111
IMPROVING THE MANAGEMENT OF VARICELLA, HERPES ZOSTER AND ZOSTER- ASSOCIATED PAIN Recommendations from the IHMF Management Strategies Workshop Editors: Dr R Johnson Dr D Patrick management strategies in herpes

IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Embed Size (px)

Citation preview

Page 1: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

IMPROVING THEMANAGEMENTOF VARICELLA,

HERPES ZOSTER AND ZOSTER-

ASSOCIATED PAIN

Recommendations from the IHMF Management Strategies Workshop

Editors: Dr R JohnsonDr D Patrick

management s trategies in herpes

Page 2: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

1

CONTENTS

Introduction 2

Recommendation Categories 3

Workshop Participants 4

Chapter 1Advances in the Pathogenesis, Molecular Biology and Immunology

of Varicella Zoster Virus 5

Chapter 2Introduction to Varicella 10

Chapter 3Varicella Vaccination 21

Chapter 4Management of Varicella in the Immunocompetent Host 33

Chapter 5Introduction to Herpes Zoster and Future Directions 46

Chapter 6Management of Herpes Zoster in the Immunocompetent Host 58

Chapter 7Varicella Zoster Infection of the Eye 69

Chapter 8Management of Varicella Zoster Virus Infections in the Immunocompromised Host 77

Chapter 9Managing Established Pain in Herpes Zoster 100

Page 3: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

2

INTRODUCTION

The International Herpes Management Forum (IHMF) was established to improve the

awareness, understanding, counselling and management of infections caused by

herpesviruses. Steered by Professor Richard Whitley, Professor Lawrence Corey, Professor

Paul Griffiths, Dr Jean-Elie Malkin, Dr Antonio Volpi, Dr David Patrick, Dr Robert Johnson,

Dr Lawrence Stanberry and Dr Anders Strand, the IHMF involves international opinion

leaders in all aspects of the management of herpesvirus infections.

A Management Strategies workshop was held on 6–7 March 2001 to discuss the

management of varicella zoster virus (VZV) infections in immunocompetent and

immunocompromised individuals. At the workshop, draft recommendations were

formulated on:

J Pre-exposure varicella vaccination

J Management of varicella and herpes zoster in immunocompromised individuals

J Management of unusual manifestations of varicella and herpes zoster in

immunocompetent and immunocompromised individuals

J Management of herpes zoster-related eye disease

J Application of new technologies and models to understanding the pathogenesis of

VZV infection.

This monograph reviews the available information on these aspects of the management of

VZV infections.

The editors would like to thank all the participants in the Management Strategies

Workshop and those who contributed to the ratification of the guidelines on the IHMF

website (www.ihmf.org).

Page 4: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

RECOMMENDATION CATEGORIES

Category 1 Consistent evidence from controlled clinical trials. For example, for an antiviral, this would

include results from at least one well-designed, randomized, controlled clinical trial, and,

in the case of laboratory studies, consistent evidence from comparative studies.

Category 2Evidence from at least one well-designed clinical trial without randomization, from cohort

or case-controlled analytical studies (preferably from more than one centre), or from

multiple time-series studies or dramatic results from uncontrolled experiments.

Category 3Evidence from opinions of respected authorities based on clinical experience, descriptive

studies or reports of expert committees.

Research NeedArea in which research is warranted.

3

Page 5: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

4

WORKSHOP PARTICIPANTS

ChairsDr R Johnson Consultant Anaesthetist, Bristol Royal Infirmary, Department of

Anaesthesia, Level 7, Marlborough Street, Bristol, BS2 8HW, UK

Dr D Patrick Associate Professor, Epidemiology & Infectious Diseases,University of British Colombia, Centre for Disease Control, 655West 12 Avenue, Vancouver, British Colombia, V5Z 4R4, Canada

ParticipantsProfessor HH Balfour Professor of Laboratory Medicine & Pathology, Professor of

Pediatrics, University of Minnesota, 15-144 PWB, 516 DelawareStreet SE, Minneapolis, Minnesota 55455-0392, USA

Dr J Breuer Reader in Virology, Dept of Medical Microbiology, StBartholomew’s & the Royal London Hospitals Medical School,Queen Mary Westfield College, 37 Ashfield Street, London, E1 1BB, UK

Professor J Gnann Associate Professor of Medicine, Division of Infectious Diseases,The University of Alabama School at Birmingham, 220 BevilBiomedical Research Building, 845 19th Street South,Birmingham, Alabama 35294-2170, USA

Dr K Higa Professor of Anesthesiology, Department of Anesthesiology,Fukuoka University, School of Medicine, 18-19-1 Nanakuma,Jonan-ku, Fukuoka 814-0180, Japan

Dr M Rowbotham Associate Professor & Director, Pain Clinical Research Center,University of California, Mount Zion, 1701 Divisadero Street,Suite 480, San Francisco, California 94117, USA

Dr J Seward Chief, Varicella Activity, Acting Chief, Child Vaccine PreventableDisease Control Branch, National Immunization Program, CDC,1600 Clifton Road, MS E-61, Atlanta, Georgia 30333, USA

Professor RJ Whitley Director of the Division of Clinical Virology & Professor ofPediatrics, Microbiology & Medicine, The University of Alabamaat Birmingham, Suite 616 Children’s Hospital, 1600 7th AvenueSouth, Birmingham, Alabama 35233-0011, USA

Dr MJ Wood Consultant Physician, Department of Infection & TropicalMedicine, Birmingham Heartlands Hospital, Bordesley GreenEast, Birmingham, B9 5ST, UK

Page 6: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

5

Management Recommendations and StatementsResearch needs J Improved and predictive animal models are needed for the study of acute disease and

of latent varicella zoster virus (VZV) infection and to assess the effect of potential new

drugs for the treatment of varicella and herpes zoster. It is recommended that the

development of suitable small animal models for studying VZV infection be continued.

J Although VZV reactivates in the dorsal root ganglia, it is unclear whether or not there

are other potential sites of reactivation. It is important to ascertain whether the virus

can reactivate in other sites and whether this reactivation can boost the immune

response. Further investigation of the hypothesis that reactivation occurs in peripheral

blood mononuclear cells is recommended, as this would offer the chance of regularly

monitoring reactivation and of predicting the onset of herpes zoster.

J Continued research is recommended to obtain a better understanding of VZV latency

and, in particular, knowledge of the viral antigens expressed during that period. This

may allow the development of approaches to prevent the reactivation of the virus and,

thus, the onset of herpes zoster.

IntroductionVZV (an alphaherpes virus) is the aetiological agent of two common diseases, varicella

(primary infection) and herpes zoster (reactivation of latent virus).1 Person-to-person

transmission of VZV is via the inhalation of aerosolized virus from

infected individuals, resulting in infection of the respiratory

mucosa.2 The pathogenesis of primary VZV infection resulting in

varicella is shown in Figure 1.

Varicella is a common disease, with a prevalence of approximately

95% in temperate countries. The majority of cases occur in children

aged <14 years.3-5 In addition to causing varicella, VZV establishes

latency during primary infection and may subsequently reactivate to

cause herpes zoster,6-8 with the attendant dermatomal rash and

associated pain.9 In tropical countries, the prevalence of VZV is

lower and varicella is usually contracted at a later age.10

The Need to Understand the MolecularMechanisms of VZV Replication and LatencyThe primary aim of treating VZV is to alleviate the acute and

chronic pain of herpes zoster and prevent the serious complications

of varicella. In addition, the current difficulty of managing both

ADVANCES IN THE PATHOGENESIS, MOLECULAR BIOLOGY

AND IMMUNOLOGY OF VARICELLA ZOSTER VIRUS1

FIGURE 1: Schematicrepresentation of thepathogenesis of varicella2

Incubation period

Acquisition by aerosol or direct contactInoculation of respiratory mucosa

Virus replication in regional nodesvirus-infected cells into capillaries

Primary viraemia replication inliver/spleen and other

reticuloendothelial sites

Secondary viraemia: mononuclear cellstransport virus to skin and mucous

membranes

Virus release into respiratory secretions

Replication in epidermal cellsVirus transported to dorsal root ganglia

Latency established

VZV-specific immunity cessationof virus replication

Acute Illness

© R

epro

duce

d w

ith p

erm

issi

on fr

om E

lsev

ier

Scie

nce

Arv

in A

M. C

hick

enpo

x (V

aric

ella

). In

:Va

ricel

la-Z

oste

r V

irus.

Mol

ecul

ar B

iolo

gy,P

atho

gene

sis

and

Clin

ical

Asp

ects

. (W

olff

MH

,Sc

hüne

man

n S,

Sch

mid

t A, e

ds).

Bas

el: K

arge

r, 19

99: 9

6-11

0.

Page 7: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

acute and chronic pain in herpes zoster11 together with the significant number of

hospitalizations caused by varicella and its attendant complications reinforce the need for

novel treatments of these conditions.

To assist in the development and testing of new agents for the prevention and treatment of

varicella and herpes zoster, improved understanding of the molecular mechanisms of VZV

pathogenesis, latency and reactivation, along with the development of suitable small

animal models of varicella and herpes zoster is required.

Animal and in vitro ModelsJ Improved and predictive animal models are needed for the study of acute disease and

of latent varicella zoster virus (VZV) infection and to assess the effect of potential new

drugs for the treatment of varicella and herpes zoster. It is recommended that the

development of suitable small animal models for studying VZV infection be continued

(research need recommendation).

The lack of suitable animal models of varicella and herpes zoster is a continuing

impediment to studying VZV infection. However, a number of new models may contribute

to our understanding of the pathogenesis of the virus. One such development is the use of

severe combined immunodeficiency disease (SCID) mice with implants of human fetal

lymphoid or skin tissue (SCID-hu), which can be infected with VZV to study virus

replication.12 Applying the technique of ‘knocking out’ individual virus genes by genetic

engineering allows the effect of those gene products to be investigated.13 However, a

problem with this is that as all these animals are immunocompromised, the role of the

immune system cannot be assessed.

A valuable contribution to the study of ocular VZV infection is the rabbit ocular VZV

infection model.14 An in vivo model of latent VZV infection in the rat was established by

Merville-Louis et al15 in 1989 using neurones from rat dorsal root ganglia and recently this

has been used to model the pain associated with herpes zoster.16

The further development of models such as these will provide greater insight into the

pathogenesis of VZV, and allow for the development of novel agents to treat varicella and

herpes zoster.

LatencyJ Although VZV reactivates in the dorsal root ganglia, it is unclear whether or not there

are other potential sites of VZV reactivation. It is important to ascertain whether virus

can reactivate in other sites and whether this reactivation can boost the immune

response. Further investigation of the hypothesis that reactivation also occurs in

peripheral blood mononuclear cells is recommended, as this would offer the chance

of regularly monitoring reactivation and of predicting the onset of herpes zoster

(research need recommendation).

J Continued research is recommended to obtain a better understanding of VZV latency

and, in particular, knowledge of the viral antigens expressed during that period. This

may allow the development of approaches to prevent the reactivation of the virus and,

thus, the onset of herpes zoster (research need recommendation).6

Page 8: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

7

Although VZV latency and reactivation are only partly understood, emerging data suggest

that virus, cellular and immune factors determine whether VZV remains in a latent state or

reactivates. Greater knowledge of these determinants may lead to the development of

novel approaches to prevent VZV reactivation, thereby avoiding herpes zoster and post-

herpetic neuralgia (PHN).

Site of latent virusIncreased understanding of how VZV establishes and maintains latency may allow the

development of new approaches to minimize reactivation of VZV, thus reducing the risk of

herpes zoster. The identity of the cells where VZV latency occurs remains controversial

despite detection of the virus genome in human sensory ganglia at autopsy using in situ

hybridization and polymerase chain reaction amplification.17-21 Cohen22 documented that

latent VZV is located primarily within neurones (at a suggested frequency of 2–5 copies

per latently infected neurone). In addition to being found in neurones,17,18,21 VZV DNA has

also been detected in the satellite cells surrounding them.19-21 However, the extensive

distribution of VZV infection in both types of cell, and the fact that herpes zoster occurs in

a dermatomal distribution, indicate ganglionic reactivation and spread from satellite

cells.20 The disparity of these findings confirms the need for further investigation into the

cellular location of VZV latency.

VZV molecular mechanisms of infection, maintenance of latency and evasionof host immune systemsIncreased knowledge of the mechanisms by which VZV infects the host, establishes latency

and evades the immune system could lead to the development of new treatments and

vaccines for varicella and herpes zoster.

The VZV protein encoded by open reading frame (ORF) 47, which is essential for

replication, could be a target for antiviral agents. Mutant VZV deficient in the ORF47 gene

is unable to replicate in either fetal lymphocyte or skin implants in SCID-hu mice. In

comparison, wild-type VZV replicates in the same model.13 This suggests that the ORF47

gene product is required for VZV replication in lymphocytes and dermal tissue.13

Therefore, the potential exists to develop agents that target this gene product and prevent

virus replication in situations such as post-transplant prophylaxis.

VZV infection of fibroblasts downregulates major histocompatibility complex (MHC)

class I antigens on the cell surface, limiting presentation of virus peptides to cytotoxic T

cells.23 Furthermore, when VZV is reactivated, upregulation of MHC class II expression by

interferon (IFN)-γ is blocked by VZV gene products.24 By reducing the expression of MHC

class I and II proteins and limiting presentation of virus peptides to T cells, VZV-infected

cells may avoid destruction by the host’s immune system.25 Agents capable of boosting the

expression of these antigens, thereby increasing virus peptide presentation to cytotoxic

T cells, would have the potential to increase substantially the cell-mediated response to

VZV.

During latency, immunity to VZV persists; immune response may be regularly boosted by

exposure to exogenous virus6 or by low-level, asymptomatic reactivation of virus in nerve

cells and possibly leucocytes.26,27 With time, cell-mediated immunity (CMI) wanes and

Page 9: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

8

may reach a critical threshold, allowing VZV to replicate and cause herpes zoster. CMI is

directed at two of the proteins expressed during latency (from the immediate early [IE]

genes IE62 and IE63).28,29 With increasing age, CMI to IE63 appears to decrease more than

CMI to IE62, indicating a possible role for IE63 in virus reactivation. Agents targeted

specifically at preventing expression of IE63 might prevent clinical reactivation of VZV,

and thus the development of herpes zoster.

VZV differs from herpes simplex virus (HSV) in that subsequent VZV reactivations following

an episode of herpes zoster are rare in immunocompetent individuals (available data suggest

only 5% experience a second herpes zoster outbreak).25 The molecular mechanisms that

maintain VZV latency differ from those controlling HSV, and analyses of these differences

could contribute to a greater understanding of latency in both viruses. Whereas latent HSV

is characterized by neuronal accumulation of latency-associated transcripts (LAT), which

appear to modulate reactivation,20 VZV has no DNA sequence homologous to LAT-coding

genes22,24 and, furthermore, VZV differs from other members of the alphaherpesvirus

subfamily in that five of its genes (ORF4, 21, 29, 62, 63) transcribed during lytic infection are

also expressed during the latent state.22 More recent experiments have shown that these

transcripts are also translated, e.g. IE63 in rats,30 and IE4, 21, 29, 62 and 63 in humans during

virus replication.31,32

The functions and interactions of the gene products expressed will need to be identified in

order to determine their role in latency. One hypothesis is that IE gene products

accumulate predominantly in the cytoplasm of latently infected cells instead of homing to

the nucleus, for example IE63,30 IE6233 and IE4.34 Finally, the observations by Lungu et al32

suggest that the intracellular localization of several IE proteins and two E proteins in

latently infected neurones is predominantly cytoplasmic and that this localization changes

during reactivation so that the proteins become detectable in the nuclei. This aberrant

localization may block or alter transcription.

Relationship between virus load and reactivation There is no clear relationship between the number of cells in which VZV establishes

latency and the risk that an individual will develop herpes zoster. In situ hybridization

studies have indicated that VZV remains latent in 0.01% to 0.03% of neurones,17,19

a significantly lower frequency than that reported for HSV (0.2% to 4.3% of the neuronal

population).17 The lower frequency of VZV latency in neurones as compared with HSV

may, however, explain the frequency of recurrences. Further research into the relationship

between the number of neurones infected, the number of copies of the VZV genome

present and the risk of reactivation is warranted. Potentially, this may allow physicians to

determine the patients most likely to develop herpes zoster within a short timeframe, and

thus allow them to implement suitable preventative measures if they become available.

SummaryIncreased knowledge of the mechanisms of VZV infection, latency and reactivation may

lead to new approaches to prevent varicella and herpes zoster. Some advances are being

made in the development of small animal models for VZV infection and reactivation,

which may provide useful insights into the pathogenesis and treatment of VZV. Continued

research in all these areas is recommended.

Page 10: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

References1. Liesegang T. Varicella zoster viral disease. Mayo Clin Proc 1999;74:983-998.2. Arvin AM. Chickenpox (Varicella). In: Varicella-Zoster Virus. Molecular Biology, Pathogenesis and Clinical

Aspects. (Wolff MH, Schünemann S, Schmidt A, eds). Basel: Karger, 1999: 96-110.3. Muench R, Nassim C, Niku S et al. Seroepidemiology of varicella. J Infect Dis 1986;153(1):153-155.4. Schneweis KE, Krentler C, Wolff MH. Varicella-zoster virus infection and the serologic determination of first

infection immunity. Dtsch Med Woschenschr 1985;110:453-457.5. Finger R, Hughes JP, Meade BJ et al. Age-specific incidence of chickenpox. Public Health Rep

1994;109(6):750-755.6. Hope-Simpson RE. The nature of herpes zoster: a long-term study and a new hypothesis. Proc R Soc Med

1965;58:9-20.7. Ragozzino MW, Melton LJ, 3rd, Kurland LT et al. Population-based study of herpes zoster and its sequelae.

Medicine (Baltimore) 1982;61(5):310-316.8. McGregor RM. Herpes zoster, chicken pox and cancer in general practice. BMJ 1957;1:84-87.9. Lungu O, Annunziato PW. Varicella-zoster virus: latency and reactivation. In: Varicella-Zoster Virus.

Molecular Biology, Pathogenesis and Clinical Aspects. (Wolff MH, Schünemann S, Schmidt A, eds). Basel:Karger, 1999: 61-75.

10. Garnett GP, Cox MJ, Bundy DA et al. The age of infection with varicella-zoster virus in St Lucia, West Indies.Epidemiol Infect 1993;110(2):361-372.

11. Portenoy RK, Duma C, Foley KM. Acute herpetic and postherpetic neuralgia: clinical review and currentmanagement. Ann Neurol 1986;20(6):651-664.

12. Arvin AM, Moffat JF, Redman R. Varicella-zoster virus: aspects of pathogenesis and host response to naturalinfection and varicella vaccine. Adv Virus Res 1996;46:263-309.

13. Moffat JF, Zerboni L, Sommer MH et al. The ORF47 and ORF66 putative protein kinases of varicella-zostervirus determine tropism for human T cells and skin in the SCID-hu mouse. Proc Natl Acad Sci USA1998;95(20):11969-11974.

14. Dunkel EC, Geary PA, Pavan-Langston D et al. Varicella-zoster virus ocular infection in the rabbit: a modelof human zoster ophthalmicus. Neurology 1995;45(Suppl 8):S21-28.

15. Merville-Louis MP, Sadzot-Delvaux C, Delree P et al. Varicella-zoster virus infection of adult rat sensoryneurons in vitro. J Virol 1989;63(7):3155-3160.

16. Kress M, Fickenscher H. Infection by human varicella-zoster virus confers norepinephrine sensitivity tosensory neurons from rat dorsal root ganglia. FASEB J 2001;15(6):1037-1043.

17. Hyman RW, Ecker JR, Tenser RB. Varicella-zoster virus RNA in human trigeminal ganglia. Lancet1983;2(8354):814-816.

18. Gilden DH, Rozenman Y, Murray R et al. Detection of varicella-zoster virus nucleic acid in neurons ofnormal human thoracic ganglia. Ann Neurol 1987;22(3):377-380.

19. Croen KD, Ostrove JM, Dragovic LJ et al. Patterns of gene expression and sites of latency in human nerveganglia are different for varicella-zoster and herpes simplex viruses. Proc Natl Acad Sci USA1988;85(24):9773-9777.

20. Meier JL, Straus SE. Comparative biology of latent varicella-zoster virus and herpes simplex virus infections.J Infect Dis 1992;166(Suppl 1):S13-S23.

21. Lungu O, Annunziato PW, Gershon A et al. Reactivated and latent varicella-zoster virus in human dorsalroot ganglia. Proc Natl Acad Sci USA 1995;92(24):10980-10984.

22. Cohen JI, Straus SE. Varicella-zoster virus and its replication. In: Fields Virology. (Fields BN, Knipe DM,Howley P, eds). Philadelphia: Lippincott-Raven Publishers, 1996: 2525-2545.

23. Cohen JI. Infection of cells with varicella-zoster virus down-regulates surface expression of class I majorhistocompatibility complex antigens. J Infect Dis 1998;177(5):1390-1393.

24. Abendroth A, Arvin A. Varicella-zoster virus immune evasion. Immunol Rev 1999;168:143-156.25. Cohen JI, Brunell PA, Straus SE et al. Recent advances in varicella-zoster virus infection. Ann Intern Med

1999;130(11):922-932.26. Mahalingam R, Wellish M, Brucklier J et al. Persistence of varicella-zoster virus DNA in elderly patients with

postherpetic neuralgia. J Neurovirol 1995;1(1):130-133.27. Devlin ME, Gilden DH, Mahalingam R et al. Peripheral blood mononuclear cells of the elderly contain

varicella-zoster virus DNA. J Infect Dis 1992;165(4):619-622.28. Arvin AM, Sharp M, Smith S et al. Equivalent recognition of a varicella-zoster virus immediate early protein

(IE62) and glycoprotein I by cytotoxic T lymphocytes of either CD4+ or CD8+ phenotype. J Immunol1991;146(1):257-264.

29. Sadzot-Delvaux C, Kinchington PR, Debrus S et al. Recognition of the latency-associated immediate earlyprotein IE63 of varicella-zoster virus by human memory T lymphocytes. J Immunol 1997;159(6):2802-2806.

30. Debrus S, Sadzot-Delvaux C, Nikkels AF et al. Varicella-zoster virus gene 63 encodes an immediate-earlyprotein that is abundantly expressed during latency. J Virol 1995;69(5):3240-3245.

31. Mahalingam R, Wellish M, Cohrs R et al. Expression of protein encoded by varicella-zoster virus openreading frame 63 in latently infected human ganglionic neurons. Proc Natl Acad Sci USA 1996;93(5):2122-2124.

32. Lungu O, Panagiotidis CA, Annunziato PW et al. Aberrant intracellular localization of varicella-zoster virusregulatory proteins during latency. Proc Natl Acad Sci USA 1998;95(12):7080-7085.

33. Baudoux L, Defechereux P, Schoonbroodt S et al. Mutational analysis of varicella-zoster virus majorimmediate-early protein IE62. Nucleic Acids Res 1995;23(8):1341-1349.

34. Defechereux P, Debrus S, Baudoux L et al. Intracellular distribution of the ORF4 gene product of varicella-zoster virus is influenced by the IE62 protein. J Gen Virol 1996;77(Pt 7):1505-1513.

9

Page 11: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

10

Management Recommendations and StatementsCategory 3J Physicians should be aware of both complications and atypical manifestations of

varicella, including varicella zoster virus (VZV) encephalitis and associated cutaneous

bacterial complications. Intravenous aciclovir is warranted for severe, atypical and

complicated VZV infections, based on anecdotal evidence.

Research needJ It is recommended that further research be conducted to assess the impact of social,

economic and demographic factors on the epidemiology of varicella. Data from such

research would facilitate public health policy decisions on vaccine deployment and

treatment of varicella.

IntroductionThis chapter reviews the epidemiology of varicella and defines its clinical signs and

symptoms and associated complications. The use of vaccination and its impact upon the

epidemiology of varicella is reviewed in Chapter 3. The management of varicella in

immunocompetent and immunocompromised individuals is discussed in Chapters 4 and 8.

Epidemiology of VaricellaJ It is recommended that further research be conducted to assess the impact of social,

economic and demographic factors on the epidemiology of varicella. Data from such

research would facilitate public health policy decisions on vaccine deployment and

treatment of varicella (research need recommendation).

