You are on page 1of 19

DISEASE MANAGEMENT CNS Drugs 2000 Aug; 14 (2): 95-113

1172-7047/00/0008-0095/$20.00/0

© Adis International Limited. All rights reserved.

Management Strategies for


Herpesvirus Infections of the CNS
Immunocompetent and Immunocompromised Patients
Paola Cinque and Adriano Lazzarin
Division of Infectious Diseases, San Raffaele Hospital, Milan, Italy

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
1. General Aspects of Herpesvirus Infections of the CNS . . . . . . . . . . . . . . . . . . . . . . . . . 96
2. Principles of Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
3. Herpes Simplex Virus (HSV) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
3.1 Immunocompetent Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
3.1.1 Herpes Simplex Encephalitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
3.1.2 HSV Type 2 Meningitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
3.2 Immunocompromised Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
4. Varicella-Zoster Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
4.1 Immunocompetent Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
4.2 Immunocompromised Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
5. Cytomegalovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
5.1 Immunocompetent Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
5.2 Immunocompromised Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
6. Epstein-Barr Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
6.1 Immunocompetent Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
6.2 Immunocompromised Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
7. Human Herpesvirus 6 (HHV-6), HHV-7 and HHV-8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
7.1 Immunocompetent and Immunocompromised Patients . . . . . . . . . . . . . . . . . . . . . 108
8. Concluding Remarks and Future Prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

Abstract The known human herpesviruses (HHVs) include herpes simplex viruses type
1 and 2, varicella-zoster virus, cytomegalovirus, Epstein-Barr virus, and HHVs
6, 7 and 8. Almost all of them may induce CNS diseases, which can vary from
mild to life threatening and are significantly different in immunocompetent and
immunocompromised patients. An aetiological diagnosis is essential for optimal
disease management, and the recently developed molecular diagnostic techniques
have provided a rapid and sensitive means for herpesvirus identification.
Effective antiherpesvirus agents have long been available, but the only regi-
men that can be recommended on the basis of proven efficacy in controlled trials
is aciclovir (acyclovir) for herpes simplex encephalitis (HSE). Nevertheless, anti-
viral agents are empirically used in the treatment of various herpes-induced CNS
complications and a number of examples of their efficacy have been reported.
Most frequently, CNS disease results from direct virus-induced cytotoxicity. How-
96 Cinque & Lazzarin

ever, immune-mediated mechanisms may be involved in some cases, and corti-


costeroids are also empirically employed.
Despite the diagnostic advances and broader availability of antiviral com-
pounds, the management of herpes-induced CNS complications is still difficult.
A significant number of patients with HSE die, fail to regain normal function or
relapse despite an initial therapeutic response. The current antiviral approaches
are often unsatisfactory in immunocompromised patients and no optimal treat-
ment has yet been established for the majority of herpes diseases of the CNS.
Information regarding new treatment strategies, including different aciclovir re-
gimens and the use of newer compounds or drug combinations, is needed and can
only be provided by large multicentre studies.

1. General Aspects of Herpesvirus most likely depend on the balance between virus
Infections of the CNS replication and the host inflammatory response. For
instance, direct virus cytotoxicity is likely to be the
The known human herpesviruses (HHVs) in- most important pathogenic mechanism in the im-
clude herpes simplex viruses type 1 and 2 (HSV-1, munocompromised patient with HSE, in whom wide-
HSV-2), varicella-zoster virus (VZV), cytomegalo- spread parenchymal destruction is associated with
virus (CMV), Epstein-Barr virus (EBV), and HHV- the presence of virus, but in whom there is a limited
6, HHV-7 and HHV-8. Almost all of these viruses inflammatory response.[2]
can induce CNS complications, but these are rela- Immune-mediated processes are also involved
tively rare in relation to the widespread nature of in cases of postinfectious encephalitis, which in-
herpesviruses. Herpesvirus infections of the CNS cludes syndromes such as acute demyelinating en-
vary greatly in terms of their pathogenic mechan- cephalitis or acute disseminated encephalomyelitis.
isms and clinico-pathological patterns. Furthermore, Postinfectious encephalitis typically follows an ex-
different forms are observed in immunocompetent anthem or a respiratory infection, caused by viruses
and immunocompromised patients. Nevertheless, or nonviral agents, e.g. Mycoplasma pneumoniae.
some common features can be recognised. Herpes- It may also be associated with vaccinations.Among
viruses are ubiquitous and usually cause asymp- herpesviruses, VZV, EBV and HSV-1 can all be in-
tomatic or mild infection (table I). However, fol- volved.[3,4] In these forms, the virus is not commonly
lowing primary infection, they remain latent in the found within the CNS lesions, although there is ev-
body and may be subsequently reactivated under idence of viral persistence in some cases, such as in
particular circumstances such as immunodeficiency. postvaricella cerebellitis.[5] CNS tissue injury usually
During primary infection or reactivation, herpes- consists of perivascular inflammation and demyelin-
viruses may enter the CNS by nerve pathways or ation. Although the underlying events are mostly un-
by crossing the blood-brain barrier. known, it is possible that viral infection of lympho-
Most frequently, the CNS injury results directly cytes is followed by deregulation of normal immune
from the herpesvirus invasion of the CNS tissues. mechanisms, resulting in an aberrant, autoimmune-
In some instances, however, a virus-induced im- like response.[3]
mune reaction may further account for tissue in- Some common diagnostic considerations can also
jury, as observed in the case of herpes simplex en- be outlined for herpesvirus infections of the CNS.
cephalitis (HSE). In this disease, tissue destruction Although the clinical presentation, neuroimaging,
is initially sustained by the direct cytotoxic effect standard CSF analysis and other examinations may
of the virus and thereafter continued by immune prove useful, optimal therapy can be introduced
mechanisms.[1] The relative importance of either only on the basis of an aetiological diagnosis. This
mechanism in determining the CNS disease will requires virus identification in the CSF or brain

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS
© Adis International Limited. All rights reserved.

Table I. Human herpesviruses and their associated infections

Virus Primary infection Reactivated infection Proven or Principal CNS diseases in Principal CNS diseases in Registered drugs for
presumed site immunocompetent patients immunocompromised patients systemic infections
of latency

HSV-1 Oral herpes Herpes labialis Sensory ganglia Herpes encephalitis (classical), Encephalitis (focal, Aciclovir (acyclovir),
herpes encephalitis (atypical ventriculoencephalitis), myelitis valaciclovir, famciclovir,
forms) foscarnet

HSV-2 Herpes genitalis Herpes genitalis Sensory ganglia Aseptic meningitis, recurrent Encephalitis (focal, Aciclovir, valaciclovir,
aseptic meningitis (Mollaret’s ventriculoencephalitis), myelitis famciclovir, foscarnet
meningitis)

VZV Varicella Herpes zoster Sensory ganglia Varicella complications (cerebellar Varicella complications Aciclovir, valaciclovir,
ataxia, meningitis, encephalitis, (encephalitis), herpes zoster famciclovir, foscarnet
myelitis, polyradiculomyelitis), complications or CNS disease
herpes zoster complications or without skin lesions (meningitis,
CNS disease without skin lesions encephalitis, myelitis, vasculitis)
(meningitis, encephalitis, myelitis,
vasculitis)

CMV CMV-mononucleosis Organ disease [e.g. Monocytes, Meningitis, encephalitis Encephalitis (focal, Ganciclovir, foscarnet,
retina, gastrointestinal endothelial cells periventricular, micronodular), cidofovir
tract (immuno- polyradiculomyelitis
compromised)]

EBV Mononucleosis Hairy leucoplakia; B lymphocytes Meningitis, encephalitis, myelitis Lymphoproliferative diseases Ganciclovir, foscarnet,
lymphoproliferative (organ transplantation), primary cidofovir
syndromesa, lymphomaa CNS lymphoma (HIV infection)
(immunocompromised)

HHV-6 Exanthema subitum Organ disease? T lymphocytes Febrile seizures, encephalitis Encephalitis None
(variants A (sixth disease) (immunocompromised)
and B)b

HHV-7 Exanthema subitum Unknown T lymphocytes Febrile seizures, encephalitis Unknown None
CNS Drugs 2000 Aug; 14 (2)

HHV-8 Kaposi’s sarcoma? Kaposi’s sarcoma? B lymphocytes Unknown Unknown None

a Resulting from EBV-induced B cell transformation.

b Variant B is associated with primary infection; either the A or B variant has been observed in CNS diseases.

CMV = cytomegalovirus; EBV = Epstein-Barr virus; HHV = human herpesviruses; HSV = herpes simplex virus; VZV = varicella-zoster virus.

