing zoster and its compli-
cations. Areas that need particular focus include:
Surveillance for zoster and its complications. Zoster is
not a notifiable condition. Other strategies will be
needed to monitor zoster and its complications include
using administrative databases, population-based sur-
veys, or active surveillance in sentinel sites. Because the
primary disease burden associated with zoster is pain,
capturing this condition will be particularly challeng-
ing using any surveillance strategy.
Durability of protection against zoster and its compli-
cations afforded by the zoster vaccine. In a persistence
substudy ongoing at 12 of 22 of the original zoster
vaccine study sites, follow up of vaccine recipients will
be extended to an observation time of 10 years. How-
ever, no concurrent randomized placebo group exists
to which these vaccine recipients can be compared, and
results will be compared against historic controls. Large
administrative databases also will be important in evalu-
ating changes in vaccine effectiveness over time. These
and other available data will help to determine changes
in vaccine policy (e.g. a booster dose). However, both
of these approaches might be confounded by secular
changes in the incidence of zoster.
Increased understanding of the epidemiology of zoster.
Better knowledge of age-adjusted changes in the inci-
dence of zoster and risk factors for any such changes
will help determine the long-term effectiveness of the
zoster vaccine and clarify whether changes in VZV cir-
culation caused by varicella vaccination might be
affecting zoster incidence. A better understanding of
the epidemiology and risk factors for zoster might also
lead to changes in policy regarding use of zoster vac-
cine (e.g., targeting the vaccine to selected risk groups
23 Vol. 57 / RR-5 MMWR
that are not now covered by the vaccine recommenda-
tions or lowering the targeted age group). Additional
information is needed to define risks for zoster in vari-
cella-vaccinated adults attributed to Oka/Merck strain
VZV from the vaccine itself or to wild-type VZV from
breakthrough varicella. Although studies involving both
immunocompromised and immunocompetent children
provided evidence that the risks for zoster are lower in
varicella-vaccinated children than in children with natu-
rally acquired varicella (3,99,100,224), characteriza-
tion of these risks in older adults will involve longer
follow up.
• Better prevention and treatment strategies for zoster
and PHN. Although licensure of zoster vaccine repre-
sents an important milestone in prevention of zoster,
the vaccine remains only partially efficacious and is not
licensed for all populations and age groups at risk.
Although available treatments for zoster and PHN have
improved, treatment of these conditions remains inad-
equate. Improved prevention and treatment strategies,
including better vaccines, are needed to reduce the dis-
ease burden of zoster. ZOSTAVAX
®
or other active or
inactive formulations of zoster vaccine should be evalu-
ated in additional cohorts of persons (e.g., persons aged
50–59 years and immunosuppressed persons at the
highest risk for zoster and its complications). Patients
infected with HIV, with or without AIDS, could
benefit substantially from the prevention of zoster. A
better understanding of immunologic correlates of pro-
tection against zoster would help facilitate the devel-
opment and evaluation of such new zoster prevention
strategies.
The epidemiology of zoster in persons with a history of
varicella vaccination. Available data suggest that chil-
dren vaccinated with varicella vaccines are at reduced
risk for Oka/Merck strain zoster as compared with the
risk for zoster from wild-type VZV in children with a
history of chickenpox. However, this evidence does not
extend to vaccine recipients as they become older. Nor
does it include decades of time after vaccination, par-
ticularly in the absence of circulating VZV that could
externally boost immunity. Data also are lacking
regarding the risk for zoster from wild-type VZV in
vaccinated persons with a history of breakthrough vari-
cella. These issues should be addressed in future stud-
ies to develop zoster vaccination policy for cohorts of
vaccine recipients as they age.
• Safety of zoster vaccination. Postlicensure studies to
evaluate further the safety of the zoster vaccine are
under development and will be conducted by the manu-
facturer. Clinical trials have been completed to assess
the safety and immunogenicity of simultaneous admin-
istration of zoster vaccine and formulations of influ-
enza vaccine. In addition, independent studies are being
developed by CDC to monitor safety through VAERS
and the CDC Vaccine Safety Datalink.
Additional Information About
Zoster and Zoster Vaccine
Additional information about zoster and zoster vaccine
is available from several sources, and new information will
be available in the future. Updated information about
zoster, PHN, and zoster vaccine is available at http://
www.cdc.gov/vaccines/vpd-vac/shingles/default.htm.
Acknowledgments
Gregory S. Wallace, MD, Mary Mulholland, MA, Immunization
Services Division, National Center for Immunization and Respiratory
Diseases, CDC, assisted in writing the section on vaccine storage and
handling. Meredith Reynolds, PhD, Division of Viral Diseases, National
Center for Immunization and Respiratory Diseases, CDC, assisted with
compiling and preparing data on economics. Aisha O. Jumaan, PhD,
MPH, Division of Viral Diseases, National Center for Immunization
and Respiratory Diseases, CDC, assisted in writing material related to
the risk of postherpetic neuralgia. Sandra S. Chaves, M.D., M.Sc.,
assisted in preparing the section on adverse events from zoster vaccine.
Jessica Leung, MPH, and Adriana Lopez, Division of Viral Diseases,
National Center for Immunization and Respiratory Diseases, CDC,
helped with graphs and tables.
References
1. CDC. Prevention of varicella: recommendations of the Advisory
Committee on Immunization Practices (ACIP). MMWR 1996;45(No.
RR-11).
2. CDC. Prevention of varicella: updated recommendations of the Advi-
sory Committee on Immunization Practices (ACIP). MMWR
1999;48(No. RR-6).
3. CDC. Prevention of varicella: updated recommendations of the Advi-
sory Committee on Immunization Practices (ACIP). MMWR
2007;56(No. RR-4).
4. Oxman MN, Levin MJ, Johnson GR, et al. A vaccine to prevent
herpes zoster and postherpetic neuralgia in older adults. N Engl J Med
2005;352:2271–84.
5. Kilgore PE, Kruszon-Moran D, Seward JF, et al. Varicella in Ameri-
cans from NHANES III: implications for control through routine
immunization. J Med Virol 2003;70 Suppl 1:S111–8.
6. Arvin A. Varicella-zoster virus. Clin Microbiol Rev 1996;9:361–81.
7. Gilden DH, Dueland AN, Devlin ME, Mahalingam R, Cohrs R. Vari-
cella-zoster virus reactivation without rash. J Infect Dis 1992;166
(Suppl 1):S30–4.
24 MMWR June 6, 2008
8. Cohrs RJ, Randall J, Smith J, et al. Analysis of individual human
trigeminal ganglia for latent herpes simplex virus type 1 and varicella-
zoster virus nucleic acids using real-time PCR. J Virol 2000;74:11464–71.
9. Levin MJ, Cai GY, Manchak MD, Pizer LI. Varicella-zoster virus
DNA in cells isolated from human trigeminal ganglia. J Virol
2003;77:6979–87.
10. Wang K, Lau TY, Morales M, Mont EK, Straus SE. Laser-capture
microdissection: refining estimates of the quantity and distribution of
latent herpes simplex virus 1 and varicella-zoster virus DNA in human
trigeminal ganglia at the single-cell level. J Virol 2005;79:14079–87.
