You are on page 1of 16

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

net/publication/227827311

West Nile virus neuroinvasive disease

Article  in  Annals of Neurology · September 2006


DOI: 10.1002/ana.20959

CITATIONS READS
318 362

10 authors, including:

Roberta L Debiasi Kathleen Y Haaland


Children's National Medical Center University of New Mexico
145 PUBLICATIONS   5,767 CITATIONS    132 PUBLICATIONS   9,538 CITATIONS   

SEE PROFILE SEE PROFILE

Steven A Pergam Molly King


Fred Hutchinson Cancer Research Center University of New Mexico
229 PUBLICATIONS   4,994 CITATIONS    14 PUBLICATIONS   576 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

CNS Infections View project

History of Neurology View project

All content following this page was uploaded by Kenneth L Tyler on 24 August 2022.

The user has requested enhancement of the downloaded file.


NEUROLOGICAL PROGRESS

West Nile Virus Neuroinvasive Disease


Larry E. Davis, MD,1,2 Roberta DeBiasi, MD,3 Diane E. Goade, MD,4 Kathleen Y. Haaland, PhD,2,5,6
Jennifer A. Harrington, MA,5,7 JoAnn B. Harnar, RN,1,2 Steven A. Pergam, MD,8 Molly K. King, MD,1,2
B. K. DeMasters, MD,9,10 and Kenneth L. Tyler, MD9,11

Since 1999, there have been nearly 20,000 cases of confirmed symptomatic West Nile virus (WNV) infection in the United
States, and it is likely that more than 1 million people have been infected by the virus. WNV is now the most common cause
of epidemic viral encephalitis in the United States, and it will likely remain an important cause of neurological disease for the
foreseeable future. Clinical syndromes produced by WNV infection include asymptomatic infection, West Nile Fever, and West
Nile neuroinvasive disease (WNND). WNND includes syndromes of meningitis, encephalitis, and acute flaccid paralysis/polio-
myelitis. The clinical, laboratory, and diagnostic features of these syndromes are reviewed here. Many patients with WNND have
normal neuroimaging studies, but abnormalities may be present in areas including the basal ganglia, thalamus, cerebellum, and
brainstem. Cerebrospinal fluid invariably shows a pleocytosis, with a predominance of neutrophils in up to half the patients.
Diagnosis of WNND depends predominantly on demonstration of WNV-specific IgM antibodies in cerebrospinal fluid. Recent
studies suggest that some WNV-infected patients have persistent WNV IgM serum and/or cerebrospinal fluid antibody re-
sponses, and this may require revision of current serodiagnostic criteria. Although there is no proven therapy for WNND, several
vaccines and antiviral therapy with antibodies, antisense oligonucleotides, and interferon preparations are currently undergoing
human clinical trials. Recovery from neurological sequelae of WNV infection including cognitive deficits and weakness may be
prolonged and incomplete.
Ann Neurol 2006;60:286 –300

West Nile Virus lie in a herringbone-like arrangement that covers the


West Nile virus (WNV) is an RNA virus belonging to lipid bilayer.3 The E protein is responsible for receptor
the Japanese encephalitis serocomplex of the genus binding and fusion with target cell membranes and
Flavivirus of the family Flaviviridae.1 WNV is an “ar- contains epitopes recognized by neutralizing antibod-
bovirus” (arthropod-borne virus) that is transmitted ies.1 The prM protein forms homotrimers that cap the
by a mosquito vector. WNV isolates are grouped into external tip of the E protein.
lineages and clades based on unique amino acid poly- The WNV genome consists of approximately 10.8kb
morphisms or deletions in their envelope proteins.1,2 single-stranded (⫹)-sense RNA. The genome serves as
WNV isolates causing significant human disease be-
messenger RNA with a single long open reading frame.
long to lineage 1, which is subdivided into four clades
The resulting 3,433-amino acid polyprotein is cleaved
(Indian, Kunjin, A, B). US WNV isolates belong to
by both host and viral proteases to form the C, prM,
clade B of lineage 1.2
WN virions are spherical, enveloped particles with a E, and 7 NS (nonstructural) proteins. NS proteins in-
50nm diameter, icosahedral symmetry, and a smooth clude enzymes involved in genome replication includ-
spikeless outer surface.3 The genome is contained ing the RNA-dependent RNA polymerase.1 Virus rep-
within a capsid composed of the viral C (capsid) pro- lication occurs in the cytoplasm in close association
tein. The capsid is surrounded by a 4nm-thick host- with rough endoplasmic reticulum. Virions are assem-
derived lipid membrane into which 180 copies of the bled within the lumen of the endoplasmic reticulum,
virus-encoded E (envelope) and prM (membrane) gly- and the nascent virions are transported within vesicles
coproteins are inserted. Homodimers of the E protein to the cell surface and released by exocytosis.1

From the 1Neurology Services, New Mexico Veterans Affairs Health Center, Denver, CO.
Care System; 2Department of Neurology, University of New Mexico,
Albuquerque, NM; 3Departments of Pediatrics, George Washington Received Apr 19, 2006, and in revised form Jul 19. Accepted for
University School of Medicine, Washington, DC, 4Department of In- publication Jul 24, 2006.
ternal Medicine, University of New Mexico; 5Psychology Services, Published online Sep 25, 2006 in Wiley InterScience
New Mexico Veterans Affairs Health Care System; Departments of (www.interscience.wiley.com). DOI: 10.1002/ana.20959
6
Psychiatry and 7Psychology, University of New Mexico, Albuquer-
que, NM; 8Department of Internal Medicine, University of Washing- Address correspondence to Dr Tyler, Neurology B-182, University
ton, Seattle, WA; Departments of 9Neurology, 10Pathology, and of Colorado Health Sciences Center, 4200 East 9th Avenue, Den-
11
Medicine, Microbiology and Immunology, University of Colorado ver, CO 80262. E-mail: ken.tyler@uchsc.edu
Health Sciences Center and the Denver Veterans Affairs Medical

286 © 2006 American Neurological Association


Published by Wiley-Liss, Inc., through Wiley Subscription Services
History largest WNV encephalitis outbreak ever reported. In
WNV was first isolated in 1937 from the blood of a 2003, 9,862 cases of West Nile infection including
woman with a febrile illness who lived in the West 2,860 (29%) cases of meningitis or encephalitis were
Nile region of Uganda.4 Field studies along the upper reported to the Centers for Disease Control and Pre-
Nile River in the 1950s helped defined the basic ecol- vention (CDC). These numbers declined to 2,539 total
ogy and pathogenesis of WNV infection. WNV was cases and 1,142 cases (45%) of meningitis or enceph-
found to cycle between Culicine mosquitoes and native alitis in 2004, and 3,000 total cases and 1,294 cases of
birds. Birds served as the natural amplifying host, and meningitis or encephalitis (43%) in 2005.13 192 cases
humans were accidental secondary hosts. Symptomatic of WNV infection from 20 states, including 84 pa-
human infection resulted in a mild dengue-like illness tients with neurological disease, have already been re-
characterized by fever, malaise, lymphadenopathy, and ported to the CDC in 2006 (as of August 8, 2006).
rash; infection resolved quickly without sequelae.5 The current seroprevalence rate of WNV in the
In the early 1950s, patients with advanced cancers United States is extremely low (⬍3%),14 although rates
were deliberately inoculated with WNV to test the po- as high as 9.5% were reported in Nebraska blood do-
tential efficacy of the virus as an oncolytic agent. nors and up to 20% in some Nebraska communities
Ninety-five patients with metastatic cancers were in- after the 2003 epidemic.15
jected intramuscularly or intraperitoneally (two cases)
with mouse brain or chick embryo suspensions of Ecology of West Nile Virus
WNV. Eleven percent of patients experienced “sugges- WNV is maintained in nature by cycling between
tive clinical signs of diffuse encephalitis” including de- more than 200 species of birds and many species of
pressed sensorium, involuntary muscle twitching, and mosquitoes.16 Mosquito species vary in their abun-
Parkinsonism. One patient experienced a transient dance in different geographic regions, their propensity
“complete flaccid paralysis of both lower extremities” to feed on mammals including humans, and the effi-
with preserved sensation and reflexes.6 ciency with which they transmit infection (“vector
WNV infection was subsequently recognized to be competence”). The species likely to be important in
endemic in Africa, Europe, and Asia.7 Fatal cases of human transmission of WNV in the United States are
encephalitis were a significant feature of WNV epi- Culex tarsalis and quinquefasciatus in the West, Culex
demics in the 1990s in Romania, Russia, and Israel. quinquefasciatus and nigripalpus in the Southeast, and
During these outbreaks, up to 60% of hospitalized pa- Culex pipiens and restuans in the Northeast.
tients had neuroinvasive disease with a 4 to 7% mor- WNV infection involves both bird-to-bird and bird-
tality rate.8 to-human transmission, and the mosquito species re-
Naturally occurring WNV infection first appeared in sponsible for these two cycles often differ. The likeli-
North America in 1999, when an outbreak in New hood that a mosquito feeding on an infected host will
York City resulted in 59 patients with encephalitis and become infected and its subsequent capacity to trans-
7 deaths.9 Comparison of the nucleotide sequence of mit infection increases dramatically as the level of vire-
the envelope gene of the New York City virus (NY99) mia increases in the host. WNV enters the mosquito in
indicated that it was most closely related (⬎99.8% infected blood, penetrates the gut, replicates in tissues
amino acid homology) to a virus that had been isolated including the nervous system and salivary glands, and
in 1998 from a goose in Israel,10 suggesting that NY99 produces a noncytopathic infection that persists for the
originated from the Middle East or in Eastern Europe, life of the insect.17 An infected Culex pipiens quinque-
where a similar virus was also subsequently shown to fasciatus mosquito injects about 104 plaque-forming
be circulating. units of virus into the host while feeding.18
Infected migratory birds,11 or possibly illicitly im- WNV must survive through winter to initiate new
ported exotic birds, may have introduced WNV into annual cycles of infection. Several species of WNV-
North America. Because humans are “dead-end hosts” infected mosquitoes, including Culex pipiens, can sur-
for WNV (except under special circumstances such as vive dormant during winter.19 Female mosquitoes can
organ transplantation or blood transfusion), an infected also transmit WNV to their eggs via transovarian trans-
person was not the source of the New York outbreak. mission.20 The infected eggs hatch in the spring into
A less likely possibility is that virus was introduced via pupa that mature into infected adults capable of trans-
infected mosquitoes inadvertently transported in an air- mitting WNV.
plane or other carrier. Mosquitoes transmit infection to susceptible birds,
After its introduction to the United States in 1999, which serve as the major amplifying hosts for WNV.
WNV spread to produce human infection throughout Insectivorous birds can also acquire WNV after being
the continental United States. WNV has been respon- fed infected mosquitoes.16 Birds may have viremia that
sible for the largest outbreaks of arboviral encephalitis lasts for more than 100 days,21 allowing for repeated
in US history,12 and the US epidemic in 2003 was the cycles of mosquito infection. The magnitude and du-