97
98 Cinque & Lazzarin

tissue, or the demonstration of a specific intrathe- replication and include nucleoside analogues and
cal immune response. In this regard, the applica- other herpesvirus DNA polymerase inhibitors. CSF
tion of the polymerase chain reaction (PCR) to CSF pharmacokinetic studies suggest that penetration
has led to revolutionary advances in the diagnosis of these drugs into the CNS is variable, although
of herpesvirus CNS infections because of its extra- CSF concentrations above inhibitory concentrations
ordinary sensitivity.[6,7] Furthermore, PCR allows are likely to be achieved in several instances (table
a result to be obtained within one day of CSF sam- II). With the exception of HSE, for which compar-
pling, thus impacting substantially on treatment de- ative trials have demonstrated the beneficial effect
cision making. of aciclovir (acyclovir),[21,22] no information based
This review briefly introduces the herpesvirus on clinical trial results is currently available for
infections of the CNS in both immunocompetent herpesvirus infections of the CNS. The relatively
and immunocompromised patients, and concentrates few cases observed, as well as diagnostic problems,
on the most relevant studies and clinical observa- have hampered research into specific treatments.
tions concerning the use of rapid diagnostic pro- Nevertheless, antiviral drugs are empirically given
cedures and treatment modalities. Treatment rec- in a number of herpes-induced CNS complications,
ommendations are provided for those diseases for with variable benefit. Recently, the use of antiviral
which a given approach has been well established drugs in combination has received attention. Drug
by clinical trials, or where experts in the field have combinations can increase drug potency and pre-
reached a consensus.[8] Further suggestions based vent the development of antiviral resistance, which
on personal or anecdotal experiences are also pro- can be of particular concern in immunocompro-
vided. Neither the encephalopathy caused by con-
mised patients. Resistance results from alterations
genital or neonatal herpesvirus infections nor her-
of viral enzymes that are critical for drug function-
pes-induced retinopathies are considered. Special
ing, and resistance to a nucleoside analogue will
mention is made of the herpesvirus CNS infections
most likely confer resistance to other drugs of the
occurring in the context of HIV infection. These
same class (table II).[23]
infections have caused severe diseases and mortal-
In cases of CNS diseases that are likely to be
ity in the past, but have become much less frequent
caused by an immunotoxic mechanism rather than
since the introduction of highly active antiretroviral
direct viral cytotoxicity, corticosteroids are empir-
therapies (HAARTs) directed against HIV, which
ically used because of their anti-inflammatory ef-
have dramatically impacted on the outcome of HIV
infection.[9] Herpesvirus CNS infections can still fects. Intravenous high dose glucocorticoids (e.g.
be observed as the first disease manifestation in methylprednisolone up to 500mg daily for 3 to 5
patients with newly recognised HIV infection, or days) has been the standard treatment for postinfec-
in those who fail to respond to, or are intolerant of, tious encephalitis,[24,25] but its efficacy has not been
HAARTs. However, because of the paucity of cases proven. Alternative treatments, including intrave-
of herpesvirus CNS disease, it is still not known nously administered immunoglobulin (IVIG)[26-28]
whether the combined use of HAART and specific and plasmapheresis,[29,30] have recently been used
antiviral therapy might favourably affect their out- in these conditions. Based on the successful res-
come, as observed in other HIV-related opportunis- ponse observed in some patients, including pati-
tic infections.[10] ents who had failed to respond to corticosteroids,
IVIG and plasmapheresis appear to be promising
2. Principles of Treatment options for the treatment of acute demyelinating
encephalitis and warrant further evaluation. At pre-
The aetiological treatment of herpesvirus infec- sent, an empirical combined use of glucocorticoids
tions of the CNS is currently based on the use of and plasmapheresis or IVIG, that takes into account
antiviral agents. These drugs act by inhibiting viral the pathogenic role of both cellular and humoral

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS
© Adis International Limited. All rights reserved.

Table II. Antiviral drugs with antiherpesvirus activity licensed for systemic use

Drug Class Highly sensitive Poorly or Administration Potentially severe CSF concentration Known basis of viral Recommended
moderately route adverse effects (% of plasma resistance regimen in CNS disease
sensitive concentration)

Aciclovir Nucleoside HSV-1, HSV-2, CMV, EBV, HHV-6, IV, oral Renal toxicity Approximately Altered or deficient 10 mg/kg every 8h for
(acyclovir)[11] analogue VZV HHV-7, HHV-8 (rare) 50[12] virus thymidine kinase 10-14 days, IVa
(HSV-1, HSV-2, VZV)

Valaciclovir[13] Nucleoside HSV-1, HSV-2, CMV, EBV, HHV-6, Oral Not reported Not available Altered or deficient Not evaluated
analogue VZV HHV-7, HHV-8 virus thymidine kinase
(aciclovir (HSV-1, HSV-2, VZV)
prodrug)

Famciclovir[14] Nucleoside HSV-1, HSV-2, CMV, EBV, HHV-6, Oral Not reported Not available Altered or deficient Not evaluated
analogue VZV HHV-7, HHV-8 thymidine kinase
(penciclovir (HSV-1, HSV-2, VZV)b
prodrug)

Ganciclovir[15] Nucleoside HSV-1, HSV-2, IV, oral Granulocytopenia, 24-67[16] Altered CMV UL97- 10 mg/kg once daily for
analogue VZV, CMV, thrombocytopenia encoded phospho- 14-21 days (IV) then 5
EBV, HHV-6, transferase or mg/kg/day
HHV-7, HHV-8 UL54-encoded (maintenance)c
polymerase

Foscarnet[17] Non- HSV-1, HSV-2, IV Renal toxicity, 5-72[18] Altered virus 180 mg/kg once daily
nucleoside VZV, CMV, electrolyte polymerase (HSV-1, for 14-21 days (IV) then
pyropho- EBV, HHV-6, imbalances, HSV-2, VZV) 90 mg/kg/day
sphate HHV-7, HHV-8 anaemia (maintenance)c
analogue

Cidofovir[19] Nucleoside HSV-1, HSV-2, IV Renal toxicity, 2-4[20]d Altered CMV UL54- 5 mg/kg once weekly
phosphonate VZV, CMV, granulocytopenia, encoded polymerase for 2 wks (IV) then 5
EBV, HHV-6, iritis (CMV) mg/kg every 2 wks
HHV-7, HHV-8 (maintenance)e

a 10 mg/kg every 8h for 10 days (IV) has been used in clinical trials[21,22] and proven to be effective.
CNS Drugs 2000 Aug; 14 (2)

b HSV-1, HSV-2 or VZV aciclovir-resistant strains frequently exhibit cross-resistance to penciclovir in vitro.

c The combination of ganciclovir and foscarnet should be considered for treatment of CMV encephalitis.

d These data were obtained 1h after cidofovir administration, and its long half-life should be considered.

e To be accompanied by aggressive IV hydration and oral administration of probenecid (2g 3h prior to cidofovir infusion and 1g at 2 and 8h after completion of the infusion).

CMV = cytomegalovirus; EBV = Epstein-Barr virus; HHV = human herpesviruses; HSV = herpes simplex virus; IV = intravenous; VZV = varicella-zoster virus.

99
100 Cinque & Lazzarin

immunity, can be recommended to treat these forms be lowered by this procedure, the oedema could be
of encephalitis. aggravated.
Along with aetiological treatments, a number of Effective control of seizures may be achieved
life-support measures are required in the clinical with the use of phenytoin at an initial dosage of 20
management of severe cases of acute encephalitis mg/kg/day, administered intravenously 3 times daily,
and meningitis caused by herpesviruses. These in- and a maintenance dosage of 7 to 10 mg/kg/day,
clude providing nutrition and controlling tempera- with concomitant monitoring of plasma drug con-
ture and electrolyte balance, as well as treatment of centrations.[32,33]
organ dysfunction other than that of the CNS. In-
tensive medical supportive care is almost invari- 3. Herpes Simplex Virus (HSV)
ably required, including control of cerebral oedema
and seizures, and assisted ventilation in those who 3.1 Immunocompetent Patients
develop deepening coma or other signs of brain-
3.1.1 Herpes Simplex Encephalitis
stem dysfunction. Computed tomography (CT) and HSE is the most common sporadic encephalitis.
magnetic resonance imaging (MRI) scans are of It may present at any age, with most of the cases
value in monitoring cerebral oedema, whereas elec- occurring during the first decade or in patients over
trophysiological monitoring and blood flow studies 50 years. Although HSE may affect young chil-
provide precious functional information. Moreover, dren, it needs to be distinguished from neonatal
intracranial pressure (ICP) monitoring should be HSV encephalitis, which differs greatly in terms of
used for management of severe cases. aetiopathogenesis, clinical presentations and out-
In order to reduce cerebral oedema, the use of come. In the neonatal disease, different treatment
corticosteroids (e.g. high doses of dexamethasone strategies may be required because of the lower rate
up to 16 to 20mg daily) is advocated by some for of responses compared with the ‘adult’ forms.[34]
the first 4 or 5 days of the illness, although this is In adults and children above the neonatal age, HSE
a matter of dispute. The immune-suppression in- is caused by HSV-1 in over 90% of the cases, and
duced by corticosteroids can theoretically favour by HSV-2 in the remaining cases. Inflammatory to
virus replication and thus worsen the outcome of haemorrhagic-necrotising lesions are characteristic-
CNS infection. However, a recent study using a rat ally located in the temporal lobes, but other adjacent
model of HSE has shown that corticosteroids do areas may also be involved.[34] The onset of HSE
not significantly inhibit the antiviral effect of aci- is typically acute, with nonspecific focal neurolog-
ical signs and symptoms and impaired conscious-
clovir on the HSV-1 load in brain tissue and are also
ness. A broader spectrum of clinical presentations
effective in decreasing the extent of brain infection
of HSV-1 CNS infections has been recognised over
if used without aciclovir.[31] These observations, if
the last few years, including mild and chronic en-
confirmed, will provide an important experimental
cephalitis, and neurological syndromes related to
rationale supporting controlled clinical evaluations specific anatomic locations.[35-38]
of corticosteroids in combination with antivirals The identification of HSV in brain tissue biop-
in acute viral CNS infections. In an acute cerebral sies used to be the only conclusive means of estab-
oedema, a rapid reduction of elevated ICP is best lishing an aetiological diagnosis of HSE, but this
achieved by hyperventilation. In order to rapidly has more recently been replaced by the PCR detec-
decrease ICP, intravenous administration of man- tion of HSV-1 DNA in the CSF as the diagnostic
nitol (1.5 to 2 g/kg, followed by 0.25 g/kg every 4 method of choice.[38] A number of retrospective and
hours, or intermittent small doses of 0.25 to 1 g/kg) prospective studies have clearly established the di-
may also be effective. The use of surgical decom- agnostic reliability of the PCR, showing a more
pression is controversial because, although ICP may than 90% sensitivity and virtually 100% specific-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 101