11. Gershon AA, LaRussa P, Steinberg S. The varicella vaccine. Clinical
trials in immunocompromised individuals. Infect Dis Clin North Am
1996;10:583-94.
12. Arvin A. Aging, immunity, and the varicella–zoster virus. N Engl J Med
2005;352:2266–7.
13. Hope-Simpson RE. Postherpetic neuralgia. J R Coll Gen Pract
1975;25:571–5.
14. Krause PR, Klinman DM. Varicella vaccination: evidence for frequent
reactivation of the vaccine strain in healthy children. Nat Med
2000;6:451–4.
15. Thomas SL, Wheeler JG, Hall AJ. Contacts with varicella or with
children and protection against herpes zoster in adults: a case-control
study. Lancet 2002;360:678–82.
16. Arvin AM, Koropchak CM, Wittek AE. Immunologic evidence of
reinfection with varicella-zoster virus. J Infect Dis 1983;148:200–5.
17. Arvin A M. Cell-mediated immunity to varicella-zoster virus. J Infect
Dis 1992;166:S35–41.
18. Wilson A, Sharp M, Koropchak CM, Ting SF, Arvin AM. Subclinical
varicella-zoster virus viremia, herpes zoster, and T lymphocyte immu-
nity to varicella-zoster viral antigens after bone marrow transplanta-
tion. J Infect Dis 1992;165:119–26.
19. Hayward A, Levin M, Wolf W, Angelova G, Gilden D. Varicella-
zoster virus-specific immunity after herpes zoster. J Infect Dis
1991;163:873–5.
20. Webster A, Grint P, Brenner MK, Prentice HG, Griffiths PD. Titra-
tion of IgG antibodies against varicella zoster virus before bone mar-
row transplantation is not predictive of future zoster. J Med Virol
1989;27:117–9.
21. Burke BL, Steele RW, Beard OW, Wood JS, Cain TD, Marmer DJ.
Immune responses to varicella–zoster in the aged. Arch Intern Med
1982;142:291–3.
22. Levin MJ, Smith JG, Kaufhold RM, et al. Decline in varicella-zoster
virus (VZV)-specific cell-mediated immunity with increasing age and
boosting with a high-dose VZV vaccine. J Infect Dis 2003;188:1336–44.
23. Hayward AR, Buda K, Jones M, White CJ, Levin MJ. Varicella zoster
virus-specific cytotoxicity following secondary immunization with live
or killed vaccine. Viral Immunol 1996;9:241–5.
24. Thomas SL, Wheeler JG, Hall AJ. Case-control study of the effect of
mechanical trauma on the risk of herpes zoster. BMJ 2004;328:439.
25. Gilden DH, Dueland AN, Cohrs R, Martin JR, Kleinschmidt-
DeMasters BK, Mahalingam R. Preherpetic neuralgia. Neurology
1991;41:1215–8.
26. Kost RG, Straus SE. Postherpetic neuralgia—pathogenesis, treatment,
and prevention. N Eng J Med 1996;335:32–42.
27. Rogers RS 3rd, Tindall JP. Geriatric herpes zoster. J Am Geriatr Soc
1971;19:495–504.
28. Glynn C, Crockford G, Gavaghan D, Cardno P, Price D, Miller J.
Epidemiology of zoster. J R Soc Med 1990;83:617–9.
29. Gnann JW, Jr. Varicella-zoster virus: atypical presentations and un-
usual complications. J Infect Dis 2002;186:S91–8.
30. Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ, Mahalingam
R, Cohrs RJ, Neurologic Complications of the reactivation of vari-
cella–zoster virus. N Engl J Med 2000;342:635–45.
31. Dworkin RH, Johnson RW, Breuer J, et al. Recommendations for the
management of herpes zoster. Clin Infect Dis 2007;44 (Suppl 1):S1–26.
32. Katz J, Cooper EM, Walther RR, Sweeney EW, Dworkin RH. Acute
pain in herpes zoster and its impact on health-related quality of life.
Clin Infect Dis 2004;39:342–8.
33. Schmader K. Herpes zoster in the elderly: issues related to geriatrics.
Clin Infect Dis 1999;28:736–9.
34. Fishbain DA. The association of chronic pain and suicide. Semin Clin
Neuropsychiatry 1999;4:221–7.
35. Schmader K. Postherpetic neuralgia in immunocompetent elderly
people. Vaccine 1998;16:1768–70.
36. Jung BF, Johnson RW, Griffin DR, Dworkin RH. Risk factors for
postherpetic neuralgia in patients with herpes zoster. Neurology
2004;62:1545–51.
37. Chidiac C, Bruxelle J, Daures JP, et al. Characteristics of patients with
herpes zoster on presentation to practitioners in France. Clin Infect
Dis 2001;33:62–9.
38. Scott FT, Leedham-Green ME, Barrett-Muir WY, et al. A study of
zoster and the development of postherpetic neuralgia in East London.
J Med Virol 2003;(Suppl 1):S24–30.
39. Ragozzino MW, Melton LJ 3rd, Kurland LT, Chu CP, Perry HO.
Population-based study of herpes zoster and its sequelae. Medicine
1982;61:310–6.
40. Shaikh S, Ta CN. Evaluation and management of herpes zoster
ophthalmicus. Am Fam Physician 2002;66:1723–30.
41. Pavan Langston D. Herpes zoster ophthalmicus. Neurology
1995;45:50–1.
42. Sweeney CJ, Gilden DH. Ramsay Hunt syndrome. J Neurol Neurosurg
Psychiatry 2001;71:149–54.
43. Adour KK. Otological complications of herpes zoster. Ann Neurol
1994;35:S62–S64.
44. Braverman DL, Ku A, Nagler W. Herpes zoster polyradiculopathy.
Arch Phys Med Rehabil 1997;78:880–2.
45. Vincent KD, Davis LS. Unilateral abdominal distension following
herpes zoster outbreak. Arch Dermatol 1998;134;1168–9.
46. Thomas JE, Howard FM. Segmental zoster paresis—a disease profile.
Neurology 1972;22:459–66.
47. Dolin R, Reichman RC, Mazur MH, Whitley RJ. Herpes zoster-
varicella infections in immunosuppressed patients. Ann Intern Med
1978;89:375–88.
48. Tyring S, Belanger R, Bezwoda W, Ljungman P, Boon R, Saltzman
RL. A randomized, double-blind trial of famciclovir versus acyclovir
for the treatment of localized dermatomal herpes zoster in
immunocompromised patients. Cancer Invest 2001;19:13–22.
49. Gnann JW, Whitley RJ. Natural history and treatment of varicella-
zoster virus in high-risk populations. J Hosp Infect 1991;18:317–29.
50. Merselis JG Jr, Kaye D, Hook EW. Disseminated herpes zoster: a
report of 17 cases. Arch Intern Med 1964;113:679–86.
51. Goffinet DR, Glatstein EJ, Merigan TC. Herpes zoster-varicella
infections and lymphoma. Ann Intern Med 1972;76:235–40.
52. Whitley RJ, Soong SJ, Dolin R, Betts R, Linnemann C Jr, Alford CA
Jr. Early vidarabine therapy to control the complications of herpes
zoster in immunosuppressed patients. N Engl J Med 1982;307:971–5.