Davis et al: WNV Neuroinvasive Disease 287


ration of viremia varies in different bird species. The isolated from blood. Viremia is typically absent at the
highest titer viremias (⬎1010 plaque-forming units per time of onset of symptomatic illness (see later).
milliliter) have been reported in passeriforme birds (eg, WNV central nervous system (CNS) disease occurs
jays, grackles, finches, crows, sparrows).21 Viremia of with increased frequency in immunocompromised in-
this magnitude leads to subsequent transmission to dividuals and the elderly,28 –30 likely reflecting a more
more than 80% of biting mosquitoes.22 Infected birds prolonged and higher titer viremia resulting, in part,
can also shed significant amounts of virus from the clo- from delayed development of neutralizing antibodies.29
aca and nasopharynx, allowing viral spread between The precise mechanism by which WNV enters the
birds independent of mosquitoes. The frequency of fa- CNS is unknown, but experimental WNV infection
tal WNV infection varies between bird species and results in elevated levels of inflammatory cytokines in-
generally parallels the magnitude of viremia.21 Interest- cluding tumor necrosis factor-␣ that can alter blood–
ingly, significant bird deaths were not a feature of brain barrier permeability.31 WNV-infected mice that
WNV epidemics before the 1998 outbreak in Israel lack Toll-like receptor 3 (Tlr 3⫺/⫺) have decreased lev-
and represent a change in the natural ecology and els of circulating inflammatory cytokines, less perme-
pathogenesis of WNV. ability of their blood–brain barrier, and reduced WNV
In birds, WNV infection involves the heart, kidney, entry into the brain (neuroinvasion).31
spleen, gut, adrenal glands, liver, and brain.23 A sud- Neurotropic flaviviruses may enter the CNS after di-
den die-off of crows or other passerine species can serve rect infection of endothelial cells in the cerebral micro-
as a sentinel event presaging subsequent human epi- vasculature or after viremic dissemination to the olfac-
demics.24 In parts of the Midwest, WNV decimated tory bulb with subsequent transneuronal spread to the
native crow populations to 10% or less of preepidemic CNS.26 Whether WNV also uses these pathways is un-
levels. Massive deaths in susceptible bird populations known. In WNV-infected mice that lack functional B
reduce the major natural amplifying reservoir of WNV cells (␮MT mice), passive transfer of WNV-specific
and may help to explain why human cases often de- antibody prevents viremia but does not completely
crease sharply in a particular year after a major WNV block viral spread to the CNS, suggesting that nonhe-
outbreak. Conversely, infected migratory birds appear matogenous pathways, such as retrograde axonal trans-
to play a major role in the spread and introduction of port from infected peripheral neurons, may contribute
WNV into new geographic regions, and migration of to CNS entry.32
uninfected susceptible birds may facilitate continued In the CNS, WNV infects neurons and, less com-
WNV transmission.11 monly, glial cells in the cerebral cortex, basal ganglia,
Besides birds, many other vertebrates are susceptible brainstem, and spinal cord.33 Both cultured neurons
to WNV infection. Naturally occurring neurological and astrocytes support WNV infection, although with
disease is rare in nonhuman mammals except for differing kinetics.34 Infection of microglial cells is abor-
horses, in which more than 15,000 cases of WNV in- tive but results in release of proinflammatory cytokines
fection have been reported in North America alone.13 (interleukin-6, tumor necrosis factor-␣), and chemo-
About 10% of infected horses develop encephalitis or kines (CXCL10, CCL2, CCL5). Infected neurons and
myelitis with a mortality rate of 28 to 45%.25 Equines astrocytes also produce chemokines including CXCL10
and vertebrates other than birds do not typically de- and CCL5, which play a critical role in recruitment of
velop high titer viremia and are unlikely to play a role virus-specific T cells into the CNS34,35 (see later).
in viral maintenance or transmission. The integrin ␣v␤3 has been identified as a putative
WNV receptor on nonneuronal cells.36,37 The WNV
West Nile Virus Human Pathogenesis neuronal receptor remains unknown. WNV attach-
WNV is injected intradermally in the infected saliva of ment results from interaction between the host cell re-
a biting mosquito and initially replicates in Langerhans ceptor and the viral envelope glycoprotein.37 Monoclo-
dendritic cells.26 The infected Langerhans cells migrate nal antibodies specific for and peptide fragment
to draining lymph nodes from which the virus enters mimetics of a subregion (domain III) of the envelope
the bloodstream (primary viremia).27 Primary viremia glycoprotein inhibit WNV infection of cultured
disseminates the virus to the reticuloendothelial system, cells.38,39 WNV infection can also be inhibited by an-
where replication further augments viremia (secondary tibodies against ␣v␤3 and by adsorbing virus with sol-
viremia).26 Virus can be detected in blood within 1 to uble ␣v␤3.36,37
2 days of a mosquito bite, and viremia typically persists WNV entry follows clathrin-mediated endocytosis.40
for up to a week. The termination of viremia coincides Virus is then transported toward the endoplasmic re-
with the appearance of serum IgM neutralizing anti- ticulum in lysosomal vesicles. As the pH inside the ves-
bodies. Human viremia is of a low level, and although icle decreases, the viral envelope fuses with the lysoso-
viral nucleic acid can be detected using sensitive nu- mal membrane releasing viral RNA into the cytoplasm,
cleic acid amplification tests, the virus is only rarely initiating genome and protein replication.

288 Annals of Neurology Vol 60 No 3 September 2006


WNV-mediated death of infected cells can occur two cohorts of Caucasian WNV-infected patients with
by either necrosis or apoptosis.41– 43 In cultured Vero symptomatic disease compared with uninfected ran-
cells, high multiplicity infection is followed by necro- dom blood donors. Homozygosity was also signifi-
sis, whereas low multiplicity infection results in cantly associated with fatal disease (OR, 13.2).49
mitochondrial-dependent apoptosis.42 Apoptosis can Innate immunity also plays a role in controlling
also be induced in a variety of cells by expression of WNV infection. Mice infected with WNV show in-
both WNV capsid proteins and the nonstructural pro- creased levels of serum interferon (IFN)-␣ and in-
tein NS3.42,44 In WNV-infected mice, apoptotic neu- creased levels of IFN-␣␤ messenger RNA in the brain.
rons are prominent in the spinal cord but rare in the WNV lethality is dramatically enhanced in mice that
brain, suggesting that the mechanisms of WNV- lack IFN-␣␤ receptor, and these mice show enhanced
induced cell death may differ within specific regions of viremia and elevated viral titers in both the brain and
the CNS.41 peripheral organs.50 Treatment of cells, including pri-
The role of host immunity in clearance of WNV mary neurons, with IFN-␣␤ before WNV infection re-
from the CNS has been studied extensively in duces viral titer and inhibits cytopathicity,50,51 provid-
mice.32,41,45 Passive transfer of WNV antibody before ing a rationale for clinical trials involving IFN-␣ in
footpad challenge with WNV protects immunocompe- humans (see later).
tent mice, but only delays disease onset and death in
mice that lack functional T and B cells (RAG1 Human West Nile Virus Transmission
mice).32 Delay in administration dramatically decreases Most human WNV infections result from mosquito
the protective efficacy of passively transferred WNV bites. Infection also has resulted from transfusion of
antibody, although some protection is still observed WNV-infected blood products, although the risk re-
even when the virus has already reached the CNS. The mains extremely low.52–54 Donors in the prodromal
appearance of neutralizing IgM antibodies plays a crit- phase of WNV infection may be asymptomatic despite
ical role in terminating viremia and preventing dissem- having high titers of infectious virus in their blood-
ination of virus to the CNS, because mice that lack streams. In 2003, 877 units of WNV-infected blood
secreted IgM (sIgM⫺/⫺) show markedly enhanced vire- were identified among 2.5 million units screened (3.5/
mia and decreased survival compared with sIgM⫹/⫹ 10,000).53 Approximately two-thirds of nucleic acid
(wild-type) mice, despite normal capacity to secrete amplification test–positive blood units lack detectable
other immunoglobulin isotypes.46 anti-WNV antibody and are likely infectious.54
Clearance of WNV from the brain and other organs WNV infection has resulted from transplacental fetal
appears to be primarily antibody-mediated, but CD8⫹ infection,55 infected breast milk,56 infected organ
cytotoxic T cells directed against peptide determinants transplants,29 and laboratory exposure.57 A recent re-
derived from WNV proteins also participate.45,47 In view of WNV infection in pregnant woman reported a
murine models of WNV infection, CD8⫹ T cells traf- 5% incidence of spontaneous abortions and 6% inci-
fic into the brain after viral infection. Mice that lack dence of premature births58; 97% (70/72) of live births
CD8⫹ T cells or major histocompatibility complex were healthy. One infant whose mother developed
class Ia antigens have up to 1,000-fold higher titers of WNV 6 days before term developed meningitis at 10
WNV in the CNS and increased mortality after foot- days of age, and another infant whose mother was in-
pad viral challenge compared with their immunocom- fected 3 weeks prepartum developed fatal WNV en-
petent counterparts.45,47 WNV-infected neurons se- cephalitis.
crete the chemokine CXCL10, which facilitates
recruitment of T cells bearing CXCR3 receptors into Clinical Features
the CNS.48 WNV-infected mice that lack CXCR3 The basic clinical features of West Nile neuroinvas-
show a phenotype similar to CD8-deficient mice, with ive disease (WNND) have been characterized exten-
enhanced viral CNS titer and mortality.48 A similar sively.8,9,28,33,59 – 69 CDC reports separate symptom-
phenotype occurs in CCR5-deficient mice, which also atic infection into West Nile fever (WNF) and
show defective T-cell recruitment into the CNS.35 WNND.13 Approximately 80% of WNV-infected pa-
About 1% of the human Caucasian population lacks tients are asymptomatic, 20% develop WNF, and less
CCR5 due to the presence of a homozygous defective than 1% develop WNND.
CCR5 allele (CCR5⌬32). CCR5 is an human immu- Common clinical features of WNF include the
nodeficiency virus coreceptor, and humans homozy- abrupt onset of fever, headache, and fatigue, with vari-
gous for CCR5⌬32 have decreased risk for human im- able malaise, anorexia, nausea, myalgia, lymphadenop-
munodeficiency virus infection after exposure, but athy, and a nonpruritic generalized maculopapular
show an enhanced risk for development of fatal WNV rash.59,60,70 When present, the rash typically begins
infection.49 The odds ratio (OR) for homozygosity of about 5 days after illness onset and lasts for a week. In
CCR5⌬32 was 4.4 to 9.1 (4.2– 8.3% prevalence) in one retrospective case series,60 the incidence and mean