Knowledge of VZV prevalence in a population would identify those at risk of developing

varicella, thus helping to determine those at risk of complications, the likely

socioeconomic impact of the disease and assisting healthcare policy makers to formulate

decisions on the need for vaccination and likely demands for treatment.

Estimates of the incidence of varicella in the USA since 1970 have been obtained from the

National Health Interview Survey.1 In the UK, the incidence of varicella is defined by cases

reported to the Royal College of General Practitioners, by sentinel practices covering a

population of approximately 700 000 in England and Wales since 1967, and in Scotland

through statutory notifications since 1988.2 For many other countries the epidemiology of

varicella has been extrapolated from smaller seroprevalence studies. However, because of

the small sample sizes, differences in populations tested, and the varying techniques used

to store and analyse sera, comparisons between data sets are difficult.

INTRODUCTION TO VARICELLA2

Page 12: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

11

Tropical and temperate countriesThe prevalence of VZV infection differs between tropical and temperate climates.2–13 In

temperate countries, the majority of primary infections occur in children.6-8 This is reflected

in its seroprevalence, which in the USA (prior to the introduction of varicella vaccine) and

Germany increases sharply in school-age children, reaching a peak prevalence of 90–95%

by adolescence (Figure 1). Only 5% of individuals remain susceptible to varicella after the

age of 15 years.6-8 An Italian study investigated the epidemiology of VZV through a sentinel

network of 39 paediatricians. Data from case reports of varicella among a monitored

population of 30 168 children were extrapolated to estimate a countrywide incidence of 5.1

cases per 100 population per year.9 However, as this study was conducted in a limited

population, the data are likely to be subject to larger error margins than the more complete

national monitoring data from France, UK and USA.

While most adults in temperate countries are VZV

seropositive, the incidence of varicella differs

between countries. For example, in France (1–

1.3 cases per 100 population per year) the incidence

is similar to that observed in the USA (1.5 cases per

100 population), but is notably higher than that

observed in the UK (0.5 cases per 100 population).11

The exact reasons for these differences between

temperate countries are not known.

In tropical countries, primary VZV infection tends to

occur much later in life than that in temperate regions.

Less than 10% of the population of St Lucia are

infected with VZV before the age of 15 years,10 there is then a gradual rise in seropositivity

until the age of 40 years, when VZV seroprevalence reaches around 70%. This figure is

considerably lower than the 95% seroprevalence observed in temperate countries over the

same age range. The mean age of infection in St Lucia is 38.3 years, compared with 10.6 years

in Germany. The high susceptibility in adults raised in tropical countries is demonstrated by

outbreaks of varicella when these individuals emigrate to countries with temperate climates.

For example, 42% of 810 US military personnel originally from Puerto Rico were found to be

seronegative, a much higher percentage than US-born recruits.12

In countries with intermediate climates, the seroprevalence of VZV falls between that

observed in tropical and temperate regions. A study in rural Nepal indicated that VZV

seroconversion rates were highest among the age groups 10–14 years and ≥15 years, with

an overall seroprevalence in adults of approximately 80%:13 a figure between that

observed in St Lucia (60%) and the USA or Europe (95%). However, the significance of

these data is difficult to assess owing to the variability observed between different

temperate countries and the different methods of data collection.

There are several possible explanations for the differences in VZV seroprevalence between

climates. The hypotheses include population size and density, exposure to the infectious agent

in confined spaces during the winter months in temperate climates, and higher ambient

temperatures and humidity in tropical climates. As such differences are not seen for measles –

also a highly contagious disease – and because VZV is heat-labile, the most likely explanation

FIGURE 1: VZVseroprevalence in St Lucia(tropical) compared withcombined data fromGermany, Spain, USA andJapan (temperate)10

1.0

0.8

0.6

0.4

0.2

00 20 40

TemperateTropical

Prop

ortio

n im

mun

e

Age (years)

60 80 100

0.1

0.3

0.5

0.7

0.9

+

+

+

++++++

+++

++

+ +

++

© R

epro

duce

d w

ith p

erm

issi

on, G

arne

tt G

P, C

ox M

J, B

undy

DA

et a

l.Th

e ag

e of

infe

ctio

n w

ith v

aric

ella

-zos

ter

viru

s in

St L

ucia

, Wes

tIn

dies

. Epi

dem

iol I

nfec

t19

93;1

10(2

):361

-372

. Cam

brid

ge U

nive

rsity

Pres

s.

Page 13: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

12

is that heat reduces the virus’ ability to survive and thus lowers the probability of transmission.

However, this hypothesis remains speculative.

Recent studies in urban populations of tropical countries demonstrate an epidemiological

picture similar to that observed in temperate countries, suggesting that other factors can

overcome the effect of climate on transmission of the virus. A study in Bengal, India found

that 96% of urban adults were VZV seropositive by the age of 25 years compared with

42% of rural adults, likely due to the differences in population density.14 The disparate

nature of the available data highlights the need for additional research to investigate the

impact of social, economic and demographic factors on the epidemiology of varicella and

consequent implications for healthcare policy.

Effect of societal changes on varicella epidemiologySentinel data from the UK indicate an increased incidence of varicella among children

younger than 5 years of age in the 1980s.15 This increase in incidence among the pre-

school population may be attributable to the larger number of children attending

organized pre-school daycare, increasing the likelihood of exposure to the virus. These

data from the UK are comparable, with a reported increase in the incidence of varicella

among pre-school children in some parts of the USA.8 These data highlight the need for

further research to assess the impact of social, economic and demographic factors on the

prevalence of varicella to guide future healthcare policy.

For a more complete review of the pre-varicella vaccine epidemiology of VZV in temperate

and tropical regions, refer to the IHMF Management Strategies in Herpes publication –

Management of Varicella.16

Clinical Signs and Symptoms of VaricellaVaricella has an incubation period of 10–21 days.17 The initial symptoms of varicella

include a prodrome consisting of fever, malaise, headache and abdominal pain. The

prodrome manifests approximately 24–48 hours prior to rash onset, and is usually more

pronounced in adolescents and adults than in children. Systemic symptoms that may

develop during the initial, viraemic phase include fever, fatigue and anorexia.

The exanthematous rash typical of varicella appears first on the face,

scalp or trunk, with a small number of isolated macropapular lesions

present. These lesions are often intensely pruritic, and change within

a few hours to the characteristic fluid-filled vesicles associated with

varicella (Figure 2). Crops of new lesions develop over the

subsequent 1–7 days and are usually located on the extremities.

Painful, ulcerative lesions of the mucous membranes frequently

accompany the evolution of the dermal rash. Varicella lesions often

involve the eyelids and bulbar conjunctivae, but serious ocular

complications are rare; unilateral anterior uveitis or corneal lesions

occasionally manifest, but usually resolve without further sequelae.17

Adolescents and adults typically develop more lesions than younger

children, as do secondary and tertiary household cases of

varicella.18,19

FIGURE 2: Typicalclinical manifestation of varicella

© R

epro

duce

d w

ith p

erm

issi

on.

Page 14: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

ComplicationsJ Physicians should be aware of both complications and atypical manifestations of

varicella, including VZV encephalitis and cutaneous bacterial complications associated

with varicella. Intravenous aciclovir is warranted for severe, atypical and complicated

VZV infections, based on anecdotal evidence (category 3 recommendation).

Although varicella is generally a benign and self-limiting disease,17 complications may

develop following initial infection, and can involve the skin, lungs, liver and nervous

system. The complications of primary VZV infection are rare in immunocompetent

children but are more frequent in adolescents, adults and individuals with suppressed cell-

mediated immunity.18-20

The risk of dying from varicella mirrors the risk of its complications. Data from the USA

gathered over the last 30 years show a higher mortality among infants aged younger than

1 year and adults 20 years of age or older compared with children aged 1–4 and 5–9

years.21 In the 1980s, mortality from varicella amongst infants (case fatality rate [CFR]

6.7/100 000 cases) and adults (CFR 17.1/100 000) was 10- to 24-fold greater than that

observed in children aged 1–4 years (CFR 0.7/100 000).21 Similar data have been reported

for the UK.2 No population-based mortality data are available from countries with tropical

climates but case reports indicate serious disease among adults.21

The mortality associated with varicella is largely due to varicella-associated pneumonia;

up to 30% of adults with varicella have some evidence of pulmonary involvement, either

radiographic or clinical.22-26 In addition, immune-altered individuals such as pregnant

women, cancer patients and neonates are at greater risk from complications of varicella,

and mortality rates in immunocompromised children with varicella range from 15% to

20% in the absence of treatment, also largely due to pneumonia.26

Complications of varicella observed in immunocompetent individualsPulmonary complicationsWhile pulmonary complications are rare in immunocompetent children with varicella,

some 5–10% of adults will have respiratory symptoms, such as cough or shortness of

breath, and a further 15–20% of adults with varicella will show radiographic evidence of

pneumonitis (Figure 3).27-29 The risk of developing varicella-associated pneumonia is

increased in men and among smokers; an Australian study indicated

the risk to smokers was 15 times higher than that of non-smokers.30

Intravenous antiviral therapy is recommended for any adolescent or

adult presenting with varicella-associated pneumonia. Although there

are no controlled clinical trials to support this indication, clinical

experience and anecdotal reports suggest that intravenous aciclovir

(10 mg/kg every 8 hours) is effective in this setting.31-33

Cutaneous complicationsSeveral cutaneous complications of varicella have been recognized,

including bacterial cellulitis and necrotizing fasciitis. They occur

more often in children than adults and bacterial cellulitis is the most

common complication leading to hospitalization, accounting for13

FIGURE 3: Chest X-rayof an individual withvaricella and evidence ofpneumonitis

© R

epro

duce

d w

ith p

erm

issi

on, P

rofe

ssor

John

Gna

nn.

Page 15: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

30% of such cases.34 It is most often caused by Staphylococcus aureus or Streptococcus

pyogenes35 and the scarring commonly associated with varicella is frequently due to

bacterial cellulitis. Measures such as the use of antibacterial soaps and trimming of

fingernails can help prevent bacterial infection. Appropriate antibacterial therapy is

recommended, but regional differences in antibiotic resistance defy global

recommendations.

Streptococcal superinfection may be associated with the use of non-steroidal anti-

inflammatory drugs (NSAIDs) for the management of the minor pain and fever associated

with varicella.36,37 For this reason, many paediatricians avoid their use in children with

varicella, and opt for drugs such as paracetamol.

Necrotizing fasciitis is also usually caused by Staphylococcus aureus or Streptococcus

pyogenes,36,37 and is exemplified by deeper fascial plane trapping of bacteria.38

Necrotizing fasciitis is a rare complication, occurring in 0.08 cases per 100 000 population

per year.39 It constitutes an emergency requiring early surgical debridement, appropriate

antibiotic administration and intensive supportive care.40,41

Atypical presentations and complications of varicellaA range of atypical presentations and complications of varicella may occur in

immunocompetent individuals, including cerebellar ataxia, transverse myelitis,

encephalitis, Guillain-Barré syndrome and Reye’s syndrome (Table 1).42-50 In general, no

controlled trials of therapy have been conducted for any of these manifestations and the

treatment recommendations are based on anecdotal reports.

14

TABLE 1: Atypicalpresentations andcomplications of varicellain the immunocompetenthost42-50

Manifestation Cases:cases of varicella Signs and symptoms Diagnosis

Cerebellar ataxia 1:4000 Headache, vomiting, lethargy. Usually clinicalAtaxia accompanied by fever, nuchal rigidity and nystagmus

VZV encephalitis 1–2:10 000 Fever, headache, vomiting, altered mental status, Laboratory diagnosisfocal neurological abnormalities, seizures CSF: Increased opening

pressure, elevated protein,normal glucoseEEG: Non-specific slowwave activityMRI: cerebral oedema

Pulmonary 5–10% of adults, Cough, shortness of breath X-raycomplications rare in children

Guillain-Barré syndrome Acute idiopathic polyneuritis

Reye’s syndrome Very rare following Vomiting, listlessness, delirium, convulsions Clinical blood chemistry:advice not to administer and loss of consciousness hypoglycaemia liveraspirin during varicella function: elevated

transaminases liver biopsy:signs of degeneration CSF: <8 cells/mm3

CSF = cerebrospinal fluid; EEG = electroencephalograph; MRI = magnetic resonance imaging

Cerebellar ataxiaCerebellar ataxia occurs with an estimated incidence of 1 case per 4000 cases of varicella

in immunocompetent children.51 It is generally benign and self-limiting, with most

individuals recovering within 1–3 weeks without further sequelae.45 Neurological signs

(headache, vomiting and lethargy) usually occur concurrently with varicella rash onset,

Page 16: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

15

but may also precede or follow the onset of dermatological symptoms.45 Fever, nuchal

rigidity and nystagmus occur in 25% of patients. In rare circumstances, seizures may also

occur.

The pathogenesis of cerebellar ataxia may involve virus replication in the cerebellum, as

indicated by the detection of VZV DNA by polymerase chain reaction (PCR) and increased

titres of VZV-specific antibodies in the CSF.52-54 However, a para-infectious, immune-

mediated mechanism may be responsible for its development.

The diagnosis of cerebellar ataxia is usually clinical, as laboratory test results are not

consistent. A low-grade pleocytosis (<100 white blood cells/mm3) in CSF is observed in

25% of patients. EEG recording detects diffuse slow-wave activity, consistent with ataxia,

in only 20% of patients with cerebellar ataxia. The role of computer tomography and MRI

scans in the diagnosis of the syndrome is uncertain, as most patients do not exhibit

abnormalities detectable by imaging.

The role of antiviral therapy in individuals presenting with varicella and cerebellar ataxia

has not been studied in a prospective or controlled fashion, but intravenous aciclovir

(10 mg/kg every 8 hours for adults, 500 mg/m2 body surface area every 8 hours for

children) is recommended.

VZV encephalitisVZV encephalitis usually has a worse prognosis than cerebral ataxia. It is characterized by

fever, headache, vomiting and an altered mental status indicative of increased intra-cranial

pressure.42,43 Other symptoms include focal neurological abnormalities, such as

hemiparesis and sensory changes. Seizures are a prominent feature of VZV encephalitis,

occurring in 29–52% of cases.55,56 The symptoms appear approximately 1 week after rash

onset, and may develop acutely or gradually.42,43 Estimates of reported mortality due to

VZV encephalitis range from 0% to 35%52,57 and neurological sequelae are observed in

10–20% of survivors.52,57

Although the absolute number of cases of varicella-associated encephalitis is highest in

children (due to the relative incidence of varicella itself in children and adults), the age-

specific incidence of encephalitis is higher in adults (1–2 cases per 10 000 cases of

varicella).55,56 In children, encephalitis following varicella (1 in 33 000 cases) is less

common than cerebellar ataxia.48,49

The role of VZV replication in the central nervous system (CNS) is not clearly defined in

the pathogenesis of VZV encephalitis. Numerous published pathology reports have

indicated a post-infectious demyelination process, whilst other findings are consistent with

direct virus cytopathology.58-60

Unlike cerebellar ataxia, laboratory diagnosis of encephalitis gives more consistent results

than clinical diagnosis. Examination of CSF indicates increased opening pressure,

pleocytosis and elevated protein with normal glucose levels. An EEG will show non-

specific slow-wave activity consistent with diffuse encephalitis.58 An MRI scan may

indicate cerebral oedema and areas of low attenuation consistent with

demyelination.58,61,62

Page 17: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

16

Although there are no prospective or controlled trials to support the use of antivirals in

VZV encephalitis (due in part to the rarity of the condition), intravenous aciclovir is

recommended based upon anecdotal evidence.

Recommended doses of aciclovir are:

• UK: 10 mg/kg every 8 hours in adults and 500 mg/m2 body surface area every 8 hours

in children. The dosage in children aged under 3 months is calculated on the basis

of body weight.

• USA: 10 mg/kg every 8 hours for 7 days in adults and 20 mg/kg every 8 hours for 7 days

in children.

Varicella in the pregnant woman, fetus and neonatePregnant womenIn the USA, where more than 95% of adults are VZV seropositive, five in 10 000 pregnant

women are estimated to develop varicella.24,63,64 The probability of infection among

women of childbearing age is likely to be greater in tropical regions where a higher

percentage of adults remain susceptible to VZV infection.65

Pregnant women are recognized as being less able to mount effective cellular immune

responses to infection compared with otherwise healthy children and adults. Thus,

varicella in the pregnant woman and neonate is more likely to have a serious outcome

than in other immunocompetent individuals, although there are no reliable population-

based studies to confirm this. The anecdotal evidence available suggests pregnant women

are at greater risk of developing severe varicella and its complications than non-pregnant

women of the same age.66-68 Based on data reported to the UK Office of National Statistics,

the risk of fatal varicella appears to be about 5-fold higher in pregnant than in non-

pregnant immunocompetent adults.69 In a study of 43 pregnant women with varicella,

nine (20%) had varicella-associated morbidity (pneumonia, premature labour, premature

delivery) and one woman died.67 If varicella pneumonia occurs during pregnancy, the

maternal mortality rate in the absence of antiviral therapy may be as high as 40%.22,23,25

The larger risk of varicella complications observed during pregnancy, and the high

mortality in the pregnant woman with varicella-associated pneumonia, highlight the need

for its prevention and management. These are discussed in Chapters 3 and 4.

Congenital varicella syndromePrimary VZV infection during pregnancy can result in a range of congenital defects in the

fetus (congenital varicella syndrome [Table 2]). The main route for congenital infection is

haematogenous spread of virus across

the placenta and it occurs irrespective

of the severity of varicella in the mother.

Congenital varicella syndrome was

initially thought to occur during the first

trimester of pregnancy only. In one

study, approximately half of the recorded cases developed following second trimester

maternal varicella.69 This indicates that the risk of congenital varicella syndrome following

maternal infection extends throughout the first half of pregnancy.70

TABLE 2: Clinicalcharacteristics ofcongenital varicellasyndrome

Defect associated with VZV infection

Skin Scarring

Limb Hypoplasia of bone and muscle

CNS Microcephaly, mental retardation, sphincter dysfunction

Eye Cataract, chorioretinitis, microphthalmia

Page 18: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

The risk of congenital varicella syndrome following maternal varicella is approximately

2–3%.71-73 In a prospective, case-controlled study,72 the pregnancy outcomes of 106

women with clinically diagnosed VZV infection in the first 20 weeks of pregnancy were

compared with 106 unexposed, age-matched controls. Only one in 86 live births (1.2%)

in the varicella group had features of congenital varicella syndrome (Table 2).

When these results were pooled with those of four previous studies, the estimated mean

risk was estimated to be 2.2%.72 In addition, this study found no difference between

infected and uninfected women in the proportion of live births (79% versus 85%),

miscarriages or elective terminations. There was a trend toward more pre-term deliveries

in the varicella-infected group (14.3% versus 5.6%; P=0.05) but gestational age at delivery

and birth weights were similar.72

These findings were corroborated by a larger, uncontrolled study of 1373 pregnant women

exposed to VZV in the first 36 weeks of pregnancy.71 In this study, the overall incidence of

congenital varicella syndrome was 0.7%, but the risk varied according to time of infection.

Congenital varicella syndrome occurred in seven out of 351 (2%) infants whose mothers

developed varicella infection between Weeks 13 and 20 of gestation, in two out of 472

(0.4%) whose mothers were infected during Weeks 8–13 and in none of 477 pregnancies

in which maternal infection was acquired after Week 20 (Table 3).71 A smaller study of 107

women who contracted varicella before 24 weeks of pregnancy recorded a risk of

congenital varicella syndrome of 2.8%.73

17

TABLE 3: Pooled resultsfrom prospective studiesmeasuring fetal riskfollowing maternal VZVinfection during pregnancy

Fetal risk* Reference

Maternal varicella in Maternal varicella in Maternal varicella infirst trimester, n (%) second trimester, n (%) third trimester, n (%)

2/27 (7.4) 0/32 (0) 2/76 (2.6) Siegel 197374

0/23 (0) 0/8 (0) 0/2 (0) Enders 198475✛

1/11 (9.0) 0/11 (0) 0/16 (0) Paryani & Arvin 198667

0/35 (0) Balducci et al. 199263

2/472 (0.4) 7/351 (2.0) 0/477 (0) Enders et al. 199471

1/86 (1.2)† 0/14 (0) Pastuszak et al. 199472

3/107 (2.8)‡ Mouly et al. 199773

*Ratio of the number of infants with congenital varicella syndrome compared with the total number of live-born infants†Estimated risk for mothers infected in the first 20 weeks of pregnancy‡Risk for mothers infected in the first 24 weeks of pregnancy✛ Figures also included in Enders et al. 199471

The potentially serious effects of congenital varicella syndrome highlight the need for

prevention and treatment of varicella during pregnancy in order to avoid congenital

varicella syndrome. These aspects are discussed in Chapters 3 and 4.

NeonatesNeonatal varicella can occur if the mother becomes infected with VZV between 4 days prior

to and 2 days after delivery. For these infections, transplacental virus transmission is the usual

route, while perinatal infection can result from contact with maternal lesions during or after

delivery.

The severity of neonatal varicella is largely dependent on the time of onset of maternal

illness in relation to delivery. The usual interval between maternal rash and onset in the

neonate is 9–15 days. Neonates are at highest risk of developing varicella when maternal

varicella begins between 3 days before and 2 days after delivery,76,77 leading to symptoms

in the neonate 5–10 days after birth. When maternal varicella develops during the perinatal

Page 19: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

18

period, the incidence of neonatal varicella can be as much as 48%.76 Approximately 20%

of infants born in this high-risk period will develop symptomatic neonatal varicella, with

disseminated infection and visceral involvement.78 For the neonate infected outside the

immediate peripartum period, the disease is generally not life-threatening.22,23

The enhanced risk of visceral dissemination in neonatal varicella and the associated high

mortality emphasize the need for effective management, which is discussed in Chapter 4.

For a more complete review of maternal, congenital and neonatal varicella, the reader is

directed to the IHMF Management Strategies in Herpes publication – Herpesvirus

Infections in Pregnancy.79

Summary Information on the epidemiology of varicella (individuals within a population at risk of

developing varicella and its attendant complications, and the socioeconomic impact of the

disease) can help public health policy makers formulate decisions on its prevention and

treatment.

The incidence of varicella varies between countries; however, in most developed

countries, the majority of cases occur in children, with only 5% of individuals remaining

susceptible after the age of 15 years. Tropical countries have a lower incidence of

childhood varicella, with peak seroprevalence being reached in adults aged 40 years or

more. Factors such as population density, economic status and social conditions may play

an important role in determining the incidence of varicella in a given country.

Varicella, typified by an exanthematous rash, is generally self-limiting due to the

development of humoral and cellular immune responses in the host. Although it is

generally a benign disease, varicella may be followed by complications, which can

involve the skin, lungs, liver or nervous system, including pneumonitis, cerebellar ataxia,

VZV encephalitis, and cutaneous bacterial superinfection. These complications are more

frequent in newborns, adolescents and adults. In addition, varicella occurring in pregnant

women and the immunocompromised is more likely to be serious in nature than disease

occurring in immunocompetent individuals of the same age.

The potentially serious nature of these complications, coupled with the number of

hospitalizations due to the disease – 11 000 per year in the USA prior to initiation of the

vaccination programme – emphasize the importance of treating varicella, particularly in

high-risk individuals such as adolescents, adults, pregnant women and the

immunocompromised.

The prevention and management of varicella are discussed in more detail in Chapters 3, 4 and 8.

References1. Prevention of varicella: Recommendations of the Advisory Committee on Immunization Practices (ACIP).

Centers for Disease Control and Prevention. MMWR Morb Mortal Wkly Rep 1996;45(RR-11):1-36.2. Fairley CK, Miller E. Varicella-zoster virus epidemiology–a changing scene? J Infect Dis 1996;174

(Suppl 3):S314-S319.3. Alavarez y Munoz MT, Torres J, Damasio-Santana L et al. Susceptibility to varicella-zoster infection in

individuals 1 to 29 years of age in Mexico. Arch Med Res 1999;30(1):60-63.4. Lieu TA, Black SB, Takahashi H et al. Varicella serology among school age children with a negative or

uncertain history of chickenpox. Pediatr Infect Dis 1998;17(2):120-125.

Page 20: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

19

5. Taylor-Wiedeman J, Yamashita K, Miyamura K et al. Varicella-zoster virus prevalence in Japan: no significantchange in a decade. Jpn J Med Sci Biol 1989;42(1):1-11.