25 Vol. 57 / RR-5 MMWR
53. Mazur MH, Dolin R. Herpes zoster at the NIH: a 20-year experience.
Am J Med 1978;65:738–44.
54. Locksley RM, Flournoy N, Sullivan KM, Meyers JD. Infection with
varicella-zoster virus after marrow transplantation. J Infect Dis
1985;152:1172–81.
55. Rogers SY, Irving W, Harris A, Russell NH. Visceral varicella zoster
infection after bone marrow transplantation without skin involvement
and the use of PCR for diagnosis. Bone Marrow Transplant
1995;15:805–7.
56. Buchbinder SP, Katz MH, Hessol NA, et al. Herpes zoster and human
immunodeficiency virus infection. J Infect Dis 1992;166:1153–6.
57. Glesby MJ, Moore RD, Chaisson RE. Clinical spectrum of herpes
zoster in adults infected with human immunodeficiency virus. Clin
Infect Dis 1995;21:370–5.
58. Vaughan-Jones SA, McGibbon DH, Bradbeer CS. Chronic verrucous
varicella-zoster infection in a patient with AIDS. Clin Exp Dermatol
1994;19:327–9.
59. Gilson IH, Barnett JH, Conant MA, Laskin OL, Williams J, Jones
PG. Disseminated ecthymatous herpes varicella zoster virus infection
in patients with acquired immunodeficiency syndrome. J Am Acad
Dermatol 1989;20:637–42.
60. Ormerod LD, Larkin JA, Margo CA, et al. Rapidly progressive her-
petic retinal necrosis: a blinding disease characteristic of advanced
AIDS. Clin Infect Dis 1998;26:34–45.
61. Siwamogstham P. Kuansuwan C. Reichart PA. Herpes zoster in HIV
infection with osteonecrosis of the jaw and tooth exfoliation. Oral
Diseases 2006;12:500–5.
62. Yawn BP, Saddier S, Wollan P, Sauver JS, Kurland M, Sy L. A population-
based study of the incidence and complications of herpes zoster
before zoster vaccine introduction. Mayo Clin Proc 2007;82:1341–9.
63. Opstelten W, van Loon AM, Schuller M, et al. Clinical diagnosis of
herpes zoster in family practice. Ann Fam Med 2007;5:305–9.
64. Rubben A, Baron JM, Grussendorf-Conen EI. Routine detection of
herpes simplex virus and varicella zoster virus by polymerase chain
reaction reveals that initial herpes zoster is frequently misdiagnosed as
herpes simplex. Br J Dermatol 1997;137:259–61.
65. Kalman CM, Laskin OL. Herpes zoster and zosteriform herpes sim-
plex virus infections in immunocompetent adults. Am J Med
1986;81:775–8.
66. Birch CJ, Druce JD, Catton MC, MacGregor L, Read T. Detection of
varicella zoster virus in genital specimens using a multiplex polymerase
chain reaction. Sex Transm Infect 2003;79:298–300
67. Nahass GT, Goldstein BA, Zhu WY, Serfling U, Penneys NS, Leonardi
CL. Comparison of Tzanck smear, viral culture, and DNA diagnostic
methods in detection of herpes simplex and varicella-zoster infection.
JAMA 1992;268:2541–4.
68. Solomon AR, Rasmussen JE, Weiss JS. A comparison of the Tzanck
smear and viral isolation in varicella and herpes zoster. Arch Dermatol
1986;122:282–5.
69. Gnann JW Jr, Whitley RJ. Clinical practice: herpes zoster. N Engl J
Med 2002;347:340–6.
70. Dahl H, Marcoccia J, Linde A. Antigen detection: the method of
choice in comparison with virus isolation and serology for laboratory
diagnosis of herpes zoster in human immunodeficiency virus-infected
patients. J Clin Microbiol 1997;35:347–9.
71. Coffin SE, Hodinka RL. Utility of direct immunofluorescence and
virus culture for detection of varicella-zoster virus in skin lesions.
J Clin Microbiol 1995;33:2792–5.
72. Morens DM, Bregman DJ, West CM, et al. An outbreak of varicella-
zoster virus infection among cancer patients. Ann Intern Med
1980;93:414–9.
73. Brunell PA, Gershon AA, Uduman SA, Steinberg S. Varicella-zoster
immunoglobulins during varicella, latency, and zoster. J Infect Dis
1975;132:49–54.
74. Harper DR, Kangro HO, Heath RB. Serological responses in varicella
and zoster assayed by immunoblotting. J Med Virol 1988;25:387–98.
75. Wittek AE, Arvin AM, Koropchak CM. Serum immunoglobulin A
antibody to varicella-zoster virus in subjects with primary varicella
and herpes zoster infections and in immune subjects. J Clin Microbiol
1983;18:1146–9.
76. Sawyer MH. Chamberlin CJ. Wu YN. Aintablian N. Wallace MR.
Detection of varicella-zoster virus DNA in air samples from hospital
rooms. J Infect Dis 1994;169:91–4.
77. Josephson A, Gombert ME. Airborne transmission of nosocomial
varicella from localized zoster. J Infect Dis 1988;158:238–41.
78. Seiler HE. A study of herpes zoster particularly in relation to
chickenpox. J Hyg 1949;47:253–62.
79. Wreghitt TG, Whipp J, Redpath C, Hollingworth W. An analysis of
infection control of varicella-zoster virus infections in Addenbrookes
Hospital Cambridge over a 5-year period, 1987–92. Epidemiol Infect
1996;117:165–71.
80. Behrman A, Schmid DS, Crivaro A, Watson B. A cluster of primary
varicella cases among healthcare workers with false-positive varicella
zoster virus titers. Infect Control Hosp Epidemiol 2003;24:202–6.
81. Brunell PA, Argaw T. Chickenpox attributable to a vaccine virus con-
tracted from a vaccinee with zoster. Pediatrics 2000;106:28.
82. Wreghitt TG, Whipp PJ, Bagnall J. Transmission of chickenpox to two
intensive care unit nurses from a liver transplant patient with zoster. J
Hosp Infect 1992;20:125–6.
83. Lopez AS, Burnett-Hartman A, Nambiar R, et al. Transmission of a
newly characterized strain of the varicella-zoster virus from a herpes
zoster patient in a long term care facility, West Virginia, 2004. J Infect
Dis 2008;197:646–53.
84. Seward JF, Zhang JX, Maupin TJ, Mascola L, Jumaan AO. Conta-
giousness of varicella in vaccinated cases: a household contact study.
JAMA 2004;292:704–8.
85. Bolyard EA, Tablan OC, Williams WW, Pearson ML, Shapiro CN,
Deitchman SD. Guideline for infection control in health care person-
nel, 1998. Available at http://www.cdc.gov/ncidod/dhqp/pdf/guide-
lines/InfectControl98.pdf.
86. Kelly PW, Petruccelli BP, Stehr-Green P, Erickson RL, Mason CJ. The
susceptibility of young adult Americans to vaccine-preventable infec-
tions: a national serosurvey of US Army recruits. JAMA
1991;266:2724–9.