ity.[39-41] This technique is rapid, which allows a In order to maintain sufficiently high exposures
diagnosis to be established in time for management in the brain to meet the inhibitory concentration for
decision making. Furthermore, it enables the diag- HSV, it is necessary that aciclovir is administered
nosis of uncommon forms of HSV CNS infection intravenously. Following oral intake, the absorb-
that may otherwise go unrecognised.[35,36] The dem- ence rate of the drug is low (15 to 30%), and plasma
onstration of an intrathecal anti-HSV antibody syn- concentrations warranting sufficient penetration in
thesis is also diagnostically useful, but can only be the CNS are not achieved by this route.[11,12]
applied in prolonged or chronic cases because a Although aciclovir is highly effective in reduc-
response can usually only be detected 7 to 10 days ing HSE-associated mortality and morbidity, a sig-
after the onset of symptoms.[39,41] nificant number of patients still die or are left with
The first antiviral agents used for the treatment severe neurological sequelae. For these reasons,
of HSE were idoxuridine, cytarabine and vidarab- more potent antiviral regimens have been propo-
ine.[42-44] The first clinical trial involved idoxurid- sed. These include increasing the duration of treat-
ine and showed that this drug had low effectiveness ment to 21 days, increasing the dosage to 15 mg/kg
and was unacceptably toxic for the treatment of every 8 hours or making use of antiviral drug com-
HSE.[45] A subsequent controlled study of vidarab- binations. In this regard, new anti-herpes compounds
ine, conducted in the late 1970s, showed that the have become available over recent years, thus pro-
rates of survival and normalisation of neurological viding additional therapy options (table II).
function were higher in patients treated with this Another important concern regarding HSE out-
come is the occurrence of relapse following a fa-
drug than in those receiving placebo.[46] Less than
vourable response to therapy.[47-51] In a recent pae-
a decade later, 2 large studies conducted indepen-
diatric series, HSE relapse was observed in 26% of
dently in Sweden and the US demonstrated that
HSE cases, usually days to weeks following the
mortality was significantly lower in patients receiv-
resolution of a first episode. Of note, relapse cases
ing aciclovir than in those treated with vidarabine
were found to be significantly more frequent in
(19 to 28% vs 50 to 54%, respectively). Further-
patients receiving lower total doses of aciclovir
more, 38 to 56% of the patients treated with aci-
during the initial episode.[51] Relapse may be asso-
clovir were left with no or only minor sequelae ciated with either a rebound of viral replication or
after 6 months, compared with 13 to 14% of those an immune-mediated mechanism. Indeed, reported
receiving vidarabine.[21,22] The efficacy of aciclo- cases show inconsistent evidence of viral replication
vir was greater in patients treated early after dis- in the CSF,[49-52] and neuropathological studies in-
ease onset and in those who were younger and had dicate that both mechanisms can be involved.[4,47,48]
less severe neurological impairment. Aciclovir, in The administration of additional aciclovir courses
contrast to vidarabine, was not associated with sig- has been shown to be useful in patients with relapse
nificant toxicity. of the disease[51] and, because of the possible virus
In these clinical trials, aciclovir 10 mg/kg was persistence in the CNS, we believe that this strat-
intravenously administered every 8 hours for 10 egy should always be used in similar cases. On the
days. In order to increase the likelihood of obtaining other hand, anti-inflammatory drugs, i.e. cortico-
complete virus suppression, however, 2 full weeks steroids, do not have proven efficacy and their use
of therapy are often used in clinical practice. Intra- remains controversial.
venous aciclovir 10 mg/kg 3 times daily for 10 to In clinical practice, aciclovir is often initiated
14 days is therefore the current recommended reg- empirically in patients with suspected HSE. How-
imen for HSE. Although the vast majority of HSE ever, a newer approach based on the use of CSF
cases are caused by HSV-1, this dosage recommen- PCR (an example of which is shown in figure 1),
dation also applies to encephalitis caused by HSV-2. is currently used in several centres and is strongly

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
102 Cinque & Lazzarin

Clinical neurological assessmenta


CT scan/MRIa Suspected Begin aciclovir
CSF chemistry HSE (acyclovir)b
Microbiology tests

CSF PCR for HSV-DNA


HSV-specific intrathecal Ab syntesisc

Other
Ab
negative diagnosis
probable
PCR PCR
positive negative
Ab
positive

Continue
aciclovir

Repeat CSF
at the end of treatment

PCR PCR
negative positive

Clinical
improvement

Stop Continue
aciclovir aciclovir

Fig. 1. Polymerase chain reaction (PCR)–based algorithm for the management of patients with suspected herpes simplex encephalitis
(HSE). (a) Before CSF sampling, to exclude the presence of mass lesions that may contraindicate the lumbar puncture. (b) Standard
treatment: 10 mg/kg/day every 8 hours intravenously for 10 to 14 days. (c) Herpes simplex virus (HSV) intrathecal antibody synthesis
detection may be of help after the first week from onset of symptoms. Ab = antibody; CT = computed tomography; MRI = magnetic
resonance imaging.

recommended.[38] In a recent decision analysis mo- ist of initially negative PCR results obtained very
del, a PCR-based approach was shown to be associ- early after the onset of HSE symptoms, which are
ated not only with a better outcome than that offered likely to reflect limited virus replication and initial
by empirical therapy, but also with a significant CNS lesions. In these cases, however, HSV DNA
reduction in the use of aciclovir.[53] These results can be detected in CSF samples drawn a few days
are due to a lower rate of inappropriate aciclovir later.[52]
discontinuation in patients with HSE and a higher On the other hand, the likelihood of finding a
rate of correct discontinuation in PCR-negative pa- positive CSF PCR result is reduced following a few
tients. In using such a PCR-based approach, how- days of aciclovir treatment, and in untreated pa-
ever, it is important that PCR results are interpreted tients from whom CSF is obtained late after the
cautiously in relation to the clinical presentation onset of neurological symptoms. A delayed exam-
and the duration of antiviral therapy. Examples ex- ination of the CSF by PCR can still be diagnosti-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 103

cally helpful, since HSV DNA has been found in may therefore be an option in patients who have
the CSF as late as 2 weeks after HSE onset, even already experienced HSV-2 meningitis episodes and
in patients who had received aciclovir.[39,40,52] More in whom an initial CNS or genital recurrence can
recently, quantitative PCR techniques have reveal- be recognised. Furthermore, oral aciclovir 400mg
ed a correlation between high CSF HSV DNA lev- twice daily,[62,63] valaciclovir 500 to 1000mg once
els and poor disease outcome and also a decrease daily or 250mg twice daily,[64] and famciclovir 250mg
in HSV DNA levels during therapy,[54] thus sug- twice daily[65] have all been proven to efficiently
gesting that HSV DNA quantitation in CSF may suppress recurrences of genital infection. Intermit-
also gain a role in the diagnostic stages and during tent or continuous prophylaxis using these regi-
HSE treatment. mens is thus likely to be also effective in the pre-
vention of CNS episodes.
3.1.2 HSV Type 2 Meningitis
HSV-2 can cause aseptic meningitis, a form that 3.2 Immunocompromised Patients
is mainly observed in women, and may be associated
with primary HSV-2 genital infection.[55] Further- CNS infections due to HSV-1 or HSV-2 are an
more, HSV-2 meningitis may recur over months or infrequent complication in both HIV-infected[66,67]
years (‘Mollaret’s meningitis’), often in association and other immunosuppressed patients.[68] In HIV-
with recurrences of genital HSV-2 infection.[56,57] infected patients with a relatively conserved im-
HSV-2 meningitis can be easily recognised when munity, the neuropathology findings and clinical
genital lesions are present; virus isolation from CSF, presentations are similar to those observed in clas-
the demonstration of an intrathecal HSV antibody sical HSE, but these are different in patients with
response and seroconversion against HSV-2 have a severely impaired immune system. Most of these
all been used to confirm such a diagnosis or to cases have combined HSV-1 or HSV-2 and CMV
formulate it when it is less obvious.[55] Over the infection, and present clinically with a subacute
last few years, the PCR detection of HSV-2 has and progressive encephalitis that is similar to that
been found to be more sensitive than these meth- observed in patients with pure CMV encephalitis.[69]
ods. Although no systematic studies of CSF PCR Extensive necrosis with no or mild inflammation,
have been reported to date, sufficient data have ac- often located in the periventricular areas, is revealed
cumulated to suggest that it may be the method of at neuropathology.[66-68] HSV encephalitis occurring
choice in both primary and recurrent forms.[57-59] in HIV-infected and other severely immunosuppre-
Primary and recurrent meningitis are benign and ssed patients is clinically difficult to recognise. How-
recover spontaneously even in the absence of ther- ever, CSF PCR has also been shown to be highly
apy.[55,57] However, published case reports and cli- reliable in these patients, with a sensitivity and spe-
nical observations indicate that intravenous aciclo- cificity of 100 and 99.5%, respectively.[69]
vir at 5 or 10 mg/kg every 8 hours is useful in In addition to aciclovir, HSV-1 and HSV-2 are
relieving the symptoms of HSV-2 meningitis and also susceptible to ganciclovir, foscarnet and cido-
shortening disease duration.[58,60] Intravenous aci- fovir, antiviral agents that are approved for the treat-
clovir at these dosages is indicated in hospitalised ment of CMV infections (table II). Although these
patients with HSV-2 meningitis. However, the use drugs are less well tolerated than aciclovir, their
of valaciclovir, an orally administered ester of aciclo- use is indicated in patients with concomitant HSV
vir, can be considered in milder recurrent cases. Fol- and CMV infections. However, HIV-infected pa-
lowing oral administration of valaciclovir 1000mg tients with HSV and CMV or pure HSV infection
every 8 hours, plasma aciclovir concentrations are respond poorly to standard aciclovir, ganciclovir
similar to those achieved after intravenous admin- or foscarnet therapy and their survival is low. Fur-
istration of aciclovir 5 mg/kg every 8 hours.[61] Val- thermore, a relapse of encephalitis following an
aciclovir at the dosage of 1000mg 3 times daily initial response has also been observed.[69] In re-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
104 Cinque & Lazzarin