6. Muench R, Nassim C, Niku S et al. Seroepidemiology of varicella. J Infect Dis 1986;153(1):153-155.7. Schneweis KE, Krentler C, Wolff MH. Varicella-zoster virus infection and the serologic determination of first

infection immunity. Dtsch Med Woschenschr 1985;110:453-457.8. Finger R, Hughes JP, Meade BJ et al. Age-specific incidence of chickenpox. Public Health Rep

1994;109(6):750-755.9. Fornaro P, Gandini F, Marin M et al. Epidemiology and cost analysis of varicella in Italy: results of a sentinel

study in the pediatric population. Italian Sentinel Group on Pediatric Infectious Diseases. Pediatr Infect Dis1999;18(5):414-419.

10. Garnett GP, Cox MJ, Bundy DA et al. The age of infection with varicella-zoster virus in St Lucia, West Indies.Epidemiol Infect 1993;110(2):361-372.

11. Deguen S, Chau NP, Flahault A. Epidemiology of chickenpox in France (1991-1995). J EpidemiolCommunity Health 1998;52 (Suppl 1):46S-49S.

12. Longfield JN, Winn RE, Gibson RL et al. Varicella outbreaks in Army recruits from Puerto Rico. Varicellasusceptibility in a population from the tropics. Arch Intern Med 1990;150(5):970-973.

13. Kubo T, Rai SK, Nakanishi M et al. Seroepidemiological study of herpes viruses in Nepal. Southeast Asian JTrop Med Public Health 1991;22(3):323-325.

14. Mandal BK, Mukherjee PP, Murphy C et al. Adult susceptibility to varicella in the tropics is a ruralphenomenon due to the lack of previous exposure. J Infect Dis 1998;178 (Suppl 1):S52-54.

15. Wharton M. The epidemiology of varicella-zoster virus infections. Infect Dis Clin North Am 1996;10(3):571-581.

16. Kroon S, Wood MJ. Management Strategies in Herpes: Management of Varicella. Worthing: PPS Europe Ltd,1996.

17. Arvin AM. Chickenpox (Varicella). In: Varicella-Zoster Virus. Molecular Biology, Pathogenesis and ClinicalAspects. (Wolff MH, Schünemann S, Schmidt A, eds). Basel: Karger, 1999: 96-110.

18. McCrary ML, Severson J, Tyring SK. Varicella zoster virus. J Am Acad Dermatol 1999;41(1):1-14; quiz 15-16.

19. Stover BH, Bratcher DF. Varicella-zoster virus: infection, control, and prevention. Am J Infect Control1998;26(3):369-381; quiz 382-364.

20. Takahashi M, Gershon AA. Varicella vaccine. In: Vaccines. 2nd edn. (Plotkin SA, Mortomer EA, eds).Philadelphia PA: WB Saunders, 1994: 387-417.

21. Seward J, Galil K, Wharton M. Epidemiology of varicella. In: Varicella Zoster Virus: Virology and ClinicalManagement. (Arvin A, Gershon A, eds). Cambridge: Cambridge University Press, 2000.

22. Miller AE, Marshall R, Vardien J. Epidemiology, outcome and control of varicella-zoster infection. Rev MedMicrobiol 1993;4:222-230.

23. Miller E, Vardien J, Farrington P. Shift in age in chickenpox. Lancet 1993;341(8840):308-309.24. Sever J, White LR. Intrauterine viral infections. Ann Rev Med 1968;19:471-486.25. Smego RA, Asperilla MO. Use of acyclovir for varicella pneumonia during pregnancy. Obstet Gynecol

1991;78:1112-1116.26. Lekstrom-Himes JA, Straus SE. Varicella-zoster virus infections in the normal and immunocompromised host.

In: Clinical Management of Herpes Viruses. (Sacks SL, Straus SE, Whitley RJ et al, eds). Amsterdam: IOSPress, 1995: 175-192.

27. Weber DM, Pellecchia JA. Varicella pneumonia. Study of prevalence in adult men. JAMA 1965;192:288-289.

28. Gogos CA, Bassaris HP, Vagenakis AG. Varicella pneumonia in adults. A review of manifestations, risk factorsand treatment. Respiration 1992;59:339-343.

29. Feldman S. Varicella-zoster virus pneumonitis. Chest 1994;106 (Suppl 1):S22-S27.30. Grayson ML, Newton-John H. Smoking and varicella pneumonia. J Infect 1988;16:312.31. Haake DA, Zakowski PC, Haake DL et al. Early treatment with acyclovir for varicella pneumonia in

otherwise healthy adults: retrospective controlled study and review. Rev Infect Dis 1990;12:788-798.32. Schlossberg D, Littman M. Varicella pneumonia. Arch Intern Med 1988;148:1630-1632.33. Wilkins EG, Leen CL, McKendrick MW et al. Management of chickenpox in the adult. A review prepared

for the UK Advisory Group on Chickenpox on behalf of the British Society for the Study of Infection. J InfectDis 1998;36 (Suppl 1):49-58.

34. Somekh E, Maharashak N, Shapira Y et al. Hospitalization for primary varicella-zoster virus infection and itscomplications in patients from Southern Israel. Infection 2000;28(4):200-204.

35. Aebi C, Ahmen A, Ramilo O. Bacterial complications of primary varicella in children. Clin Infect Dis1996;23:698-705.

36. Zerr DM, Alexander ER, Duchin JS et al. A case-control study of necrotizing fasciitis during primaryvaricella. Pediatrics 1999;103:783-790.

37. Choo PW, Galil K, Donahue JG et al. Risk factors for postherpetic neuralgia. Arch Intern Med1997;157(11):1217-1224.

38. Cazorla C. What data is needed today to deal with cellulitis and necrotizing fasciitis? Ann DermatolVenereol 2001;128(3):443-451.

39. Laupland KB, Davies HD, Low DE et al. Invasive group A streptococcal disease in children and associationwith varicella-zoster virus infection. Ontario Group A Streptococcal Study Group. Pediatrics2000;105(5):E60.

40. Wilson GJ, Talkington DF, Gruber W et al. Group A streptococcal necrotizing fasciitis following varicella inchildren: case reports and review. Clin Infect Dis 1995;20(5):1333-1338.

41. Brogan TV, Nizet V, Waldhausen JH et al. Group A streptococcal necrotizing fasciitis complicating primaryvaricella: a series of fourteen patients. Pediatr Infect Dis 1995;14:588-594.

42. Applebaum E, Rachelson MH, Dolgopol VB. Varicella encephalitis. Am J Med 1953;15:223-230.43. Boughton CR. Varicella-zoster in Sydney. II. Neurological complications of varicella. Med J Aust

1966;2:444-447.

Page 21: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

44. Heppleston JD, Pearch KM, Yates PO. Varicella encephalitis. Arch Dis Child 1959;34:318-321.45. Johnson R, Milbourn PE. Central nervous system manifestations of chickenpox. Can Med Assoc J

1970;102:831-834.46. Underwood EA. The neurological complications of varicella: a clinical and epidemiological study. Br J Child

Dis 1935;32:376-378.47. Welch RG. Chickenpox and the Guillian-Barré syndrome. Arch Dis Child 1962;37:557-559.48. Choo PW, Donahue JG, Manson JE et al. The epidemiology of varicella and its complications. J Infect Dis

1995;172:706-712.49. Preblud SR. Age-specific risks of varicella complications. Pediatrics 1981;68:14-17.50. Calvani M. Reye’s syndrome: the death of a syndrome? (Or death by a syndrome?) Recent Prog Med

2000;91(12):675-680.51. Guess HA. Population-based studies of varicella complications. Pediatrics 1986;78:723-730.52. Echevarria JM, Casas I, Martinez-Martin P. Infections of the nervous system caused by varicella-zoster virus:

a review. Intervirology 1997;40:72-84.53. Puchhammer-Stockl E. Detection of varicella-zoster virus DNA by polymerase chain reaction in the

cerebrospinal fluid of patients suffering from neurological complications associated with chickenpox orherpes zoster. J Clin Microbiol 1991;29:1513-1516.

54. Dangood F, Engle E, Yessayan L et al. Pre-eruptive varicella cerebellitis confirmed by PCR. Pediatr Neurol1993;172:706-712.

55. Gibbs FA, Gibbs EL, Spies HW et al. Common types of childhood encephalitis. Arch Neurol1964;10:15-25.

56. Griffith JF, Salam MV, Adams RD. The nervous system diseases associated with varicella. Acta Neurol Scand1970;46:279-300.

57. Preblud SR, D’Angelo LJ. Chickenpox in the United States. J Infect Dis 1979;140:257-260.58. Bauman ML, Bergman I. Post varicella encephalitis. Arch Neurol 1984;41:556-558.59. McCormick WR, Rodnitzky RL, Schochet SS et al. Varicella-zoster encephalomyelitis: a morphologic and

virologic study. Arch Neurol 1969;21:559-570.60. Takashima S, Becker LE. Neuropathology of fatal varicella. Arch Pathol Lab Med 1979;103:209-213.61. Tenorio G, Whitaker JN. Steriod-dependent post varicella encephalomyelitis. Child Neurol 1991;6:45-48.62. Hurst DL, Mehta S. Acute cerebellar swelling in varicella encephalitis. Pediatr Neurol 1988;2:122.63. Stagno S, Whitley RJ. Herpesvirus infections of pregnancy. Part II: Herpes simplex virus and varicella-zoster

virus infections. N Engl J Med 1985;313:1327-1330.64. Balducci J, Rodis JF, Rosengren S et al. Pregnancy outcome following first-trimester varicella infection.

Obstet Gynecol 1992;79:5-6.65. Weller TH. Varicella and herpes-zoster. Changing concepts of the natural history, control, and importance

of a not-so-benign virus (Part 1). N Engl J Med 1983;309:1362-1368.66. Gray F, Mohr M, Rozenberg F. Varicella-zoster virus encephalitis in acquired immunodeficiency syndrome:

a report of four cases. Neuropathol Appl Neurobiol 1992;18:502-514.67. Paryani SG, Arvin AM. Intrauterine infection with varicella-zoster virus after maternal varicella. N Engl J Med

1986;314(24):1542-1546.68. Cox SM, Cunningham FG, Luby J. Management of varicella pneumonia complicating pregnancy. Am J

Perinatol 1990;7:300-301.69. Enders G, Miller E. Varicella and herpes zoster in pregnancy and the newborn. In: Varicella-Zoster Virus:

Virology and Clinical Management. (Arvin A, Gershon A, eds). Cambridge: Cambridge University Press,2000.

70. Gershon AA. Varicella-zoster virus: prospects for control. Adv Pediatr Infect Dis 1995;10:93-124.71. Enders G, Miller E, Cradock-Watson J et al. Consequences of varicella and herpes zoster in pregnancy:

prospective study of 1739 cases. Lancet 1994;343(8912):1548-1551.72. Pastuszak AL, Levy M, Schiek B et al. Outcome after maternal varicella infection in the first 20 weeks of

pregnancy. N Engl J Med 1994;330:901-905.73. Mouly M, Mirlesse V, Meritet JF. Prenatal diagnosis of fetal varicella-zoster virus infection with polymerase

chain reaction of amniotic fluid in 107 cases. Am J Obstet Gynecol 1997;177:894-898.74. Siegel M. Congenital malformations following chickenpox, measles, mumps and hepatitis: results of a

cohort study. JAMA 1973;226:1521-1524.75. Enders G. Varicella-zoster virus infection in pregnancy. Prog Med Virol 1984;29:166-196.76. Miller E, Cradock-Watson JE, Ridehalgh MK. Outcome in newborn babies given anti-varicella-zoster

immunoglobulin after perinatal maternal infection with varicella-zoster virus. Lancet 1989;2(8659):371-373.

77. Gershon A. Infections in the fetus and newborn infant. New York: Alan R Liss, 1975.78. Meyers JD. Congenital varicella in term infants: risk reconsidered. J Infect Dis 1974;129(2):215-217.79. Pass R, Weber T, Whitley RJ. Management Strategies in Herpes: Herpesvirus Infections in Pregnancy.

Worthing: PAREXEL MMS Europe Ltd, 2000.

20

Page 22: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

21

VARICELLA VACCINATION3

Management RecommendationsCategory 2J If there is a policy of routine childhood vaccination, all children and adolescents

should be immunized. Immunizing only a subset of the paediatric population is likely

to increase the risk of creating a population of adults vulnerable to varicella and its

attendant morbidity.

J Considerations as to whom to vaccinate may differ between temperate regions (where

varicella is almost exclusively a disease of young children) and tropical regions (where

a larger proportion of the population contracts varicella at an older age).

J To facilitate the uptake of universal vaccination in the USA and other countries, it is

recommended that proof of varicella vaccination or positive varicella zoster virus

(VZV) serostatus should be considered a requirement for school entry.

J Targeted vaccination of at-risk groups is recommended in countries not considering

universal vaccination programmes. Such programmes should include teachers,

healthcare workers, identified seronegative adults, seronegative women of

childbearing age who are not pregnant and patients undergoing haematopoietic stem

cell transplantation.

J Programmes should be established to screen for VZV-seropositivity in healthcare

workers, teachers and other adults likely to come into sustained contact with VZV-

infected children, and to vaccinate susceptible individuals. This would serve to protect

healthcare workers from VZV-infected patients, protect susceptible patients from

healthcare workers with varicella, and protect seronegative teachers from VZV-

infected children.

J Clinicians caring for women of childbearing age should be encouraged to determine

the VZV serological status of their patients. Women with no history of varicella should

undergo serological testing and susceptible women should be vaccinated. Vaccination

of susceptible women will reduce the risk of severe maternal morbidity caused by the

varicella-related complications that can occur during pregnancy and obviate the need

for varicella zoster immune globulin (VZIG) prophylaxis during pregnancy. Pregnant

women with no history of varicella should not be vaccinated while pregnant but

should undergo serological testing to determine susceptibility.

Page 23: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

22

Research needsJ More data are required on the long-term outcomes (prevalence, frequency of

complications, economic factors) following a varicella vaccination programme. As the

USA moves toward universal varicella vaccination, the effect of this programme on the

epidemiology of varicella and herpes zoster must be monitored. In addition, the

economic impact of the programme should be assessed. Analysis of data from

immunocompromised individuals will provide valuable information but should not

replace monitoring of the general vaccinated population in the USA.

J Comparative analyses of health-outcome benefits should be conducted comparing the

USA with countries that do not have universal varicella vaccination programmes. To

facilitate policy decisions on the implementation of varicella vaccination programmes,

health policy decision-makers will require the maximum amount of data possible on

the medical and economic impact of varicella and varicella vaccination.

IntroductionVaricella has generally been regarded as a mild, self-limiting disease in otherwise healthy

children. However, in the USA, before the licensing of the varicella vaccine in 1995, there

were approximately 4 million cases per year, resulting in nearly 9300 hospitalizations1 and

100 deaths each year.2 The impact was greatest on children, who accounted for more than

90% of cases, 66% of hospitalizations and 45% of deaths.1 However, the risk of severe

complications and death was highest among infants and adults2 (see Chapter 2). Maternal

infection and infection of the newborn can also have severe sequelae. A further group at

risk from varicella is immunocompromised patients with impaired cell-mediated

immunity.3,4

A live attenuated varicella vaccine (VZVoka vaccine) was developed and clinically tested

in Japan in 1974.5 The VZVoka strain is the basis for all currently available varicella vaccines

(produced by Biken; Merck & Co Inc; SmithKline Beecham [now GlaxoSmithKline];

Aventis Pasteur). Experience with the live attenuated VZVoka vaccine for prevention of

varicella has accumulated over the last 25 years in Japan and the USA. The vaccine is well

tolerated and effective in healthy children and adults. Thus, this highly infectious disease

and its complications are preventable by vaccination.

Efficacy and Safety of Varicella VaccineVaccination in immunocompetent individualsEfficacy of varicella vaccineA number of retrospective and prospective clinical studies have demonstrated that

varicella vaccination is effective in immunocompetent individuals.6-11 However, of all

these trials, only two have been placebo-controlled. One trial, using the Merck vaccine,

was conducted in children (1–14 years of age), 468 of whom were immunized with a dose

of 17430 plaque-forming units (pfu) of virus and 446 were administered placebo.9 During

the initial 9 months following vaccination, 39 of the placebo recipients developed

varicella, compared with no cases in the varicella vaccine group. Thus, the vaccine

efficacy was 100%. During the second year of the study, one vaccinated child developed

varicella, giving an efficacy rate of 98%.9 Over a 7-year follow-up period, the efficacy of

the vaccine was estimated at 95% in immunocompetent children. The vaccine was well

tolerated, although it had a much higher titre than those commercially produced.9

Page 24: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

23

In the second placebo-controlled trial,12 performed with the SmithKline Beecham vaccine,

513 healthy children (10–30 months old) were randomized to receive either:

• High-titre vaccine dose (10 000 or 15 850 pfu/dose)

• Low-titre vaccine dose (630 or 1260 pfu/dose)

• Placebo.

Between 94% and 100% of previously seronegative children developed varicella-specific

antibodies. The attack rate during an average of 29 months follow-up was 3% (5 cases) in

the high-titre group and 11.4% (5 cases) in the low-titre group. Both these attack rates were

significantly lower than the 26% observed with placebo (41 cases; P≤0.005).

Placebo-controlled trials of the varicella vaccine in two groups at high risk of varicella

complications, adults and immunocompromised children, have not yet been conducted.

Trials conducted in healthy young adults and leukaemic children indicate that the vaccine

is less effective in these two groups.13,14 This may be due to both groups having impaired

responses to VZV, as humoral and cell-mediated immunity was lower in these vaccinees

than in individuals who had experienced natural varicella infection. In general, two doses

of the vaccine are required to achieve a seroconversion rate of 95–98%. In adults, the

efficacy of the vaccine was estimated to be 65–70% following household exposure.15

The two placebo-controlled trials indicate that dose is important in defining efficacy.

Currently, the VZVoka-Merck vaccine (Varivax; Merck & Co Inc) used in the USA provides a

dose of approximately 2500 pfu but high levels of protection are still observed. Data from

the American trials of the VZVoka-Merck vaccine, in which vaccinated individuals of all ages

were followed up for up to 9 years, demonstrated seroconversion rates of 95.6–99% per

year depending on vaccine batch and time since vaccination.16 A prospective follow-up

study in the USA demonstrated that protection lasted at least 10 years following

vaccination;17 similar studies in Japan have shown protection persisting for 20 years.18

In the USA, trials with no placebo control have demonstrated that vaccination of adults

and children significantly lowers the attack rate among household exposures.10,11 The

incidence of varicella following household exposure in vaccinees was approximately

12%; household contact historically results in 87% infection. Nearly all of the vaccinees

who had varicella after vaccination had a clinically modified disease. Varicella infections

were milder than wild-type infections in the unvaccinated (Table 1).10

TABLE 1:Seroconversion andduration of protectionobserved in clinical trialswith varicella vaccine10

Study group Follow-up (years) Rash or fever (%) Seroconversion (%) Protection 1–3 years (%) Protection >3 years (%)

Children 2–16 5 95 89–94 88 Adults na 10 >90 (two doses) 70 86

na=not applicable

A potential weakness of the available data on the persistence of protection in the US

populations is that these studies were conducted prior to the adoption of universal

vaccination. High levels of vaccine coverage may reduce the chance of exposure to wild-

type VZV, thereby lowering the opportunities for immunological boosting from subclinical

infection with exogenous VZV. Periodic boosting of cellular immunity by such exposure

may be a factor in maintaining protection against varicella. Further studies will establish

whether vaccination provides protection which is as durable as that of natural infection or

whether booster doses will be required to maintain lifelong protection.

Page 25: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Safety of vaccination Varicella vaccination is generally well tolerated in healthy adults and children. In healthy

children, the frequency of rash after immunization is approximately 5% and there are very

few skin lesions observed.8,19 Other common adverse events include fever (15%),

temporary discomfort at the injection site (19–24%) and rash at the injection site

(3–4%).8,19 Similar adverse event rates are seen in healthy adolescents and adults, in

whom two doses of vaccine are required. However, the rate of vaccine-associated rash in

adults (10%) is twice that seen in healthy children.8,19

Vaccination for women of childbearing ageJ Clinicians caring for women of childbearing age should determine the VZV

serological status of their patients. Women with no history of varicella should undergo

serological testing and susceptible women should be vaccinated. Vaccination will

reduce the risk of severe maternal morbidity caused by the varicella-related

complications that can occur during pregnancy and obviate the need for VZIG

prophylaxis during pregnancy. Pregnant women with no history of varicella should not

be vaccinated while pregnant but should undergo serological testing to determine

susceptibility (category 2 recommendation).

Vaccination of susceptible women of childbearing age may potentially also prevent

congenital varicella syndrome or neonatal varicella. Seronegative women are at risk of

exposure to varicella from children in their household as most cases occur in those under

5 years of age. In addition, the attack rate of varicella is extremely high following

household contact. Thus, vaccination should be considered for non-pregnant seronegative

women of childbearing age. The US Advisory Committee on Immunization Practices

(ACIP) has recently strengthened its recommendations for varicella so that vaccination is

recommended for susceptible, non-pregnant women of childbearing age.20

Pregnancy is a contraindication for varicella vaccination. No epidemiological data are

available on exposure to the vaccine during pregnancy and the effects on the fetus are

unknown. A theoretical risk is that a vaccinated woman may develop varicella infection

from the attenuated virus with the consequence of fetal varicella infection. Therefore, in

the USA, the Varivax manufacturer (Merck & Co Inc) advises a 3-month interval between

vaccination and conception, although the ACIP and Centers for Disease Control suggest

that vaccinated non-pregnant women should avoid becoming pregnant for 1 month

following each injection of vaccine.16 However, inadvertent vaccination immediately

prior to or early in pregnancy is not a justification for termination of pregnancy. In the

USA, a Varivax Pregnancy Registry (telephone: ++1 800 986 8999) has been established

to monitor the maternal and fetal outcomes of pregnant women who receive varicella

vaccine up to 3 months before, or at any time during, pregnancy.

Potential of Vaccine to Prevent Herpes ZosterThe availability of a safe and effective varicella vaccine presents the opportunity to

determine whether it may also be effective for preventing herpes zoster in the elderly. The

initial suggestion that vaccination may reduce the likelihood of herpes zoster was

prompted by a study in children with leukaemia who received the VZVoka vaccine. Among

these, the incidence of herpes zoster was lower than in children who had experienced

natural VZV infection.2124

Page 26: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

25

Based on the results of the study described above, investigators have hypothesized that

administration of live attenuated VZV vaccine to older adults (who are likely to be already

latently infected with VZV) may stimulate their waning cellular immune responses, thereby

preventing herpes zoster during the last decades of life when individuals are at highest

risk.22,23 Administration of a VZVoka vaccine (at a higher dose than that routinely used to

immunize children) boosts the number of circulating T-lymphocytes that are responsive to

VZV antigens; the duration of this booster effect is at least 6 years.22 A large-scale

serological study has provided evidence that VZVoka can reactivate in vaccinees who have

declining anti-VZV antibody titres.24 Reactivation of VZVoka may result in a mild varicella-

like illness and also in a substantial boost in anti-VZV antibody titres, thus extending the

duration of protection against clinically significant varicella. Such boosting of the VZV

immune responses in older adults has the potential to reduce the incidence of herpes

zoster or at least attenuate the severity of the disease.

To determine the clinical value of vaccination in older adults, a double-blind, placebo-

controlled trial of varicella vaccine investigating its effects on the severity and

complications of a first occurrence of herpes zoster is underway in the USA (the Shingles

Prevention Trial). Its target population was 37 200 volunteers over the age of 60 years. With

a 3-year follow-up period, the study will last approximately 5 years.

Prophylactic administration of the inactivated varicella vaccine has also reduced the risk

of zoster in patients undergoing haematopoietic stem cell transplantation (see Chapter 8

for further details).25

Vaccination StrategiesJ Comparative analyses of health-outcome benefits should be conducted comparing the

USA with countries that do not have universal varicella vaccination programmes. To

facilitate policy decisions on the implementation of varicella vaccination programmes,

health policy decision-makers will require the maximum amount of data possible on

the medical and economic impact of varicella and varicella vaccination (research

need recommendation).

J More data are required on the long-term outcomes following varicella vaccination

with the aim of optimizing the target population. As the USA moves toward universal

varicella vaccination, the effect of this programme on the epidemiology of varicella

and herpes zoster must be monitored. In addition, the economic impact of the

programme should be investigated. Analysis of data from immunocompromised

individuals will provide valuable information but should not replace monitoring of the

general vaccinated population in the USA (research need recommendation).