87. Alter SJ, Hammond JA, McVey CJ, Myers MG. Susceptibility to vari-
cella-zoster virus among adults at high risk for exposure. Am J Infect
Control 1986;7:448–51.
88. Ferson MJ, Bell SM, Robertson PW. Determination and importance
of varicella immune status of nursing staff in a childrens hospital. J
Hosp Infect 1990;15:347–51.
89. Struewing JP, Hyams KC, Tueller JE, Gray GC. The risk of measles,
mumps, and varicella among young adults: a serosurvey of US Navy
and Marine Corps recruits. Am J Public Health 1993;83:1717–20.
90. McKinney WP, Horowitz MM, Battiola RJ. Susceptibility of hospi-
tal-based health care personnel to varicella-zoster virus infections. Am
J Infect Control 1989;17:26–30.
26 MMWR June 6, 2008
91. Brunell PA, Kotchmar GS Jr. Zoster in infancy: failure to maintain
virus latency following intrauterine infection. J Pediatr 1981;98:71–3.
92. Baba K, Yabuuchi H, Takahashi M, et al. Increased incidence of
herpes zoster in normal children infected with varicella zoster virus
during infancy: community-based follow-up study. Pediatr
1986;108:372–7.
93. Terada K, Kawano S, Yoshihiro K, Miyashima H, Morita T. Charac-
teristics of herpes zoster in otherwise normal children. Pediatr Infect
Dis J. 1993;12:960–1.
94. Nikkels AF, Nikkels-Tassoudji N, Pierard GE. Revisiting childhood
herpes zoster. Pediatr Dermatol 2004;21:18–23.
95. Thomas SL, Hall AJ. What does epidemiology tell us about risk
factors for herpes zoster? Lancet Infect Dis 2004;4:26–33.
96. Ross RT, Nicolle LE, Cheang M. Varicella zoster virus and multiple
sclerosis in a Hutterite population. J Clin Epidemiol 1995;48:1319–24.
97. Takayama N, Takayama M, Takita J. Herpes zoster in healthy chil-
dren immunized with varicella vaccine. Pediatr Infect Dis J
2000;19:169–70.
98. Hardy IB, Gershon A, Steinberg S, et al. The incidence of zoster after
immunization with live attenuated varicella vaccine. A study in chil-
dren with leukemia. N Engl J Med 1991;325:1545–50.
99. Gershon AA. Live-attenuated varicella vaccine. Infect Dis Clin N Amer
2001;15:65–81.
100. Black S, Shinefield H, Ray P, et al. Postmarketing evaluation of the
safety and effectiveness of varicella vaccine. Pediatr Infect Dis J
1999;18:1041–6.
101. Merck. VARIVAX
®
Package Insert. 2006. Available at http://
www.merck.com/product/usa/pi_circulars/v/varivax/varivax_pi.pdf.
102. Fleming DM, Cross KW, Cobb WA, Champion RS. Gender differ-
ence in the incidence of zoster. Epidemiol Infect 2004;132:1–5.
103. Insinga RP, Itzler RF, Pellissier JM. Acute/subacute herpes zoster:
healthcare resource utilisation and costs in a group of US health
plans. Pharmacoeconomics 2007;25:155–69.
104. Jumaan AO, Yu O, Jackson LA, Bohlke K, Galil K, Seward JF.
Incidence of herpes zoster, before and after varicella-vaccination-
associated decreases in the incidence of varicella, 1992–2002.
J Infect Dis 2005;191:2002–7.
105. Brisson M, Edmunds WJ, Law B, et al. Epidemiology of varicella
zoster virus infection in Canada and the United Kingdom. Epidemiol
Infect 2001;127,305–14.
106. Schmader K. Herpes zoster in older adults. Clin Infect Dis
2001;32:1481–6.
107. Berger R, Florent G, Just M. Decrease of the lymphoproliferative
response to varicella-zoster virus antigen in the aged. Infect Immun
1981;32:24–7.
108. Berger R, Luescher D, Just M. Enhancement of varicella-zoster-
specific immune responses in the elderly by boosting with varicella
vaccine. J Infect Dis 1984;149:647.
109. Choo PW, Galil K, Donahue JG, Walker AM, Spiegelman D, Platt
R. Risk factors for postherpetic neuralgia. Arch Intern Med
1997;157:1217–24.
110. De Moragas JM, Kierland RR. The outcome of patients with herpes
zoster. Arch Dermatol 1957;75:193–6.
111. Opstelten W, Van Essen GA, Schellevis F, Verheij TJ, Moons K.
Gender as an independent risk factor for herpes zoster: a population-
based prospective. Ann Epidemiol 2006;1006;16:692–5.
112. Parker CJ, Morgan K, Dewey ME. Physical illness and disability
among elderly people in England and Wales: The Medical Research
Council Cognitive Function and Ageing Study. J Epidemiol Commu-
nity Health 1997;51:494–501.
113. Di Luzio Paparatti U, Arpinelli F, Visona G. Herpes zoster and its
complications in Italy: an observational survey. J Infect 1999;38:116.
114. Donahue JG. Choo PW. Manson JE. Platt R. The incidence of
herpes zoster. Arch Intern Med 1995;155:1605–9.
115. Schmader K., George L.K., Burchett B.M, Pieper CF. Racial differ-
ences in the occurrence of herpes zoster. J Infect Dis 1995;171:701–4.
116. Schmader K. George LK. Burchett BM. Hamilton JD. Pieper CF.
Race and stress in the incidence of herpes zoster in older adults. J Am
Geriatr Soc 1998;46:973–7.
117. Garnett GP, Grenfell BT. The epidemiology of varicella-zoster virus
infections: the influence of varicella on the prevalence of herpes zoster.
Epidemiol Infect 1992;108:513–28.
118. Zak-Prelich M, Borkowski JL, Alexander F, Norval M. The role of
solar ultraviolet irradiation in zoster. Epidemiol Infect 2002;129:593–7.
119. Gallerani M, Manfredini R. Seasonal variation in herpes zoster infec-
tion. Br J Dermatol 2000;142:588–9.
120. Rusthoven JJ, Ahlgren P, Elhakim T, et al. Varicella-zoster infection
in adult cancer patients. A population study. Arch Intern Med
1988;148:1561–6.
121. Dunst J, Steil B, Furch S, Fach A, Bormann G, Marsch W. Herpes
zoster in breast cancer patients after radiotherapy. Strahlenther Onkol
2000;176:513–6.
122. Guinee VF, Guido JJ, Pfalzgraf KA, et al. The incidence of herpes
zoster in patients with Hodgkins disease: an analysis of prognostic
factors. Cancer 1985;56:642–8.
123. Schimpff S, Serpick A, Stoler B, et al. Varicella-zoster infection in
patients with cancer. Ann Intern Med 1972;76:241–54.
124. Sokal JE. Firat D. Varicella-zoster infection in Hodgkins disease:
clinical and epidemiological aspects. Am J Med 1965;39:452–63.
125. Rusthoven JJ. The risk of varicella-zoster infections in different
patient populations: a critical review. Transfus Med Rev 1994;8:
96–116.