lapses of HSE observed in HIV-infected patients, antibody production is also diagnostically useful
the neuropathological findings and the persistence for VZV-induced neurological complications, but
of HSV DNA in the CSF suggest that CNS damage reliable results appear only days to weeks after the
results principally from residual virus replication. onset of neurological symptoms.[41,75]
It is likely that standard antiviral regimens are not Intravenous aciclovir is usually given to pati-
sufficient to clear extensive CNS infections in the ents with VZV disease of the CNS and there is ev-
absence of adequate host immune control. How- idence for the efficacy of this approach.[72,76,77] On
ever, drug resistance to aciclovir has also been re- the basis of reported and direct experience with
ported.[70] For these reasons, the use of drug com- these patients, and taking into account the informa-
binations may represent a reasonable strategy in tion collected in the HSE treatment trials, intrave-
this particular patient population. nous aciclovir at 10 mg/kg 3 times daily for 10 to
14 days appears indicated for the treatment of VZV-
4. Varicella-Zoster Virus associated neurological diseases. Because the in-
hibitory aciclovir concentrations required for VZV
4.1 Immunocompetent Patients are higher than those required for HSV-1 or HSV-
2,[12] the use of 15 mg/kg every 8 hours has also
VZV CNS disease may complicate both chicken- been suggested to treat the most severe forms.[71]
pox and herpes zoster, but may also occur in the Corticosteroids are frequently employed in the
absence of skin lesions. The most frequent compli- treatment of cerebellar ataxia, but antiviral therapy
cation of varicella is acute and usually benign cer- is also indicated because of the possible association
ebellar ataxia, whereas other CNS localisations are of this complication with virus replication in the
less commonly observed (table I). Cerebellar atax- CNS. However, cerebellar ataxia is usually a self-
ia, as well as some cases of encephalomyelitis, are limiting disease and the benefit of anti-inflamma-
thought to originate from postinfectious immune- tory or antiviral regimens has not yet been estab-
mediated mechanisms. Herpes zoster–related neu- lished.
rological disease generally occurs in the elderly and
may be severe. This consists of vasculitis, enceph- 4.2 Immunocompromised Patients
alitis, meningitis, myelitis or mixed forms.[71] VZV
vasculitis is now a well defined syndrome that re- In HIV-infected patients, different clinico-path-
sults from infection of large vessels in the CNS ological forms of VZV infections of the CNS have
(granulomatous arteritis) and leads to thrombosis been recognised, including multifocal leucoence-
and cerebral infarcts.[72] phalitis, ventriculo-encephalitis, meningomyelo-
CSF PCR is the most sensitive method for diag- radiculitis, myelitis and vasculitis.[78] Unlike vas-
nosing VZV CNS infections, whereas other viro- culitis of the immunocompetent host, in whom VZV
logical techniques are rarely successful.[73] With the infection principally involves large arteries, a small
use of CSF PCR, VZV DNA has been found in the vessel vasculitis is observed in HIV-infected or other
majority of patients with herpes zoster and neurolo- immunosuppressed patients.[72] Herpes zoster pre-
gical complications, in patients with VZV CNS in- cedes or accompanies CNS disease in approxim-
fection and no cutaneous lesions, but also in patients ately half of the cases, whereas no cutaneous le-
with herpes zoster and no CNS symptoms.[5,41,61,73,74] sions are otherwise observed.[78,79] VZV infection
Remarkably, the VZV genome is also found in the can also spread to the visceral organs (including the
CSF of approximately half of the children with cer- CNS), in other immunocompromised patients.[80]
ebellar ataxia,[5] suggesting that VZV persistence This was of great concern in children with leukae-
in the CNS might play a role in sustaining the im- mia or other malignancies who contracted varice-
mune processes presumably associated with this lla; however, this risk has now been drastically re-
disease. The detection of intrathecal VZV-specific duced by preventive strategies based on the use of

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 105

vaccination, passive immunisation and the treat- 5. Cytomegalovirus


ment of varicella with intravenous aciclovir.[81] In
HIV-infected patients, VZV DNA can be found by 5.1 Immunocompetent Patients
CSF PCR in the case of proven VZV CNS dis- CNS infections due to CMV, including menin-
ease,[79,82,83] although the virus has also been re- gitis or encephalitis, are rare in immunocompetent
covered in the CSF of patients in whom there was patients.[93-95] The serological demonstration of a
no clear evidence of VZV CNS involvement.[79] primary CMV infection, i.e. immunoglobulin (Ig)
Clinical responses to aciclovir, at the doses used G (IgG) seroconversion or the demonstration of
in immunocompetent patients, have also been repor- specific IgM, concomitant with neurological symp-
ted in HIV-infected patients with encephalitis or toms strongly supports a CMV-induced CNS dis-
ease. Although a number of methods have been
myelitis.[72,79,84-86] Furthermore, VZV DNA is clear-
used to achieve a definite diagnosis, including the
ed from the CSF in the majority of treated cases.[79,83]
demonstration of intrathecal antibody production,
Intravenous high dose aciclovir can therefore be virus isolation from CSF or virus detection by im-
recommended for the treatment of VZV neurolog- munohistochemistry, CSF PCR is the most sensi-
ical complications in immunocompromised pati- tive diagnostic technique for the demonstration of
ents. CMV DNA.[94-96] Furthermore, the application of
However, clinical failure is not uncommon and CSF PCR has been of major importance in defining
may be associated with virus persistence in the the clinical spectrum of CMV-associated neurolog-
CSF.[79,83] Lack of clinical and virological response ical manifestations.[94]
might result from inadequate CNS drug concentra- The outcome of CNS complications due to CMV
tions, which might support the use of increased is usually good, although the symptoms may occa-
sionally persist for some months.[94] In anecdotal
aciclovir dosages, i.e. 15 mg/kg 3 times daily, for
cases of CMV encephalitis, the use of intravenous
treating severe VZV neurological disease. How- ganciclovir at standard doses has been followed by
ever, treatment failure might also result from the a recovery of neurological symptoms.[97] Because
development of aciclovir resistant VZV strains.[87] of the paucity of clinical experiences, there is no
Alternative approaches, for example, the use of other clear indication on whether or not to treat CMV
drugs or drug combinations, therefore need to be complications in the CNS in immunocompetent pa-
considered. New antiviral agents that are highly tients. Both disease severity and possible adverse
effective against VZV, such as famciclovir and so- effects of anti-CMV drugs, that are more relevant
rivudine, have recently been developed, but no in- than those caused by aciclovir, should be taken into
formation is so far available on their use in CNS account in treatment decision making (table II).
infections. Famciclovir, an orally administered es- 5.2 Immunocompromised Patients
ter of penciclovir, has been proven to be at least as
effective as aciclovir for the treatment of herpes CMV infections of the CNS in HIV-infected pa-
zoster in immunocompetent patients.[88,89] Sorivu- tients include the well described syndromes of en-
cephalitis and polyradiculomyelitis.[95,98,99] CMV
dine, an extremely potent inhibitor of VZV DNA
encephalitis used to be observed in up to 25% of
polymerase, was even more effective than aciclovir
autopsied cases, although it has become infrequent
in accelerating cutaneous healing of herpes zoster in since the introduction of HAARTs. Its main neu-
HIV-infected patients.[90,91] This drug, however, has ropathological forms are ventriculo-encephalitis,
not been approved for clinical use because it cau- focal encephalitis and diffuse micronodular ence-
sed lethal toxicity in patients concomitantly treated phalitis, which all present clinically as a subacute
with fluorouracil.[92] encephalopathy.[98] The prognosis is poor, with death

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
106 Cinque & Lazzarin

ensuing within weeks despite antiviral treatment. More recently, clinical improvement or stabil-
CMV polyradiculomyelitis is due to infection of isation of CNS CMV disease has been reported in
the cauda equina and lumbosacral rootlets. It is cha- more than half of the patients receiving a combina-
racterised by a subacute onset of leg weakness and tion of ganciclovir and foscarnet, whereas therapy
sensory loss, with sphincter dysfunction, rapidly discontinuation due to adverse effects was observed
progressing to ascending paraparesis.[99] in approximately one-third of the patients treated
Although infrequent, other immunocomprom- with this regimen.[110] Unlike encephalitis, CMV
ised patients such as liver, bone marrow, heart and polyradiculitis responds to ganciclovir in approxi-
kidney transplant recipients, may also develop CMV mately 50% of cases, and responses have also been
encephalitis. Symptoms may be severe in these pa- reported following foscarnet therapy.[99,111]
tients, but the extensive necrosis commonly obser- The failure of anti-CMV drugs in the treatment
ved in HIV-infected patients is rarely observed at of CMV infections of the CNS may be due to inad-
histopathological examination.[95] equate CNS drug concentrations in relation to the
CSF viral isolation and antigen detection in po- viral burden or to the presence of irreversible tissue
lymorphonuclear leucocytes have low sensitivity damage caused by extensive necrosis. However, dev-
in cases of encephalitis. These techniques, how- elopment of drug-resistant CMV strains may also
ever, may be useful in cases of polyradiculopathy, occur. In this regard, viral strains resistant to gan-
in which a typical CSF polymorphonuclear pleo- ciclovir and CMV-UL 97 gene mutations confer-
cytosis is observed.[99] CSF PCR for the detection ring resistance to this drug have been detected in
of CMV DNA is highly sensitive and specific for the CSF of patients with CMV neurological dis-
both polyradiculomyelitis and encephalitis, with a eases.[112,113]
diagnostic sensitivity of 80 to 95% and specificity An induction course of ganciclovir, foscarnet or
of 90 to 100% in HIV-infected patients.[7,100-103] cidofovir, followed by lifelong maintenance treat-
Since CMV DNA can be found in the CSF of pati- ment, at the dosage recommended for treatment of
ents with either mild or extensive CNS lesions, fur- extracerebral CMV infections (table II), is the stan-
ther investigations using quantitative PCR can help dard treatment for CMV-induced neurological com-
to distinguish forms with different severity.[104,105] plications. However, more potent regimens are clearly
Although there is considerably less experience in necessary in the majority of cases. A combination
this area, CSF PCR has also been shown to be help- of ganciclovir and foscarnet, each used at standard
ful in other immunocompromised patients.[106] doses, has been proposed for the treatment of se-
Although no information from controlled studies vere CMV infections of the CNS, and is strongly
is available, there is abundant clinical experience recommended in patients who have received pre-
with the use of ganciclovir and, to a lesser extent, vious anti-CMV therapy.[110,114,115] The 2 drugs have
foscarnet in HIV-infected patients. Neither is very a synergistic effect in vitro[116] and different resis-
effective in cases of CMV encephalitis, the occur- tance and toxicity profiles,[15,17] and their combi-
rence of which is actually common in patients under- nation has been shown to provide some benefit in
going maintenance treatment with these drugs.[107] patients with CNS CMV disease.[110] Along with
However, CSF CMV DNA titres often decrease fol- the clinical assessment, it is important to monitor
lowing ganciclovir therapy and may also become un- the local virological response to treatment, and this
detectable; in some cases these findings have been can be best accomplished by CSF quantitative nu-
associated with partial neurological improvement.[104] cleic acid amplification techniques. Studies of se-
A good clinical response has also been shown in 2 quential CSF specimens collected before and after
patients with CMV encephalitis following cidofo- treatment with ganciclovir, foscarnet or both have
vir, but further experience is clearly needed to de- shown that CMV replication is not adequately su-
fine the effectiveness of this drug.[108,109] ppressed in the CSF following 3 weeks of treat-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 107