J Considerations as to whom to vaccinate may differ between temperate regions (where

varicella is almost exclusively a disease of young children) and tropical regions (where

a larger proportion of the population contracts varicella at an older age) (category 2

recommendation).

Varicella vaccination strategies may be either universal for all children or targeted at those

susceptible individuals who are considered to be at high risk of exposure. The choice of

which strategy to adopt is influenced by many factors including cost effectiveness and

Page 27: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

perceptions of the seriousness of varicella. The USA is the first country in which routine

varicella vaccination for all children has been recommended.20,26 The vaccine has been

licensed in other countries, where it is mainly used for high-risk individuals and their

contacts. Gathering data on the long-term medical, epidemiological and socioeconomic

outcomes following the introduction of universal childhood vaccination in the USA will allow

critical analysis of the benefits of this strategy. Should similar data become available for targeted

vaccination, a comparison of the benefits of the two different strategies will be possible.

The choice of strategy employed may also be influenced by the differences in the

epidemiology of varicella between temperate and tropical regions. There may be a greater

need for universal vaccination in tropical countries, where a greater proportion of adults

remain susceptible to varicella, and a relatively high complication rate is expected in this

group. However, the implementation of such a strategy should be viewed in the context of

other, more serious, childhood diseases preventable by vaccinations. For example, in

Africa, vaccine coverage for measles (a far more life-threatening illness) falls below the

World Health Organization global goals; thus, varicella vaccination may be considered

neither feasible nor a priority.

Universal vaccinationThe objective of a universal varicella vaccination programme in children is to achieve an

overall reduction in varicella cases and complications among both children and adults. In

theory, a universal programme has the potential to eradicate clinical varicella and reduce

the severity of herpes zoster, should it develop.

A major argument for the routine varicella programme in the USA comes from a series of

detailed cost-effectiveness analyses conducted since 1985.27-29 The first of the analyses,

performed, found that universal vaccination was cost-effective if parents’ loss of income

from time taken off work to care for their sick children was taken into account along with

the medical costs.27 The second analysis, in 1994, compared universal vaccination with no

intervention.28 Direct medical costs of US$2.00 per case of varicella prevented were

calculated. In terms of overall cost to society (including direct medical costs and the cost of

pay-loss by parents), for every US$1.00 spent on vaccination, US$5.00 was saved. Another

study conducted at the same time also had similar conclusions for universal vaccination.30

Epidemiology of varicella in the USA following vaccine introductionSince the introduction of the varicella vaccine in the USA in 1995, active varicella

surveillance has been conducted in the states of California, Pennsylvania and Texas.

Reporting sources include schools, childcare centres, pre-school groups, private

physicians, public-health clinics, hospitals, large companies and the general public.

Vaccine used in these geographical areas has been monitored in private healthcare

providers’ offices, immunization registries, and through the Vaccines for Children Program.

In children aged 12–23 months, vaccine coverage in the three US states increased from

30–50% in 1997 to 60–80% in 1999. The surveillance has highlighted a declining trend

in the number of varicella cases and hospitalizations due to varicella-related illness. In

these three areas, there was a 77–84% reduction in the number of cases of varicella or

associated complications and a 79% reduction in hospitalizations due to varicella in all

age groups (including infants and adults) in 1999 compared with 1995. 26

Page 28: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

27

Strategies to improve vaccination rates in the USA J To facilitate the uptake of universal vaccination, it is recommended that proof of

varicella vaccination or positive VZV serostatus should be considered a requirement

for school entry (category 2 recommendation).

Despite the availability of varicella vaccine in the USA, significant numbers of preventable

deaths from varicella continue to occur among both children and adults.31-33 The US

Department of Health and Human Services has set an objective to achieve >90% vaccine

coverage of children 19–35 months of age, and >95% at school entry by the year 2010.34

To increase vaccine uptake, it is proposed that proof of varicella vaccination or positive VZV

serostatus should be considered a requirement for school entry.

Mathematical models of varicella incidence following universal childhoodvaccinationJ If a policy of routine childhood vaccination is pursued, it is recommended that all

children should be immunized, with countries following the model used in the USA.

Immunizing only a subset of the paediatric population is likely to increase the risk of

creating a population of adults vulnerable to varicella and its attendant morbidity

(category 2 recommendation).

The initial evidence suggests that the introduction of universal childhood vaccination in

the USA is reducing the overall morbidity of varicella. However, the data are from the first

5 years of the programme and it remains to be seen what levels of coverage will be

achieved in the longer term. Mathematical modelling suggests that the success of universal

vaccination is heavily dependent on the level of vaccination coverage achieved. It predicts

that if the coverage were too low, there would be an ongoing circulation of wild-type virus,

albeit at a reduced level. This would probably lead to a gradual increase in the age of

varicella disease onset and move it into older, more susceptible age groups who are at

greater risk of varicella disease complications.35,36 However, higher levels of coverage

would reduce the overall incidence of varicella and its complications.

Halloran and colleagues (1994)37 used an age-structured transmission model to investigate

the effects of routine varicella immunization of pre-school children over a period of

10–50 years. It compared the age distribution and overall morbidity of varicella in a

number of scenarios, which differed according to:

• Level of vaccine coverage (50%, 70%, 97%); the target coverage in each case being

attained 6 years after introduction of the immunization programme, and vaccination

at this level continued for 70 years37

• Whether a catch-up programme was instituted; 12-year-old children who had neither

a history of varicella, nor prior varicella vaccination, were vaccinated. The catch-up

programme was initiated 11 years after the introduction of the vaccine to pre-school

children and continued for an 11-year period.

Page 29: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

28

The model predicted that 30 years after initiation of the vaccine programme, the average

number of varicella cases and hospitalizations per year would decrease irrespective of the

level of vaccine coverage and the utilization of a catch-up programme. However, the

greatest reduction was seen with the 97% vaccine coverage which was larger than if a

catch-up programme accompanied the 70% or 50% coverage scenarios (Table 2).37

Despite these results, varicella is occurring in targeted cities, suggesting that the model

may have flaws. It does, however, highlight the importance of attaining a high level of

coverage when implementing a routine vaccination programme. Currently, varicella

vaccine coverage is only 50% in the USA, although a higher percentage is likely when a

combined varicella-measles-mumps-rubella vaccine becomes available.26

The model demonstrated that a varicella vaccine programme would shift the age

distribution of varicella cases upwards for those who were not vaccinated. Although older

individuals are at high risk of the complications of varicella, the overall reduction in cases,

especially with high vaccine coverage, resulted in decreased overall morbidity as

measured by the number of primary cases and hospitalizations.37 The model is limited by

the unknown relationship between incidence of herpes zoster and varicella; exposure to

varicella may boost immune response and provide some protection against the former.

The Halloran model is being used by many countries to evaluate the cost-effectiveness of

varicella vaccination. However, there are limitations of the model, one being that the

vaccination efficacy parameters were optimistic.38 Brisson and colleagues39 revisited the

Halloran model to quantify key parameters describing varicella vaccine efficacy. These were

identified by finding the best fit of the predicted number of annual breakthrough infections

(defined as that occurring in seroconverted individuals, which is clinically modified and less

severe than natural varicella) that was observed in three trials conducted in the USA on

different batches of the varicella vaccine. Their model used the assumption that immunity

wanes over time, although this has not been observed to date. The findings suggest that

vaccination appears to result in a high proportion (97%) of individuals who are initially

protected, but that they lose this protective immunity at a rate of 3% per year. Consequently,

these individuals would be likely to develop breakthrough varicella if exposed to VZV later

in life. Brisson and colleagues39 concluded that studies conducted using Halloran et al’s

data might overestimate the effectiveness of varicella immunization.

Other limitations of the Halloran model include the fact that its age structure did not

accurately reflect the epidemiology of varicella, no sensitivity analysis was performed on

patterns of mixing, and it did not explore the possible effects of immunization on herpes

zoster occurrences. Addressing these aspects, Brisson et al,38 therefore, developed a model

to simulate the incidence of varicella and herpes zoster before and after universal

vaccination, using Canada as an example.

TABLE 2: Predictedannual average number of varicella cases andnumber of hospitalizationsby vaccine coverage andpre-vaccineimplementation37

Without catch-up programme With catch-up programme

Coverage (%) Number of cases (mean per year) Hospitalizations Number of cases (mean per year) Hospitalizations

Pre-vaccine 3960000 9870 3960000 987050 2125000 6268 1698000 471770 1190000 3958 786000 247997 240000 610 202000 504

Page 30: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

The modelling by Brisson et al38 demonstrated that varicella incidence and morbidity

(measured by in-patient days) would be reduced by mass vaccination of 12-month-old

children. This reduction in overall morbidity occurred despite a shift in the average age of

infection and an increase in morbidity with age. In agreement with the earlier work by

Halloran et al,37 the authors concluded that the overall effectiveness of immunization is

highly dependent upon the level of vaccine coverage achieved. Varicella morbidity was

predicted to decrease if coverage was more than 70%, with >80% having the greatest

impact. However, even at such high levels, epidemics are likely to occur because not all

would have been vaccinated. The model showed that use of catch-up campaigns could

reduce epidemics as well as minimize the overall number of varicella cases. The successful

catch-up strategies vaccinated children aged 1–11 years in the first year of a vaccination

programme or implemented an annual catch up of 11-year-olds for the first 11 years of the

programme. Which of these choices is preferable depends on the decision-makers’ attitudes

to current versus future health benefits.

The Brisson model also predicted that an effective vaccination strategy would lead to a

short- to medium-term increase in the incidence of herpes zoster. The assumption used in

the model was that exposure to wild-type varicella prevents, or delays, the development

of herpes zoster.39 However, vaccination may itself prevent herpes zoster, a possibility that

was not modelled in the simulations. Data from a study of 192 children with leukaemia,

where half received vaccine and half did not, support this theory.40,41 The incidence of

herpes zoster was significantly lower in the vaccinated children than in those who had

experienced natural infection. Continued, long-term follow-up in the USA, and the results

of the ongoing trial of the effect of vaccination on herpes zoster, will determine exactly

how varicella vaccination will influence the epidemiology of varicella and herpes

zoster.1,42-44

Targeted Vaccination J Programmes should be established to screen for VZV-seropositivity in healthcare

workers, teachers and other adults likely to come into sustained contact with VZV-

infected children and to vaccinate susceptible individuals. This would serve to protect

healthcare workers from VZV-infected patients, protect susceptible patients from

healthcare workers with varicella and protect seronegative teachers from VZV-infected

children (category 2 recommendation).

J Targeted vaccination of at-risk groups is recommended in countries not considering

universal vaccination programmes. Such programmes should include teachers,

healthcare workers, identified seronegative adults, and seronegative women of

childbearing age who are not pregnant (category 2 recommendation).

In addition to recommending universal vaccination of children, the ACIP strongly

recommends vaccination of all susceptible individuals aged ≥13 years at high risk of

exposure to VZV20 (see text box page 30). In Germany, varicella vaccination has been

recommended for susceptible individuals at risk of varicella and their contacts.45 In the UK,

it has been proposed that immunocompromised patients and susceptible healthcare workers

be immunized, but such an approach has not, as yet, gained widespread acceptance.46

Targeted vaccination has the advantage that it is less costly than universal and protects high-

risk individuals, and also that immunological boosting from circulating wild-type VZV in the

29

Page 31: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

community is not forfeited, as would happen with a universal vaccination programme. The

major difficulty encountered with targeted vaccination is in its implementation, as it depends

largely on the vigilance and commitment of each medical practitioner. In addition, many of

the at-risk populations require two doses of vaccine, adding to the logistical problems. There

may also be people who have severe immunosuppressive disorders, making them unsuitable

for vaccination, but who are still at risk from naturally acquired varicella. Finally, as with

universal vaccination, there is the issue of potential waning immunity over time.

Just as universal vaccination has been shown to be cost-effective, so targeted vaccination

of at-risk groups has been shown to yield economic benefits. For example, in a study of

472 leukaemic children, vaccination was calculated to be 11–13 times cheaper than

treatment of varicella.29

The targeted vaccination of women of childbearing age has been estimated to cost

US$7000 per case of varicella prevented.47 In the model used, it was assumed that 95%

of women were seropositive, the incidence of varicella in pregnancy was three women per

1000 and that all women were screened regardless of disease history. The intervention

might have been cost-saving if the costs of diagnosis and treatment of varicella, and those

relating to congenital varicella, were taken into account. A similar analysis using data from

a clinical population in the UK found that vaccination of women was cost-saving. In the

population considered, 80% of women were VZV-seropositive and the incidence of

varicella in pregnancy was 7.7 cases per 1000 women.48 Thus, vaccination of women may

be cost-effective if the seroprevalence in that element of the population is low.

A strategy of serological screening of healthcare workers with no history of varicella (or

herpes zoster), followed by vaccination of non-immune workers, is a cost-effective

strategy.49 Savings accrue from reduced absenteeism by employees with varicella and

reduced medical expenses for patients who contract varicella from healthcare workers.49,50

The American Medical Association has recommended that all susceptible healthcare

workers receive the varicella vaccine.51

SummaryVaricella is generally regarded as a benign, self-limiting disease in otherwise healthy children;

however, infants, adults, adolescents, pregnant women and the immunocompromised are at

risk of more severe disease with serious outcomes and are more likely to experience

complications.

A live, attenuated varicella vaccine (VZVoka vaccine) was developed in Japan in 1974. All

currently available varicella vaccines are based on the VZVoka strain, and experience of their use

for the prevention of varicella has accumulated over the past 25 years in Japan and the USA.

30

High-risk groups appropriate for varicella vaccination (susceptible individuals)20

• VZV-seronegative persons living or working in environments where transmission of VZV is likely (e.g. healthcare workers, teachers ofyoung children, child daycare employees)

• VZV-seronegative persons living and working in environments where transmission can occur (e.g. college students, inmates and staffmembers of correctional institutions, military personnel)

• VZV-seronegative non-pregnant women of childbearing age

• VZV-seronegative adolescents and adults living in households with children

• VZV-seronegative international travellers

Page 32: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Varicella vaccination is effective in children, susceptible adolescents and adults, including

selected groups of immunocompromised children, such as those with leukaemia in

remission and selected transplant recipients. Whilst vaccination of certain

immunocompromised children is beneficial, there is a lack of data regarding the use of

varicella vaccine in immunocompromised adults.

The effectiveness in a country of a universal vaccination programme in reducing the

overall morbidity due to varicella is largely dependent upon the actual coverage achieved.

If it is low, there will be an ongoing circulation of wild-type virus which, in the presence

of some vaccinated individuals, may lead to an increase in the age of onset of varicella.

This will shift the disease burden into older, more susceptible age groups who are at

greater risk of complications.

Targeted vaccination of at-risk individuals, such as healthcare workers, teachers and non-

pregnant women of childbearing age, is less costly than universal vaccination whilst still

providing protection for those individuals at greatest risk of complications. The main

disadvantages of targeted vaccination are that it relies upon the vigilance of individual

medical practitioners and that there may be those, such as immunocompromised adults,

who are unsuitable for vaccination but who are at risk from naturally acquired varicella.

Routine vaccination of children is recommended as are screening programmes to identify

susceptible adolescents and adults. Healthcare workers, teachers and non-pregnant

women of childbearing age should be considered priority groups for screening and

vaccination as necessary. Continuing surveillance of the vaccinated is required to assess

long-term efficacy, safety and effect on the occurrence of herpes zoster.

References1. Meyer PA, Seward JF, Jumaan AO et al. Varicella mortality: trends before vaccine licensure in the United

States, 1970-1994. J Infect Dis 2000;182(2):383-390.2. Preblud SR. Age-specific risks of varicella complications. Pediatrics 1981;68:14-17.3. Feldman S, Hughes WT, Daniel CB. Varicella in children with cancer: Seventy-seven cases. Pediatrics

1975;56(3):388-397.4. Perronne C, Lazanas M, Leport C et al. Varicella in patients infected with the human immunodeficiency

virus. Arch Dermatol 1990;126(8):1033-1036.5. Asano Y. Varicella vaccine: the Japanese experience. J Infect Dis 1996;174 (Suppl 3):S310-S313.6. Asano Y, Nagai T, Miyata T. Long-term protective immunity of recipients of Oka strain of live varicella

vaccine. Pediatrics 1985;75:667-671.7. Gershon A, Steinberg S, Gelb L. A multicentre trial of live attenuated varicella vaccine in children with

leukaemia in remission. Postgr Med J 1985;61(Suppl):S73-S78.8. Weibel RE, Neff BJ, Kuter BJ. Live attenuated varicella virus vaccine. Efficacy trial in healthy children. N Engl

J Med 1984;310:1409-1415.9. Kuter BJ, Weibel RE, Guess HA. Oka/Merck varicella vaccine in healthy children: final report of a 2-year

efficacy study and 7-year follow-up studies. Vaccine 1991;9:643-647.10. Gershon A, LaRussa P, Hardy I. Varicella vaccine: the American experience. J Infect Dis 1992;166

(Suppl 1):S63-S68.11. White CJ, Kuter BJ, Hildebrand CS. Varicella vaccine (VARIVAX) in healthy children and adolescents: results

from clinical trials 1987–1989. Pediatrics 1991;87:604-610.12. Varis T, Vesikari T. Efficacy of high-titer live attenuated varicella vaccine in healthy young children. J Infect

Dis 1996;174 (Suppl 3):S330-S334.13. Kuter BJ, Ngai A, Patterson CM et al. Safety, tolerability, and immunogenicity of two regimens of Oka/Merck

varicella vaccine (Varivax) in healthy adolescents and adults. Oka/Merck Varicella Vaccine Study Group.Vaccine 1995;13(11):967-972.

14. Gershon AA, Steinberg SP, National Institute of Allergy and Infectious Diseases Varicella VaccineCollaborative Study Group. Live attenuated varicella vaccine: Protection in healthy adults compared withleukemic children. J Infect Dis 1990;161:661-666.

15. Gershon AA, Steinberg SP, LaRussa P et al. Immunization of healthy adults with live attenuated varicellavaccine. J Infect Dis 1988;158(1):132-137.

16. Prevention of varicella: Recommendations of the Advisory Committee on Immunization Practices (ACIP).Centers for Disease Control and Prevention. MMWR Morb Mortal Wkly Rep 1996;45(RR-11):1-36.

17. Johnson CE, Stancin T, Fattlar D et al. A long-term prospective study of varicella vaccine in healthy children.Pediatrics 1997;100(5):761-766.

31

Page 33: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

18. Asano Y, Suga S, Yoshikawa T et al. Experience and reason: twenty-year follow-up of protective immunity ofthe Oka strain live varicella vaccine. Pediatrics 1994;94(4 Pt 1):524-526.

19. Gershon AA. Varicella-zoster virus: prospects for control. Adv Pediatr Infect Dis 1995;10:93-124.20. Prevention of varicella. Update recommendations of the Advisory Committee on Immunization Practices

(ACIP). MMWR Morb Mortal Wkly Rep 1999;48(RR-6):1-5.21. Takahashi M. Current status and prospects of live varicella vaccine. Vaccine 1992;10(14):1007-1014.22. Levin MJ, Barber D, Goldblatt E et al. Use of a live attenuated varicella vaccine to boost varicella-specific

immune responses in seropositive people 55 years of age and older: duration of booster effect. J Infect Dis1998;178 Suppl 1:S109-112.

23. Raeder CK, Hayney MS. Immunology of varicella immunization in the elderly. Ann Pharmacother2000;34(2):228-234.

24. Krause PR, Klinman DM. Varicella vaccination: evidence for frequent reactivation of the vaccine strain inhealthy children. Nat Med 2000;6(4):451-454.

25. Hata A, Asanuma H, Rinki M et al. Use of an inactivated varicella vaccine in recipients of hematopoietic-cell transplants. N Engl J Med 2002;347:26-34.

26. American Academy of Pediatrics. Committee on Infectious Diseases. Varicella vaccine update. Pediatrics2000;105(1 Pt 1):136-141.

27. Preblud SR, Orenstein WA, Koplan JP et al. A benefit-cost analysis of a childhood varicella vaccinationprogramme. Postgrad Med J 1985;61(Suppl 4):S17-S22.

28. Lieu TA, Cochi SL, Black SB et al. Cost-effectiveness of a routine varicella vaccination program for USchildren. JAMA 1994;271(5):375-381.

29. Lieu TA, Finkler LJ, Sorel ME et al. Cost-effectiveness of varicella serotesting versus presumptive vaccinationof school-age children and adolescents. Pediatrics 1995;95(5):632-638.

30. Huse DM, Meissner HC, Lacey MJ et al. Childhood vaccination against chickenpox: an analysis of benefitsand costs. J Pediatr 1994;124(6):869-874.

31. Spingarn RW, Benjamin JA. Universal vaccination against varicella [letter]. N Engl J Med 1998;338(10):683;discussion 684.

32. From the Centers for Disease Control and Prevention. Varicella-related deaths among children–UnitedStates, 1997. JAMA 1998;279(22):1773-1774.

33. Varicella-related deaths among children—United States, 1997. Can Commun Dis Rep 1998;24(13):108-111.

34. Healthy People 2010 Objectives: Draft for Public Comment. Immunizations and infectious diseases.1998:22-36.

35. Halloran ME. Epidemiologic effects of varicella vaccination. Infect Dis Clin North Am 1996;10(3):631-655.36. Ferguson NM, Anderson RM, Garnett GP. Mass vaccination to control chickenpox: the influence of zoster.

Proc Natl Acad Sci USA 1996;93(14):7231-7235.37. Halloran ME, Cochi SL, Lieu TA et al. Theoretical epidemiologic and morbidity effects of routine varicella

immunization of preschool children in the United States. Am J Epidemiol 1994;140(2):81-104.38. Brisson M, Edmunds WJ, Gay NJ et al. Modelling the impact of immunization on the epidemiology of

varicella zoster virus. Epidemiol Infect 2000;125(3):651-669.39. Brisson M, Edmunds WJ, Gay NJ et al. Analysis of varicella vaccine breakthrough rates: implications for the

effectiveness of immunisation programmes. Vaccine 2000;18(25):2775-2778.40. Hardy I, Gershon AA, Steinberg SP et al. The incidence of zoster after immunization with live attenuated

varicella vaccine. A study in children with leukemia. Varicella Vaccine Collaborative Study Group. N Engl JMed 1991;325(22):1545-1550.

41. Lawrence R, Gershon AA, Holzman R et al. The risk of zoster after varicella vaccination in children withleukemia. N Engl J Med 1988;318(9):543-548.

42. Nowgesic E, Skowronski D, King A et al. Direct costs attributed to chickenpox and herpes zoster in BritishColumbia–1992 to 1996. Can Commun Dis Rep 1999;25(11):100-104.

43. Deguen S, Chau NP, Flahault A. Epidemiology of chickenpox in France (1991–1995). J EpidemiolCommunity Health 1998;52 (Suppl 1):46S-49S.

44. Fairley CK, Miller E. Varicella-zoster virus epidemiology–a changing scene? J Infect Dis 1996;174 (Suppl3):S314-S319.

45. Impfempfehlungen der Ständigen Impfkomission (STIKO) am Robert-Koch-Institut. EpidemiologischesBulletin 15/97 1997.

46. Jones EM, Reeves DS. Controlling chickenpox in hospitals. BMJ 1997;314(7073):4-5.47. Seidman DS, Stevenson DK, Arvin AM. Varicella vaccine in pregnancy. BMJ 1996;313(7059):701-702.48. Griffiths PD, Volpi A. Management Strategies in Herpes: Progress with Diagnostic Tests and Vaccines for

Alphaherpesviruses. Worthing, PAREXEL MMS Ltd, 1998.49. Gray AM, Fenn P, Weinberg J et al. An economic analysis of varicella vaccination for health care workers.

Epidemiol Infect 1997;119(2):209-220.50. Weinstock DM, Rogers M, Lim S et al. Seroconversion rates in healthcare workers using a latex agglutination

assay after varicella virus vaccination. Infect Control Hosp Epidemiol 1999;20(7):504-507.51. Lyznicki JM, Bezman RJ, Genel M. Report of the Council on Scientific Affairs, American Medical

Association: immunization of healthcare workers with varicella vaccine. Infect Control Hosp Epidemiol1998;19(5):348-353.