126. Feld R. Evans WK. DeBoer G. Herpes zoster in patients with carci-
noma of the lung. Am J Med 1982;73:795–801.
127. Wilson JF. Marsa GW. Johnson RE. Herpes zoster in Hodgkins
disease. Clinical, histologic, and immunologic correlations. Cancer
1972;29:461–5.
128. Arvin AM. Varicella-zoster virus: pathogenesis, immunity, and clini-
cal management in hematopoietic cell transplant recipients. Biol Blood
Marrow Transplant 2000;6:219–30.
129. Schuchter LM, Wingard JR, Piantadosi S, Burns WH, Santos. GW,
Saral R. Herpes zoster infection after autologous bone marrow trans-
plantation. Blood 1989;74:1424–7.
130. Gourishankar S, McDermid JC, Jhangri GS, Preiksaitis JK. Herpes
zoster infection following solid organ transplantation: incidence, risk
factors and outcomes in the current immunosuppressive era. Am J
Transplant 2004;4:108–15.
131. Luby JP. Ramirez-Ronda C. Rinner S. Hull A. Vergne-Marini P. A
longitudinal study of varicella-zoster virus infections in renal trans-
plant recipients. J Infect Dis 1977;135:659–63.
132. Rand KH. Rasmussen LE. Pollard RB. Arvin A. Merigan TC. Cellu-
lar immunity and herpesvirus infections in cardiac-transplant patients.
N Engl J Med 1977;296:1372–7.
27 Vol. 57 / RR-5 MMWR
133. McNulty A, Li Y, Radtke U, et al. B. Herpes zoster and the stage and
prognosis of HIV-1 infection. Genitourin Med 1997;73:467–70.
134. Veenstra J, Krol A, van Praag RM, et al. Herpes zoster, immunologi-
cal deterioration and disease progression in HIV-1 infection. AIDS
1995;9:1153–8.
135. Glesby MJ, Hoover DR, Tan T, et al. Herpes zoster in women with
and at risk for HIV: data from the Womens Interagency HIV Study.
J Acquir Immune Defic Syndr 2004;37:1604–9.
136. Gebo KA, Kalyani R, Moore RD, Polydefkis M. The incidence of,
risk factors for, and sequelae of herpes zoster among HIV patients in
the HAART era. JAIDS 2005;40:169–74.
137. Gershon AA, Mervish N, LaRussa P, et al. Varicella-zoster virus
infection in children with underlying human immunodeficiency virus
infection. J Infect Dis 1997;176:1496–500.
138. Kahl LE. Herpes zoster infections in systemic lupus erythematosus:
risk factors and outcome. J Rheumatol 1994;21:84–6.
139. Nagasawa K, Yamauchi Y, Tada Y, Kusaba T, Niho Y, Yoshikawa H.
High incidence of herpes zoster in patients with systemic lupus erythe-
matosus: an immunological analysis. Ann Rheumatic Dis
1990;49:630–3.
140. Kang TY, Lee HS, Kim TH, Jun JB, Yoo DH. Clinical and genetic
risk factors of herpes zoster in patients with systemic lupus erythe-
matosus. Rheumatol Int 2005;25:97–102.
141. Manzi S, Kuller LH, Kutzer J, et al. Herpes zoster in systemic lupus
erythematosus. J Rheumatol 1995;22:1254–8.
142. Moutsopoulos HM, Gallagher JD, Decker JL, Steinberg AD. Her-
pes zoster in patients with systemic lupus erythematosus. Arthritis
Rheum 1978;21:789–802.
143. Pope JE, Krizova A, Ouimet JM, Goodwin JL, Lankin M. Close
association of herpes zoster reactivation and systemic lupus erythe-
matosus (SLE) diagnosis: case-control study of patients with SLE or
noninflammatory nusculoskeletal disorders. J Rheumatol
2004;31:274–9.
144. Antonelli MA, Moreland LW, Brick JE. Herpes zoster in patients
with rheumatoid arthritis treated with weekly, low-dose methotrex-
ate. Am J Med 1991;90:295–8.
145. Smitten AL, Choi HK, Hochberg MC, et al. The risk of herpes zoster
in patients with rheumatoid arthritis in the United States and the
United Kingdom. Arthritis Rheum 2007;57:1431–8.
146. Wung PK, Holbrook JT, Hoffman GS, et al. Herpes zoster in
immunocompromised patients: incidence, timing, and risk factors.
Am J Med 2005;118:1416.e9–e18.
147. Gupta G, Lautenbach E, Lewis JD. Incidence and risk factors for
herpes zoster among patients with inflammatory bowel disease. Clin
Gastroenterol Hepatol 2006;4:1483–90.
148. Brown GR. Herpes zoster: correlation of age, sex, distribution,
neuralgia, and associated disorders. South Med J 1976;69:576–8.
149. Heymann AD, Chodick G, Karpati T, et al. Diabetes as a risk factor
for herpes zoster infection: results of a population-based study in
Israel. Infection 2008;May 3. [Epub ahead of print].
150. Arndt V, Vine MF, Weigle K. Environmental chemical exposures and
risk of herpes zoster. Environ Health Perspect 1999;107:835–41.
151. Ragozzino MW, Melton LJ, Kurland LT. Herpes zoster and diabetes
mellitus: an epidemiological investigation. J Chronic Dis
1983;36:501–5.
152. Ross RT, Cheang M, Landry G, Klassen L, Doerksen K. Herpes
zoster and multiple sclerosis. Can J Neurol Sci 1999;26:29–32.
153. Palmer SR, Caul EO, Donald DE, Kwantes W, Tillett H. An out-
break of zoster? Lancet 1985;2:1108–11.
154. Su WP. Muller SA. Herpes zoster. Case report of possible accidental
inoculation. Arch Dermatol 1976;112:1755–6.
155. Brisson M, Gay NJ, Edmunds WJ, Andrews NJ. Exposure to varicella
boosts immunity to herpes-zoster: implications for mass vaccination
against chickenpox. Vaccine 2002;20:2500–7.
156. Gershon A, LaRussa P, Steinberg S, Lo SH, Mervish N, Meier P. The
protective effect of immunologic boosting against zoster: an analysis
in leukemic children who were vaccinated against chickenpox. J Infect
Dis 1996;173:450–3.
157. Schmader K, Studenski S, MacMillan J, Grufferman S, Cohen HJ.
Are stressful life events risk factors for herpes zoster? J Am Geriatr
Soc 1990;38:1188–94.
158. Massad MG, Navarro RA, Rubeiz H, et al. Acute postoperative
zoster after thoracic sympathectomy for hyperhidrosis. Ann Thorac
Surg 2004;78:2159–61.
159. Gilden DH, Katz RI. Surgical induction of zoster in a contralateral
homologous dermatomal distribution. Arch Neurol 2003;60:616–7.
160. Andrews TR, Perdikis G, Shack RB. Herpes zoster as a rare compli-
cation of liposuction. Plast Reconstr Surg 2004;113:1838–40.
161. Walter R, Hartmann K, Fleisch F, Reinhart WH, Kuhn M. Reactiva-
tion of herpesvirus infections after vaccinations? Lancet 1999;353:810.