Davis et al: WNV Neuroinvasive Disease 289


duration of commonly reported symptoms included fa- (OR, 2.7; range, 1.2– 6.5), alcohol abuse (OR, 3.4;
tigue (96%, 36 days), fever (81%, 5 days), headache range, 0.9 –12.9), diabetes (OR, 3.1; range, 1.1–9.2),
71% (10 days), muscle weakness (61%, 28 days), and and immunosuppression (OR, 2.2; range, 0.7–7.6)
cognitive impairment (53%, 14 days). were all more commonly associated with development
of encephalitis compared with WNF (risk ratio and
West Nile Neuroinvasive Disease 95% confidence interval from univariate analysis).71
WNND includes meningitis, encephalitis, and acute WNND is rarely reported in children or individuals
flaccid paralysis (AFP)/poliomyelitis. The distinctions younger than 30 years.72 However, from 2002 to
are at times arbitrary, and mixed patterns of disease are 2004, more than 1,000 cases of WNV infection were
common. Although the CDC does not subcategorize reported nationwide in children younger than 19 years,
WNND, approximately 40% of patients with WNND and 30% of the cases (n ⫽ 317) were categorized as
have meningitis and about 60% encephalitis,9,62,63,71 WNND. The impact of childhood WNV infection on
with a subgroup in both categories having AFP/polio- growth, development, and cognition remains un-
myelitis (see later). Patients with fever, stiff neck, head- known.
ache, photophobia, and cerebrospinal fluid (CSF) pleo-
cytosis, but without altered mental status or focal Acute West Nile Encephalitis
weakness, are classified as having meningitis; patients WNV encephalitis is more common in the elderly.9,71
showing signs, symptoms, or laboratory evidence of In the original New York City outbreak in 1999, 88%
brain parenchymal involvement are classified as having of those affected were older than 50 years and 51%
encephalitis.9,33,61 were older than 70.9 In a small series of WNND from
WNND occurs in approximately 1% of infected pa- Louisiana, the median age was 70 years for patients
tients, although this frequency is significantly higher in with encephalitis, 56 for those with AFP, and 35 for
the elderly and immunocompromised individuals.9,71 those with meningitis.61
In one series of 221 hospitalized WNV patients, 32% The most commonly reported signs and symptoms in
of those with encephalitis and 16% with meningitis patients with WNV encephalitis8,61– 63,65– 67,71 include:
were immunosuppressed, organ transplant recipients, fever (85–100%), diffuse weakness/fatigue (42– 85%),
or had autoimmune disease.71 Age 50 years or older headache (47–90%), and confusion/altered mental status

Table 1. Clinical Features Seen in West Nile Neuroinvasive Disease

Signs and Symptoms

Common (⬎50%) Fever (⬎38°C) Generalized nonfocal weakness


Anorexia Trouble walking
Nausea/vomiting Numbness in limb or body
Fatigue Tremors
Headache Myalgias
Trouble concentrating Blurry vision
Memory problems Myoclonic jerks
Confusion, delirium, lethargy Marked sleepiness
Stiff neck or neck pains
Less common (5–50%) Chills Focal sensory loss
Dizziness Sensitivity to light
Imbalance Nonpruritic maculopapular rash
Back pains Dysphagia
Somnolence Nystagmus
Slurred speech Babinski sign
Diarrhea Stupor
Arthralgias Uncoordinated gait/ataxia
Focal arm or leg weakness Joint pains
Paresthesias in limbs
Uncommon (1–4%) Seizure/status epilepticus Paraplegia or quadriplegia
Lymphadenopathy Urinary/fecal incontinence
Pharyngitis Parkinsonism
Conjunctivitis Monocular visual loss
Coma Diplopia
Facial palsy Marked chorioretinitis
Data are from References 8, 9, 61, 63– 66, and 103.

290 Annals of Neurology Vol 60 No 3 September 2006


(46 –74%) (Table 1). Systemic signs and symptoms are acute infection have provided formal assessment of
common and can include gastrointestinal complaints mental status, and only one study reported Glasgow
(nausea, vomiting: 31–74%; diarrhea and abdominal Coma Scores.61
pain: 7–21%),8,9,61,62,66,67 rash (19 –57%),8,9,63,65,67,71 Among patients admitted to an acute rehabilitation
and arthralgia and myalgia (15– 61%).9,62,66,71 Less facility after WNV encephalitis who underwent neuro-
commonly reported are pharyngitis (5%),9 lymphade- psychological testing,74 persisting cognitive deficits still
nopathy (2– 4%),9,66 and conjunctivitis (3%).9 The present at discharge included language/social commu-
frequency of tremor has varied remarkably from as low nication deficits, memory impairment, executive dys-
as 12%9 to as high as 80 to 100%.61 Tremor may be function, and defects in attention and concentration
present at rest or kinetic and typically is asymmetric (all present in ⬎65%). Approximately 25% of patients
and predominates in the upper extremities. Myoclonus had slowed processing speed. The pattern of cognitive
was reported to occur in one-third of cases in one se- impairment resembled that seen in frontal subcortical
ries,61 and when present, it is a clue to the diagnosis of dementias, and deficits were significantly greater on the
WNND because it is distinctly unusual in nonflavivi- frontal assessment battery than the MMSE. Neuropsy-
rus encephalitis. Parkinsonian features (rigidity, brady- chiatric symptoms were found in greater than 75% and
kinesia, postural instability) have been reported in up included emotional dysregulation (depression, anxiety,
to two-thirds of WNND cases.61 Meningeal signs oc- irritability) and psychomotor abnormalities (apathy, ag-
cur in a variable percentage (19 –57%). Seizures are itation) resembling those seen in subcortical dementias
unusual (1–16%). The frequency of cerebellar abnor- or human immunodeficiency virus encephalopathy.
malities (eg, incoordination, gait ataxia) varies greatly In addition to cognitive impairment, patients with
in different series from 11%62 to 57%.65 encephalitis have a wide range of additional clinical
Cranial nerve palsies are common in WNND, al- signs and symptoms that reflect the propensity of
though their exact frequency is difficult to ascertain WNV to involve areas as diverse as the cerebral cortex,
from published case series. Reports of cranial nerve in- basal ganglia, brainstem, and spinal cord (see Table 1).
volvement in 11%62 of cases are likely an underesti- Visual problems in patients with WNND have become
mate. The seventh cranial nerve is most frequently in- increasingly recognized. Patients often describe blurry
volved, producing unilateral or bilateral facial vision, trouble seeing, and photophobia. Clinically re-
weakness, which when bilateral may be synchronous or duced vision, active nongranulomatous uveitis, vitritis
sequential. Involvement of the eighth cranial nerve or multifocal chorioretinitis, and optic neuritis have all
inner ear may account for frequent reports of dizziness, been described.75,76 In one study of 29 consecutive pa-
vertigo, or the presence of nystagmus.61,63 Signs or tients with symptomatic WNV infection who under-
symptoms of brainstem or cranial nerve involvement, went complete funduscopic examinations and fluores-
or both, can include dysarthria9,65 and dysphagia.6 Un- cein angiography, 69% had abnormalities, including
usual syndromes associated with WNV infection have intraretinal hemorrhages, focal retinal vascular sheath-
included opsoclonus-myoclonus65 and stiff-person syn- ing, retinal vascular leakage, and optic disc swelling.76
drome.73
Abnormalities on the neurological examination par- Acute Flaccid Paralysis/Poliomyelitis
allel the symptoms described earlier. Patients frequently WNV can infect neurons in the spinal cord, producing
have an altered mental status, cranial nerve abnormal- true poliomyelitis77– 80 (Fig 1). Patients experience de-
ities as noted, and generalized weakness. Asymmetric velopment of an AFP that may appear abruptly and
lower motor neuron weakness with preserved sensation progress rapidly to result in asymmetric weakness that
may occur in patients with AFP/poliomyelitis (see involves one or more limbs, especially the legs.61,67,78 – 80
later). Movement disorders can be either hyperkinetic The weakness may develop concomitant with signs of
(eg, tremor, myoclonus) or hypokinetic (Parkinsonian meningitis or encephalitis or develop several days after
bradykinesia and rigidity). The tendon reflexes may be their onset. Patients may report back or muscle pains
reduced or hyperactive. Decreased or absent deep- at or just before the onset of weakness. Areflexia or
tendon reflexes have been reported in 3 to 94% of hyporeflexia is common, and loss of bladder and bowel
cases.9,63 Conversely, hyperreflexia or extensor plantar function may develop. Some patients will develop re-
responses have been reported in 6 to 38% of cases.9,61 spiratory distress requiring intubation and mechanical
Incoordination suggestive of cerebellar involvement oc- ventilation.78,79 Patients may experience subjective par-
curs variably. esthesias in limbs, but objective sensory loss is rare.
Despite the frequency of cognitive deficits in pa- WNV AFP results from viral infection and injury to
tients with WNND, mental status abnormalities have motor neurons in the anterior horns of the spinal cord.
not been comprehensively characterized. Some studies Electrophysiological studies are consistent with injury
emphasize the presence of either confusion8,63,64,66 or to anterior horn cells or anterior roots with absent or
change in level of consciousness.61,63,66 No studies of minimal sensory abnormalities. After a few weeks,