32

Page 34: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

33

MANAGEMENT OF VARICELLA IN THE

IMMUNOCOMPETENT HOST4

Management RecommendationsCategory 2Antiviral treatmentJ Oral aciclovir (20 mg/kg up to 800 mg four times daily for 5 days) is recommended as

the treatment of choice for otherwise healthy children up to the age of 12 years with

varicella. Valaciclovir and famciclovir are likely to be as effective as aciclovir, but have

not been studied in controlled clinical trials and no paediatric formulation of

valaciclovir or famciclovir exists.

J Complications are more likely and frequently more serious in nature in adults and

adolescents than in children. They are also more likely and often more severe in

secondary cases in a household than in the index case. Therefore, treatment with oral

aciclovir (800 mg four to five times daily for 5–7 days) should be offered to all adults

and adolescents with varicella presenting within 48 hours of rash onset.

Category 3Antiviral treatmentJ Valaciclovir (1000 mg three times daily) and famciclovir (250 mg or 500 mg three

times daily) are likely to be as effective as aciclovir; although they have not been

studied in controlled clinical trials, pharmacokinetic data support their use.

J It is recommended that all patients presenting with varicella-associated pneumonia,

whether pregnant or not, should receive intravenous aciclovir (10 mg/kg every

8 hours).

J It is recommended that post-exposure prophylaxis of pregnant women and neonates

be provided with varicella zoster immune globulin (VZIG). Prophylactic aciclovir

(800 mg five times daily) and varicella vaccine could play a role in these settings but

have not been the subject of controlled studies.

J It is recommended that treatment of more severe cases of varicella in children, and

varicella in adults, adolescents and other at-risk individuals, is maintained as a priority

even in the presence of a varicella vaccination programme. For example, post-

exposure prophylaxis with aciclovir in pregnant women should theoretically be

effective but this strategy is currently not advocated, at least during the first

20 weeks of pregnancy.

Varicella vaccineJ Varicella vaccine may be considered for post-exposure prophylaxis in

immunocompetent hosts as it will effectively prevent or ameliorate varicella when

Page 35: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

34

given to a susceptible individual within 3–5 days after exposure to varicella zoster

virus (VZV). Although post-exposure vaccination and post-exposure aciclovir (800 mg

fives times daily) administration have not been compared directly, they are likely to

have similar efficacy, while the former is probably less expensive.

J Post-exposure prophylaxis with aciclovir (800 mg five times daily) may remain a

viable option when the VZV exposure is not recognized until more than 5 days later.

J The use of oral aciclovir (800 mg five times daily), valaciclovir or famciclovir for

pregnant women who contract varicella in their second or third trimester is

recommended. However, it is important to note that this recommendation is based on

anecdotal evidence, and that patients should be advised that these drugs are not

licensed for use during pregnancy.

J Physicians should be aware of rare but serious complications and atypical

manifestations of varicella, such as cerebellar ataxia, varicella-associated pneumonia,

VZV encephalitis and cutaneous bacterial complications. The use of intravenous

aciclovir (10 mg/kg every 8 hours) is warranted in these cases, based on anecdotal

evidence.

J Pre-pregnancy serological screening of non-pregnant women of childbearing age and

varicella vaccination are recommended. This approach has the potential to prevent

maternal varicella, congenital varicella syndrome and neonatal varicella where the

infant acquires their infection from the mother.

J Neonates (<28 days old) who are exposed to varicella post-natally should be

administered prophylactic VZIG.

Research needsJ It is recommended that further investigation be conducted to assess whether pregnant

women who contract varicella during the first trimester of pregnancy should be

administered intravenous aciclovir (10 mg/kg every 8 hours). There is currently no

evidence that this treatment results in fetal malformations or that it prevents congenital

varicella.

J It is recommended that the efficacy of valaciclovir and famciclovir be assessed in

adults with varicella. It may be applicable for such studies to be conducted in tropical

countries due to the larger potential adult study population.

Although varicella is a generally benign and self-limiting disease, as discussed in

Chapter 2, there are potentially serious complications associated with it. These are more

likely to manifest in neonates, pregnant women, adolescents and adults, but can, in rare

instances, occur in otherwise healthy children. The potentially serious nature of these

complications highlights the need for effective prevention and management of varicella in

high-risk individuals.

Page 36: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

35

Pre-Exposure Varicella VaccinationAs discussed in Chapter 3, experience with the live attenuated VZVoka vaccine for

prevention of varicella has accumulated over the last 25 years in Japan and the USA. The

vaccine is well tolerated and effective in healthy children and strongly indicated in VZV-

seronegative adolescents and adults.1,2 The vaccine offers long-term protection against

clinically severe varicella.3 For a detailed discussion, see Chapter 3.

Post-Exposure ProphylaxisJ Varicella vaccine may be considered for post-exposure prophylaxis in

immunocompetent hosts as it will effectively prevent or ameliorate varicella when

given to a susceptible individual within 3–5 days after exposure to VZV (category 3

recommendation). Although post-exposure vaccination and post-exposure aciclovir

(800 mg fives times daily) administration have not been compared directly, they are

likely to have similar efficacy, while the former is probably less expensive.

J Post-exposure prophylaxis with aciclovir (800 mg five times daily) may remain a viable

option when the VZV exposure is not recognized until more than 5 days later (category 3

recommendation).

Due to the potentially serious nature of the complications of varicella, and the costs

associated with treating and managing patients presenting with severe disease, prevention

of varicella in susceptible individuals following exposure to VZV is likely to be both

beneficial to the patient and cost-effective.

Post-exposure prophylaxis with varicella vaccine A small, controlled study of 40 immunocompetent children indicates that the varicella

vaccine is likely to be effective for post-exposure prophylaxis of varicella in children,

provided it is administered sufficiently soon after exposure to VZV.4 In the study,

experimental VZVoka vaccines of varying doses administered within 72 hours of initial

exposure to VZV prevented the development of varicella symptoms; all of the

unvaccinated controls developed clinical varicella.4 Case studies also support the concept

of post-exposure immunization to prevent varicella. In a study of 10 susceptible siblings

given varicella vaccine within 72 hours of lesion onset in the index case, five did not

develop varicella and five developed only mild disease (four of five exhibited <20

lesions).5 In a recent study, post-exposure administration of varicella vaccine limited

spread of varicella among children and adults in a women’s refuge.6 In the study, residents

of the refuge with no history of varicella were offered vaccination when one of the

residents and her child developed varicella. This was administered within 3 days of the

rash onset. None of 25 adults and only two of 42 children developed illness. However, the

two children with varicella were siblings of the index case, and were housed in the same

room. The illness that developed in the children was brief, afebrile and was characterized

by fewer than 20 lesions (Table 1). The mildness of the illness in the secondary cases

suggests that the vaccine ameliorated the disease as, untreated, varicella is usually more

severe in secondary and tertiary household contacts compared with the index case.7

However, as there was no control group, the actual effect of vaccination cannot be

determined.

Page 37: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

36

Despite issues with some of these studies, including the small numbers of subjects, the

different vaccine formulations used, and the inadequate assessment of susceptibility, the

US Advisory Committee on Immunization Practices (ACIP) now recommends varicella

vaccine for susceptible persons following exposure to varicella. Vaccine should be

administered within 3–4 days of exposure.2 In terms of cost, post-exposure vaccination is

less expensive than post-exposure prophylaxis with aciclovir; both of these options are less

expensive than VZIG administration.8

Post-exposure prophylaxis with antiviralsA small number of studies have investigated post-exposure prophylaxis with aciclovir for

susceptible immunocompetent children following VZV exposure. Aciclovir may be

effective in preventing the development of varicella.

In 50 exposed infants and children, oral aciclovir (40–80 mg/kg/day) initiated 7–9 days

after exposure to VZV and administered for 7 days prevented the development of clinical

varicella in 21 out of 25 treated children; all of the 25 untreated children in the control

group developed varicella.9 In another study, in which children were given oral aciclovir

(40 mg/kg/day) 1–3 days after exposure to VZV, 10 out of 13 children developed varicella

within 28–42 days.10 These studies were performed in different populations, some of

whom received different doses of aciclovir and were followed up for different lengths of

time, making it difficult to draw direct comparisons. Despite this, the data suggest that

post-exposure prophylaxis with aciclovir is less effective if administered too soon

following VZV exposure. Up to 20% of individuals receiving aciclovir for post-exposure

prophylaxis may not seroconvert; this may be due to lack of actual exposure to VZV, or

there may be blunting of the immune response following the termination of VZV

replication by aciclovir.9,11,12

If susceptible patients can be identified within 5 days of varicella exposure, post-exposure

vaccination would be a more efficient approach than aciclovir administration.13 However,

there is minimal and conflicting evidence regarding protection when vaccine is received

4 to 5 days after exposure.4

Antiviral TreatmentJ Oral aciclovir (20 mg/kg up to 800 mg four times daily for 5 days) is recommended as

the treatment of choice for otherwise healthy children up to the age of 12 years with

varicella (category 2 recommendation). Valaciclovir and famciclovir are likely to be as

effective as aciclovir, but have not been studied in controlled clinical trials and no

paediatric formulation exists.

TABLE 1: Summary ofstudies of varicellavaccine used for post-exposure prophylaxis

Population Study Vaccine administered Results Reference

40 children Vaccination versus Within 72 hours of No cases of varicella in the Asano Y et al. 19774

no vaccination initial exposure to VZV vaccinated group; all developed varicella in the group that was not vaccinated

10 children Susceptible siblings Within 72 hours of lesion 5 had no disease; 5 developed mildwere given varicella vaccine onset in the index case disease (4 of these had <20 lesions) Salzman M, et al. 19985

42 children and Individuals with no Within 3 days of rash onset None of the adults developed Watson B et al. 20006

25 adults history of varicella were in mother and her child illness; 2 children developed offered vaccination mild illness

Page 38: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

37

J Complications are more likely, and frequently more serious, in adults and adolescents

than in children. They are also more likely and often more severe in secondary

household cases than the index case. Therefore, treatment with oral aciclovir (800 mg

four to five times daily for 5–7 days) should be offered to all adults and adolescents

with varicella presenting within 48 hours of rash onset (category 3 recommendation).

J Valaciclovir (1000 mg three times daily) and famciclovir (250 or 500 mg three times

daily) are likely to be as effective as aciclovir, but have not been studied in controlled

clinical trials (category 3 recommendation).

J It is recommended that all patients presenting with varicella-associated pneumonia (or

symptoms or signs of respiratory involvement), whether pregnant or not, should

receive intravenous aciclovir (10 mg/kg every 8 hours) (category 3 recommendation).

J Physicians should be aware of rare but serious complications and atypical

manifestations of varicella, such as cerebellar ataxia, VZV encephalitis and cutaneous

bacterial complications. The use of intravenous aciclovir (10 mg/kg every 8 hours) is

warranted in these cases, based on anecdotal evidence (category 3 recommendation).

J It is recommended that studies of valaciclovir and famciclovir be conducted in adults

with varicella to assess efficacy. It may be applicable for such studies to be conducted

in tropical countries due to the larger potential adult study population (research need

recommendation).

Treatment options available are summarized in Table 2.

Prospective, controlled, large-scale

clinical trials document the efficacy of

oral aciclovir for the treatment of

varicella in immunocompetent

children, adolescents and adults.14-17

It increases the rate of rash resolution,

decreases the duration of febrile illness

and reduces the systemic symptoms of

varicella. The limited scale of these benefits means that the case for aciclovir treatment of

varicella, at least in otherwise healthy children, has not been universally accepted.

In immunocompetent children aged 2–12 years of age, oral aciclovir (20 mg/kg four times

daily for 5 days) initiated within 24 hours of rash onset resulted in a shorter duration of

febrile illness, fewer skin lesions (294 versus 347), and accelerated lesion healing (2.7 days

versus 3.2 days) compared with placebo (Table 3).16 Similarly, in adolescents, oral

aciclovir (800 mg four times daily for 5 days) initiated within 24 hours of rash onset was

more effective than placebo in reducing the duration of febrile illness and resulted in more

rapid cessation of lesion formation (by 0.5 days).15 Because the treatment provides a

relatively modest reduction in duration and severity of illness, the decision to treat children

should be individualized.

TABLE 2: Treatmentoptions for immuno-competent individualswith varicella8

Patient group Treatment options

Neonates Intravenous aciclovir for 10 days Children <12 years of age Symptomatic care only; oral aciclovir may

be considered

Adolescents/adults Oral valaciclovir, famciclovir or aciclovir for 5–7 days

Women in last trimester of pregnancy Oral aciclovir for 5–7 days

Patients with pneumonitis or other Intravenous aciclovir for 7–10 dayssevere infection

Page 39: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

38

TABLE 3: Cutaneousevents in children withvaricella treated withaciclovir or placebo16

Aciclovir group Placebo group P-value

Median number of lesions 294 (277) 347 (386) <0.001 Number with >500 lesions (%) 78 (21.3) 137 (38.4) <0.001 Median number of residual lesions on D28 13 (6) 33 (13) <0.001

In adults, oral aciclovir (800 mg five times daily for 7 days) administered within 24 hours

of rash onset was beneficial compared with placebo. Aciclovir accelerated the time to

cutaneous healing, decreased the duration of fever (by 1.8 days), and lessened systemic

symptoms (Figure 1).17 However, if therapy was administered after 24 hours following rash

onset, there was no appreciable benefit.

The results of these trials have been confirmed by data from a recent study of aciclovir

versus placebo in 177 children, adolescents and adults with varicella.18 In this study,

maximum clinical benefit was achieved when aciclovir therapy was initiated within 24

hours of rash onset, although children and adolescents (but not adults) showed significant

improvement in clinical end-points when aciclovir therapy was initiated 48 hours after

rash onset.18 The populations studied in these trials

were not sufficiently large, or the incidence of

complications was too low, to assess the impact of

aciclovir therapy on the incidence of varicella

complications.

Valaciclovir and famciclovir are likely to be as

effective as aciclovir for treatment of varicella, as

pharmacokinetic data show that they achieve serum

levels of aciclovir or penciclovir at least as high as

the parent compounds.19,20 No data from

controlled, prospective trials have yet been

published to support this indication. However, a

family case study indicates that valaciclovir has potential as a therapy for adult varicella.21

As a consequence of the limited clinical benefit of antiviral therapy, many clinicians view

it as an optional treatment for otherwise healthy children with varicella. For example, the

Committee on Infectious Disease of the American Academy of Pediatrics (AAP) does not

consider the prescribing of aciclovir to healthy children to be of sufficient benefit to justify

routine administration.22 However, it acknowledges that aciclovir use is justified in some

cases, as complications and severe varicella may occur in adolescents and adults, or in

secondary-case patients who live in the households of infected children.1

The role of antiviral therapy in individuals presenting with rarer complications of varicella,

such as cerebellar ataxia and encephalitis, has not been studied in a prospective or

controlled fashion. Despite this, administration of intravenous aciclovir (10 mg/kg every

8 hours) to such patients is likely to be appropriate, based upon anecdotal evidence.

(J Gnann, RJ Whitley, personal communication.)

As there are no published data from randomized, controlled trials available on the efficacy

of valaciclovir or famciclovir for the treatment of varicella in the immunocompetent host,

specific recommendations regarding their use, especially in high-risk populations, cannot

be made. However, following valaciclovir administration, significantly higher plasma

FIGURE 1: Dailyconstitutional illnessscores of adults withvaricella treated withaciclovir or placebowithin 24 hours of rashonset17

1 2Day of study3 4 5

2.0

1.6

1.2

0.8

0.4

0

Illne

ss s

core

s

0.2

0.6

1.0

1.4

1.8

6 7

AciclovirPlacebo

P=0.141

P=0.049

P=0.002

P=0.087

P=0.082P >0.2

P >0.2

© R

epro

duce

d w

ith p

erm

issi

on, W

alla

ce M

R, B

owle

r W

A, M

urra

yN

Bet

al.

Trea

tmen

t of a

dult

vari

cella

with

ora

l acy

clov

ir. A

rand

omiz

ed, p

lace

bo-c

ontr

olle

d tr

ial.

Ann

Inte

rn M

ed19

92;1

17(5

):358

-363

. Am

eric

an C

olle

ge o

f Phy

sici

ans.

Page 40: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

39

levels of aciclovir are achieved than are possible following administration of oral

aciclovir.19 Similarly, famciclovir delivers high levels of penciclovir.20 Both valaciclovir

and famciclovir are likely to be effective for the treatment of varicella, and at least as

effective as aciclovir at preventing complications.

Prevention and Management of Varicella in the Pregnant Womanand NeonateJ It is recommended that post-exposure prophylaxis of pregnant women and neonates is

based on passive immunization with VZIG. Prophylactic aciclovir (800 mg five times

daily) and varicella vaccine could play a role in these settings but have not been the

subject of controlled studies (category 3 recommendation).

J The use of oral aciclovir (800 mg five times daily) for pregnant women who contract

varicella in their second or third trimester is recommended. It is important to note that

this recommendation is based on anecdotal evidence, and that patients should be

advised that antiviral drugs are not licensed for use during pregnancy (category 3

recommendation). The roles of valaciclovir and famciclovir for the treatment of

varicella infection in the pregnant woman remain to be evaluated in clinical trials.

J Pre-pregnancy serological screening of non-pregnant women of childbearing age and

varicella vaccination are recommended. This approach has the potential to prevent

maternal varicella, congenital varicella syndrome and neonatal varicella where the

infant acquires infection from the mother (category 3 recommendation).

J Neonates (<28 days old) who are exposed to varicella post-natally should be

administered prophylactic VZIG (category 3 recommendation).

J It is recommended that further investigation be conducted to assess whether pregnant

women who contract varicella during the first trimester of pregnancy should be

administered intravenous aciclovir (10 mg/kg every 8 hours). There is currently no evidence

that this treatment results in fetal malformations (research need recommendation).

The high risk of complications in the pregnant woman with varicella, coupled with the

potentially devastating fetal abnormalities caused by congenital varicella syndrome and

the risks to neonates if maternal varicella occurs immediately before or after birth,

emphasize the need for effective prevention and management.

Varicella vaccination in the pregnant womanAs discussed in Chapter 3, vaccination of pregnant women is not recommended because

of the theoretical risk to both the fetus and the mother.

Prevention of maternal varicella may be achieved by pre-pregnancy serological screening

of women of childbearing age and subsequent varicella vaccination as appropriate. This

approach would also help minimize the risk of congenital varicella syndrome and

neonatal varicella. The US ACIP recommends varicella vaccination for susceptible, non-

pregnant women of childbearing age.2

Page 41: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

40

For a detailed discussion on the use of varicella vaccination in pregnant women, see

Chapter 3.

Post-exposure prophylaxis in the pregnant womanThe aim of post-exposure prophylaxis in the pregnant woman is to prevent or modify

illness, and to reduce the risk of intrauterine VZV infection. It should be offered to

susceptible pregnant women with significant virus exposure (e.g. from their own family)

during the first 20 weeks of gestation, because secondary household cases may be more

severe and there is a 1–2% risk of congenital varicella.23 Before administering post-

exposure prophylaxis, it is important to ascertain the type of VZV exposure and the

previous varicella history of the pregnant woman. If there is any uncertainty regarding

infection history, VZV antibody status should be determined rapidly.

Unfortunately, in this time-critical scenario, the true VZV serological status of pregnant

women without a history of varicella is often not known. The clinician may be faced with

a decision as to whether to initiate prophylactic treatment empirically or wait for

serological test results. The ideal time to determine VZV serological status would be before

pregnancy, when vaccination can be offered to women confirmed to be VZV seronegative.

Antiviral prophylaxisJ Post-exposure prophylaxis with aciclovir in pregnant women should theoretically be

effective but this strategy is currently not advocated, at least not during the first 20

weeks of pregnancy (category 3 recommendation).

VZIG prophylaxis in pregnancyAs maternal varicella can result in fetal and neonatal morbidity, susceptible pregnant

women who have a close exposure to VZV should receive passive immunotherapy with

VZIG. In the UK, recommendations are that VZIG is effective in the pregnant woman if

given up to 10 days after initial exposure and to neonates up to 28 days post-partum.23

The American Academy of Pediatrics

(AAP) recommendations are shown in

Table 4; differences in UK and US

recommendations arose because the

findings did not agree. This may have

been due to differences in the VZIG

preparations used. Similarly, as antibody

content varies between countries, a

particular dose of VZIG cannot be

recommended.1

Although the purpose of VZIG administration in early pregnancy is principally the prevention

of maternal VZV infection, the possible attenuation of the disease with VZIG is also valuable.

In a study of 212 seronegative pregnant women, VZIG was administered either

intramuscularly (for a 70 kg woman: 2100 IU) or intravenously (for a 70 kg woman: 1750 IU)

and within 1–3 days of significant exposure to VZV. Forty-one per cent of the women

developed clinical varicella, and a further 5% developed subclinical infection (Table 5).22

TABLE 4: AAPrecommendations forimmunocompetentcandidates benefitingfrom VZIG administrationfollowing varicellaexposure22

AAP recommendations

• Susceptible pregnant women

• Newborn infants whose mothers have varicella within 5 days before or48 hours after delivery

• Hospitalized premature infants (>28 weeks gestation) whose mothers have nohistory of varicella

• Hospitalized premature infants (<28 weeks gestation or <1000 g) irrespective ofmaternal history

Page 42: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

41

TABLE 5: Outcome ofvaricella exposure in 212seronegative pregnantwomen followingadministration of VZIG23

Outcome

VZIG administration Number treated No infection Subclinical infection Modified/normal varicella (Days after exposure) n (%) n (%) n (%)

1–3 153 83 (54) 7 (5) 63 (41) 4–5 46 27 (59) 1 (2) 18 (39) 6–10 13 4 (31) 3 (23) 6 (46) Total 212 114 (54) 11 (5) 87 (41)

In summary, VZIG should be administered as soon as possible to susceptible (seronegative)

women following exposure to VZV in the first 20 weeks of gestation. VZIG prophylaxis for

women exposed later in pregnancy is a lower priority.23-29

There is limited evidence that VZIG benefits the fetus and reduces the risk of intrauterine

varicella infection.23,30 In a prospective study of 97 pregnant women who contracted

varicella despite receiving VZIG, there were no cases of congenital varicella syndrome,

and VZV-specific IgM antibodies were detectable at birth in only one infant (maternal

varicella in Week 36 of gestation).23 However, this study did not demonstrate a statistically

significant protective effect of VZIG to the fetus because the numbers were too small.31 In

another prospective study, 6 (5%) of 106 varicella-infected women received VZIG after

exposure in early pregnancy.32 The only infant with congenital varicella syndrome was

born to a woman who received VZIG. The VZV-susceptible mother received VZIG 4 days

after her exposure to varicella at Week 11 of pregnancy; she developed varicella 2 weeks

later. These two studies demonstrate the need for a large prospective trial to clarify whether

passive prophylaxis can prevent congenital varicella syndrome.

Post-exposure vaccination in pregnancyAs discussed above and in Chapter 3, post-exposure prophylaxis with vaccine is not advocated

for pregnant women, but is recommended for non-pregnant women of childbearing age.

Management of the pregnant woman with varicellaAntiviral treatment of varicella in pregnancyTreatment of varicella with oral aciclovir in adults, including pregnant women with a

normal course of illness, differs from country to country. In the UK and Australia, oral

aciclovir for 7 days (800 mg five times daily) is offered to individuals, including pregnant

women past 20 weeks of gestation, presenting within 24 hours of rash onset. This

procedure reduces the severity of disease in non-pregnant adults,1,15,17,33 but no studies

have prospectively assessed the efficacy of antiviral therapy in pregnant women. A

retrospective analysis of aciclovir in pregnant women with varicella pneumonia noted

reduced morbidity and mortality when varicella infection occurred during the last two

trimesters; aciclovir reduced mortality rates to 14% compared with 41% in untreated

historic controls.34

In pregnant women, the potential benefits of treatment and post-exposure prophylaxis

should be balanced against potential adverse outcomes in the fetus. Although aciclovir is

not licensed for use in pregnancy, the prospective follow-up of 1205 women treated during

the first (n=739), second (n=188) and third (n=278) trimester of pregnancy did not

demonstrate an increase in the number of birth defects when compared with that expected

in the general population.35,36 In addition, there is no consistent pattern of defects among

Page 43: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

42

prospective or retrospective reports of aciclovir use. Although the data from the Pregnancy

Registry are not strong enough to exclude a teratogenic effect of aciclovir, this treatment

should not be withheld in early pregnancy if clinically indicated.