ment.[117] Longer induction courses than those cur- strating specific intrathecal antibody production or
rently recommended would probably be required by PCR detection of EBV DNA in CSF.[124-126] How-
to clear the infection from the CSF, but in this re- ever, in line with the hypothesis of immunotoxic
gard, drug toxicity is expected to be an important damage to the CNS, EBV amplification from CSF
limiting factor. has failed in a number of cases.[123] Of note, EBV
Patients with CMV retinitis receiving HAART has also been found in the CSF of patients with
show prolonged responses to anti-CMV therapy[10] other herpesvirus CNS infections.[127] This finding
and, currently, maintenance anti-CMV therapy is suggests that EBV might be locally reactivated by
no longer recommended in HAART-treated patients other CNS infections;[126] however, the clinical sig-
in whom immunological conditions have signifi- nificance of such CSF coinfection, if any, is poorly
cantly improved.[115] If practicable, therapeutic ap- understood.
proaches combining anti-CMV and anti-HIV drugs The prognosis of mild EBV disease is generally
appear to be a reasonable option for the manage- good, but neurological sequelae can be observed in
ment of CMV complications of the CNS. Experience up to 40% of patients with meningoencephalitis,
with CMV encephalitis is limited in immunosup- and fatal cases have also been reported.[121,122] No
pressed patients other than those with HIV infec- treatment for EBV complications of the CNS has
tion. In these patients, CMV encephalitis is also yet been validated and treatment decisions are fur-
a severe, potentially life-threatening complication. ther hampered by an incomplete knowledge of the
Therefore, treatment with anti-CMV agents is strong- underlying pathogenic mechanisms. Both aciclo-
ly recommended. Nevertheless, the fact that their vir and corticosteroids have been used in the treat-
immunosuppression may be transitory and less pro- ment of CNS EBV diseases, although the benefit
nounced than in patients with AIDS suggests that of these approaches is uncertain.[128-131] The use of
they may respond better to anti-CMV agents.[106,118] aciclovir and corticosteroids, alone or in combina-
tion, has been found to offer no substantial clinical
6. Epstein-Barr Virus benefit in clinical trials involving patients with un-
complicated acute mononucleosis.[132,133] Further-
6.1 Immunocompetent Patients more, neurological complications have been repor-
ted in patients with acute mononucleosis receiving
Headache and CSF pleocytosis are not infrequ- corticosteroids.[134] In vitro, ganciclovir has been
ent in patients with uncomplicated acute mononu- shown to be more efficient than aciclovir in inhibit-
cleosis, which suggests possible CNS involvement ing EBV replication. Therefore, the use of this drug
at the time of primary EBV infection.[119] More se- deserves consideration in the treatment of poten-
vere CNS involvement has been shown in the form tially severe EBV-induced CNS complications.[135]
of meningoencephalitis, although cerebellitis and
myelitis have also been described.[120,121] The di- 6.2 Immunocompromised Patients
rect role of EBV replication in causing brain dam-
age is not obvious, and a number of the reported EBV encephalitis or meningitis are exceptional
cases of EBV-associated encephalopathies are con- in the setting of HIV infection. However, EBV is
sistent with an immune-mediated postinfectious me- constantly associated with HIV-related primary CNS
chanism.[122,123] EBV-induced neurological compli- lymphomas.[136] Although EBV detection in the CSF
cations are classically diagnosed by means of the or brain of patients with lymphomas may have diag-
serological demonstration of a recent primary in- nostic implications,[137] management of this neo-
fection, proven by the detection of serum IgG and plasm is beyond the scope of this article.
IgM against replicative EBV antigens, in the ab- In patients who have had a transplant, neurolog-
sence of antibodies to the EBV nuclear antigens. A ical involvement can be observed in the context of
more reliable diagnosis can be obtained by demon- EBV-related lymphoproliferative disorders. EBV

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
108 Cinque & Lazzarin

encephalitis or meningoencephalitis has also been ease, suggesting that this test should be used cau-
observed, and treatment with aciclovir or ganciclovir tiously for diagnostic purposes.[147] HHV-7 has spo-
has been followed by clinical improvement.[135,138] radically been found in the CSF of children with
febrile seizures.[151]
7. Human Herpesvirus 6 (HHV-6), HHV-7 There is no established treatment for HHV-6 in-
and HHV-8 fections of the CNS. Febrile seizures of childhood
are usually benign and antiviral agents are not em-
7.1 Immunocompetent and ployed. However, different considerations are re-
Immunocompromised Patients quired for HHV-6 encephalitis. There is increasing
evidence showing successful responses to ganci-
Febrile convulsions and other neurological symp- clovir or foscarnet in immunocompetent and bone
toms can occur in young children with exanthe- marrow transplant patients with encephalitis.[145-147]
ma subitum or unrecognised primary HHV-6 infec- On the basis of these preliminary observations, and
tion.[139,140] Furthermore, encephalitis presumably on the high susceptibility of A and B HHV-6 vari-
due to HHV-6 has recently been described in imm- ants to both ganciclovir and foscarnet,[153] the use
unocompetent adults[141,142] and cases of CNS dis- of these drugs at the doses employed in the treat-
ease resulting from HHV-6 are increasingly being ment of CMV infections (table II), is indicated in
recognised among bone marrow transplant recip- cases of HHV-6 encephalitis. These same drugs are
ients.[143-146] The virus does not seem to play a pro- also effective against HHV-7[153] but, in this regard,
minent role in inducing clinical neurological disease no clinical data have yet been reported.
in HIV-infected adults,[147] although CNS compli-
cations of primary HHV-6 infections may be fatal 8. Concluding Remarks and
in HIV-infected children.[148] Two distinct HHV-6 Future Prospects
variants exist: A and B. Variant B is associated with
uncomplicated primary HHV-6 infection and is also Progress towards the optimal management of her-
identified in the CSF or brain of children with neu- pesvirus infections of the CNS has continued since
rological complications of primary infections.[149] the dramatic decline in mortality and morbidity fol-
Both variants can be found in paediatric and adult lowing the introduction of aciclovir for HSE. New
immunocompetent patients with encephalitis, as well clinical manifestations have been defined, and mo-
as in immunocompromised patients.[144,147,149,150] lecular techniques have provided clinicians with an
HHV-7 infection of the CNS might be responsi- invaluable diagnostic tool to be used in combina-
ble for febrile seizures or other focal neurological tion with classical examinations. New compounds
signs in association with HHV-7 exanthema.[151] have been developed and mechanisms of action,
Currently, there is no strong evidence of an aetio- pharmacokinetics properties and patterns of viral
logical association of HHV-8 with any CNS dis- resistance further delineated.
ease. However, despite these optimistic remarks, much
In children with seizures, CNS involvement by remains to be done. A number of herpes diseases of
HHV-6 can be suggested by serological evidence the CNS are still poorly characterised, especially
of acute HHV-6 infection.[140,141] Furthermore, HHV- in terms of virus-host interactions, and diagnostic
6 DNA can be detected in the CSF by PCR, al- techniques, particularly those based on CSF molec-
though this finding is not specific.[152] HHV-6 has ular analysis, still suffer from a lack of standard-
also been identified, through the use of CSF PCR, isation. In cases of less frequently encountered dis-
in transplant recipients with encephalitis and in HIV- eases, even the real diagnostic reliability of CSF
infected patients. In these latter patients, however, PCR is poorly defined. Frequently, treatment reco-
HHV-6 detection in the CSF was not associated mmendations cannot be formulated because of lim-
with clinically significant HHV-6–induced CNS dis- ited clinical experience. Moreover, the current treat-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 109