Davis et al: WNV Neuroinvasive Disease 291


Mild depression of hemoglobin (Male ⬍ 13.5gm/dl;
Female ⬍ 12gm/dl) was reported in 41% of cases in
an Israeli outbreak.66 Hyponatremia (⬍135mM/L) is
seen in 33 to 50% of patients.62,64,66 Abnormal liver
function tests (aspartate aminotransferase, alanine ami-
notransferase, alkaline phosphatase, bilirubin) occur in
up to 21 to 24% of patients.62,66 Elevated creatine ki-
nase levels also can occur.67,82 In one small series, 65%
of patients were reported to have transiently elevated
serum lipase levels, with 24% having levels more than
two times normal. None of the patients had signs or
symptoms of pancreatitis.83

CEREBROSPINAL FLUID. CSF findings in a large series


(250 cases) of serologically confirmed WNND recently
have been described.84 Ninety-seven percent of patients
Fig 1. Anterior horn of the spinal cord (Luxol fast blue–peri- with meningitis had a CSF pleocytosis (⬎5 cells/mm3)
odic acid–Schiff stain) showing virtually complete destruction with a mean cell count of 226/mm3. Results were
of the anterior horn in a patient with West Nile virus acute
nearly identical in patients with encephalitis, with 95%
flaccid paralysis. Original magnification ⫻40.
having a pleocytosis with a mean cell count of 227/
mm3. Forty-eight percent of meningitis and 41% of
some patients note fasciculations in involved muscles encephalitis cases had a neutrophil predominance on
with later atrophy of involved muscles. Patients who their initial CSF examination. Among all patients, the
experience development of AFP are often considerably mean percentage of neutrophils was 45% for meningi-
younger than those who develop encephalitis.61,65,80 tis and 41% for encephalitis cases. A glucose level less
Overall, the incidence of flaccid limb weakness with than 40mg/dl was seen in only 1 of 238 cases. Ninety
atrophy has been difficult to estimate, but is likely to percent of meningitis and 99% of encephalitis patients
be at least 5 to 10% of patients developing neuroinva- had elevated protein levels (⬎40mg/dl), with a mean
sive disease80 with an incidence of about 4 cases/ value of 76mg/dl in meningitis and 101mg/dl in en-
100,000 population during a WNV epidemic.80 cephalitis cases. In smaller series, the mean cell count has
West Nile–associated AFP must be distinguished ranged between 24 and 566 cells/mm3 and mean pro-
from reversible muscle weakness. The muscle weakness tein concentration between 57 and 126mg/dl.8,9,61– 63,67
may be initially severe and involves one or more limbs, Cells resembling plasma85 and Mollaret cells67,86 have
but it is typically transient and completely reversible been found in CSF (Fig 2).
and not associated with fasciculations, muscle atrophy,
or electromyographic (EMG) changes suggesting de- Virological and Serological Tests for Diagnosis
nervation.79 The cause of this transient weakness re- WNV infection can be diagnosed by virus isolation, by
mains unknown but may reflect mild myelitis with spi- detection of viral nucleic acid or antigens, or by serol-
nal cord edema and transient anterior horn cell ogy (Table 2, Fig 3). Virus isolation is rarely performed
dysfunction. WNV is also associated with a Guillain– due to biosafety issues. Detection of viral nucleic acid
Barré–like involvement of peripheral nerves, although has been extremely useful in screening donated blood
true cases of Guillain–Barré–like syndrome appear to products for potential WNV infection (see earlier). Se-
be rare in comparison with cases of poliomyelitis.79,80 rum nucleic acid amplification tests are of only limited
utility in diagnosis of WNND, because viremia is typ-
Laboratory Findings ically over at the onset of symptomatic CNS disease.
CHEMISTRY AND HEMATOLOGY. Elevated white blood Less than 15% of blood samples from patients with
cell counts (WBCs; ⬎10,800/mm3) were reported in WNND are polymerase chain reaction–positive.87 In
about 40% of cases in a recent Israeli outbreak, and patients with WNND, CSF polymerase chain reaction
several US series have reported mild elevations in mean is 100% specific but relatively insensitive (57–70%)
WBC (10,400 –10,800/mm3).8,62 In one small series, compared with detection of WNV IgM in CSF.87
patients with meningitis had a higher mean WBC Diagnosis of WNND typically is based on detection
(13,900/mm3) compared with those with AFP of WNV-specific antibodies in serum, CSF, or both
(11,800/mm3) and encephalitis (7,600/mm3).61 WBCs using commercially available enzyme-linked immu-
greater than 20,000/mm3 are not usually seen.61,62 nosorbent assays88 (see Table 2, Fig 3). WNV enzyme-
Prolonged lymphocytopenia has been described,81 and linked immunosorbent assays can detect heterologous
thrombocytopenia (⬍135,000/mm3) occurs in 15%.66 cross-reacting antibodies resulting from infection with

292 Annals of Neurology Vol 60 No 3 September 2006


positive and IgM-positive, 41% were IgG-positive but
IgM-negative, and only about 1% were IgM-positive
but IgG-negative.90
Serum WNV IgM may persist for more than 2
months in more than 50% of patients, although titers
decline over time.90 In one small study, 7 of 12 pa-
tients with WNV encephalitis still had IgM antibody
detectable at more than 500 days after onset of ill-
ness.91 Because of persistence of IgM antibody even in
asymptomatic individuals, it may be necessary to dem-
onstrate an increase in IgG antibodies between acute
and convalescent sera or obtain sequential paired IgG
and IgM serum and/or CSF serology to unequivocally
establish acute infection.

Electrodiagnosis
Seizures, including rare cases of status epilepticus, are
reported in less than 10% of cases of WNND.8,9,63,67
Fig 2. Cerebrospinal fluid cytospin preparation (Wright–Gi-
Despite the rarity of clinical seizures, electroencephalo-
emsa stain) from a patient with West Nile virus meningoen-
cephalitis showing the wide variety of cells that can be seen,
graphic abnormalities have been reported in 57 to 86%
including neutrophils, lymphocytes, small mononuclear cells of cases.9,61– 63,65,92 In one selected series of 13 hospi-
with a plasmacytoid appearance, and larger mononuclear cells. talized patients with WNND, the electroencephalo-
gram was abnormal in 85% (11/13), with the most
common abnormalities being generalized slowing with
other flaviviruses, including St Louis encephalitis, Jap- anterior (n ⫽ 8) or temporal (n ⫽ 2) prominence and
anese encephalitis, dengue 2, yellow fever, and Powas- intermittent temporal slowing (n ⫽ 1).92 In another
san viruses.88 In addition, individuals recently vacci- small series, electroencephalographic abnormalities
nated with yellow fever or Japanese encephalitis were found in 57% of cases (4/7), with the most com-
vaccines may develop WNV cross-reacting antibodies. mon patterns being generalized slowing with anterior
It may be necessary to confirm positive enzyme-linked or temporal predominance.8 In a third small series of
immunosorbent assay tests by plaque reduction neu- seven patients with encephalitis, diffuse, irregular slow
tralization waves were noted in 86%, and one patient each had
assays (performed by many state public health labora- electrographic seizures and focal sharp waves.61
tories and the CDC) for definitive diagnosis to exclude EMG and nerve conduction velocity studies are ab-
cross-reactions.
The kinetics of CSF WNV IgM and IgG responses
have not been completely characterized. Immunosup-
pressed patients may exhibit delayed serum or CSF se-
roconversion, or both.30 False-positive serum WNV
IgM antibody tests can occur in patients with rheuma-
toid factor or other inflammatory processes.88 WNV
IgM may occasionally appear in CSF before serum in
patients with WNND.89,90 The cumulative percentage
of patients who are seropositive increases by approxi-
mately 10% per day during the first week of illness,90
and specimens obtained early after onset may need to
be repeated to avoid misdiagnosis. In one study,90 the
relative frequency of IgG and IgM antibodies was ex-
amined as a function of time after illness onset. Fifty-
eight percent of seropositive CSF specimens and 51%
of seropositive serum specimens obtained at days 2 to 7
of illness had WNV IgM but not IgG (the remainder Fig 3. Schematic of typical time course for development of
were IgG⫹ and IgM⫹). During days 8 to 20 of illness, West Nile virus (WNV) IgG and IgM antibodies in humans
98% of seropositive serum specimens have both WNV after WNV infection. (Courtesy CDC’s Arbovirus Disease
IgG and IgM. Fifty-one percent of seropositive speci- Branch, Diagnostic and Reference Laboratory, Fort Collins,
mens obtained at days 21 to 98 of illness were IgG- CO).