162. Thomas SL, Wheeler JG, Hall AJ. Micronutrient intake and the risk
of herpes zoster: a case-control study. Int J Epidemiol 2006;35:307–14.
163. Haanpaa M, Nurmikko T, Hurme M. Polymorphism of the IL-10
gene is associated with susceptibility to herpes zoster. Scand J Infect
Dis 2002;34:112–4.
164. Richards P. Shingles in one family practice. Arch Fam Med 1996;5:42–6.
165. Mullooly JP, Riedlinger K, Chun C, Weinmann S, Houston H.
Incidence of herpes zoster, 1997–2002. Epidemiol Infect
2005;133:245–53.
166. Yih WK, Brooks DR, Lett SM, et al. The incidence of varicella and
herpes zoster in Massachusetts as measured by the Behavioral Risk
Factor Surveillance System (BRFSS) during a period of increasing
varicella vaccine coverage, 1998–2003. BMC Public Health
2005;5:68.
167. Russell ML, Schopflocher DP, Svenson L, Virani SN. Secular trends
in the epidemiology of shingles in Alberta. Epidemiol Infect
2007;135:908–13.
168. Perez-Farinos N, Ordobas M, Garcia-Fernandez C, et al. Varicella
and herpes zoster in Madrid, based on the Sentinel General Practi-
tioner Network: 1997–2004. BMC Infect Dis 2007;7:59.
169. Molin L. Aspects of the natural history of herpes zoster: a follow-up
investigation of outpatient material. Acta Derm-Venereol
1969;49:569–83.
170. Coplan P, Black S, Rojas C, et al. Incidence and hospitalization rates
of varicella and herpes zoster before varicella vaccine introduction: a
baseline assessment of the shifting epidemiology of varicella disease.
Pediatr Infect Dis J 2001;20:641–5.
171. Brisson M, Edmunds WJ. Epidemiology of varicella-zoster virus in
England and Wales. J Med Virol 2003;70(Suppl 1):9–14.
172. Lin F, Hadler JL. Epidemiology of primary varicella and herpes zoster
hospitalizations: the pre-varicella vaccine era. J Infect Dis
2000;181:1897–905.
28 MMWR June 6, 2008
173. MacIntyre CR, Chu CP, Burgess MA. Use of hospitalization and
pharmaceutical prescribing data to compare the prevaccination bur-
den of varicella and herpes zoster in Australia. Epidemiol Infect
2003;131:675–82.
174. Chant KG, Sullivan EA, Burgess MA, et al. Varicella-zoster virus
infection in Australia. Aust N Z J Public Health 1998;22:413–8.
175. Oh HM, Ho AY, Chew SK, Monteiro EH. Clinical presentation of
herpes zoster in a Singapore hospital. Singapore Med J 1997;38:471–4.
176. Schmader, KE. Herpes Zoster Epidemiology. In: Arvin A, Gershon
A., eds. Varicella-Zoster Virus, Cambridge, Massachusetts:
Cambridge University Press, 2007.
177. Dworkin RH, Boon RJ, Griffin DR, Phung D. Postherpetic neural-
gia: impact of famciclovir, age, rash severity and acute pain in herpes
zoster patients. J Infect Dis 1998;178:76–80S.
178. Wood MJ, Kay R, Dworkin RH, Soong SJ, Whitley RJ. Oral acyclovir
therapy accelerates pain resolution in patients with herpes zoster: a
meta-analysis of placebo-controlled trials. Clin Infect Dis
1996;22:341–7.
179. Whitley RJ, Weiss H, Gnann JW Jr, et al. Acyclovir with and without
prednisone for the treatment of herpes zoster: a randomized, placebo-
controlled trial. Ann Intern Med 1996;125:376–83.
180. Wood MJ, Johnson RW, McKendrick MW, Taylor J, Mandal BK,
Crooks J. A randomized trial of acyclovir for 7 days or 21 days with
and without prednisolone for treatment of acute herpes zoster. N Engl
J Med 1994;330:896–900.
181. Dubinsky RM, Kabbani H, El-Chami Z, Boutwell C, Ali H. Practice
parameter: treatment of postherpetic neuralgia: an evidence-based
report of the Quality Standards Subcommittee of the American Acad-
emy of Neurology. Neurology 2004; 63:959–65.
182. Hempenstall K, Nurmikko TJ, Johnson RW, A’Hern RP, Rice AS.
Analgesic therapy in postherpetic neuralgia: a quantitative system-
atic review. PLoS Med 2005;2:e164.
183. CDC. A new product (VariZIG) for postexposure prophylaxis of
varicella available under an investigational new drug application
expanded access protocol. MMWR 2006;55:209–10.
184. Takahashi M, Otsuka T, Okuno Y, Asano Y, Yazaki T, Isomura S.
Live vaccine used to prevent the spread of varicella in children in
hospital. Lancet 1974;2:1288–90.
185. Schmader K, Saddier P, Johnson G, et al. The effect of a zoster
vaccine on interference of herpes zoster with activities of daily living
(ADL) [Abstract 859]. 44th Annual Meeting of IDSA, Oct. 12–15,
2006, Toronto..
186. Merck. ZOSTAVAX
®
Package Insert. 2006. Available at http://
www.merck.com/product/usa/pi_circulars/z/zostavax/zostavax_pi.pdf.
187. Food and Drug Administration. Biological Products Advisory Com-
mittee, 2005. Available at http://www.fda.gov/ohrms/dockets/ac/05/
slides/5-4198S2_1.pdf .
188. Merck. An overview of herpes zoster/PHN and the Shingles Preven-
tion Study for Zostavax
®
. Presentation to the Advisory Committee
on Immunization Practices (ACIP) meeting; February 2006.
189. Food & Drug Administration, Vaccines & Related Biological Prod-
ucts Advisory Committee. 2005 http://www.fda.gov/OHRMS/
DOCKETS/ac/05/briefing/5-4198B2_1.pdf .
190. Schlienger K, Lange J, Tyring SK, et al. Immunogenicity, kinetics of
VZV-Specific CD4+ T-cell g-IFN production and safety of a live
attenuated Oka/Merck zoster vaccine in healthy adults >60 years of
age [Abstract 857]. 44th Annual Meeting of IDSA, Oct. 12–15,
2006, Toronto.
191. Sperber SJ, Smith BV, Hayden FG.Serologic response and
reactogenicity to booster immunization of healthy seropositive adults
with live or inactivated varicella vaccine. Antiviral Res 1992;17:213–22.
192. Oxman M, Levin M, Johnson G, Zhang J, Caulfield M. The effect of
age on clinical and immunologic responses to a herpes zoster vaccine
[Abstract 858]. 44th Annual Meeting of IDSA, Oct. 12–15, 2006,
Toronto.
193. Sutradhar SC, Wang WWB, Schlienger K, et al. Comparison of
immunogenicity and safety of Zostavax
®
in adults 50 to 59 years old
and those >60 years old [Abstract S27]. 10th Annual Conference on
Vaccine Research, April 30–May 2, 2007, Baltimore MD.
194. Levin MJ, Barber D, Goldblatt E, et al. Use of a live attenuated
varicella vaccine to boost varicella-specific immune responses in
seropositive people 55 years of age and older: duration of booster
effect. J Infect Dis 1998;17(Suppl 1)8:109–12.