ment strategies are unsatisfactory in a number of apy: a 2 year prospective study. Br J Ophthalmol 1999; 83:
652-5
instances. 11. Wagstaff AJ, Faulds D, Goa KL. Aciclovir. A reappraisal of its
The first steps towards the evaluation of new antiviral activity, pharmacokinetic properties and therapeutic
efficacy. Drugs 1994; 47: 153-205
treatment strategies will involve quality control stu-
12. Whitley RJ, Blum MR, Barton N, et al. Pharmacokinetics of
dies of the current diagnostic procedures and stan- acyclovir in humans following intravenous administration.
dardisation of the diagnostic criteria for neurolog- Am J Med 1982; 73 Suppl.: 165-71
13. Perry CM, Faulds D. Valaciclovir. A review of its antiviral ac-
ical complications of herpesvirus infections. New tivity, pharmacokinetics properties and therapeutic efficacy
aciclovir regimens, as well as newer antiviral agents in herpesvirus infections. Drugs 1996; 52: 754-72
or drug combinations, clearly warrant evaluation 14. Perry CM, Wagstaff AJ. Famciclovir. A review of its pharma-
cological properties and therapeutical efficacy in herpesvirus
in the context of controlled trials. Given the low infections. Drugs 1995; 50: 396-415
frequency of herpesvirus CNS infections, clinical 15. Markham A, Faulds D. Ganciclovir. An update of its therapeutic
use in cytomegalovirus infection. Drugs 1994; 48: 455-84
assessments of antiviral regimens can only be done
16. Fletcher C, Sawchuck R, Chinnok B, et al. Human pharmaco-
in the context of large multicentre studies. These kinetics of the antiviral drug DHPG. Clin Pharmacol Ther
will not only require close collaboration among phy- 1986; 40: 281-6
17. Chrisp P, Clissold SP. Foscarnet. A review of its antiviral activity,
sicians, virologists and pathologists, but also sub- pharmacokinetic properties and therapeutic use in immuno-
stantial support from health programmes and drug compromised patients with cytomegalovirus retinitis. Drugs
companies. 1991; 41: 104-29
18. Hengge UR, Brockmeyer NH, Malessa R, et al. Foscarnet pen-
etrates the blood-brain barrier: rationale for therapy of cyto-
Acknowledgements megalovirus encephalitis. Antimicrob Agents Chemother 1993;
37: 1010-4
We wish to thank Lucia Angelini and the members of the 19. Lea AP, Bryson HM. Cidofovir. Drugs 1996; 52: 225-30
20. De Wit S, Snoeck R, Rossi C, et al. Treatment of progressive
European Union Concerted Action on Viral Encephalitis and
multifocal leukoencephalopathy with cidofovir in an AIDS
Meningitis for helpful advice. patient [abstract P106]. Third International Congress on Drug
Therapy in HIV Infection: 1996 Nov 3-7; Birmingham, UK
21. Skoldenberg B, Forsgren M, Alestig K, et al. Acyclovir versus
References vidarabine in herpes simplex encephalitis. Randomised mul-
1. Esiri MM. Herpes simplex encephalitis. An immunohistologi-
ticentre study in consecutive Swedish patients. Lancet 1984;
cal study of the distribution of viral antigen within the brain.
II: 707-11
J Neurol Sci 1982; 54: 209-26
22. Whitley RJ, Alford CA, Hirsch MS, et al. Vidarabine versus
2. Price R, Chernik NL, Horta-Babosa L, et al. Herpes simplex
acyclovir therapy in herpes simplex encephalitis. N Engl J
encephalitis in an anergic patient. Am J Med 1973; 54: 222-8
Med 1986; 314: 144-9
3. Johnson RT. Acute encephalitis. Clin Infect Dis 1996; 23: 219-26
23. Field AK, Biron KK. ‘The end of innocence’ revisited: resis-
4. Koenig H, Rabinowitz SG, Day E, et al. Post-infectious ence-
tance of herpesviruses to antiviral drugs. Clin Microbiol Rev
phalomyelitis after successful treatment of herpes simplex
1994; 7: 1-13
encephalitis with adenine-arabinoside: ultrastructural obser-
vations. N Engl J Med 1979; 300: 1089-93 24. Pasternak JF, De Vivo DC, Prensky AL. Steroid-responsive en-
5. Puchhammer-Stöckl E, Popow-Kraupp T, Heinz FX, et al. De- cephalomyelitis in childhood. Neurology 1980; 30: 481-6
tection of varicella-zoster virus DNA by polymerase chain 25. Hawley RJ. Early high-dose methylprednisolone in acute dis-
reaction in the cerebrospinal fluid from patients suffering from seminated encephalomyelitis. Neurology 1998; 51: 644-5
neurological complications associated with chickenpox or her- 26. Hahn JS, Siegler DJ, Enzmann D. Intravenous gammaglobulin
pes zoster. J Clin Microbiol 1991; 29: 1513-6 therapy in recurrent acute disseminated encephalomyelitis.
6. Tyler KL. Polymerase chain reaction and the diagnosis of viral Neurology 1996; 46: 1173-4
central nervous system diseases. Ann Neurol 1994; 6: 809-11 27. Daaboul Y, Vern BA, Blend MJ. Brain SPECT imaging and
7. Cinque P, Scarpellini P, Vago L, et al. Diagnosis of central nerv- treatment with IVIg in acute post-infectious cerebellar ataxia:
ous system complications in HIV-infected patients: cerebro- case report. Neurol Res 1998; 20: 85-8
spinal fluid analysis by the polymerase chain reaction. AIDS 28. Assa A, Watemberg N, Bujanover Y, et al. Demyelinative brain-
1997; 11: 1-17 stem encephalitis responsive to intravenous immunoglobulin
8. Gross PA, Barrett TL, Dellinger EP, et al. Purpose of quality therapy. Pediatrics 1999; 104: 301-4
standards for infectious diseases. Infectious Diseases Society 29. Kanter DS, Horensky D, Sperling RA, et al. Plasmapheresis in
of America. Clin Infect Dis 1994; 18 (3): 421 fulminant acute disseminated encephalomyelitis. Neurology
9. Palella Jr FJ, Delaney KM, Moorman AC, et al. Declining mor- 1995; 45: 824-7
bidity and mortality among patients with advanced human 30. Balestri P, Grosso S, Acquaviva A, et al. Plasmapheresis in a
immunodeficiency virus infection. HIV Outpatient Study In- child affected by acute disseminated encephalomyelitis. Brain
vestigators. N Engl J Med 1998; 338: 853-60 Dev 2000; 22: 123-6
10. Mitchell SM, Membrey WL, Youle MS, et al. Cytomegalovirus 31. Thompson KA, Blessing WW, Wesselingh SL. Herpes simplex
retinitis after the initiation of highly active antiretroviral ther- replication and dissemination is not increased by corticoste-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
110 Cinque & Lazzarin

roids treatment in a rat model of focal herpes encephalitis. J 50. Dennett C, Klapper PE, Cleator GM. Polymerase chain reaction
Neurovirol 2000; 6: 25-32 in the investigation of ‘relapse’ following herpes simplex en-
32. Bhabha SK, Bharucha NE, Bharucha EP. Viral infections. In: cephalitis. J Med Virol 1996; 48: 129-32
Bradley WG, Daroff RB, Ferrichel GM, et al., editors. Neuro- 51. Ito Y, Kimura H, Yabuta Y, et al. Exacerbation of herpes simplex
logy in clinical practice. 2nd ed. Boston (MA): Butterworth- encephalitis after successful treatment with acyclovir. Clin
Heineman, 1996: 1259-75 Infect Dis 2000; 30: 185-7
33. Wald SL. Disorders of cerebrospinal fluid circulation and brain 52. Rozenberg F, Lebon P. Amplification and characterization of
edema. In: Bradley WG, Daroff RB, Ferrichel GM, et al., herpesvirus DNA in cerebrospinal fluid from patients with
editors. Neurology in clinical practice. 2nd ed. Boston (MA): acute encephalitis. J Clin Microbiol 1991; 29: 2412-7
Butterworth-Heineman, 1996: 1431-58 53. Tebas P, Nease RF, Storch GA. Use of the polymerase chain
34. Whitley RJ. Herpes simplex virus. In: Scheld WM, Whitley RJ, reaction in the diagnosis of herpes simplex encephalitis: a
Durack DT, editors. Infections of the central nervous system. decision analysis model. Am J Med 1998; 105: 287-95
2nd ed. New York (NY): Raven Press, 1997: 73-90 54. Domingues RB, Lakeman FD, Mayo MS, et al. Application of
35. Domingues RB, Tsanaclis AM, Pannuti CS, et al. Evaluation of competitive PCR to cerebrospinal fluid samples from patients
the range of clinical presentations of herpes simplex enceph- with herpes simplex encephalitis. J Clin Microbiol 1998; 36:
alitis by using polymerase chain reaction assay of cerebrospi- 2229-34
nal fluid samples. Clin Infect Dis 1997; 25: 86-91 55. Bergstrom T, Vahlne A, Alestig K, et al. Primary and recurrent
36. Fodor PA, Levin MJ, Weinberg A, et al. Atypical herpes simplex herpes simplex virus type 2-induced meningitis. J Infect Dis
virus encephalitis diagnosed by PCR amplification of viral 1990; 162: 322-30
DNA from CSF. Neurology 1998; 51: 554-9 56. Mollaret P. La Méningite endothélio-leucocytaire multirécur-
37. Nakajima H, Furutama D, Kimura F, et al. Herpes simplex virus rent benigne: syndrome nouveau or maladie nouvelle? Rev
myelitis: clinical manifestations and diagnosis by the poly- Neurol (Paris); 1944; 76: 57-76
merase chain reaction method. Eur Neurol 1998; 39: 163-7
57. Tedder DG, Ashley R, Tyler KL, et al. Herpes simplex virus
38. Cinque P, Cleator GM, Weber T, et al., and The European Union infection as a cause of benign recurrent lymphocytic menin-
Concerted Action on Viral Encephalitis and Meningitis Group. gitis. Ann Intern Med 1994; 121: 334-8
The role of laboratory investigation in the diagnosis and man-
58. Schlesinger Y, Tebas P, Gaudreault-Keener M, et al. Herpes
agement of patients with suspected herpes simplex encepha-
simplex virus type 2 meningitis in the absence of genital le-
litis. A consensus report. J Neurol Neurosurg Psychiatry 1996;
sions: improved recognition with use of the polymerase chain
61: 339-45
reaction. Clin Infect Dis 1995; 20: 842-8
39. Aurelius E, Johansson B, Sköldenberg B, et al. Rapid diagnosis
59. Read SJ, Kurtz JB. Laboratory diagnosis of common viral in-
of herpes simplex encephalitis by nested polymerase chain
fections of the central nervous system by using a single multi-
reaction assay of cerebrospinal fluid. Lancet 1991; 337: 189-92
plex PCR screening assay. J Clin Microbiol 1999; 37: 1352-5
40. Lakeman FD, Koga J, Whitley RJ. Detection of antigen to her-
pes simplex virus in cerebrospinal fluid from patients with 60. Bergstrom T, Alestig K. Treatment of primary and recurrent
herpes simplex encephalitis. J Infect Dis 1987; 155 (6): 1172-8 herpes simplex virus type 2 induced meningitis with acyclo-
vir. Scand J Infect Dis 1990; 22: 239-40
41. Linde A, Klapper PE, Monteyne P, et al. Specific diagnostic
methods for herpesvirus infections of the central nervous sys- 61. Haanpaa M, Dastidar P, Weinberg A, et al. CSF and MRI find-
tem: a consensus review by the European Union Concerted ings in patients with acute herpes zoster. Neurology 1998; 51:
Action on Virus Meningitis and Encephalitis. Clin Diagn Virol 1405-11
1997; 8: 83-104 62. Straus SE, Takiff HE, Seidlin M, et al. Suppression of frequently
42. Nolan DC, Lauter CB, Lerner AM. Idoxuridine in herpes sim- recurring genital herpes. A placebo-controlled double-blind
plex virus (type 1) encephalitis. Experience with 29 cases in trial of oral acyclovir. N Engl J Med 1984; 310: 1545-50
Michigan, 1966 to 1971. Ann Intern Med 1973; 78: 243-6 63. Douglas JM, Critchlow C, Benedetti J, et al. A double-blind
43. Hryniuk W, Foerster J, Shojania M, et al. Cytarabine for herpes- study of oral acyclovir for suppression of recurrences of gen-
virus infections. JAMA 1972; 219: 715-8 ital herpes simplex virus infection. N Engl J Med 1984; 310:
44. Aronson MD, Phillips CF, Gump DW, et al. Vidarabine therapy 1551-6
for severe herpesvirus infections. An unusual syndrome of 64. Reitano M, Tyring S, Lang W, et al. Valaciclovir for the sup-
chronic varicella and transient immunologic deficiency. JAMA pression of recurrent genital herpes simplex virus infection: a
1976; 235: 1339-42 large-scale dose range-finding study. International Valaciclo-
45. Boston Interhospital Virus Study Group and the NIAID Spon- vir HSV Study Group. J Infect Dis 1998; 178: 603-10
sored Cooperative Antiviral Clinical Study. Failure of high 65. Diaz-Mitoma F, Sibbald RG, Shafran SD, et al. Oral famciclovir
dose 5-iodo-2’-deoxyuridine in the therapy of herpes simplex for the suppression of recurrent genital herpes: a randomized
virus encephalitis. Evidence of unacceptable toxicity. N Engl controlled trial. Collaborative Famciclovir Genital Herpes Re-
J Med 1975; 292: 599-603 search Group. JAMA 1998; 280: 887-92
46. Whitley RJ, Soong SJ, Dolin R, et al. Adenosine arabinoside 66. Chretien F, Belec L, Hilton DA, et al. Herpes simplex virus type
therapy of biopsy proven herpes simplex encephalitis. N Engl 1 encephalitis in acquired immunodeficiency syndrome. Neu-
J Med 1977; 297: 289-94 ropathol Appl Neurobiol 1996; 22: 394-404
47. Barthez MA, Billard C, Santini JJ, et al. Relapse of herpes sim- 67. Vago L, Sala E, Nebuloni M, et al. Coinfection of the central
plex encephalitis. Neuropediatrics 1987; 18: 3-7 nervous system (CNS) by cytomegalovirus and herpes sim-
48. VanLandingham KE, Marsteller HB, Ross GW, et al. Relapse plex virus type 1 or 2 in AIDS patients: autopsy study on 82
of herpes simplex encephalitis after conventional acyclovir cases by immunocytochemistry and polymerase chain reac-
therapy. JAMA 1988; 259: 1051-3 tion. Acta Neuropathol (Berl) 1996; 92: 404-8
49. Kimura H, Aso K, Kuzushima K, et al. Relapse of herpes sim- 68. Schiff D, Rosenblum MK. Herpes simplex encephalitis (HSE)
plex encephalitis in children. Pediatrics 1992; 89: 891-4 and the immunocompromised: a clinical and autopsy study of