Davis et al: WNV Neuroinvasive Disease 293


Table 2. Criteria for Clinical and Definite Diagnoses of West Nile Neuroinvasive Disease

Possible Clinical Diagnosis of West Nile Neuroinvasive Disease (before serology results available)
Requirements: 1, 2, 3 (A, B, or C), 5 with 4 supportive
New onset of:
1. Fever ⬎38°C or subjective fever by patient when temperature never taken
2. Acute systemic symptoms (⬎1 lasting ⬎ 48 hours)
Headache
Nausea or vomiting
Myalgias
Transient erythematosus maculopapular rash
3. Acute neurological signs and symptoms (A, B, or C required)
A. Altered mental status for ⬎48 hours (⬎1 below are required)
Marked lethargy that often incapacitates the individual
Delirium or agitation
Marked disorientation or confusion that often incapacitates the individual
Profound sleepiness (sleeping all day for ⬎2 days)
Stupor or coma
B. Brainstem or spinal cord signs that persist ⬎48 hours (⬎1 below are required)
Cranial nerve palsies
Myelitis
i. New focal weakness in one or more limbs that is not due to arthritis or limb pains lasting ⬎ 48 hours
ii. Respiratory failure requiring intubation that is not solely due to pneumonia
iii. Electromyographic evidence of recent denervation in more than one nerve root
C. Cerebrospinal fluid (CSF) (all required if examined)
Pleocytosis (rarely may be absent if lumbar puncture performed within first 2 days of central nervous system symptoms)
Cerebrospinal fluid IgM antibody for West Nile virus
Negative cultures for bacteria and negative Gram stain (if mycobacterial, fungal, or viral cultures were done, they
should be negative)
4. Supportive criteria
A. Focal neurological signs of acute onset (ⱖ1 desired but not required)
Hemiparesis
Visual field loss or chorioretinitis
Hyperactive deep-tendon reflexes or Babinski sign
Seizure
Tremor or myoclonus
Bradykinesia, spasticity, or rigidity
Photophobia
Severe generalized weakness and fatigue that keeps patient in bed
B. Mosquitoes as vector (1 desired unless patient received transfusion or transplant organ)
Positive West Nile virus (WNV) mosquito pools or infected horses in region within past 3 weeks or
Recent cases of acute WNV infection in region or
Travel within past 3 weeks to regions with positive WNV mosquito pools or cases of WNV infection
5. Exclusion criteria (required)
Underlying dementia, congestive heart failure, or chronic obstructive pulmonary disease that could cause altered mental
status and no known other neurological disease that could cause the neurological signs
Definite or Confirmed Diagnosis of West Nile Neuroinvasive Disease
Above criteria plus WNV serology/virology (A and B plus/or C or D)
A. Demonstration of WNV IgM antibody in serum without vaccination with yellow fever or Japanese B viral vaccines in
past 5 years or recent infection with other flavivirus, such as St Louis encephalitis virus. Note: If WNV has occurred
in region in prior years, criterion B is needed because previously infected individuals may have prolonged persistence of
IgM in serum.
B. Fourfold or greater increase in serum WNV IgG or IgM antibody titer between acute and convalescent samples taken
10 to 28 days apart.
C. Demonstration of WNV IgM antibody in CSF
D. Identification of WNV in CSF by viral culture or of WNV nucleic acid by polymerase chain reaction

normal in patients with myelitis. During the first 2 no motor nerve conduction block, although motor
weeks after onset, patients may have normal studies; conduction velocities may be slowed in patients with
subsequently, signs of denervation including fibrilla- significant reduction in compound muscle action po-
tions and positive sharp waves are found that predom- tential amplitudes. Sensory nerve action potentials are
inate in the weakest muscles.79,93–95 Compound mus- normal. EMG results show reduced recruitment in in-
cle action potential amplitudes are decreased and volved muscles, as well as signs of denervation (fibril-
parallel the location and severity of weakness. There is lation potentials, positive sharp waves). Studies months

294 Annals of Neurology Vol 60 No 3 September 2006


to years after illness show evidence of collateral sprout-
ing with reduced numbers of large-amplitude, long-
duration polyphasic motor unit action potentials.94

Neuroimaging
Neuroimaging studies are frequently normal in patients
with WNND. Cranial computed tomography abnor-
malities are not seen in patients with WNND.9,61,63,67
Surprisingly, magnetic resonance imaging (MRI) is also
relatively insensitive, and abnormalities are reported in
only 20 to 70% of patients with acute WNND, the
variability depending, in part, on the timing and na-
ture of the imaging sequences used.61,63,67,96,97 In one
recent series, 29% (5/17) of patients had completely
normal studies and an additional 24% (4/17) had ab-
normalities seen only on diffusion-weighted imaging.97
Although not studied systematically, the frequency of
lesions identified by MRI appears to increase over the
first week of illness, and late studies may show abnor-
malities not identified acutely. Lesions, when present,
typically are hyperintense on T2-weighted and fluid-
attenuated inversion recovery images and of normal in-
tensity and nonenhancing on T1-weighted images67,96
(Fig 4). Restricted water diffusion may be seen on
diffusion-weighted images in up to 50% of patients97
and can assist in confirmation of subtle abnormalities
seen on T2- or fluid-attenuated inversion recovery se-
quences.67,96,97 When present, lesions have a predilec-
tion for deep gray matter structures including the basal
ganglia and thalami, as well as the brainstem and cer-
ebellum.61,96,97 Some patients with parkinsonism have
Fig 4. Magnetic resonance images at day 6 of illness from a
had striking abnormalities in the substantia nigra. patient with West Nile virus (WNV) encephalitis showing
Periventricular hyperintensity also has been reported in increased signal (representing restricted diffusion confirmed by
6 to 14% of cases.62,63 In one retrospective review of apparent diffusion coefficient maps) in the thalamus (A) and
17 WNV cases, 5 of 8 cases with MRI abnormalities substantia nigra (B).
had involvement of the mesial temporal lobes(s), and
in 3 cases, it was an isolated abnormality.96 Cerebral
leptomeningeal enhancement on MRI has been re- 60% (6/10). The severity of inflammation was also
ported in 6 to 31% of cases.9,62,63,96,97 typically greater in brainstem compared with cortex or
MRI abnormalities involving the anterior horns and cerebellum.33 Inflammatory changes were also found in
anterior roots have been reported in some patients with all (17/17) spinal cords examined.33 Within the spinal
WNV-associated AFP, although neuroimaging studies cord, inflammatory changes were more prominent in
are commonly normal.65,67,78,96 –98 In some patients, the anterior horn in 53% and equal in anterior and
there is gadolinium enhancement of the conus med- posterior horns in 47%.33 Twenty-two percent of pa-
ullaris, cauda equine, or both.61,67,78,96,97 tients had lymphocytic infiltrates involving cranial or
spinal nerves.33 CD8⫹ T cells predominated in the pa-
Neuropathology renchymal and glial nodules and CD20⫹ B cells in
The gross appearance of the brain is normal in patients perivascular cuffs.64,101
with WNND. Microscopically, almost all patients with WNV antigen was detected by immunohistochemis-
encephalitis are found to have glial nodules, perivascu- try in only 60% of brainstem, 12.5% of cortex, and
lar cuffing with mononuclear cells, and a variable de- 41% of spinal cords examined,33 and was often focal
gree of neuronal loss.33 Neuronophagia and, rarely, fo- and sparse when found. The intensity of antigen stain-
cal areas of brain necrosis occasionally can be ing was maximal in patients dying during the first
seen.33,99,100 In a review of 23 autopsy cases, inflam- week of encephalitis, but positive staining was seen as
mation was noted in the brainstem in 100% (20/20), late as 3 weeks after onset in two patients who were
in the cortex in 69% (11/16), and in the cerebellum in immunosuppressed.33 In contrast with neurons, glial

Davis et al: WNV Neuroinvasive Disease 295


cells only rarely contain detectable WNV antigen, and turned home with assistance, and 80% returned home
endothelial and meningeal cells are antigen-negative. with no assistance.71 Risk factors for fatal outcome in
patients with encephalitis included intubation (OR,
Prognosis and Outcome 12.7; range, 1.2–139), previous stroke (OR, 42.7;
Patients with WNND often experience prolonged re- range, 2.4 –756), immunosuppression (OR, 26.5;
covery times from both constitutional and neurological range, 3–234), and age 50 years or older (OR of 1.14
symptoms. In one study, patients were interviewed 5 to for each year over age 50; range, 1.02–1.29).71
6 months after symptoms, and they reported it took 60 One recent series reviewed outcome in 11 patients
days (median) to return to normal.102 Fifty-three per- with AFP.104 The degree of motor recovery at 18
cent of those surveyed reported difficulty concentrating months after illness was quite variable. Improvement
for 2 weeks post-infection (median). Telephone inter- correlated with motor unit number estimation
views of patients infected during the New York City (MUNE), as determined on initial EMG study. Pa-
outbreak in 1999 indicated that only 37% had tients with more than 50% of normal MUNE made
achieved a full recovery at 1 year after illness.103 better recoveries than those with less than 50%. Inter-
Younger patients tended to make better recoveries than estingly, MUNE did not always correlate with degree
older ones.103 Physical sequelae found to be signifi- of acute clinical weakness, and patients with severe
cantly more prevalent at 1 year after infection com- weakness but preserved MUNE had a generally good
pared with before infection baseline included: fatigue prognosis for recovery.104
(64%), muscle weakness (56%), insomnia (47%), dif- The severity of MRI abnormalities may also predict
ficulty walking (42%), muscle pain (39%), and head- outcome. Patients with normal studies or lesions seen
ache (36%). Cognitive sequelae at 1 year included: only on diffusion-weighted imaging sequences appear
memory loss (44%), depression (44%), irritability to have a higher likelihood of full recovery than those
(39%), lightheadedness (37%), loss of concentration with abnormalities seen on T1- and T2-weighted and
(33%), and confusion (31%). These physical and cog- fluid-attenuated inversion recovery sequences.97
nitive signs and symptoms impacted on activities of
daily living. About 30 to 56% of patients reported im- Treatment
pairments in ability to shop, prepare meals, do laundry Currently, there is no treatment of proven efficacy for
and housekeeping, and use transportation.103 WNV infection. In murine models of WNV, IFN
In the study of 15 surviving patients with treatment reduced mortality when administered before
WNND,61 at 8 months after infection, 73% (11/15) viral challenge, but not when given 2 days after infec-
were home and functioning independently, 20% tion.105 Human studies with IFN treatment of WNV
(3/15) were home but dependent on assistance, and infection are limited. Two patients with chronic hepa-
7% (1/15) remained in rehabilitation. Surprisingly, the titis C infection developed WNF despite receiving
severity of initial illness did not correlate well with out- IFN-␣2b and ribavirin for treatment of their chronic
come, and five of seven cases with severe encephalitis hepatitis C.106 Noncontrolled and nonblinded anec-
(Glasgow Coma Score ⬍ 12) returned to their premor- dotal reports of treatment65,66,107 include examples of
bid functional level. In general, recovery occurred both improvement and no benefit in patients receiving
within 4 months of illness. Persistent symptoms were IFN-␣2b. A randomized, double-blind, clinical trial of
common among the WNND patients and included fa- IFN-␣-n3 (alferon; 3 million units administered intra-
tigue (67%), cognitive difficulties (27%), myalgia venously once, then subcutaneously every day for 7
(13%), and headache (13%). Neurological examination days) for treatment of WNV meningoencephalitis has
was normal at 8 months in all five patients with men- been initiated,108 although accrual has been limited.
ingitis. The three patients with AFP showed no im- Ribavirin, a guanosine analogue, inhibits WNV rep-
provement in limb weakness and remained wheelchair lication and cytopathic effect in tissue culture.51,109
dependent for ambulation. Parkinsonism was still However, WNV infection has developed in patients
present at 8 months in 45% (5/11) and tremor in 33% being treated with ribavirin for hepatitis C.106 In a
(5/15) of patients in whom it was initially noted. Two noncontrolled, nonrandomized trial during an out-
of five patients had persisting myoclonus. break of WNV infection in Israel in 2000,66 mortality
In a large series of 221 hospitalized patients from was 41% (15/37) in patients receiving ribavirin com-
Colorado in 2003, 18% of encephalitis patients died, pared with 10% (18/178) among those not receiving
46% were discharged to rehabilitation or long-term ribavirin (OR of 6.7 for death; p ⬍ 0.0001), although
care facilities, 15% returned home with assistance, and this negative result may have been biased by selection
20% returned home with no assistance.71 Results were of more seriously ill patients for treatment.
significantly better in the meningitis group, in which Both polyclonal antisera and monoclonal antibodies
there were no deaths, 9% of patients were discharged directed against the WNV envelope glycoprotein can
to rehabilitation or long-term care facilities, 11% re- protect mice against WNV encephalitis.32,39,46,110 –112