As the complications of varicella are more common in adults, and given the limited

Pregnancy Registry data available, there is no apparent reason to withhold aciclovir at any

time during pregnancy. The dosage and route of administration are determined by the

severity of disease. If a woman has severe or complicated disease (e.g. pneumonitis),

intravenous aciclovir should be given (10 mg/kg every 8 hours for 7 days or longer).

Pregnant women with less severe disease should be treated with oral aciclovir (800 mg five

times per day for 7 days or longer). More data are required on long-term follow-up of

children exposed to aciclovir in utero. The roles of valaciclovir and famciclovir for the

treatment of varicella infection in pregnancy remain to be evaluated in clinical trials.

The pregnant woman with varicella should avoid contact with all other pregnant women

and neonates until her lesions are crusted.

Pregnant women exposed to herpes zosterFollowing exposure to herpes zoster, passive prophylaxis with VZIG is recommended only

for VZV-seronegative pregnant women up to 21 weeks of gestation intimately exposed to

patients with extensive herpes zoster lesions. Local guidelines for treating herpes zoster in

adults should be followed. Women who develop localized herpes zoster in pregnancy

should be reassured that the risk to the fetus is negligible. In addition, passive

immunization of neonates whose mothers develop perinatal herpes zoster is not indicated.

Post-exposure prophylaxis in the neonateVZIG prophylaxis in neonatesAdministration of VZIG to the infant is advised if the mother develops varicella within

7 days before or after delivery. The neonate of a mother with active varicella should be

isolated while in hospital, from birth to Day 21 (or Day 28 if the infant has been given

VZIG). Early discharge of the mother should be considered where feasible.

Despite VZIG prophylaxis, approximately one-half to two-thirds of infants exposed to

maternal varicella around the time of delivery will become infected.24 In the UK, VZIG is

also recommended for infants with non-maternal post-natal exposure during the first

7 days of life who test negative for VZV antibodies.

VZIG does not appear to reduce the infection rate for varicella in neonates although it may

reduce the risk of serious infection. In a study in which VZIG was administered to 41

neonates born to women whose varicella lesions began 4 days before to 2 days after

delivery, the neonatal attack rate was 51% despite VZIG therapy.37 There were no fatalities

and only two cases of severe disseminated varicella. In a larger UK study of 280 infants who

received VZIG after being born to mothers with varicella or herpes zoster in the perinatal

period, the attack rate was 48%, with the infection being severe in 19 infants (6.8%).38

Management of the neonate with varicellaAlthough in most cases neonatal VZV infection is mild, fatal outcomes in VZIG-treated

infants have occured in those whose mothers developed varicella rash in a narrow window

Page 44: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

43

of time (4 days before to 2 days after delivery).39-41 See also Table 4, the AAP

recommendations for VZIG administration to newborn infants whose mothers develop

varicella within 5 days before to 48 hours after delivery.

Neonates exposed to varicella should be observed closely. If they develop vesicles, they

should be treated as early as possible with intravenous aciclovir (10 mg/kg every 8 hours).

Therapy with aciclovir should be considered in neonates developing varicella despite

receiving VZIG prophylaxis.

For a more detailed discussion of varicella and its management in pregnant women and

neonates, the reader is directed to the previous IHMF Management Strategies in Herpes

publication – Herpesvirus Infections in Pregnancy.

SummaryAlthough generally benign and self-limiting, varicella can have serious complications

associated with it. These complications are more common and serious in neonates,

pregnant women, adolescents and adults; however, complications can occur, albeit rarely,

in otherwise healthy children. The potentially serious nature of these complications

highlights the need for effective treatment of varicella especially in high-risk individuals.

Varicella vaccine may be effective as post-exposure prophylaxis in immunocompetent hosts

when administered to a susceptible individual within 3–5 days of exposure to VZV.

Additionally, post-exposure prophylaxis with oral aciclovir (800 mg five times daily) may

remain a viable option when the VZV exposure is not recognized until more than 5 days later.

Oral aciclovir (20 mg/kg up to 800 mg four times daily for 5 days) is the treatment of

choice when antiviral therapy is indicated in otherwise healthy children less than 12 years

of age with varicella. Valaciclovir and famciclovir are likely to be as effective as aciclovir,

but have not been studied in controlled clinical trials.

Due to the increased severity of varicella and increased likelihood of complications in

adolescents and adults compared with children, oral aciclovir (800 mg four or five times

daily for 5–7 days) should be offered to all adults and adolescents with varicella presenting

within 48 hours of rash onset. Valaciclovir (1000 mg three times daily) and famciclovir

(250 mg or 500 mg three times daily) are likely to be as effective as aciclovir, but have not

been studied in controlled clinical trials.

Patients presenting with varicella-associated pneumonia or complications of varicella,

such as cerebellar ataxia and VZV encephalitis, should receive intravenous aciclovir

(10 mg/kg every 8 hours).

Post-exposure prophylaxis of pregnant women and neonates should be based on passive

immunization with VZIG. Prophylactic aciclovir and varicella vaccine are not advocated

for either population.

The use of oral aciclovir (800 mg five times daily for 7 days) for the treatment of pregnant

women who contract varicella in their second or third trimester is recommended.

However, it is important to note that this recommendation is based on anecdotal evidence,

Page 45: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

44

and that patients should be advised that aciclovir is not licensed for use during pregnancy.

The roles of valaciclovir and famciclovir for the treatment of varicella infection in the

pregnant woman remain to be evaluated in clinical trials.

Neonates exposed to varicella should be observed closely; if vesicle formation is apparent,

treatment with intravenous aciclovir (10 mg/kg every 8 hours) should be initiated as early

as possible.

References1. Prevention of varicella: Recommendations of the Advisory Committee on Immunization Practices (ACIP).

Centers for Disease Control and Prevention. MMWR Morb Mortal Wkly Rep 1996;45(RR-11):1-36.2. Prevention of varicella. Update recommendations of the Advisory Committee on Immunization Practices

(ACIP). MMWR Morb Mortal Wkly Rep 1999;48(RR-6):1-5.3. White CJ. Varicella-zoster virus vaccine. Clin Infect Dis 1997;24(5):753-761.4. Asano Y, Nakayama H, Yazaki T. Protection against varicella in family contacts by immediate inoculation

with live varicella vaccine. Pediatrics 1977;59:3-7.5. Salzman MB, Garcia C. Postexposure varicella vaccination in siblings of children with active varicella.

Pediatr Infect Dis J 1998;17(3):256-257.6. Watson B, Seward J, Yang A et al. Postexposure effectiveness of varicella vaccine. Pediatrics 2000;105

(1 Pt 1):84-88.7. Arvin AM. Chickenpox (Varicella). In: Varicella-Zoster Virus. Molecular Biology, Pathogenesis and Clinical

Aspects (Wolff MH, Schünemann S, Schmidt A, eds). Basel: Karger, 1999: 96-110.8. Cohen JI, Brunell PA, Straus SE et al. Recent advances in varicella-zoster virus infection. Ann Intern Med

1999;130(11):922-932.9. Asano Y, Yoshikawa T, Suga S et al. Postexposure prophylaxis of varicella in family contact by oral acyclovir.

Pediatrics 1993;92(2):219-222.10. Suga S, Yoshikawa T, Ozaki T et al. Effect of oral acyclovir against primary and secondary viraemia in

incubation period of varicella. Arch Dis Child 1993;69(6):639-642; discussion 642-633.11. Kumagai T, Kamada M, Igarashi C et al. Varicella-zoster virus-specific cellular immunity in subjects given

acyclovir after household chickenpox exposure. J Infect Dis 1999;180(3):834-837.12. Yoshikawa T, Suga S, Kozawa T et al. Persistence of protective immunity after postexposure prophylaxis of

varicella with oral aciclovir in the family setting. Arch Dis Child 1998;78(1):61-63.13. Ogilvie MM. Antiviral prophylaxis and treatment in chickenpox. A review prepared for the UK Advisory Group

on Chickenpox on behalf of the British Society for the Study of Infection. J Infect 1998;36 (Suppl 1):S31-S38.14. Balfour HH, Jr., Kelly JM, Suarez CS et al. Acyclovir treatment of varicella in otherwise healthy children.

J Pediatr 1990;116(4):633-639.15. Balfour HH, Jr., Rotbart HA, Feldman S et al. Acyclovir treatment of varicella in otherwise healthy

adolescents. The Collaborative Acyclovir Varicella Study Group. J Pediatr 1992;120(4 Pt 1):627-633.16. Dunkle LM, Arvin AM, Whitley RJ et al. A controlled trial of acyclovir for chickenpox in normal children.

N Engl J Med 1991;325(22):1539-1544.17. Wallace MR, Bowler WA, Murray NB et al. Treatment of adult varicella with oral acyclovir. A randomized,

placebo-controlled trial. Ann Intern Med 1992;117(5):358-363.18. Balfour HH, Jr., Edelman CK, Anderson RS et al. Controlled trial of acyclovir for chickenpox evaluating time

of initiation and duration of therapy and viral resistance. Pediatr Infect Dis J 2001;20:919-926.19. Weller S, Blum MR, Doucette M et al. Pharmacokinetics of the acyclovir pro-drug valaciclovir after escalating

single- and multiple-dose administration to normal volunteers. Clin Pharmacol Ther 1993;54:595-605.20. Filer CW, Allen GD, Brown TA et al. Metabolic and pharmacokinetic studies following oral administration

of 14C-famciclovir to healthy subjects. Xenobiotica 1994;24(4):357-368.21. Walsh JB, Geaney M. Valaciclovir in secondary and tertiary cases of adult chickenpox. J Eur Acad Dermatol

Venereol 1996;6(3):290-291.22. American Academy of Pediatrics: Committee on infectious diseases. Pediatrics 1994:510-517.23. Enders G, Miller E, Cradock-Watson J et al. Consequences of varicella and herpes zoster in pregnancy:

prospective study of 1739 cases. Lancet 1994;343(8912):1548-1551.24. Miller AE, Marshall R, Vardien J. Epidemiology, outcome and control of varicella-zoster infection. Rev Med

Microbiol 1993;4:222-230.25. Enders G. Varicella-zoster virus infection in pregnancy. Prog Med Virol 1984;29:166-196.26. Prober CG, Gershon AA, Grose C. Consensus: varicella-zoster infections in pregnancy and the perinatal

period. Pediatr Infect Dis 1990;9:865-869.27. Enders G. Management of varicella-zoster contact and infection in pregnancy using a standardized varicella-

zoster ELISA test. Postgrad Med 1985;61:23-30.28. Gilbert GL. Chickenpox during pregnancy. BMJ 1993;306:1079-1080.29. Greenspoon JS, Masaki DI. Fetal varicella syndrome. J Pediatr 1988;112:505-506.30. Brunell PA. Varicella in pregnancy, the fetus and the newborn: problems of management. J Infect Dis

1992;166 (Suppl 1):S42-S47.31. Grose C. Varicella infection during pregnancy. Herpes 1999;6:633-637.32. Pastuszak AL, Levy M, Schiek B et al. Outcome after maternal varicella infection in the first 20 weeks of

pregnancy. N Engl J Med 1994;330:901-905.33. Al-Nakib W, Al-Kandari S, El-Khalik DM et al. A randomised controlled study of intravenous acyclovir

(Zovirax) against placebo in adults with chickenpox. J Infect 1983;6(1 Suppl):49-56.

Page 46: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

45

34. Smego RA, Asperilla MO. Use of acyclovir for varicella pneumonia during pregnancy. Obstet Gynecol1991;78:1112–1116.

35. From the Centers for Disease Control and Prevention. Varicella-related deaths among children – UnitedStates, 1997. JAMA 1998;279(22):1773-1774.

36. Chung CS, Myrianthopoulos NC. Factors affecting risks of congenital malformations. I. Epidemiologicalanaylsis. New York: Stratton Intercontinental, 1975.

37. Hanngren K, Grandier M, Grandstrom C. Effect of zoster immunoglobulin for varicella prophylaxis in thenewborn. Scand J Infect Dis 1985;17:343-347.

38. Miller E, Cradock-Watson JE, Ridehalgh MK. Outcome in newborn babies given anti-varicella-zosterimmunoglobulin after perinatal maternal infection with varicella-zoster virus. Lancet 1989;2(8659):371-373.

39. Bakshi SS, Miller TC, Kaplan M. Failure of varicella-zoster immunoglobulin in modification of severecongenital varicella. Pediatr Infect Dis 1986;5:699-702.

40. King SM, Gorensek M, Ford-Jones EL. Fatal varicella-zoster infection in a newborn treated with varicella-zoster immunoglobulin. Pediatr Infect Dis 1986;5:588-589.

41. Holland P, Isaacs D, Moxon ER. Fatal neonatal varicella infection. Lancet 1986;2:1156.42. Pass R, Weber T, Whitley RJ. Management Strategies in Herpes. Herpesvirus Infections in Pregnanacy:

PAREXEL MMS Europe Ltd, 2002.

Page 47: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

46

Management RecommendationsCategory 3J It is important that clinicians recognize that acute and chronic pain and different

chronic pain subtypes are engendered by different mechanisms.

J Physicians should be aware of the existence of rare but serious complications and

atypical manifestations of herpes zoster, such as delayed contralateral hemiparesis,

and neurological complications. The use of intravenous aciclovir (10 mg/kg every

8 hours for adults, 500 mg/m2 or 20 mg/kg every 8 hours for children) is warranted in

the majority of these cases, based on anecdotal evidence.

Research needsJ A trial is recommended using polymerase chain reaction (PCR) techniques to estimate the

peak concentration of virus in serum during the course of herpes zoster (encompassing

prodrome, acute rash and post-herpetic neuralgia [PHN] where possible).

J The further application of statistical models to understanding pain in herpes zoster is

encouraged, particularly in the assessment of new treatment modalities.

IntroductionHerpes zoster is known to be the cause of considerable morbidity, especially among

elderly patients.1 Over a lifetime, estimates of the proportions of individuals experiencing

herpes zoster vary from 10% to 20%1 and up to 23%,2 with the incidence increasing with

age. The most debilitating symptom of herpes zoster is the associated pain, which may be

both acute and chronic in nature and can persist for months or even years.3-5 This has a

major impact on patients’ quality of life and, once established, can be difficult to manage

effectively,6 making this the most compelling reason for early treatment of herpes zoster.7

Analysis of several studies indicates that antiviral treatment initiated within 72 hours of

rash onset increases the rate of rash healing and also speeds the resolution of zoster-

associated pain in some patients.8

Clinical FeaturesHerpes zoster is a manifestation of the reactivation of varicella zoster virus (VZV).

Following initial infection, the virus remains latent in dorsal sensory ganglia until it

reactivates and replicates. Virus replication and transmission in nerves and ganglia,

together with the subsequent development of skin rash, contribute to the prodromal and

acute-phase pain of herpes zoster. Subsequent to this, there can be chronic pain. Thus,

pain associated with herpes zoster is typically compartmentalized into three phases –

prodromal, eruptive or rash, and PHN.

INTRODUCTION TO HERPES ZOSTER AND FUTURE

DIRECTIONS5

Page 48: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

47

Prodromal phaseThe prodrome may precede rash onset by 48–72 hours, but can occasionally occur a week

or more prior to the rash becoming visible. During this phase, the patient may experience

acute neuritis characterized by pain and paraesthesia. The type of pain during the

prodromal phase, often accompanied by malaise, can vary widely but the most common

description is throbbing. It can be intermittent or constant.

Eruptive or rash phaseClinical diagnosis of herpes zoster is

usually achieved during the eruptive or

rash phase, when the typical

papulovesicular rash appears. The

vesicles are typically clustered in a

unilateral dermatomal distribution. In the

immunocompetent host, new lesions appear for

3–5 days, and healing usually occurs over a period

of 2–4 weeks (see Table 1 and Figure 1). Extensive

cutaneous dissemination is unusual and visceral

dissemination rare in the immunocompetent host.

In immunocompromised individuals and the elderly,

the healing process is likely to be slower with new

vesicle formation continuing for a week or more.9

Pain accompanies the rash in 60–90% of

immunocompetent individuals. Pain occurs in

fewer than 20% of individuals aged less than

20 years, but in more than 80% of individuals over

50 years old.3 The pain, which can be constant or

intermittent, has been variously described as

shooting, stabbing, tender, itching or hot.

PHN and zoster-associated painAlthough the pain experienced during the acute phase usually resolves as the rash heals,

chronic pain can persist for prolonged periods (e.g. months or even years). The term PHN

has been variously used to describe:

• Pain that persists or occurs after resolution of the cutaneous rash10

• Pain persisting more than 30 days after rash onset

• Pain at 3–6 months after the acute episode.10

Thus, there is a continuum of pain – from the pain of the prodrome through to the

persisting pain. This continuum is referred to as zoster-associated pain.

Pathogenesis of chronic painIn normal pain perception, noxious stimuli activate nerve endings in the skin and generate

signals which pass to the dorsal root ganglia and then to the dorsal horn (see Figure 2).

Spinal cord neurones are inhibited by powerful descending signals from the brain,

mediated by biogenic amines such as serotonin. Drugs which potentiate such chemicals

may act by enhancing these descending inhibitory pathways. The net result of peripheral

afferent input and descending inhibitory input is perceived pain.

TABLE 1: Stages ofhealing of acute herpeszoster in theimmunocompetent

Time Stage of healing

3–5 days Cessation of new vesicle formation 4–6 days Total pustulation 7–10 days Total scabbing 2–4 weeks Complete healing

Primary infection withVZV

(varicella)

Latency

Reactivation Zoster sineherpete

Herpes zoster

Myelitis Large-vesselgranulomatous

arteritis

PHN Myelitis Small-vesselencephalitis

Immunocompromisedpatients

Immunocompetentpatients

FIGURE 1: Naturalhistory of herpes zoster

Page 49: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

48

In the case of herpes zoster,

damage to peripheral nerves by

virus reactivation and

inflammation in the skin, and

altered signal processing in the

central nervous system (CNS),

can lead to the development of

chronic neuropathic pain.12

Injured neurones have lower

activation thresholds and may

discharge spontaneously and

frequently in response to weak,

non-noxious stimuli. Noxious

stimuli cause more pain than

normal (primary hyperalgesia).

In addition to peripheral sensitization, central sensitization may also occur, in which

abnormal signal amplification in the CNS causes hyperalgesia. As the damaged axons

repair, the growth cone and nerve sprout are also sensitive and may be the source of

abnormal discharges, particularly when their growth becomes interrupted or blocked by

scarring. The discharges give rise to pain sensations and maintain a state of

hyperexcitability in the dorsal horn, and this in turn causes exaggerated responses in the

CNS to all stimuli.

Epidemiology of Herpes ZosterThere have been few recent reports on the incidence of herpes zoster in general

populations; the figures that are most often quoted are based on surveys conducted

between 1949 and 1959 in the USA by Ragozzino et al4 and in the UK from 1947 to 1962

by Hope-Simpson.13 More recent studies have explored the incidence of herpes zoster in

large cohorts in the USA and Iceland. The overall incidence of herpes zoster in these

studies is 120–350 per 10 0000 person-years (Table 2).

FIGURE 2: Normalpain perception11

Skin or mucousmembrane

Descending noradrenergic andserotoninergic inhibitory fibres

Ascendingspinothalamic fibres

Dorsal-rootganglion

TABLE 2: Incidence ofherpes zoster in largecohort studies4,13-18

Age range (years) Location (date of study) Incidence (per 100 000 patient-years)

0–99 Cirencester, UK (1947–1962)13 340

0–75+ Rochester, Minnesota, USA (1949–1959)4 131 < 14 40 25–30 100 ≥75 400

0–19 Rochester, Minnesota, USA (1960–1981)14 42

0–75+ Boston, USA (1990–1992)15 215 (unadjusted)287 (age adjusted to represent the same age

distribution as in Ragozzino et al4 study)

65–104 North Carolina, USA (1987–1990)16 710

0–80+ Iceland (1990–1995)17 460

≥ 15 Italy (1995)18 410

Comparison of these studies reveals marked differences in the incidence of herpes zoster

in the USA4 and UK.13 The incidence of herpes zoster in the Ragozzino et al4 study has

been adjusted to the 1970 population structure in the USA to give a rate of 131/100 000

person-years. This incidence rate is lower than that reported in England by Hope-Simpson3

© R

epro

duce

d w

ith p

erm

issi

on, K

ost R

G, S

trau

s SE

. Pos

ther

petic

neu

ralg

ia—

path

ogen

esis

, tre

atm

ent,

and

prev

entio

n. N

Eng

l J M

ed19

96;3

35(1

):32-

42.

Mas

sach

uset

ts M

edic

al S

ocie

ty.

Page 50: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

49

in 1965 (340/100 000 person-years) during a similar time period. The reason for this is

uncertain but may be due partly to differences in study methodology as well as true

population differences.

Studies conducted in the USA suggest that the incidence of herpes zoster is increasing.

In the study by Ragozzino et al,4 the rate increased from 112 to 150/100 000 person-years

over the 10-year period of the survey (1949–1959). A later study by Donahue et al (1995)15

surveyed the population of a health maintenance organization in the USA from 1990 to

1992. It found a higher incidence (287 per 100 000 patient-years) after age adjustment to

the same standard population as described in the earlier study by Ragozzino et al

(Table 1). The rise reported over the period of the Ragozzino et al study, if it continued,

may explain the higher incidence in the Donahue et al study. The difference in rates may,

however, be explained by differences in the ascertainment of herpes zoster between the

two studies.

Relationship between risk of herpes zoster and age A striking feature of herpes zoster is its increase in incidence with ageing. It is relatively

uncommon in the immunocompetent individual less than 40 years old, but has a marked

rise in the over 50-year-olds and an even higher

incidence in those over 75 years (Figure 3).4 This

relationship between age and risk of herpes zoster is

illustrated by the fact that although the overall

lifetime probability of developing herpes zoster in

the USA and UK is 10–20%, the risk for those living

to ≥80 years increases to 50%. However, the age

structure of most populations means that even

though the incidence is highest among those over

75 years, the prevalence is greatest in the 55–

75-year age group. Changing population

demographics and increasing longevity are likely to

lead to an increase in the number of individuals who will experience herpes zoster.

Prodromal symptoms are more frequent in the elderly, and are likely to be of greater

duration than in younger individuals (Figure 4).3 In a UK study of herpes zoster in

individuals aged over 60 years (n=205), 40% of the study participants experienced

prodromal pain for more than 4 days.19,20 The rash

phase is also frequently extended in elderly patients

compared with younger patients, with new lesions

continuing to develop for a number of days. The

median time to loss of vesicles is 7 days in the

elderly, with full crusting occurring more than 10 days

after rash onset.9 This compares with a median time

to loss of vesicles of 48 hours in younger

individuals.8 Studies have indicated that elderly

patients experience prolonged pain more often than

younger patients.3 PHN is one of the most severe

chronic pain syndromes experienced, and one of the

worst seen at a pain clinic.9

FIGURE 3: Theincidence of herpeszoster increases with age4

Age (years)

500

400

300

200

100

0

Inci

denc

e/10

0 00

0 pe

rson

– y

ears

≤14 15–24 25–34 35–44 45–54 55–64 65–74 ≥75

FemalesMales

FIGURE 4: Duration ofzoster-associated pain indifferent age groups3

≥70

60–69

50–59

40–49

30–39

20–29

0–19

Age

(yea

rs)

Percentage with pain0 10 20 30 40 50 60 70 80 90 100

<1 month 1–6 months 6–12 months >1 year

© R

epro

duce

d w

ith p

erm

issi

on, d

e M

orag

as JM

, Kie

rlan

d R

R. T

heou

tcom

e of

pat

ient

s w

ith h

erpe

s zo

ster

. Arc

h D

erm

atol

1957

;75:

193-

196.

Cam

brid

ge M

edic

al P

ublic

atio

ns.

© R

epro

duce

d w

ith p

erm

issi

on R

agoz

zino

MW

, Mel

ton

LJ,

3rd,

Kur

land

LT

et a

l.Po

pula

tion-

base

d st

udy

of h

erpe

szo

ster

and

its

sequ

elae

. Med

icin

e (B

altim

ore)

1982

;61(

5):3

10-3

16.