195. Tyring SK, Diaz-Mitoma F, Padget LG, et al. Safety and tolerability
of a high-potency zoster vaccine in adults >50 years of age. Vaccine
2007;25:1877–83.
196. Ortega-Sanchez IR. Projected cost-effectiveness of vaccinating US
elderly to prevent shingles. Oral presentation to the Advisory Com-
mittee for Immunization Practices (ACIP) Meeting, June 2006.
197. Pellissier JM, Brisson M, Levin MJ. Evaluation of the cost-
effectiveness In the United States of a vaccine to prevent herpes
zoster and postherpetic neuralgia in older adults. Vaccine
2007;25:8326–37.
198. Insinga RP, Itzler RF, Pellissier JM, Saddier P, Nikas AA. The inci-
dence of herpes zoster in a United States administrative database.
J Gen Intern Med 2005;20:748–53.
199. Rothberg MB, Virapongse A, Smith KJ. Cost-effectiveness of a vac-
cine to prevent herpes zoster and postherpetic neuralgia in older
adults. Clin Infect Dis 2007;44:1280–8.
200. Dworkin RH, White R, O’connor AB, Baser O, Hawkins K.
Healthcare costs of acute and chronic pain associated with a diagno-
sis of herpes zoster. J Am Geriatr Soc 2007;55:1168–75.
201. Dworkin RH, White R, O’Connor AB, Hawkins K. Health care
expenditure burden of persisting herpes zoster pain. Pain Medicine
2008;9:348–53.
202. Hornberger J, Robertus K. Cost-effectiveness of a vaccine to prevent
herpes zoster and postherpetic neuralgia in older adults. Ann Intern
Med 2006;145:317–25.
203. Ho A, Coplan PM, Lee A, et al. Cost-effectiveness of varicella zoster
vaccination in older adults. Canadian J Anesthesia 2006 (Suppl 1):
26394.
204. Gold MR, Siegel JE, Russell LB, MC Weinstein. Cost-effectiveness
in health and medicine. New York, New York: Oxford University
Press, 1996.
205. Schwartz B, Ortega-Sanchez IR. Economic analyses of vaccines and
vaccination programs. In: Levine MM, Woodrow GC, Kaper JB,
Cobon GA, eds. New Generation Vaccines. New York, New York:
Marcel Dekker, 2004.
206. World Health Organization. Choosing interventions that are cost-
effective. Available at www.who.int/choice/costs/CER_levels/en/
index.html.
207. Neumann PJ, Rosen AB, Weinstein MC. Medicare and cost-
effectiveness analysis. N Engl J Med 2005;353:1516–22.
208. The CEA Registry. Tufts-New England Medical Center Institute for
Clinical Research and Health Policy Studies. Available at www.tufts-
nemc.org/cearegistry.
29 Vol. 57 / RR-5 MMWR
209. CDC. General recommendations on immunization: recommenda-
tions of the Advisory Committee on Immunization Practices. MMWR
2006;55(No. RR-15).
210. Ihara T, Kamiya H, Torigoe S, Sakurai M, Takahashi M. Viremic
phase in a leukemic child after live varicella vaccination. Pediatrics
1992;89:147–9.
211. Levin MJ, Oxman MN, Zhang JH, et al. VZV-specific immune
responses in elderly recipients of a herpes zoster vaccine. J Infect Dis
2008;197:825–35.
212. Bohlke K, Galil K, Jackson LA, et al. Postpartum varicella vaccina-
tion: is the vaccine virus excreted in breast milk? Obstet Gynecol
2003;102:970–7.
213. National Center for Health Statistics. Health, United States, 2005.
Available at http://www.cdc.gov/nchs/data/hus/hus05.pdf#088.
214. Cheney C, Ramsdell JW. Effect of medical records’ checklists
on implementation of periodic health measures. Am J Med 1987;83:
129–36.
215. Cohen DI, Littenberg B, Wetzel C, Neuhauser D. Improving physi-
cian compliance with preventive medicine guidelines. Med Care
1982;20:1040–5.
216. Briss PA, Rodewald LE, Hinman AR, et al. Reviews of evidence
regarding interventions to improve vaccination coverage in children,
adolescents, and adults: The Task Force on Community Preventive
Services. Am J Prev Med 2000;18:97–140.
217. Gyorkos TW, Tannenbaum TN, Abrahamowicz M, et al. Evaluation
of the effectiveness of immunization delivery methods. Can J Public
Health 1994;85(Suppl 1):S14-30.
218. Buffington J, Bell KM, LaForce FM, et al. A target-based model for
increasing influenza immunizations in private practice. J Gen Intern
Med 1991;6:204–9.
219. CDC. Assessing adult vaccination status at age 50 years. MMWR
1995;44:561–3.
220. CDC. Adult immunization programs in nontraditional settings: quality
standards and guidance for program evaluation—a report of the
National Vaccine Advisory Committee. MMWR 2000;49(No. RR-1).
221. Weibel RE, Neff BJ, Kuter BJ, et al. Live attenuated varicella virus
vaccine. Efficacy trial in healthy children. N Eng J Med
1984;310:1409–15.
222. Shinefield H, Black S, Digilio L, et al. Evaluation of a quadrivalent
measles, mumps, rubella and varicella vaccine in healthy children.
Pediatr Infect Dis J 2005;24:665–9.
223. Shinefield H, Black S, Williams WR, et al. Dose-response study of a
quadrivalent measles, mumps, rubella and varicella vaccine in healthy
children. Pediatr Infect Dis J 2005;24:670–5.
224. Civen RH, Maupin TJ, Xiao H, Seward JF, Jumaan AO, Mascola L.
A population based study of herpes zoster in children and adoles-
cents post-varicella licensure, Antelope Valley 2000–2003. 38th
National Immunization Conference, Memphis, TN, May 2004.
30 MMWR June 6, 2008
Advisory Committee on Immunization Practices Shingles Work Group
Chair: John Treanor, MD, Rochester, New York.
Members: Members: William L. Atkinson, MD, MPH, Atlanta, Georgia; Jeffrey I. Cohen, MD, Bethesda, Maryland; Robert H. Dworkin, PhD,
Rochester, New York; Sandra Gambescia, Atlanta, Georgia; Paul M. Gargiullo, PhD, Atlanta, Georgia; Anne A. Gershon, MD, New York, New York; John
W. Glasser, PhD, MPH, Atlanta, Georgia; Dalya Güris, MD, MPH, Atlanta, Georgia; Penina Haber, MPH, Atlanta, Georgia; Rafael Harpaz, MD, MPH,
Atlanta, Georgia; Beth F. Hibbs, MPH, Atlanta, Georgia; John K. Iskander, MD, MPH, Atlanta, Georgia; Samuel L. Katz, MD, Durham, North Carolina;
Philip R. Krause, MD, Bethesda Maryland; Phillip S. LaRussa, MD, New York, New York; Myron J. Levin, MD, Denver, Colorado; Tracy A. Lieu, MD,
MPH, Boston, Massachusetts; Mona E. Marin, MD, MPH, Atlanta, Georgia; Kathleen M. Neuzil, MD, MPH, Seattle Washington; Kristin Nichol, MD,
MPH, MBA, Minneapolis, Minnesota; Ismael R. Ortega- Sánchez, PhD, Atlanta, Georgia; Gregory A. Poland, MD, Rochester, Minnesota; Sara
Rosenbaum, JD, Washington, DC; Tammy A. Santibanez, PhD; William Schaffner, MD, Nashville, Tennessee; Kenneth E. Schmader, MD, Durham, North
Carolina; D. Scott Schmid, PhD, Atlanta, Georgia; Jane Seward, MBBS, MPH, Atlanta, Georgia; Heather Stafford, Philadelphia, Pennsylvania; Ray
Strikas, MD, Washington, DC; Gregory S. Wallace, MD, Atlanta, Georgia; Barbara Watson, MB ChB, Philadelphia, Pennsylvania.