296 Annals of Neurology Vol 60 No 3 September 2006


Protection also occurs in mice after passive transfer of humans include inactivated subunit vaccines; attenu-
human intravenous immunoglobulin containing high ated WNV vaccines; chimeric vaccines based on vac-
titers of anti-WNV antibody.111 In murine models of cine strains of YFV, dengue, measles, or canarypoxvi-
WNV infection, protection depends on the dose, fre- rus; and naked DNA vaccines.122–126 Chimeric
quency of administration, and timing of antibody vaccines typically contain the WNV genes encoding
treatment. Protection is maximal when antibody is the E and PrM/M proteins inserted into a vaccine virus
given before virus has reached the CNS, although some genome. A chimeric WNV/dengue vaccine was shown
protection still occurs even after the virus has entered to protect rhesus monkeys against WNV challenge.125
the CNS.112 An intravenous immunoglobulin from Is- A randomized, double-blind, placebo-controlled phase
rael (Omr-IgG-am) contains high titers of anti-WNV II trial in 200 subjects of a chimeric vaccine
IgG and has been reported to be of benefit in treat- (ChimeriVax-West Nile) constructed from the YFV
ment of WNV encephalitis in some,112–114 but not 17D vaccine strain and containing WNV prM/E genes
all,115 noncontrolled, nonblinded case reports. These was recently initiated after phase I safety and immuno-
studies prompted the National Institutes of Health/ genicity studies found the vaccine to be safe and to
Collaborative Antiviral Study Group (CASG) to insti- generate detectable WNV antibody titers in greater
tute a phase I/II multicenter, randomized, double- than 96% of immunized subjects by 28 days after im-
blind, placebo-controlled trial (CASG 210) of the munization.127 A phase I trial of a chimeric dengue
safety and efficacy of Omr-IgG-am in treatment of 4-WNV vaccine also has been initiated.128
WNV encephalitis and myelitis. Enrollment in the trial
began in September 2003 and will be finished in 2006.
The role, if any, for steroid treatment in patients Chronic/Persistent and Postinfectious Immune-
with WNV infection is unknown. There is one case Mediated West Nile Virus Central Nervous
report of a patient with AFP who improved after treat- System Disease
ment with intravenous methylprednisolone (500mg qd WNV can persist in the brains of infected monkeys
for 4 days).116 who recovered from encephalitis for 5 to 6 months af-
Antisense oligomers that bind to WNV RNA can ter peripheral inoculation, even in the absence of clin-
inhibit the translation of viral proteins and viral RNA ically overt illness.129 Brain tissue and CSF from fatal
replication. Small, interfering RNA directed against WNV cases has contained virus even 3 weeks after
WNV envelope protein sequences afforded partial pro- acute infection,33 and in one immunocompromised pa-
tection to mice when injected intravenously 24 hours tient, virus RNA and antigens were detectable at au-
before viral challenge, but not when injected 24 hours topsy 99 days after illness onset.130 To date, there are
after viral challenge.117 A proprietary WNV antisense no proven cases of chronic or relapsing WNV CNS
oligomer (AVI-4020; AVI BioPharma, Portland, OR; disease in immunocompetent individuals, nor are there
see the AVI BioPharma Web site for more informa- definitive cases of WNV-associated postinfectious
tion: http://www.avibio.com)118 has undergone phase I immune-mediated encephalomyelitis.
safety evaluation in a group of nine WNV-infected pa-
tients, with no reports of significant toxicity and effi-
cacy (phase II) trials planned. References
1. Brinton M. The molecular biology of West Nile virus: a new
invader of the western hemisphere. Annu Rev Microbiol 2002;
Prevention of West Nile Virus Infection 56:371– 402.
Prevention of WNV infection involves reduction in 2. Kuno G, Chang GJ, Tsuchiya KR, et al. Phylogeny of the
risk for exposure to infected mosquitoes both through genus flavivirus. J Virol 1998;72:73– 83.
personal protection and community-wide mosquito 3. Mukhopadhyay S, Kim B, Chipman P, et al. Structure of
West Nile virus. Science 2003;302:248.
eradication measures. Personal protection includes re- 4. Smithburn K, Hughes T, Burke A, Paul J. A neurotropic virus
ducing outdoor exposure time during peak mosquito isolated from the blood of a native of Uganda. Am J Trop
biting periods at dusk and dawn, using mosquito re- Med Hyg 1940;20:471– 492.
pellent when outdoors for more than 30 minutes, and 5. Goldblum N, Sterk VV, Paderski B. The clinical features of
covering exposed skin. Personal protective measures the disease and the isolation of West Nile virus from the blood
of nine human cases. Am J Hyg 1954;59:89 –103.
can reduce the risk for WNV infection during an epi- 6. Southam CM, Moore AE. Induced virus infections in man by
demic by 50%.119 N,N-diethyl-m-toluamide (DEET) the Egypt isolates of West Nile virus. Am J Trop Med Hyg
and picaridin applied to exposed skin are effective and 1954;3:19 –50.
relatively nontoxic mosquito deterrents. Unfortunately, 7. Murgue B, Zeller H, Deubel V. The ecology and epidemiol-
utilization of personal protection is suboptimal despite ogy of West Nile virus in Africa, Europe and Asia. Curr Top
Microbial Immunol 2002;267:195–221.
intensive publicity during WNV outbreaks.119 8. Klein C, Kimiagar I, Pollak L, et al. Neurological features of
Two WNV vaccines have been approved for equine West Nile virus infection during the 2000 outbreak in a re-
use.120,121 Vaccines under development and testing in gional hospital in Israel. J Neurol Sci 2002;200:63– 66.