Page 51: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

50

The vulnerability of elderly individuals to herpes zoster is most likely due to the

senescence of their cellular immune system.21 Rates of positive skin tests for VZV antigens

and in vitro measurements of the lymphoproliferative response to VZV reduce with

increasing age. This reduction in immune response to the virus is inversely correlated with

the frequency of reactivation of latent VZV.22

Although rare in children, herpes zoster has been reported in some aged less than 2 years

whose mothers contracted varicella during pregnancy and also in children who contracted

varicella during their first year.23 The incidence of herpes zoster is less where contact with

varicella is greater. Hope-Simpson13 originally observed that herpes zoster was less likely

to occur during epidemic years for varicella, leading him to postulate that each time a

person who has had varicella (i.e. is VZV seropositive) comes into contact with the virus,

their immunity is boosted. This may reverse the decline in antibodies that normally occur

with age, and therefore delay the chance of an episode of herpes zoster.

Rare Complications in the ImmunocompetentHostJ Physicians should be aware of the existence of rare but serious

complications and atypical manifestations of herpes zoster, such

as delayed contralateral hemiparesis, and neurological

complications. The use of intravenous aciclovir (10 mg/kg every

8 hours for adults, 500 mg/m2 or 20 mg/kg every 8 hours for

children) is warranted in the majority of these cases, based on

anecdotal evidence (category 3 recommendation).

PHN is the most commonly observed complication of herpes zoster.

However, a number of other rare but distinct neurological syndromes

are also associated with it, including encephalitis (both large and

small vessel) (Figure 5), ophthalmic zoster (with delayed contralateral

hemiparesis, myelitis, polyradiculitis), and numerous cranial and

peripheral nerve palsies (including Bell’s palsy and Ramsay Hunt

syndrome).24 Ophthalmic zoster is discussed in Chapter 7.

Delayed contralateral hemiparesisDelayed contralateral hemiparesis, involving the first division of the trigeminal nerve, is a

rare but serious complication of herpes zoster that can occur weeks to months (average

7 weeks) after an episode of herpes zoster.25-27 Hemiparesis has been reported in both

immunocompetent and immunocompromised individuals. The mortality rate associated

with the condition is 20–25% and there is a high probability of permanent neurological

sequelae among survivors.25,28

The pathogenesis of hemiparesis is thought to be direct VZV invasion of cerebral arteries

along intracranial branches of the trigeminal nerve, resulting in inflammation of the

internal carotid artery or one of its branches on the same side as the rash.29

The typical presentation of delayed contralateral hemiparesis is headache and hemiplegia

occurring in a patient with a recent history of herpes zoster ophthalmicus. Examination of

cerebrospinal fluid (CSF) reveals mononuclear cell pleocytosis (usually <100 cells per cm3)

FIGURE 5: Encephalitisassociated with thereactivation of VZV24

© 2

002.

Rep

rodu

ced

with

per

mis

sion

, Gild

en D

H, K

lein

schm

idt-

DeM

aste

rs B

K, L

aGua

rdia

JJet

al.

Neu

rolo

gic

com

plic

atio

ns o

f the

rea

ctiv

atio

n of

var

icel

la-z

oste

r vi

rus.

N E

ngl J

Med

2000

;342

(9):6

35-

645.

Mas

sach

uset

ts M

edic

al S

ocie

ty.

Page 52: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

51

and increased protein.28 Arteriography is usually diagnostic and demonstrates

inflammation, narrowing and thrombosis of the proximal branches of the anterior or

middle cerebral artery.30,31

Management of delayed contralateral hemiparesisBoth aciclovir and corticosteroids have been used to treat this syndrome, although neither

therapy has been evaluated in controlled trials.32,33 Antiviral therapy is warranted because

of the demonstrated presence of VZV in the inflamed arteries. However, the benefit of

therapy is difficult to assess, since irreversible cerebral infarction has often occurred by the

time the diagnosis is made.

Zoster sine herpeteClinicians have frequently noted patients who present with herpes zoster-like neuropathic pain,

but do not develop the characteristic dermatomal rash. This presentation is known as zoster sine

herpete, and is characterized by radicular neuropathic pain in a dermatomal distribution

without cutaneous eruption. Recent studies of patients presenting with dermatomal pain

without rash have established that some of these cases are due to VZV reactivation.34

Patients with zoster sine herpete have raised titres of VZV-specific antibody (up to 4-fold

increase) compared with other immunocompetent individuals, and raised levels of VZV

DNA in the CSF and peripheral blood mononuclear cells.34,35 The lack of a simple and

accurate diagnostic method for zoster sine herpete means that its prevalence remains

unknown. Some anecdotal reports indicate that it responds to intravenous aciclovir, but

these reports are not consistent.

Prolonged Zoster-Associated PainThe incidence of PHN is unclear because of the lack of good prospective population-based

studies and the absence of a standardized definition. Information on the incidence of

prolonged zoster-associated pain comes from retrospective population surveys and the

placebo arms of clinical trials.

Retrospective population-based studies have reported that, in the absence of therapy,

8–14% of patients with herpes zoster develop pain persisting for longer than

4 weeks.4,13,17,36 The pain generally improves and, overall, these studies suggest that 3–7%

of patients with herpes zoster have pain that persists for more than 3 months and only

2–5% have pain after 1 year. The data from prospective trials is more robust than that from

retrospective studies; prospective data on the incidence of PHN have been supplied by

clinical trials of aciclovir. They suggest an incidence of 50–60% at 1 month, which

decreases with time. For example, in a placebo-controlled trial of aciclovir in patients aged

over 60 years, the incidence of PHN in the placebo recipients fell from 61% at 1 month

after the onset of rash to 13% at 6 months.20 Although this is a low estimate for the over-

60s age group, this trend is also observed in other placebo-controlled studies; in one

aciclovir study of acute herpes zoster in patients >16 years of age, the incidence of PHN

in the placebo recipients fell from 40% at 1 month after rash onset to 20% at

6 months.37 Overall, these placebo-controlled trials give higher incidence figures than the

population-based studies. This may be due to differences in study design and because

patients in clinical trials may differ from those in the general population.

Page 53: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

52

Predicting patients at greater risk of PHNEstablished PHN remains difficult to treat. Antiviral therapy (and possibly tricyclic antidepressants

or sympathetic nerve blocks) can accelerate the resolution of zoster-associated pain. However,

optimal use of antiviral therapy requires accurate prediction of herpes zoster patients at risk of

developing PHN, and prompt initiation of therapy in the acute phase.

Age has a major influence on the incidence and duration of PHN, for example, the study by

Hope-Simpson13 demonstrated an incidence of 74 cases per 100 000 for children aged

0–9 years, rising to 292 per 100 000 for those aged 40–49 years. The reliability of age as a

predictor of PHN was confirmed in three recent analyses.38-40 One of these studies was a large,

observational study.38 The second study analysed baseline data of 1778 patients with herpes

zoster collected prior to randomization into antiviral drug studies.39 In the third, Cox’s

proportional hazard modelling was used to identify a range of factors influencing pain outcome

in controlled trials of antiviral therapies in herpes zoster (total number of subjects = 2367).40

The severity of the acute pain during herpes zoster is predictive of prolonged pain,40,41 and

is illustrated by a study comparing aciclovir with prednisolone; patients who presented with

severe or incapacitating pain and a large number of lesions were less likely to achieve

resolution of both acute neuritis and zoster-associated pain (risk ratio [RR], 18.0; 95%

confidence interval [CI], 6.6–48.6, and RR, 5.3; 95% CI, 4.2–17.2, respectively).40 Similarly,

in an analysis of data from six randomized, controlled double-blind studies, severe pain at

presentation was a highly significant prognostic factor for prolonged zoster-associated

pain.41 The same analysis reported that the presence of a prodrome is also a significant risk

factor for the prolonged pain in patients 50 years of age or older (hazard ratio [HR], 1.30;

CI, 1.06–1.56; P=0.008) and approached significance in those younger than 50 years

(HR, 1.32; CI, 0.99–1.72; P=0.06). The importance of both presence of a prodrome and

intensity of acute pain was confirmed further in a large observational study (Figure 6).38

Thus, the three main risk factors that predict PHN following herpes zoster are:41,42

• Advancing age

• Presence of a prodrome

• Acute pain severity.

Other hypothetical factors predictive for PHN

include viraemia, rash severity, and adverse

psychosocial factors.

As well as being related to the incidence of PHN,

there is a strong association between age and the

duration of pain following herpes zoster. This is

shown by a study in the USA conducted during the

1950s in which 20% of those aged 50–59 had short-

lived PHN (defined as any subjective, painful

sensation that persisted after the acute phase of

infection), whereas in the over-60s, more than 30%

had PHN that persisted for many months.42 Another

study in the USA, also conducted in the 1950s,

showed that individuals under the age of 40 years

FIGURE 6: Time tocessation of painfollowing herpes zoster ina) subjects aged 18–50 years and >50 years;b) subjects with no/mild,moderate and severeprodromal pain.38 Allsubjects receivedvalaciclovir treatment.

1.00.90.80.70.60.50.40.30.20.1

00 20 40 60 80 100 120 140 160 180

Prop

ortio

n of

cas

esw

ith z

oste

r-as

soci

ated

pai

n

Time (days)

Median duration (days)Pain at 6 months (%)HR (95% CI)

18–50 years

921.91 (1.71, 2.12)

>50 years

2311P < 0.0001

1.00.90.80.70.60.50.40.30.20.1

00 20 40 60 80 100 120 140 160 180

Prop

ortio

n of

cas

esw

ith z

oste

r-as

soci

ated

pai

n

Time (days)

Age (years)18–50>50

Maximum intensity of prodromal painNon/noticeable/mildModerateSevere/very severe

a)

b)

© R

epro

duce

d w

ith p

erm

issi

on, D

ecro

ix J,

Par

tsch

H, G

onza

lez

Ret

al.

Fact

ors

influ

enci

ng p

ain

outc

ome

inhe

rpes

zos

ter:

an

obse

rvat

iona

l stu

dy w

ith v

alac

iclo

vir.

Val

acic

lovi

r In

tern

atio

nal Z

oste

r A

sses

smen

t Gro

up(V

IZA

). J E

ur A

cad

Der

mat

ol V

ener

eol2

000;

14(1

):23-

33. B

lack

wel

l Sci

ence

Ltd

.

Page 54: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

53

rarely had pain lasting beyond 1 month but that 47% of patients aged over 70 years of age

still had pain at 1 year.3 These early studies have been confirmed by a recent, large,

observational study in which pain resolved almost twice as rapidly in patients aged

between 18 and 49 years compared with those older than 50 years (P<0.0001; Figure 6).38

Application of PCR to Understanding Pathogenesis and RiskFactors for Prolonged Zoster-Associated PainJ A trial is recommended using PCR techniques to estimate the peak level of virus in

blood during the course of herpes zoster (encompassing prodrome, acute rash and

PHN where possible) (research need recommendation).

The importance of detectable VZV in peripheral blood cells on the development of PHN is

uncertain. It has been reported that the occurrence of PHN was not correlated with the

presence of VZV DNA in lymphocytes. However, the existence of VZV DNA was correlated

significantly with increasing age (P<0.05).43 More recent data have suggested that there is

a link between detectable viraemia at presentation and development of PHN.44

A 52-week prospective study determined whether VZV DNA in lymphocytes at

presentation predicted the likelihood of PHN development.45 One hundred and sixty five

patients with herpes zoster were asked to quantify the severity of their pain at presentation

using the Visual Analogue Scale (VAS), the McGill Pain Questionnaire and Active Daily

Living Scores. Blood samples were collected from 119 of the patients and the presence of

VZV DNA in lymphocytes from these was assessed by nested PCR directed at open reading

frame 29.45 Forty per cent of patients received antiviral medication. Amitriptyline was

prescribed for any patient who experienced pain persisting for more than 6 weeks.45

The presence of PCR-detectable VZV viraemia at presentation was significantly associated

with prolonged pain of >6 months’ duration (P<0.05). This association was more significant

in those patients aged >50 years (P<0.01).

Age and pain severity are independent predictors of the development of PHN. In keeping with

the findings of the studies discussed on page 52, a significantly larger proportion of patients

aged >50 years reported experiencing prolonged pain compared with those aged <50 years.

Moreover, patients with more severe pain at presentation (VAS score >6) were more likely to

experience prolonged pain than patients with less severe pain (VAS score <6).45

These data suggest that patients at greatest risk of developing prolonged zoster-associated

pain are likely to be over 50 years old, have high pain scores at presentation and have

detectable viraemia at presentation. This may offer a virological explanation for previously

identified risk factors for pain persisting after herpes zoster and may in turn lead to the

development of laboratory tests for the clinical evaluation of patients at highest risk of

prolonged pain.

Mathematical Modelling of Zoster-Associated PainJ It is important that clinicians recognize that acute and chronic pain and different chronic

pain subtypes are engendered by different mechanisms (category 3 recommendation).

Page 55: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

54

J The further application of statistical models to understanding pain in herpes zoster is

encouraged, particularly in the assessment of new treatment modalities (research need

recommendation).

Background to model Existing methods to analyse the clinical trials of antiviral therapy can lead to bias. In an

effort to increase the understanding of the mechanisms of zoster-associated pain and

improve the design of future herpes zoster studies, a mathematical model of the

progression and resolution of pain associated with herpes zoster has been developed.44

Currently, considering the effect of treatments on PHN means that only a subset of patients

who actually develop persistent pain will be included in any given analysis. Because this

subset is chosen after randomization, early treatment effects cannot be assessed and

baseline characteristics may be unbalanced among the different groups. Similarly,

considering pain over a follow-up period of 1 year may be rigorous, as it accounts for the

early effects of lesion healing and termination of acute pain. However, this approach has

the major disadvantage that it looks at all pain (both acute and chronic pain together).

Therefore, treatment effects specific to PHN cannot be evaluated.

To overcome these limitations, the model assumes that zoster-associated pain manifests,

and resolves, in three distinct phases:

• Acute pain, observed in most patients, caused by inflammation of nerve tissue and

damage to skin, occurring at the time of acute zoster VZV reactivation and relating to

ongoing viral replication

• Subacute pain, observed in a proportion of patients, caused by mixed aetiology

• Chronic pain observed in a smaller subset of patients, brought about by pathological

changes in the CNS and peripheral nervous system (PNS).

In considering pain as three phases, the model allows the change points of the phases

(i.e. the change from acute to subacute; change from subacute to chronic pain) and the

rates of pain resolution of each phase to be determined. It also permits examination of the

effect on each phase of any proposed treatment regimen to alleviate zoster-associated

pain, while controlling for its effect on the other phases.

Findings from the model of zoster-associated painThe model provides insight into the natural history of zoster-associated pain. Based on data

from trials of aciclovir, the change points in zoster-associated pain (time of transition from

acute to subacute pain; time of change from subacute to chronic pain) had previously

been proposed to be 30 and 120 days, respectively, but this hypothesis had not been

tested. The model yielded estimates of change points that were similar to those estimated

from historical data (Table 3).44,46

TABLE 3: Zoster-associated pain changepoints in immuno-competent and immuno-compromised individuals44

Trial Acute to subacute pain Standard error Subacute to chronic Standard error change point (days) pain change point (days)

Aciclovir and steroids in 46.2 9.8 138.3 21.4immunocompetent hosts ≥50 years

Aciclovir versus sorivudine in cancer 36.9 13.5 136.4 19.9patients

Aciclovir versus sorivudine in 26.9 8.4 124.3 23.6individuals with HIV

Page 56: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

55

In addition, by understanding the rate of resolution of the different phases of zoster-

associated pain, the model can further contribute to an understanding of its natural history.

The model demonstrated that for each patient group, there were differences in the rates of

pain resolution between the different phases, but that these differences were not consistent.

For example, the rates of resolution of subacute and chronic phases were not significantly

different for either the immunocompetent adults or the cancer patients but were different for

the HIV-infected cohort. This difference can be important when translating or evaluating

treatment regimens when herpes zoster occurs in patients with different underlying diseases.

Barriers to the Treatment of Herpes ZosterDespite available evidence as to the benefits of treating patients who have herpes zoster (see

Chapters 6, 7 and 9), a number of barriers to treatment persist (see Table 4). One such barrier

is the perception among some physicians that treating herpes zoster is expensive. However,

an analysis of valaciclovir and famciclovir, using a Markov decision model, indicated that

treatment of severe acute herpes zoster

was cost-effective from a third-party

payer perspective.47 In addition,

treatment of mildly symptomatic acute

herpes zoster was cost-effective in

situations where there was an increased

risk of PHN, e.g. in the elderly and

immunocompromised.47

SummaryHerpes zoster is caused by the reactivation of latent VZV infection. It is characterized by

the presence of dermatomal pain, and usually a papulovesicular rash also in a dermatomal

distribution, although the rash is not always accompanied by pain and vice versa. The rash

associated with herpes zoster typically resolves within 2–4 weeks, although this can take

significantly longer in immunocompromised individuals. Some 9–15% of patients will

experience prolonged pain that may continue for months or even years.

Complications of herpes zoster may include delayed contralateral hemiparesis, myelitis,

polyradiculitis and nerve palsies.

Prevention and management of PHN are the main objectives of therapy. In addition,

treatment of herpes zoster aims to prevent complications and increase the rate of rash

healing. The treatment and management of patients with herpes zoster and PHN are

discussed in Chapters 6, 7 and 9.

The continued application of mathematical modelling and detection techniques, such as

PCR, will increase understanding of the pathogenesis and risk factors involved and provide

the potential to develop new treatment methodologies to manage the pain associated with

herpes zoster.

TABLE 4: Barriers to thetreatment of herpes zoster

• Lack of clear initial symptoms due to the variable nature of prodromal pain

• Time taken to obtain a consultation once rash appears due to waiting lists andappointment systems, and patients’ fear of illness – especially in the elderly

• Time between consultation and dispensing of prescription

• Lack of awareness by some physicians of the suffering experienced by patients withherpes zoster

• Perception among some physicians that treatments are expensive

Page 57: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

56

References1. Liesegang T. Varicella zoster viral disease. Mayo Clin Proc 1999;74:983-998.2. Miller E, Vardien J, Farrington P. Shift in age in chickenpox. Lancet 1993;341(8840):308-309.3. de Moragas JM, Kierland RR. The outcome of patients with herpes zoster. Arch Dermatol 1957;75:

193-196.4. Ragozzino MW, Melton LJ, 3rd, Kurland LT et al. Population-based study of herpes zoster and its sequelae.

Medicine (Baltimore) 1982;61(5):310-316.5. Rogers RS, Tindall JP. Geriatric herpes zoster. J Am Geriatr Soc 1971;19(6):495-504.6. Tyring SK, Beutner KR, Tucker BA et al. Antiviral therapy for herpes zoster: randomized, controlled clinical

trial of valacyclovir and famciclovir therapy in immunocompetent patients 50 years and older. Arch FamMed 2000;9(9):863-869.

7. Balfour HH, Jr. Antiviral drugs. N Engl J Med 1999;340(16):1255-1268.8. Wood MJ, Shukla S, Fiddian AP et al. Treatment of acute herpes zoster: effect of early (< 48 h) versus late

(48-72 h) therapy with acyclovir and valaciclovir on prolonged pain. J Infect Dis 1998;178 (Suppl 1):S81-S84.

9. Wood MJ, Easterbrook P. Shingles, Scourge of the elderly. In: Clinical Management of Herpes Viruses. (SacksSL, Straus SE, Whitley RJ et al., eds). Amsterdam: IOS Press, 1995: 193-209.

10. Dworkin RH, Carrington D, Cunningham A et al. Assessment of pain in herpes zoster: lessons learned fromantiviral trials. Antiviral Res 1997;33(2):73-85.

11. Kost RG, Straus SE. Postherpetic neuralgia—pathogenesis, treatment, and prevention. N Engl J Med1996;335(1):32-42.

12. Wall PD. Neuropathic pain and injured nerve: central mechanisms. Br Med Bull 1991;47(3):631-643.13. Hope-Simpson RE. The nature of herpes zoster: a long-term study and a new hypothesis. Proc R Soc Med

1965;58:9-20.14. Guess HA, Broughton DD, Melton LJ, 3rd et al. Epidemiology of herpes zoster in children and adolescents:

a population- based study. Pediatrics 1985;76(4):512-517.15. Donahue JG, Choo PW, Manson JE et al. The incidence of herpes zoster. Arch Intern Med

1995;155(15):1605-1609.16. Schmader K. Management of herpes zoster in elderly patients. Infect Dis Clin Pract 1995;4(293-299):9.17. Helgason S, Sigurdsson J, Gudmundsson S. The clinical course of herpes zoster: a prospective study in

primary care. Eur J Gen Prac 1996;2:12-16.18. di Luzio Paparatti U, Arpinelli F, Visona G. Herpes zoster and its complications in Italy: an observational

survey. J Infect 1999;38:116-120.19. Terada K, Kawano S, Yoshihiro K et al. Characteristics of herpes zoster in otherwise normal children. Pediatr

Infect Dis J 1993;12(11):960-961.20. McKendrick GD, McGill J, White CJ et al. Oral acyclovir in acute herpes zoster. BMJ 1986;293:1529-1532.21. Wood MJ. Treatment of herpes zoster in the elderly. Herpes 1998;5:60-63.22. Miller AH. Selective decline in cellular immune response to varicella zoster in the elderly. Neurology

1980;30:582-587.23. Enders G, Miller E, Cradock-Watson J et al. Consequences of varicella and herpes zoster in pregnancy:

prospective study of 1739 cases. Lancet 1994;343(8912):1548-1551.24. Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ et al. Neurologic complications of the reactivation of

varicella-zoster virus. N Engl J Med 2000;342(9):635-645.25. Hilt DC, Buchholz D, Krumholz A et al. Herpes zoster ophthalmicus and delayed contralateral hemiparesis

caused by cerebral angiitis: diagnosis and management approaches. Ann Neurol 1983;14(5):543-553.26. Verghese A, Sugan AM. Herpes zoster ophthalmicus and granulomalous angitis. J Am Geriatr Soc

1986;34:309-312.27. Gasperetti C, Song SK. Contralateral hemiparesis following herpes zoster ophthalmicus. J Neurol Neurosurg

Psych 1985;48:338-341.28. Reshef E, Greenberg SB, Jankovic J. Herpes zoster ophthalmicus followed by contralateral hemiparesis:

report of two cases and review of literature. J Neurol Neurosurg Psych 1985;48:122-127.29. Melanson M, Chalk C, Georgevich L. Varicella-zoster virus DNA in CSF and arteries in delayed contralateral

hemiplegia: evidence for viral invasion of cerebral arteries. Neurology 1996;47:569-570.30. Eidelberg D, Sotrel A, Horoupian DS et al. Thrombotic cerebral vasculopathy associated with herpes zoster.

Ann Neurol 1986;19:7-14.31. Mackenzie RA, Forbes GS, Karnes WE. Angiographic findings in herpes zoster arteritis. Ann Neurol

1981;10:458-464.32. Booss J, Haak BB, Levoy RF. Delayed contralateral hemiplegia following herpes zoster ophthalmicus: should

antiviral therapy be used? Eur Neurol 1985;24:225-228.33. Friedman MS, MacDonald RD. Herpes zoster ophthalmicus with delayed cerebral infarction and

meningoencephalitis. Can J Neurol Sci 1987;14:12-14.34. Gilden DH, Wright RR, Schneck SA et al. Zoster sine herpete, a clinical variant. Ann Neurol 1994;35:

530-533.35. Easton HG. Zoster sine herpete causing acute trigeminal neuralgia. Lancet 1970;2(7682):1065-1066.36. Choo PW, Galil K, Donahue JG et al. Risk factors for postherpetic neuralgia. Arch Intern Med

1997;157(11):1217-1224.37. Morton P, Thomson AN. Oral acyclovir in the treatment of herpes zoster in general practice. N Z Med J

1989;102(863):93-95.38. Decroix J, Partsch H, Gonzalez R et al. Factors influencing pain outcome in herpes zoster: an observational

study with valaciclovir. Valaciclovir International Zoster Assessment Group (VIZA). J Eur Acad DermatolVenereol 2000;14(1):23-33.

39. Dworkin RH, Johnson RW, Griffin DRJ. Acute pain in herpes zoster. In: 9th World Congress on Pain, 2000.

Page 58: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

57

40. Whitley RJ, Weiss HL, Soong SJ et al. Herpes zoster: risk categories for persistent pain. J Infect Dis1999;179(1):9-15.