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 111

HSE in the settings of cancer and human immunodeficiency in an acquired immunodeficiency syndrome patient. J Med
virus-type 1 infection. Hum Pathol 1998; 29: 215-22 Virol 1994; 42: 338-47
69. Cinque P, Vago L, Marenzi R, et al. Herpes simplex viruses 88. Beutner KR, Friedman DJ, Forszpaniak C, et al. Valaciclovir
infections of the nervous system in human immunodeficiency compared with aciclovir for improved therapy for herpes zos-
virus-infected patients: clinical management by polymerase ter in immunocompetent adults. Antimicrob Agents Chemo-
chain reaction analysis of cerebrospinal fluid. Clin Infect Dis ther 1995; 39: 1546-53
1998; 27: 303-9 89. Tyring S, Barbarash RA, Nahlik JE, et al. Famciclovir for the
70. Gateley A, Gander RM, Johnson PC, et al. Herpes simplex virus treatment of acute herpes zoster: effects on acute disease and
type 2 meningoencephalitis resistant to acyclovir in a patient postherpetic neuralgia. A randomized, double-blind, placebo-
with AIDS. J Infect Dis 1990; 161: 711-5 controlled trial. Collaborative Famciclovir Herpes Zoster Study
71. Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ, et al. Group. Ann Intern Med 1995; 123: 89-96
Neurologic complications of the reactivation of varicella-zos- 90. Bodsworth NJ, Boag F, Burdge D, et al. Evaluation of sorivudine
ter virus. N Engl J Med 2000; 342: 635-45 (BV-araU) versus acyclovir in the treatment of acute localized
72. Amlie-Lefond C, Kleinschmidt-DeMasters BK, Mahalingam herpes zoster in human immunodeficiency virus-infected ad-
R, et al. The vasculopathy of varicella-zoster virus encepha- ults. The Multinational Sorivudine Study Group. J Infect Dis
litis. Ann Neurol 1995; 37: 784-90 1997; 176: 103-11
73. Echevarria JM, Casas I, Martinez-Martin P. Infections of the 91. Gnann Jr JW, Crumpacker CS, Lalezari JP, e al. Sorivudine
nervous system caused by varicella-zoster virus: a review. versus acyclovir for treatment of dermatomal herpes zoster in
Intervirology 1997; 40: 72-84 human immunodeficiency virus-infected patients: results from
74. Echevarria JM, Casas I, Tenorio A, et al. Detection of varicella- a randomized, controlled clinical trial. Collaborative Antivi-
zoster virus-specific DNA sequences in cerebrospinal fluid of ral Study Group/AIDS Clinical Trials Group, Herpes Zoster
patients with acute aseptic meningitis and no cutaneous le- Study Group. Antimicrob Agents Chemother 1998; 42: 1139-45
sions. J Med Virol 1994; 43: 331-5 92. Okuda H, Ogura K, Kato A, et al. A possible mechanism of
75. Echevarria JM, Martinez-Martin P, Tellez A, et al. Aseptic men- eighteen patient deaths caused by interactions of sorivudine,
ingitis due to varicella-zoster virus: serum antibody levels and a new antiviral drug, with oral 5-fluorouracil prodrugs. J
local synthesis of specific IgG, IgM, and IgA. J Infect Dis Pharmacol Exp Ther 1998; 287: 791-9
1987; 155: 959-67 93. Bale Jr JF. Human cytomegalovirus infection and disorders of
76. Johns DR, Gress DR. Rapid response to acyclovir in herpes the nervous system. Arch Neurol 1984; 41: 310-20
zoster-associated encephalitis. Am J Med 1987; 82: 560-2 94. Studahl M, Ricksten A, Sandberg T, et al. Cytomegalovirus
77. Peterslund NA. Herpes zoster associated encephalitis: clinical infection of the CNS in non-compromised patients. Acta Neu-
findings and acyclovir treatment. Scand J Infect Dis 1988; 20: rol Scand 1994; 89: 451-7
583-92
95. Arribas JR, Storch GA, Clifford DB, et al. Cytomegalovirus
78. Gray F, Belec L, Lescs MC, et al. Varicella-zoster virus infec- encephalitis. Ann Intern Med 1996; 125: 577-87
tion of the central nervous system in the acquired immune
96. Prosch S, Schielke E, Reip A, et al. Cytomegalovirus (HCMV)
deficiency syndrome. Brain 1994; 117: 987-99
encephalitis in an immunocompetent young person and diag-
79. Cinque P, Bossolasco S, Vago L, et al. Varicella-zoster virus
nostic reliability of HCMV DNA PCR using cerebrospinal
(VZV) DNA in cerebrospinal fluid of patients infected with
fluid of nonimmunosuppressed patients. Clin Microbiol 1998;
human immunodeficiency virus: VZV disease of the central
36: 3636-40
nervous system or subclinical reactivation of VZV infection?
Clin Infect Dis 1997; 25: 634-9 97. Pantoni L, Inzitari D, Colao MG, et al. Cytomegalovirus en-
cephalitis in a non-immunocompromised patient: CSF diag-
80. Peterson LR, Ferguson RM. Fatal central nervous system infec-
nosis by in situ hybridization cells. Acta Neurol Scand 1991;
tion with varicella-zoster virus in renal transplant recipients.
84: 56-8
Transplantation 1984; 37: 366-8
81. Arvin AM. Varicella-zoster virus. Clin Microbiol Rev 1996; 9: 98. McCutchan JA. Cytomegalovirus infections of the nervous sys-
361-81 tem in patients with AIDS. Clin Infect Dis 1995; 20: 747-54
82. Burke DG, Kalayjian RC, Vann VR, et al. Polymerase chain 99. Anders HJ, Goebel FD. Cytomegalovirus polyradiculopathy in
reaction detection and clinical significance of varicella-zoster patients with AIDS. Clin Infect Dis 1998; 27: 345-52
virus in cerebrospinal fluid from human immunodeficiency 100. Cinque P, Vago L, Brytting M, et al. Cytomegalovirus infection
virus-infected patients. J Infect Dis 1997; 176: 1080-4 of the central nervous system in patients with AIDS: diagnosis
83. Iten A, Chatelard P, Vuadens P, et al. Impact of cerebrospinal by DNA amplification from the cerebrospinal fluid. J Infect
fluid PCR on the management of HIV-infected patients with Dis 1992; 166: 1408-11
varicella-zoster virus infection of the central nervous system. 101. Wolf DG, Spector SA. Diagnosis of human cytomegalovirus
J Neurovirol 1999; 5: 172-80 central nervous system disease in AIDS patients by DNA am-
84. Poscher ME. Successful treatment of varicella zoster virus me- plification from cerebrospinal fluid. J Infect Dis 1992; 166:
ningoencephalitis in patients with AIDS: report of four cases 1412-5
and review. AIDS 1994; 8: 1115-7 102. Gozlan J, Salord JM, Roullet E, et al. Rapid detection of cyto-
85. Lionnet F, Pulik M, Genet P, et al. Myelitis due to varicella-zos- megalovirus DNA in cerebrospinal fluid of AIDS patients
ter virus in two patients with AIDS: successful treatment with with neurologic disorders. J Infect Dis 1992; 166: 1416-21
acyclovir. Clin Infect Dis 1996; 22: 138-40 103. Fox DJ, Brink NS, Zuckermann MA, et al. Detection of herpes-
86. de Silva SM, Mark AS, Gilden DH, et al. Zoster myelitis: im- virus DNA by nested PCR in CSF of HIV-infected persons
provement with antiviral therapy in two cases. Neurology with neurological disease: a prospective evaluation. J Infect
1996; 47: 929-31 Dis 1995; 172: 1087-90
87. Snoeck R, Gerard M, Sadzot-Delvaux C, et al. Meningoradi- 104. Cinque P, Baldanti F, Vago L, et al. Ganciclovir therapy for
culoneuritis due to acyclovir-resistant varicella-zoster virus cytomegalovirus (CMV) infection of the central nervous sys-