Davis et al: WNV Neuroinvasive Disease 297


9. Nash D, Mostashari F, Fine A, et al. The outbreak of West 30. Kleinschmidt-DeMasters BK, Marder BA, Levi ME, et al.
Nile virus infection in the New York City area in 1999. Naturally acquired West Nile virus encephalomyelitis in trans-
N Engl J Med 2001;344:1807–1814. plant recipients: clinical, laboratory, diagnostic, and neuro-
10. Lanciotti R, Roehrig J, Deubel V, et al. Origin of the West pathological features. Arch Neurol 2004;61:1120 –1220.
Nile virus responsible for an outbreak of encephalitis in the 31. Wang T, Town T, Alexopoulou L, et al. Toll-like receptor 3
Northeastern United States. Science 1999;286:2333–2337. mediates West Nile virus entry into the brain causing lethal
11. Rappole JH, Derrickson SR, Hubalek Z. Migratory birds and encephalitis. Nat Med 2004;10:1366 –1372.
spread of West Nile virus in the western hemisphere. Emerg 32. Engle MJ, Diamond MS. Antibody prophylaxis and therapy
Infect Dis 2000;6:319 –327. against West Nile virus infection in wild-type and immuno-
deficient mice. J Virol 2003;77:12941–12949.
12. Solomon T. Flavivirus encephalitis. N Engl J Med 2004;351:
33. Guarner J, Shieh W, Hunter ST, et al. Clinicopathologic
370 –378.
study and laboratory diagnosis of 23 cases with West Nile vi-
13. Centers for Disease Control and Prevention West Nile virus.
rus encephalomyelitis. Hum Pathol 2004;35:983–990.
Available at: http://www.cdc.gov/ncidod/dvbid/westnile/
34. Cheeran MC-J, Hu S, Sheng WS, et al. Differential responses
surv&control.htm. Accessed July 15, 2006. of human brain cells to West Nile virus infection. J Neurovi-
14. Hayes EB, Komar N, Nasci RS, et al. Epidemiology and rol 2005;11:512–524.
transmission dynamics of West Nile virus disease. Emerg In- 35. Glass WG, Lim JK, Cholera R, et al. Chemokine receptor
fect Dis 2005;11:1167–1173. CCR5 promotes leukocyte trafficking to the brain and survival
15. Schweitzer BK, Kramer WL, Sambol AR, et al. Geographic in West Nile virus infection. J Exp Med 2005;202:
factors contributing to a high seroprevalence of West Nile 1087–1098.
virus-specific antibodies in humans following an epidemic. 36. Chu JJ, Ng ML. Interaction of West Nile virus with alpha v
Clin Vaccine Immunol 2006;13:314 –318. beta 3 integrin mediatesviral entry into cells. J Biol Chem
16. van der Meulen KM, Pensaert MB, Nauwynck HJ. West Nile 2004;279:54533–54541.
virus in the vertebrate world. Arch Virol 2005;150:637– 657. 37. Lee JW, Chu JJ, Ng ML. Quantifying the specific binding
17. Girard YA, Popov V, Wen J, et al. Ultrastructural study of between West Nile virus envelope domain III protein and the
West Nile virus pathogenesis in Culex pipiens quinquefasciatus cellular receptor alpha v beta 3 integrin. J Biol Chem 2006;
(Diptera: Culicidae). J Med Entomol 2005;42:429 – 444. 281:1352–1360.
18. Van Landingham DL, Schneider BS, Klinger K, et al. Real- 38. Chu JJ, Rajamanonmani R, Li J, et al. Inhibition of West Nile
time reverse transcriptase-polymerase chain reaction quantifi- virus entry by using a recombinant domain III from the en-
cation of West Nile virus transmitted by Culex Pipiens Quin- velope glycoprotein. J Gen Virol 2005;86:405– 412.
quefasciatus. Am J Trop Med Hyg 2004;71:120 –123. 39. Nybakken GE, Oliphant T, Johnson S, et al. Structural basis
of West Nile virus neutralization by a therapeutic antibody.
19. Nacsi R, Savage H, White D, et al. West Nile virus in over-
Nature 2005;437:764 –769.
wintering Culex mosquitoes, New York City, 2000. Emerg In-
40. Chu J, Ng M. Infectious entry of West Nile virus occurs
fect Dis 2001;7:1–3.
through a clathrin-mediated endocytic pathway. J Virol 2004;
20. Miller B, Nasci R, Godsey M, et al. First field evidence for
78:10543–10555.
natural vertical transmission of West Nile virus in Culex 41. Shrestha B, Gottlieb D, Diamond M. Infection and injury of
Univittatus Complex mosquitoes from Rift Valley Province, neurons by West Nile encephalitis virus. J Virol 2003;77:
Kenya. Am J Trop Med Hyg 2000;62:240 –246. 13203–13213.
21. Komar N, Langevin S, Hinten S, et al. Experimental infection 42. Yang J-S, Ramanathan MP, Muthumani K, et al. Induction of
of North American birds with New York 1999 strain of West inflammation by West Nile virus capsid through the caspase-9
Nile virus. Emerg Infect Dis 2003;9:311–322. apoptotic pathway. Emerg Infect Dis 2002;12:1379 –1384.
22. Turell M, O’Guinn M, Oliver J. Potential for New York mos- 43. Parquet MC, Kumatori A, Hasebe F, et al. West Nile virus-
quitoes to transmit West Nile virus. Am J Trop Med Hyg induced bax-dependent apoptosis. FEBS Lett 2001;500:
2000;62:413– 414. 17–24.
23. Steele KE, Linn MJ, Schoepp RJ, et al. Pathology of fatal 44. Ramanathan MP, Chambers JA, Pankhong P, et al. Host cell
West Nile virus infections in native and exotic birds during killing by the West Nile virus NS2B-NS3 proteolytic complex:
the 1999 outbreak in New York City, New York. Vet Pathol NS3 alone is sufficient to recruit caspases-8-based apoptotic
2000;37:208 –224. pathway. Virology 2006;345:56 –72.
24. Guptill SC, Julian KG, Campbell GL, et al. Early-season avian 45. Wang Y, Lobigs M, Lee E, Mullbacher A. CD8 T cells me-
deaths from West Nile virus as warnings of human infection. diate recovery and immunopathology in West Nile virus en-
Emerg Infect Dis 2003;9:483– 484. cephalitis. J Virol 2003;77:13323–13334.
25. Schuler LA, Khaitsa ML, Dyer NW, Stoltenow CL. Evalua- 46. Diamond MS, Sitati EM, Friend LD, et al. A critical role for
induced IgM in the protection against West Nile virus infec-
tion of an outbreak of West Nile virus infection in horses: 569
tion. J Exp Med 2003;12:1853–1862.
cases (2002). J Am Vet Med Assoc 2004;225:1084 –1089.
47. Shrestha B, Diamond MS. Role of CD8⫹ T cells in control
26. Chambers TJ, Diamond MS. Pathogenesis of flavivirus en-
of West Nile virus infection. J Virol 2004;78:8312– 8321.
cephalitis. Adv Virus Res 2003;60:273–342.
48. Klein RS, Lin E, Zhang B, et al. Neuronal CXCL10 directs
27. Johnston L, Halliday G, King N. Langerhans cells migrate to CD8⫹ recruitment and control of West Nile virus encepha-
local lymph nodes following cutaneous infection with an ar- litis. J Virol 2005;79:11457–11466.
bovirus. J Invest Dermatol 2000;114:560 –568. 49. Glass WG, McDermott DH, Lim JK, et al. CCR5 deficiency
28. Hayes EB, Gubler DJ. West Nile virus: epidemiology and increases risk of symptomatic West Nile virus infection. J Exp
clinical features of an emerging epidemic in the United States. Med 2006;203:35– 40.
Annu Rev Med 2006;57:181–194. 50. Samuel MA, Diamond MS. Alpha/beta interferon protects
29. Iwamoto M, Jernigan D, Guasch A, et al. Transmission of against lethal West Nile virus infection by restricting cellular
West Nile virus from an organ donor to four transplant recip- tropism and enhancing neuronal survival. J Virol 2005;79:
ients. N Eng J Med 2003;348:2196 –2203. 13350 –13361.

298 Annals of Neurology Vol 60 No 3 September 2006


51. Anderson JF, Rahal JJ. Efficacy of interferon alpha-2b and 74. Arcienagas DB, Anderson CA. Viral encephalitis: neuropsychi-
ribavirin against West Nile virus in vitro. Emerg Infect Dis atric and neurobehavioral aspects. Curr Psychiatry Rep 2004;
2002;8:107–108. 6:372–379.
52. Pealer L, Marfin A, Petersen L, et al. Transmission of West 75. Anninger W, Lubow W. Visual loss with West Nile virus
Nile virus through blood transfusion in the United States in infection: a wider spectrum of a “new” disease. Clin Infect Dis
2002. N Eng J Med 2003;349:1236 –1246. 2004;38:e55–e56.
53. Kleinman S, Glynn SA, Busch M, et al. The 2003 West Nile 76. Khairallah M, Yahia SB, Ladjimi A, et al. Chorioretinal in-
virus United States epidemic: the America’s Blood Centers ex- volvement in patients with West Nile virus infection. Oph-
perience. Transfusion 2005;45:469 – 479. thalmology 2004;111:2065–2070.
54. Stramer SL, Fang CT, Foster GA, et al. West Nile virus 77. Doron SI, Dashe JF, Adelman LS, et al. Histopathologically
among blood donors in the United States, 2003 and 2004. proven poliomyelitis with quadriplegia and loss of brainstem
N Engl J Med 2005;353:451– 459. function due to West Nile virus. Clin Infect Dis 2003;37:
55. CDC. Intrauterine West Nile virus infection—New York, 374 –377.
2002. MMWR Morb Mortal Wkly Rep 2002;51:1135–1136. 78. Sejvar J, Leis A, Stokic D, et al. Acute flaccid paralysis and
56. CDC. Possible West Nile virus transmission to an infant West Nile virus infection. Emerg Infect Dis 2003;9:788 –793.
through breast-feeding—Michigan, 2002. MMWR Morb 79. Leis A, Stokic D, Webb R, et al. Clinical spectrum of muscle
Mortal Wkly Rep 2002;51:877– 878. weakness in human West Nile virus infection. Muscle Nerve
57. CDC. Laboratory-acquired West Nile virus infections— 2003;28:302–308.
United States, 2002. MMWR Morb Mortal Wkly Rep 2002; 80. Sejvar JJ, Bode AV, Marfin AA, et al. West Nile virus-
51:1133–1135. associated flaccid paralysis. Emerg Infect Dis 2005;11:
58. O’Leary DR, Kuhn S, Kniss KL, et al. Birth outcomes follow- 1021–1027.
ing West Nile virus infection of pregnant women in the 81. Cunha BA, Minnaganti V, Johnson DH, Klein NC. Profound
United States: 2003-2004. Pediatrics 2006;117:e537–e545. and prolonged lymphocytopenia with West Nile encephalitis.
59. Campbell G, Marfin A, Lanciotti R, Gubler D. West Nile Clin Infect Dis 2000;31:1116 –1117.
virus. Lancet Infect Dis 2002;2:519 –529. 82. Medarov BI, Multz AS, Brown W, Lempert L. West Nile me-
60. Watson JT, Pertel PE, Jones RC, et al. Clinical characteristics ningoencephalitis and rhabdomyolysis. Lancet Infect Dis
and functional outcome of West Nile fever. Ann Intern Med 2005;5:2.
83. Batuello JT, Youngwerth J, Gabel R. Increased serum lipase in
2004;141:360 –365.
west Nile virus infection. N Engl J Med 2005;352:420 – 421.
61. Sejvar J, Haddad M, Tierney B, et al. Neurologic manifesta-
84. Tyler KL, Pape J, Goody R, et al. CSF findings in 250 pa-
tions and outcome of West Nile virus infection. JAMA 2003;
tients with serologically confirmed West Nile virus meningitis
290:511–515.
and encephalitis. Neurology 2006;66:361–365.
62. Weiss D, Carr D, Kellachan J, et al. Clinical findings of West
85. Carson P, Steidler T, Patron R, et al. Plasma cell pleocytosis
Nile virus infection in hospitalized patients, New York and
in cerebrospinal fluid in patients with West Nile virus enceph-
New Jersey, 2000. Emerg Infect Dis 2001;7:654 – 658.
alitis. Clin Infect Dis 2003;37:e12–e15.
63. Brilla R, Block M, Geremia G, Wichter. Clinical and neuro-
86. Procop GW, Yen-Lieberman B, Prayson RA, Gordon SM.
radiologic features of 39 cases of West Nile virus meningoen-
Mollaret-like cells in patients with West Nile virus infection.
cephalitis. J Neurol Sci 2004;220:37– 40.
Emerg Infect Dis 2004;10:753–754.
64. Burton J, Kern R, Halliday W, et al. Neurological manifesta-
87. Lanciotti RS, Kerst AJ, Nasci RS, et al. Rapid detection of
tions of West Nile virus infection. Can J Neurol Sci 2004;31: West Nile virus from human clinical specimens, field collected
185–193. mosquitoes, and avian samples by a TaqMan reverse
65. Sayao AL, Suchowersky O, Al-Khathaami A, et al. Calgary transcriptase-PCR assay. J Clin Microbiol 2000;38:4066 – 4071.
experience with West Nile virus neurological syndrome during 88. Shi P-Y, Wong SJ. Serologic diagnosis of West Nile virus in-
the late summer of 2003. Can J Neurol Sci 2004;31:194 –203. fection. Expert Rev Mol Diagn 2003;3:733–741.
66. Chowers MY, Lang R, Nassar F, et al. Clinical characteristics 89. Roehrig JT, Nash D, Maidin B, et al. Persistence of virus-
of the West Nile fever outbreak, Israel, 2000. Emerg Infect reactive serum immunoglobulin M antibody in confirmed
Dis 2001;7:675– 678. West Nile virus encephalitis cases. Emerg Infect Dis 2003;9:
67. Jeha L, Sila C, Lederman R, et al. West Nile virus infection. A 376 –379.
new acute paralytic illness. Neurology 2003;61:55–59. 90. Tardei G, Ruta S, Chitu V, et al. Evaluation of immunoglob-
68. Tyler K. West Nile virus infection in the United States. Arch ulin M (IgM) and IgG enzyme immunoassays in serologic di-
Neurol 2004;61:1190 –1195. agnosis of West Nile virus infection. J Clin Microbiol 2000;
69. DeBiasi RL, Tyler KL. West Nile virus meningoencephalitis. 38:2232–2239.
Nat Clin Pract Neurol 2006;5:264 –275. 91. Kapoor H, Sigus K, Somsel P, et al. Persistence of West Nile
70. Ferguson DD, Gershman K, LeBailly A, Petersen LR. Char- virus (WNV) IgM antibodies in cerebrospinal fluid from pa-
acteristics of the rash associated with West Nile virus fever. tients with CNS disease. J Clin Virol 2004;31:289 –291.
Clin Infect Dis 2005;41:1204 –1207. 92. Gandelman-Marton R, Kimiagar I, Itzhaki A, et al. Electroen-
71. Bode V, Sejvar JJ, Pape WJ, et al. West Nile virus disease: a cephalography findings in adult patients with West Nile virus-
descriptive study of 228 patients hospitalized in a 4-county associated meningitis and meningoencephalitis. Clin Infect
region of Colorado in 2003. Clin Infect Dis 2006;42: Dis 2003;37:1573–1578.
1234 –1240. 93. Marciniak C, Sorosky S, Hynes C. Acute flaccid paralysis as-
72. Yim R, Posfay-Barbe KM, Nolt D, et al. Spectrum of clinical sociated with West Nile virus: motor and functional improve-
manifestations of West Nile virus infection in children. Pedi- ment in 4 patients. Arch Phys Med Rehabil 2004;85:
atrics 2004;114:1673–1675. 1933–1938.
73. Hassin-Baer S, Kirson ED, Shulman L, et al. Stiff-person syn- 94. Al-Shekhlee A, Katirji B. Electrodiagnostic features of acute
drome following West Nile fever. Arch Neurol 2004;61: paralytic poliomyelitis associated with West Nile infection.
938 –941. Muscle Nerve 2004;29:376 –380.