Advisory Committee on Immunization Practices Membership List, June 2007
Chairman: Jon S. Abramson, MD, Wake Forest University School of Medicine, Winston-Salem, North Carolina.
Executive Secretary: Larry K. Pickering, MD, CDC, Atlanta, Georgia.
Members: Ban Mishu Allos, MD, Vanderbilt University School of Medicine, Nashville, Tennessee; Carol Baker, MD, Baylor College of Medicine,
Houston, Texas; Robert L. Beck, JD, Palmyra, Virginia; Janet R. Gilsdorf, MD, University of Michigan, Ann Arbor, Michigan; Harry Hull, MD, Minnesota
Department of Health, Minneapolis, Minnesota; Susan Lett, MD, MPH, Massachusetts Department of Public Health, Jamaica Plain, Massachusetts;
Tracy Lieu, MD, MPH, Harvard Pilgrim Health Care and Harvard Medical School, Boston, Massachusettes; Dale L. Morse, MD, New York State
Department of Health, Albany, New York; Julia Morita, MD, Chicago Department of Public Health, Chicago, Illinois; Kathleen Neuzil, MD, MPH,
University of Washington, Seattle, Washington; Patricia Stinchfield, Childrens Hospitals and Clinics, St. Paul, Minnesota; Ciro Valent Sumaya, MD,
MPH, Texas A&M University System Health Science Center, College Station, Texas; John J. Treanor, MD, University of Rochester, Rochester, New York;
and Robin J. Womeodu, MD, University of Tennessee Health Science Center, Memphis, Memphis, Tennessee.
Ex-Officio Members: James Cheek, MD, Indian Health Service, Albuquerque, New Mexico; Wayne Hachey, DO, Department of Defense, Falls Church,
Virginia; Geoffrey S. Evans, MD, Health Resources and Services Administration, Rockville, Maryland; Bruce Gellin, MD, National Vaccine Program
Office, Washington, DC; Linda Murphy, Centers for Medicare and Medicaid Services, Baltimore, Maryland; George T. Curlin, MD, National Institutes
of Health, Bethesda, Maryland; Norman Baylor, PhD, U.S. Food and Drug Administration, Rockville, Maryland; and Kristin Lee Nichol, MD, Department
of Veterans Affairs, Minneapolis, Minnesota.
Liaison Representatives: American Academy of Family Physicians, Jonathan Temte, MD, Madison, Wisconsin, and Doug Campos-Outcalt, MD, Phoenix,
Arizona; American Academy of Pediatrics, Keith Powell, MD, Akron, Ohio, and Carol Baker, MD, Houston, Texas; Americas Health Insurance Plans,
Andrea Gelzer, MD, Hartford, Connecticut; American College Health Association, James C. Turner, MD, Charlottesville, Virginia; American College of
Obstetricians and Gynecologists, Stanley Gall, MD, Louisville, Kentucky; American College of Physicians, Kathleen M. Neuzil, MD, Seattle, Washington;
American Medical Association, Litjen Tan, PhD, Chicago, Illinois; American Pharmacists Association, Stephan L. Foster, PharmD, Memphis, Tennessee;
Association of Teachers of Preventive Medicine, W. Paul McKinney, MD, Louisville, Kentucky; Biotechnology Industry Organization, Clement Lewin,
PhD, Cambridge, Massachusetts; Canadian National Advisory Committee on Immunization, Monica Naus, MD, Vancouver, British Columbia; Healthcare
Infection Control Practices Advisory Committee, Steve Gordon, MD, Cleveland, Ohio; Infectious Diseases Society of America, Samuel L. Katz, MD,
Durham, North Carolina; London Department of Health, David Salisbury, MD, London, United Kingdom; National Association of County and City
Health Officials, Nancy Bennett, MD, Rochester, New York, and Jeffrey S. Duchin, MD, Seattle, Washington; National Coalition for Adult Immunization,
David A. Neumann, PhD, Alexandria, Virginia; National Foundation for Infectious Diseases, William Schaffner, MD, Nashville, Tennessee; National
Immunization Council and Child Health Program, Romeo S. Rodriquez, Mexico City, Mexico; National Medical Association, Patricia Whitley-Williams,
MD, New Brunswick, New Jersey; National Vaccine Advisory Committee, Gary Freed, MD, Swiftwater, Pennsylvania, and Peter Paradiso, PhD, Collegeville,
Pennsylvinia; Society for Adolescent Medicine, Amy B. Middleman, MD, Houston, Texas; Pharmaceutical Research and Manufacturers of America,
Damian A. Araga, Swiftwater, Pennsylvania.
MMWR
The Morbidity and Mortality Weekly Report (MMWR) Series is prepared by the Centers for Disease Control and Prevention (CDC) and is available free of charge
in electronic format. To receive an electronic copy each week, send an e-mail message to listserv@listserv.cdc.gov. The body content should read SUBscribe mmwr-
toc. Electronic copy also is available from CDC’s Internet server at http://www.cdc.gov/mmwr or from CDC’s file transfer protocol server at ftp://ftp.cdc.gov/pub/
publications/mmwr. Paper copy subscriptions are available through the Superintendent of Documents, U.S. Government Printing Office, Washington, DC
20402; telephone 202-512-1800.
Data in the weekly MMWR are provisional, based on weekly reports to CDC by state health departments. The reporting week concludes at close of business on
Friday; compiled data on a national basis are officially released to the public on the following Friday. Data are compiled in the National Center for Public Health
Informatics, Division of Integrated Surveillance Systems and Services. Address all inquiries about the MMWR Series, including material to be considered for
publication, to Editor, MMWR Series, Mailstop E-90, CDC, 1600 Clifton Rd., N.E., Atlanta, GA 30333 or to [email protected].
All material in the MMWR Series is in the public domain and may be used and reprinted without permission; citation as to source, however, is appreciated.
Use of trade names and commercial sources is for identification only and does not imply endorsement by the U.S. Department of Health and Human Services.
References to non-CDC sites on the Internet are provided as a service to MMWR readers and do not constitute or imply endorsement of these organizations or
their programs by CDC or the U.S. Department of Health and Human Services. CDC is not responsible for the content of these sites. URL addresses listed in
MMWR were current as of the date of publication.
U.S. Government Printing Office: 2008-723-026/41097 Region IV ISSN: 1057-5987