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
112 Cinque & Lazzarin

tem in AIDS patients: monitoring by CMV DNA detection in 122. Schellinger PD, Sommer C, Leithauser F, et al. Epstein-Barr
cerebrospinal fluid. J Infect Dis 1995; 171: 1603-6 virus meningoencephalitis with a lymphoma-like response in
105. Arribas JR, Clifford DB, Fichtenbaum CJ, et al. Level of cyto- an immunocompetent host. Ann Neurol 1999; 45: 659-62
megalovirus (CMV) DNA in cerebrospinal fluid of subjects 123. Imai S, Usui N, Sugiura M, et al. Epstein-Barr virus genomic
with AIDS and CMV infection of the central nervous system. sequences and specific antibodies in cerebrospinal fluid in
J Infect Dis 1995; 172: 527-31 children with neurologic complications of acute and reacti-
106. Bamborschke S, Wullen T, Huber M, et al. Early diagnosis and vated EBV infections. J Med Virol 1993; 40: 278-84
successful treatment of acute cytomegalovirus encephalitis in 124. Landgren M, Kyllerman M, Bergstrom T, et al. Diagnosis of
a renal transplant recipient. J Neurol 1992; 239: 205-8 Epstein-Barr virus-induced central nervous system infections
107. Berman SM, Kim RC. The development of cytomegalovirus by DNA amplification from cerebrospinal fluid. Ann Neurol
encephalitis in AIDS patients receiving ganciclovir. Am J Med 1994; 35: 631-5
1994; 96 (5): 415-9 125. Kaji M, Shoji H. Detection of Epstein-Barr virus genome in
108. Sadler M, Morris-Jones S, Nelson M, et al. Successful treatment peripheral leucocytes and CSF by the polymerase chain reac-
of cytomegalovirus encephalitis in an AIDS patient using ci- tion in two patients with Epstein-Barr virus related to aseptic
dofovir. AIDS 1997; 11: 1293-4 meningitis [letter]. J Neurol Neurosurg Psychiatry 1995; 59
109. Blick G, Garton T, Hopkins U, et al. Successful use of cidofovir (1): 99
in treating AIDS-related cytomegalovirus retinitis, encepha- 126. Tselis A, Duman R, Storch GA, et al. Epstein-Barr virus enceph-
litis, and esophagitis. J Acquir Immune Defic Syndr Hum alomyelitis diagnosed by polymerase chain reaction: detec-
Retrovirol 1997; 15: 84-5 tion of the genome in the CSF. Neurology 1997; 48: 1351-5
110. Anduze-Faris BM, Fillet AM, Gozlan J, et al. Induction and 127. Studahl M, Hagberg L, Rekabdar E, et al. Hematogenously spread
maintenance therapy of cytomegalovirus central nervous sys- herpesviruses are detected as frequently as neuronally spread
tem infection in HIV-infected patients. AIDS 2000; 14: 517-24 herpesviruses in cerebrospinal fluid by polymerase chain re-
111. Cohen BA. Prognosis and response to therapy of cytomegalo- action assay. Clin Infect Dis 1999; 29: 216-8
virus encephalitis and meningomyelitis in AIDS. Neurology 128. Frenkel EP. Shiver CB, Berg P, et al. Meningoencephalitis in
1996; 46: 444-50 infectious mononucleosis: report of a case treated with corti-
112. Smith IL, Shinkai M, Freeman WR, et al. Polyradiculopathy sone. JAMA 1956; 162: 885-6
associated with ganciclovir-resistant cytomegalovirus in an 129. Mizutani T, Murashima A, Shiozawa R, et al. Unusual neuro-
AIDS patient: phenotypic and genotypic characterisation of logic manifestations associated with Epstein-Barr virus infec-
sequential virus isolates. J Infect Dis 1996; 173: 1481-4 tion. Intern Med 1993; 32: 36-8
113. Wolf DG, Lee DJ, Spector SA. Detection of human cytomega- 130. Paskavitz JF, Anderson CA, Filley CM, et al. Acute arcuate fiber
lovirus mutations associated with ganciclovir resistance in demyelinating encephalopathy following Epstein-Barr virus
cerebrospinal fluid of AIDS patients with central nervous sys- infection. Ann Neurol 1995; 38: 127-31
tem disease. Antimicrob Agents Chemother 1995; 39: 2552-4 131. Corssmit EP, Leverstein-van Hall MA, Portegies P, et al. Severe
114. Cinque P, Cleator GM, Weber T, et al. Diagnosis and clinical neurological complications in association with Epstein-Barr
management of neurological disorders caused by cytomega- virus infection. J Neurovirol 1997; 3 (6): 460-4
lovirus in AIDS patients. European Union Concerted Action 132. Andersson J, Britton S, Ernberg I, et al. Effect of acyclovir on
on Virus Meningitis and Encephalitis. J Neurovirol 1998; 4: infectious mononucleosis: a double-blind, placebo-controlled
120-32 study. J Infect Dis 1986; 153: 283-90
115. Whitley RJ, Jacobson MA, Friedberg DN, et al. Guidelines for 133. Tynell E, Aurelius E, Brandell A, et al. Acyclovir and pred-
the treatment of cytomegalovirus diseases in patients with nisolone treatment of acute infectious mononucleosis: a mul-
AIDS in the era of potent antiretroviral therapy: recommen- ticenter, double-blind, placebo-controlled study. J Infect Dis
dations of an international panel. International AIDS Society 1996; 174: 324-31
– USA. Arch Intern Med 1998; 158: 957-69 134. Waldo RT. Neurologic complications of infectious mononucle-
116. Manischewitz JF, Quinnan GV, Lane HC, et al. Synergistic ef- osis after steroid therapy. South Med J 1981; 74: 1159-60
fect of ganciclovir and foscarnet on cytomegalovirus replica- 135. Dellemijn PL, Brandenburg A, Niesters HG, et al. Successful
tion in vitro. Antimicrob Agents Chemother 1990; 34: 373-5 treatment with ganciclovir of presumed Epstein-Barr meningo-
117. Cinque P, Pisa E, Racca S, et al. Efficacy of different antiviral encephalitis following bone marrow transplant. Bone Marrow
treatments in cytomegalovirus neurological compliocations: Transplant 1995; 16: 311-2
evaluation by quantitative polymerase chain reaction on cere- 136. MacMahon EM, Glass JD, Hayward SD, et al. Epstein-Barr
brospinal fluid [abstract OP3.3]. 3rd International Congress virus in AIDS-related primary central nervous system lym-
on Drug Therapy in HIV Infection: 1996 Nov 3-7; Birming- phoma. Lancet 1991; 338: 969-73
ham, UK 137. Cinque P, Brytting M, Vago L, et al. Epstein-Barr virus DNA in
118. Van Droogenbroeck J, De Ceuninck M, Snoeck HW, et al. Suc- cerebrospinal fluid of patients with AIDS-related primary lym-
cessful treatment of cytomegalovirus encephalitis in a patient phoma of the central nervous system. Lancet 1993; 342: 398-401
with Hodgkin’s disease in remission. Ann Hematol 1998; 76: 138. Davis K, Hinrichs S, Fidler J, et al. Post-transplant Epstein-Barr
179-81 virus-associated meningoencephalitis and lymphoid intersti-
119. Gautier-Smith PC. Neurological complications of glandular fe- tial pneumonitis. Bone Marrow Transplant 1999; 24: 443-4
ver (infectious mononucleosis). Brain 1965; 88: 323-34 139. Suga S, Yoshikawa T, Asano Y, et al. Clinical and virological
120. Grose C, Henle W, Henle G, et al. Primary Epstein-Barr-virus analyses of 21 infants with exanthem subitum (roseola in-
infections in acute neurologic diseases. N Engl J Med 1975; fantum) and central nervous system complications. Ann Neu-
292: 392-5 rol 1993; 33: 597-603
121. Domachowske JB, Cunningham CK, Cummings DL, et al. Acute 140. Kondo K, Nagafiji H, Hata A, et al. Association of human her-
manifestations and neurologic sequelae of Epstein-Barr virus pesvirus 6 infection of the central nervous system with recur-
encephalitis in children. Pediatr Infect Dis J 1996; 15: 871-5 rence of febrile convulsions. J Infect Dis 1993; 167: 1197-200

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)
Herpesvirus Infections of the CNS 113

141. McCullers JA, Lakeman FD, Whitley RJ. Human herpesvirus 148. Knox KK, Harrington DP, Carrigan DR. Fulminant human her-
6 is associated with focal encephalitis. Clin Infect Dis 1995; pesvirus six encephalitis in a human immunodeficiency vi-
21: 571-6 rus-infected infant. J Med Virol 1995; 45: 88-92
142. Kimberlin DW, Whitley RJ. Human herpesvirus-6: neurologi- 149. Braun DK, Dominguez G, Pellet PE. Human herpesvirus 6. Clin
cal implications of a newly described herpesvirus. J Neu- Microbiol Rev 1997; 10: 521-67
rovirol 1998; 4: 474-85 150. Hall CB, Caserta MT, Schnabel KC, et al. Persistence of human
143. Drobyski W, Knox KK, Majewski D, et al. Brief report: fatal herpesvirus 6 according to site and variant: possible greater
encephalitis due to variant B human herpesvirus-6 infection neurotropism of variant A. Clin Infect Dis 1998; 26: 132-7
in a bone marrow-transplant recipient. N Engl J Med 1994; 151. Torigoe S, Koide W, Yamada M, et al. Human herpesvirus-7
330: 356-60 infection associated with central nervous system manifesta-
144. Wang F-Z, Linde A, Hägglund H, et al. Human herpesvirus-6 tions. J Pediatr 1996; 129 (2): 301-5
DNA in cerebrospinal fluid from allogeneic bone marrow trans- 152. Caserta MT, Hall CB, Schnabel K, et al. Neuroinvasion and
plant patients: does it have a clinical significance? Clin Infect persistence of human herpesvirus 6 in children. J Infect Dis
Dis 1999; 28: 562-8 1994; 170: 1586-9
145. Mookerjee BP, Vogelsang G. Human herpesvirus-6 encephalitis 153. Yoshida M, Yamada M, Tsukazaki T, et al. Comparison of anti-
after bone-marrow transplantation: successful treatment with viral compounds against human herpesvirus 6 and 7. Antiviral
Res 1998; 40: 73-84
ganciclovir. Bone Marrow Transplant 1997; 20: 905-6
146. Bethge W, Beck R, Jahn G, et al. Successful treatment of human
herpesvirus-6 encephalitis after bone marrow transplantation.
Bone Marrow Transplant 1999; 24: 1245-8
147. Bossolasco S, Marenzi R, Dahl H, et al. Human herpesvirus 6 Correspondence and offprints: Dr Paola Cinque, Division of
in cerebrospinal fluid of patients infected with HIV: frequ- Infectious Diseases, San Raffaele Hospital, Via Stamira
ency and clinical significance. J Neurol Neurosurg Psychiatry d’Ancona, 20, 20127 Milan, Italy.
1999’ 67 (6): 789-92 E-mail: cinque.paola@hsr.it

 Adis International Limited. All rights reserved. CNS Drugs 2000 Aug; 14 (2)

You might also like