Davis et al: WNV Neuroinvasive Disease 299


95. Li J, Loeb JA, Shy ME, et al. Asymmetric flaccid paralysis: a 113. Hamdan A, Green P, Mendelson E, et al. Possible benefit of
neuromuscular presentation of West Nile virus infection. Ann intravenous immunoglobulin therapy in a lung transplant re-
Neurol 2003;53:703–710. cipient with West Nile virus encephalitis. Transpl Infect Dis
96. Petropoulou KA, Gordon SM, Prayson RA, Ruggierri PM. 2002;4:160 –162.
West Nile virus meningoencephalitis: MR imaging findings. 114. Shimoni Z, Niven MJ, Pitlick S, Bulvik S. Treatment of West
AJNR Am J Neuroradiol 2005;26:1986 –1995. Nile virus encephalitis with intravenous immunoglobulin.
97. Ali M, Sagriel Y, Sohi J, et al. West Nile virus infection: MR Emerg Infect Dis 2001;7:759.
imaging findings in the nervous system. AJNR Am J Neuro- 115. Haley M, Retter AS, Fowler D, et al. The role for intravenous
radiol 2005;26:289 –297. immunoglobulin in the treatment of West Nile virus enceph-
98. Kraushaar G, Patel R, Stoneham GW. West Nile virus: a case alitis. Clin Infect Dis 2003;37:e88 –e90.
report with flaccid paralysis and cervical spinal cord MR im- 116. Pyrgos V, Younus F. High-dose steroids in the management of
aging findings. AJNR Am J Neuroradiol 2005;26:26 –29. acute flaccid paralysis due to West Nile virus infection. Scand
99. Bouffard J, Riudavets M, Holman R, Rushing E. Neuropa- J Infect Dis 2004;36:509 –512.
117. Bai F, Wang T, Pal U, et al. Use of RNA interference to
thology of the brain and spinal cord in human West Nile virus
prevent lethal murine West Nile virus infection. J Infect Dis
infection. Clin Neuropathol 2004;23:59 – 61.
2005;191:1148 –1154.
100. Fratkin J, Leis A, Stokic D, et al. Spinal cord neuropathology
118. Deas TS, Binduga-Gajewska I, Tilgner M, et al. Inhibition of
in human West Nile virus infection. Arch Pathol Lab Med
flavivirus infections by antisense oligomers specifically sup-
2004;128:533–537. pressing viral translation and RNA replication. J Virol 2005;
101. Sampson B, Armbrustmacher V. West Nile encephalitis: the 79:4599 – 4609.
neuropathology of four fatalities. Ann N Y Acad Sci 2001; 119. Loeb M, Elliott SJ, Gibson B, et al. Protective behavior and
951:172–178. West Nile virus risk. Emerg Infect Dis 2005;11:1433–1436.
102. Watson JT, Pertel PE, Jones RC, et al. Clinical characteristics 120. Ng T, Hathaway D, Jennings N, et al. Equine vaccine for
and functional outcomes of West Nile fever. Ann Intern Med West Nile virus. Dev Biol 2003;114:221–227.
2004;141:360 –365. 121. Minke JM, Siger L, Karaca K, et al. Recombinant canarypox-
103. Klee AL, Maidin B, Edwin B, et al. Long-term prognosis for virus vaccine carrying the prM/E genes of West Nile virus
clinical West Nile virus infection. Emerg Infect Dis 2004;10: protects horses against a West Nile virus-mosquito challenge.
1405–1411. Arch Virol Suppl 2004;221–230.
104. Cao NJ, Ranganathan C, Kupsky WJ, Li J. Recovery and 122. Yamshchikov G, Borisevich V, Seregin A, et al. An attenuated
prognosticators of paralysis in West Nile virus infection. West Nile prototype virus is highly immunogenic and protects
J Neurol Sci 2005;236:73– 80. against the deadly NY99 strain: a candidate for live WN vac-
105. Morrey JD, Day CW, Julander JG, et al. Effect of interferon- cine development. Virology 2004;330:304 –312.
alpha and interferon-inducers on West Nile virus in mouse 123. Lustig S, Olshevsky U, Ben-Nathen D, et al. A live attenuated
and hamster animal models. Antivir Chem Chemother 2004; West Nile virus strain as a potential veterinary vaccine. Viral
15:101–109. Immunol 2000;13:401– 410.
106. Hrnicek MJ, Mailliard ME. Acute West Nile virus in two pa- 124. Arroyo J, Miller C, Catalan J, et al. ChemiVax-West Nile vi-
tients receiving interferon and ribavirin for chronic hepatitis rus live-attenuated vaccine: preclinical evaluation of safety, im-
C. Am J Gastroenterol 2004;99:957. munogenicity, and efficacy. J Virol 2004;78:12497–12507.
107. Kalil AC, Devetten MP, Singh S, et al. Use of interferon-␣ in 125. Pletnev AG, Claire MS, Elkins R, et al. Molecularly engi-
patients with West Nile encephalitis: report of 2 cases. Clin neered live-attenuated chimeric West Nile/dengue virus vac-
Infect Dis 2005;40:764 –766. cines protect rhesus monkeys from West Nile virus. Virology
108. Rahal JJ, Wehbeh WA. Double blinded, placebo controlled 2003;314:190 –195.
126. Despres P, Combredet C, Frenkiel M-P, et al. Live measles
trial of alpha interferon therapy for West Nile meningoen-
vaccine expressing the secreted form of the West Nile virus
cephalitis. Protocol WN-102. Available at: www.nyhq.org/
envelope glycoprotein protects against West Nile virus enceph-
posting/rahal.html. Accessed July 15, 2006.
alitis. J Infect Dis 2005;191:207–214.
109. Jordan I, Briese T, Fischer N, et al. Ribavirin inhibits West
127. Acambis. Acambis becomes first company to enter Phase II
Nile virus replication and cytopathic effect in neural cells. testing of vaccine against West Nile virus. Available at: http://
J Infect Dis 2000;182:1214 –1217. www.acambis.co.uk/print.asp?id⫽1446. Accessed July 15,
110. Diamond M, Shrestha B, Marri A, et al. B cells and antibody 2006.
play critical roles in the immediate defense of disseminated 128. Durbin A. Safety of and immune response to a West Nile
infection by West Nile encephalitis virus. J Virol 2003;77: virus vaccine (WN/DEN4 –3⬘delta30) in healthy adults. Avail-
2578 –2586. able at: http://www.clinicaltrials.gov/show/NCT00094718.
111. Ben-Nathan D, Lustig S, Tam G, et al. Prophylactic and ther- Accessed July 15, 2006.
apeutic efficacy of human intravenous immunoglobulin in 129. Pogodina V, Frolova M, Malenko G, et al. Study on West
treating West Nile virus infection in mice. J Infect Dis 2003; Nile Virus persistence in monkeys. Arch Virol 1983;75:
188:5–12. 71– 86.
112. Agrawal AG, Peterson LR. Human immunoglobulin as a 130. Penn RG, Guarner J, Sejvar JJ, et al. Persistent neuroinvasive
treatment for West Nile virus infection. J Infect Dis 2003; West Nile virus infection in an immunocompromised patient.
188:1– 4. Clin Infect Dis 2006;42:680 – 683.

300 Annals of Neurology Vol 60 No 3 September 2006

View publication stats

You might also like