41. Whitley RJ, Shukla S, Crooks RJ. The identification of risk factors associated with persistent pain followingherpes zoster. J Infect Dis 1998;178 (Suppl 1):S71-S75.

42. Burgoon CFJ, Burgoon JS, Bladridge GD. The natural history of herpes zoster. JAMA 1957;164:265-269.43. Devlin ME, Gilden DH, Mahalingam R et al. Peripheral blood mononuclear cells of the elderly contain

varicella- zoster virus DNA. J Infect Dis 1992;165(4):619-622.44. Arani RB, Soong SJ, Weiss H et al. Phase specific analysis of herpes zoster associated pain data: a new

statistical approach. Stat Med 2001;30:2429-2439.45. Breuer J, Scott F, Leedham Green M et al. Virological evidence to identify patients at high risk of zoster-

associated pain persisting following herpes zoster [abstract & poster]. Fourth International Conference onVaricella, Herpes Zoster and Post-Herpetic Neuralgia 2001; La Jolla, California, USA.

46. Dworkin RH, Portenoy RK. Proposed classification of herpes zoster pain. Lancet 1994;343(8913):1648.47. Smith KJ, Roberts MS. Cost effectiveness of newer antiviral agents for herpes zoster: is the evidence spotty?

J Infect Dis 1998;178 (Suppl 1):S85-S90.

Page 59: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Management RecommendationsCategory 1J Antiviral therapy for immunocompetent adults over 50 years of age with herpes zoster

is recommended, particularly if therapy can be instituted within 72 hours of lesion

onset. As a result of their improved pharmacokinetic profiles and simpler dosing

regimens, valaciclovir (1000 mg three times a day for 7 days) or famciclovir (250 mg

or 500 mg three times a day) have replaced aciclovir (800 mg five times a day) as the

preferred treatment.

J It is recommended that physicians consider the use of steroids to reduce the

inflammation that may be contributing to acute pain, provided there are no contra-

indications and the potential risk of excess side-effects is explained. Although they do

not prevent post-herpetic neuralgia (PHN), steroids reduce acute symptoms and may

facilitate return to normal quality of life.

J There is no indication for the use of topical antiviral agents in the treatment of

cutaneous herpes zoster.

Category 2J Early use of tricyclic antidepressants should be considered as they may prevent or

reduce PHN in elderly patients who present with acute herpes zoster.

J Intravenous aciclovir (10 mg/kg every 8 hours) remains the drug of choice for those

rare immunocompetent patients with herpes zoster who develop evidence of

pneumonitis or disseminated infection (e.g. varicella zoster virus [VZV] encephalitis).

Category 3J It is important that patients with herpes zoster are encouraged to present to physicians

as early as possible for prompt medical care. Both public and medical education

efforts may be beneficial.

J The presence of risk factors for the development of PHN should be assessed and

documented for each patient.

J If the patient has severe acute pain, the following treatments may be beneficial:

sympathetic nerve blocks, corticosteroids, antidepressants, gabapentin and opioids.

J There is no published evidence for the use of famciclovir 750 mg once daily for the

treatment of herpes zoster, and no follow-up data on its use beyond 1 month.

MANAGEMENT OF HERPES ZOSTER IN THE

IMMUNOCOMPETENT HOST

58

6

Page 60: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

59

Research needsJ Studies are required to establish the role of gabapentin and other treatment modalities

in the prevention of PHN.

J Severity of acute pain may be predictive of long-term persistence of pain. Severity of pain

may, therefore, be an appropriate decision factor in initiating antiviral treatment. Further

research is recommended to evaluate the benefit and applicability of this approach.

J Although every effort should be made to encourage early treatment, research is needed

to assess the effectiveness of initiating antiviral treatment in patients presenting more

than 72 hours after rash onset.

IntroductionJ It is important that patients with herpes zoster are encouraged to present to physicians

as early as possible for prompt medical care. Both public and medical education

efforts may be beneficial (category 3 recommendation).

For the majority of immunocompetent individuals, herpes zoster manifests as a rash with

pain of relatively short duration. In others there is the risk of complications (e.g. herpes

zoster ophthalmicus) and, particularly in the elderly, of prolonged pain. The goals of

therapy in immunocompetent individuals with herpes zoster include accelerating

cutaneous healing, reducing acute pain

and, more importantly, preventing

complications such as persistent pain

and herpes zoster ophthalmicus. A

number of therapeutic options have

been assessed, including antiviral

therapy, corticosteroids and anti-

depressants, each of which has a

different rationale for its use (Table 1).

Antiviral Therapy in Herpes ZosterJ Antiviral therapy for immunocompetent adults >50 years of age with herpes zoster is

recommended, particularly if therapy can be instituted within 72 hours of lesion onset.

As a result of their improved pharmacokinetic profiles and simpler dosing regimens,

valaciclovir (1000 mg three times a day for 7 days) or famciclovir (250 mg or 500 mg

three times a day) have replaced aciclovir (800 mg five times a day) as the preferred

treatment (category 1 recommendation).

J There is no published evidence for the use of famciclovir 750 mg once daily for the

treatment of herpes zoster, and no follow-up data on its use beyond 1 month (category

3 recommendation).

J There is no indication for the use of topical antiviral agents in the treatment of

cutaneous herpes zoster (category 1 recommendation).

J Intravenous aciclovir (10 mg/kg every 8 hours) remains the drug of choice for those

rare immunocompetent patients with herpes zoster who develop evidence of

pneumonitis or disseminated infection (e.g. varicella zoster virus [VZV] encephalitis)

(category 2 recommendation).

TABLE 1: Summary oftreatment options forimmunocompetentindividuals with herpeszoster1

Patient group Treatment options

Age <50 years with mild pain Symptomatic care only

Ophthalmic rash Oral valaciclovir, famciclovir oraciclovir for 7 daysOphthalmological examination

Age ≥50 years or moderate-to-severe pain Oral valaciclovir, famciclovir oraciclovir for 7 daysConsider corticosteroidsConsider tricyclic antidepressants

Page 61: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

60

The clinical manifestations of herpes zoster result from reactivation of VZV and, therefore,

to reduce disease severity, it is logical to inhibit such replication. Three antiviral drugs –

aciclovir (800 mg five times a day for 7 days), valaciclovir (1000 mg three times a day) and

famciclovir (250 mg or 500 mg three times a day) – are widely available for treatment of

VZV infection. All three are effective, have excellent safety profiles, and are approved for

the treatment of herpes zoster in many countries.

AciclovirAciclovir was the first antiviral therapy investigated in herpes zoster patients. Intravenous

aciclovir (5–10 mg/kg three times daily for 5 days) was effective in several randomized,

placebo-controlled trials.2 All the trials demonstrated that aciclovir reduced acute pain,

providing therapy was initiated within 72 hours of rash onset. The impracticalities of

intravenous therapy led to the investigation of oral aciclovir. A series of randomized,

placebo-controlled studies were conducted with an aciclovir dose of 800 mg five times

daily for 7–10 days administered within 72 hours of rash onset. Oral aciclovir reduced the

duration of virus shedding and new lesion formation, and significantly accelerated healing

of the rash. A meta-analysis of four trials demonstrated that aciclovir treatment significantly

shortened the duration of zoster-associated pain.3 Benefit was particularly evident in

patients aged 50 years and older, in whom pain relief was on average twice as fast as those

receiving placebo.

A further meta-analysis of the placebo-controlled trials of aciclovir compared the efficacy

of early therapy (started within 48 hours of rash onset) with late therapy (not started until

48–72 hours after rash onset).4 Irrespective of when aciclovir therapy was initiated, within

72 hours of rash onset, there was a statistically significant reduction in median time to

resolution of acute pain versus placebo. These data have been discussed fully in the

previous IHMF Management Strategies in Herpes publication – Reducing the Burden of

Zoster-Associated Pain – Update.5

Aciclovir, therefore, is effective in the treatment of herpes zoster and has been the

benchmark for the assessment of valaciclovir and famciclovir.

ValaciclovirValaciclovir (1000 mg three times daily for 7 or 14 days) was compared with aciclovir

(800 mg five times daily for 7 days) for the treatment of herpes zoster in 1141 patients aged

over 50 years (mean age 68 years).6 In an intent-to-treat analysis, all regimens had similar

effects on rash healing but both valaciclovir regimens alleviated pain significantly faster

than aciclovir.

The 7-day valaciclovir regimen accelerated the resolution of zoster-associated pain 34%

faster than aciclovir (hazard ratio [HR] 1.34, 95% confidence interval [CI] 1.12, 1.60;

TABLE 2: Relative risksfor the duration of pain inpatients with herpeszoster treated withvalaciclovir 1000 mgthree times daily for 7 or 14 days or aciclovir800 mg five times dailyfor 7 days6

Intent-to-treat analysis (n=1141) P-value Ophthalmic zoster subset (n=119) P-value

Valaciclovir 7 day 1.34 (1.12, 1.60) 0.001 1.27 (0.72, 2.22) ns versus aciclovir

Valaciclovir 14 day 1.22 (1.03, 1.46) 0.03 1.15 (0.66, 2.00) nsversus aciclovir

Valaciclovir 7 day 1.10, (0.92, 1.30) ns 1.10 (0.63, 1.93) nsversus valaciclovir 14 day

ns=not significant

Page 62: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

61

P=0.001) (Table 2, Figure 1).6 The median time to cessation of pain was shorter in the 7- and

14-day valaciclovir groups (38 and 44 days, respectively) than in the aciclovir group

(51 days). However, 14-day administration did not have additional clinical benefit over the

7-day regimen. Treatment with valaciclovir (pooled

7- and 14-day treatment data) resulted in a lower

incidence of prolonged pain greater than 6 months’

duration than aciclovir treatment (19.3% versus

25.7%; P=0.02).7,8

A subsequent analysis of these data has shown that

the benefit of valaciclovir over aciclovir was

maintained whether treatment was started within

48 hours of rash onset or 48–72 hours after rash

onset.4

A Japanese study compared valaciclovir (1000 mg

three times daily for 7 days) with aciclovir (800 mg five times daily for 7 days) for the

treatment of herpes zoster in 200 patients.9,10 There were no significant differences

between valaciclovir and aciclovir with respect to time to 50% vesicle crusting or time to

complete healing. However, valaciclovir showed a significant improvement over aciclovir

in terms of time to cessation of new lesion formation (P=0.0039). PHN – defined by the

authors as pain persisting for more than 2 weeks following initiation of therapy – was

observed in 28% and 35% of valaciclovir-treated and aciclovir-treated patients,

respectively. The median times to resolution of PHN were 58 days and 86 days for

valaciclovir-treated and aciclovir-treated patients, respectively.10 Formal statistical analysis

was not possible as only those patients experiencing PHN were followed.

FamciclovirFamciclovir (500 mg and 750 mg three times daily for 7 days) has undergone evaluation

in a placebo-controlled study in patients with herpes zoster, aged 18 years or over,

presenting within 72 hours of rash onset. Analysis of patients (44%) who developed PHN

(pain persisting after rash healing) showed a significant reduction in its duration in the

famciclovir arm compared with placebo.11 In terms of acute pain, neither famciclovir dose

showed benefit compared with placebo.11 An analysis of prolonged zoster-associated pain

was not presented by Tyring et al.11 Importantly, the study evaluated a famciclovir dose of

500 mg three times daily, the dose that is approved in the USA; however, no connection

can be inferred between this and the 250 mg three times daily dose approved for use in

many other countries.12

An international study of 545 patients with herpes zoster, presenting within 72 hours of

rash onset, evaluated famciclovir 250 mg, 500 mg or 750 mg three times daily for 7 days

compared with aciclovir 800 mg three times daily for 7 days.13 This study showed no

difference between any of the three doses of famciclovir and aciclovir with respect to

cutaneous healing parameters. In a subset analysis of all patients treated within 48 hours

of rash onset, famciclovir 500 mg three times daily reduced the duration of zoster-

associated pain significantly faster than aciclovir; however, no significant difference was

observed between either the 250 mg or 750 mg famciclovir dose and aciclovir.13 A further

subset analysis – patients treated within 48 hours of rash onset, who did not have crusts at

enrolment (39% of the original sample size) – demonstrated faster resolution of zoster-

FIGURE 1: Time tocessation of pain in herpeszoster patients treatedwith valaciclovir or oralaciclovir6

Time from first dose (days)

1.0

0.8

0.6

0.4

0.2

0

Prop

ortio

n of

pat

ient

sex

peri

enci

ng p

ain

Cox regression P≤0.03

Aciclovir (7 day)

Valaciclovir (7 day)

0 20 40 60 80 100 120 140 160 180

© 2

002

Rep

rodu

ced

with

per

mis

sion

from

Els

evie

r Sc

ienc

e. B

eutn

er K

R, F

ried

man

DJ,

Fors

zpan

iak

Cet

al.

Val

acic

lovi

r co

mpa

red

with

acy

clov

ir fo

r im

prov

ed th

erap

y fo

r he

rpes

zost

er in

imm

unoc

ompe

tent

adu

lts. A

ntim

icro

b A

gent

s C

hem

othe

r19

95;3

9(7)

:154

6-15

53.

Page 63: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

62

associated pain for all three doses of famciclovir compared with aciclovir 800 mg five

times daily.13 However, when these data were age-adjusted, only the famciclovir 250 mg

three times daily dosage was statistically significant.13

Valaciclovir compared with famciclovir Valaciclovir (1000 mg three times daily for 7 days)

was compared with famciclovir (500 mg three times

daily for 7 days) for the treatment of herpes zoster in

597 patients.12 No differences (either statistical or

clinical) were observed between famciclovir

(n=300) and valaciclovir (n=297) with respect to

either rash healing, acute pain resolution or

resolution of PHN (Figure 2).12 At treatment

initiation, prodromal pain was absent in more

patients in the famciclovir group than in the

valaciclovir group (30 versus 22, P=0.03). Of those

patients experiencing prodromal pain, a higher

proportion in the valaciclovir group experienced

severe or very severe prodromal pain compared with the famciclovir group (34/78 versus

24/70 respectively, P=0.03).12 More intense or prolonged prodromal pain and more severe

acute pain are important risk factors for prolonged zoster-associated pain, so it is possible

that the baseline disease characteristics were biased.

With their superior pharmacokinetics and convenience of administration, valaciclovir7

and famciclovir11 are preferable to aciclovir for cases of uncomplicated herpes zoster in

otherwise healthy adults.

Effect of time of initiation of antiviral therapyJ Although every effort should be made to encourage early treatment, research is needed

to assess the effectiveness of initiating antiviral treatment in patients presenting more

than 72 hours after rash onset (research need recommendation).

Valaciclovir and aciclovir are equally effective for

herpes zoster when treatment initiation is 48–72 hours

after rash onset or within 48 hours of rash onset

(Figure 3).4,14,8

An observational, non-comparative study enrolled

1897 patients treated with valaciclovir (1000 mg

three times daily for 7 days) from the day of

presentation.15 As the impact of valaciclovir on pain

outcome may not be limited to patients whose

treatment is initiated within 72 hours of rash onset,

patients who presented after this were included in

the study. All patients were assessed for time to

cessation of zoster-associated pain, time to

complete cessation of abnormal sensations, and

time to complete rash healing on Days 1, 8 and 29,

and then every 28 days up to 24 weeks. Resolution

FIGURE 2: Resolutionof pain in patients withherpes zoster treated withvalaciclovir (1000 mgthree times daily) orfamciclovir (500 mg threetimes daily)12

1.00.90.80.70.60.50.40.30.20.1

00 50 75

Prop

ortio

n of

pat

ient

s w

ith p

ain

17515012510025

ValaciclovirFamciclovir

l f h h l l

Duration of zoster-associated pain (days)

FIGURE 3: Effect of timeto treatment initiation onduration of zoster-associated pain in patientstreated with valaciclovir oraciclovir for 7 days.8

<72 <48

Time to treatment after rash onset (hours)

48–72

50

40

30

20

10

0Med

ian

dura

tion

of z

oste

r-as

soci

ated

pai

n (d

ays)

Valaciclovir (1000 mg 3x/day) n=384

Aciclovir (800 mg 5x/day) n=376

***

*

**

*P=0.03, **P=0.02, ***P=0.001 versus aciclovir

At least 60% of patients in each group began treatment within 48 hours of rash onset.

© R

epro

duce

d w

ith p

erm

issi

on, T

yrin

g SK

, Beu

tner

KR

, Tuc

ker

BAet

al.

Ant

ivir

al th

erap

y fo

r he

rpes

zos

ter:

ran

dom

ized

, con

trol

led

clin

ical

tria

lof

val

acyc

lovi

r an

d fa

mci

clov

ir th

erap

y in

imm

unoc

ompe

tent

pat

ient

s50

yea

rs a

nd o

lder

. Arc

h Fa

m M

ed20

00;9

(9):8

63-8

69. A

mer

ican

Med

ical

Ass

ocia

tion.

© R

epro

duce

d w

ith p

erm

issi

on, O

rmro

d D

, Goa

K. V

alac

iclo

vir:

a r

evie

w o

f its

use

in th

em

anag

emen

t of h

erpe

s zo

ster

. Dru

gs20

00;5

9(6)

:131

7-13

40.

Page 64: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

63

of zoster-associated pain and abnormal sensations was similarly rapid in patients whose

treatment was initiated more than 72 hours after rash onset compared with patients whose

treatment was initiated earlier (Table 3).

This finding is important as it indicates that valaciclovir may have a benefit even when

treatment is commenced outside what is

considered to be the ‘normal’

therapeutic window. It also shows that

the duration of rash is not a predictor of

the outcome of zoster-associated pain.

As the study demonstrated that patients

with moderate-to-severe prodromal pain or acute pain during the rash phase were at

greater risk of zoster-associated pain, it showed that prior duration or severity of pain

should be used to select or stratify cases in future studies.15 There are no published data

available on the efficacy of famciclovir when treatment is initiated more than 72 hours

after rash onset.

Corticosteroid Therapy in Herpes ZosterJ It is recommended that physicians consider the use of steroids to reduce the

inflammation that may be contributing to acute pain, provided there are no

contraindications and the potential risk of excess side-effects is explained. Although

they do not prevent PHN, steroids reduce acute symptoms and may facilitate return to

normal quality of life (category 1 recommendation).

The premise that the pain experienced by people with herpes zoster is primarily due to

inflammation and necrosis of neurones led to the suggestion that, by reducing

inflammation, corticosteroids would reduce pain. Small studies provided conflicting data

on the efficacy of corticosteroids. However, the debate on the role of corticosteroid therapy

in herpes zoster has largely been resolved by two large prospective clinical trials.16,17 While

both larger studies demonstrated their benefit in reducing the duration of acute pain, neither

study showed any reduction in the incidence or duration of PHN among corticosteroid

recipients.

In one of the studies, which was placebo-controlled, 349 patients received aciclovir (800 mg

five times daily) for 7 or 21 days with either prednisolone (40 mg daily tapering to

5 mg daily) or placebo for 21 days.17 Steroid treatment had a significant effect on rash healing

at Days 7 and 14 (P=0.02), and significantly reduced the proportion of patients experiencing

pain on Days 7 and 14 (P<0.01). However, there was no significant benefit in the time to

complete cessation of pain with steroid administration compared with placebo.17

In the second study, 208 immunocompetent herpes zoster patients aged over 50 years

were randomized to one of four treatment groups: either aciclovir (800 mg five times daily)

alone, prednisolone (60 mg per day on Days 1–7, 30 mg per day on Days 8–14, 15 mg

per day on Days 15 to 21) alone, aciclovir in combination with prednisolone, or placebo

alone.16 Patients receiving aciclovir plus prednisolone had significant improvements

compared with placebo recipients with respect to time to:

TABLE 3: Time to loss of abnormal sensationsand zoster-associatedpain in patients treatedwith valaciclovir ≤72 hours and >72 hoursafter rash onset15

Treatment initiation in relation to rash onset

Parameter ≤72 hours >72 hours

Time to cessation of abnormal 24 days 22 dayssensations

Time to loss of zoster-associated pain 16 days 15 days

Page 65: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

64

• Total crusting

• Lesion healing

• Cessation of acute neuritis

• Return to normal activities (i.e. uninterrupted sleep, usual daily activity)

• Cessation of analgesic therapy (P<0.05 for all) (Table 4).16

TABLE 4: Diseaseresolution based uponCox regression model foraciclovir withprednisolone andaciclovir alone in thetreatment of herpeszoster16

Dependent variable risk ratio (95% CI)

Aciclovir plus prednisolone Aciclovir aloneversus placebo n (range) versus placebo n (range)

1-month evaluation of cutaneous healingTime to total crusting 2.27 (1.46–3.55)* 1.51 (0.98–2.33) Time to total healing 2.07 (1.26–3.38)* 1.57 (0.97–2.53)

1-month evaluation of quality of life Time to cessation of acute neuritis 3.02 (1.42– 6.41)* 1.47 (0.67–3.21) Time to uninterrupted sleep 2.12 (1.25–3.58)* 1.16 (0.58–2.05) Time to return to 100% usual activity 3.22 (1.92–5.40)* 1.63 (0.96–2.76) Time to no use of analgesic agents 3.15 (1.69–5.39)* 1.27 (0.66–2.49)

6-month evaluation of pain Time to cessation of zoster-associated pain 1.56 (0.92–2.66) 1.39 (0.84–2.32)

* P<0.05

Although benefits were shown for resolution of acute pain, there was no significant

difference in zoster-associated pain at 6 months between patients receiving prednisolone

plus aciclovir and those receiving placebo.16

In a third, large study of aciclovir with and without prednisolone,17 treatment with

aciclovir for 21 days or the addition of prednisolone to aciclovir therapy conferred only

slight benefits over standard 7-day treatment with aciclovir. Additional treatment with

prednisolone did not reduce the frequency of PHN, and the steroid recipients reported

more adverse events.

The data support the use of corticosteroids plus an antiviral compound in older individuals

(≥50 years of age) with significant pain at presentation. Corticosteroid therapy without co-

administration of an antiviral compound to limit viral replication is not recommended.

Since corticosteroid therapy has a range of adverse effects, its potential short-term benefit

on acute herpes zoster needs to be weighed against possible risks. Published studies have

examined only the combination of aciclovir and corticosteroids, although there are no

theoretical reasons why similar benefits would not be seen with corticosteroids used in

combination with valaciclovir or famciclovir.

Antidepressant Treatment for Herpes ZosterJ It is recommended that early use of tricyclic antidepressants in elderly patients with

acute herpes zoster should be considered as it may prevent or reduce PHN (category 2

recommendation).

Tricyclic antidepressant treatment initiated early in the clinical course of herpes zoster has

the potential to reduce the number of patients with prolonged pain, and shorten the

duration of pain in patients experiencing prolonged zoster-associated pain.18 In addition

to their effect of alleviating depression, these agents also have analgesic properties. This is

most likely due to their action of blocking re-uptake of monoamine neurotransmitters

released by descending axons from the brainstem.

Page 66: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

65

In elderly patients (>60 years of age), amitriptyline (25 mg once daily for 90 days) alone and

in combination with aciclovir (800 mg five times daily for 7 days) given within 48 hours of

rash onset was compared with placebo for the treatment of herpes zoster.18 Patients entering

the study were randomized to either amitriptyline or placebo; they received aciclovir

(800 mg five times daily for 7 days) according to their physician’s discretion.

When data from all patients receiving amitriptyline (amitriptyline alone and amitriptyline plus

aciclovir groups) were compared with those receiving placebo only, a significantly higher

proportion of the amitriptyline recipients were pain-free at 6 months (84.2% versus 64.7%;

P=0.05) .18 This controlled trial suggests that treatment of herpes zoster with aciclovir (or another

effective antiviral) and low-dose amitriptyline reduces the duration of PHN. As the efficacy of

amitriptyline may be dose-dependent, further studies are needed to confirm this finding,

evaluate other doses of the antidepressant and assess the benefit of other antiviral therapies.

Sympathetic Nerve Blocks and Other Agents to Treat Acute Painin Herpes Zoster and for Prevention of PHNJ The presence of risk factors for the development of PHN should be assessed and

documented for each patient (category 3 recommendation).

J If the patient has severe acute pain, the following treatments may be beneficial:

sympathetic nerve blocks, antidepressants, gabapentin and opioids (category 3

recommendation).

J Studies are required to establish the role of gabapentin and other treatment modalities

in the prevention of PHN (research need recommendation).

J Severity of acute pain may be predictive of long-term persistence of pain. Severity of pain

may, therefore, be an appropriate decision factor in initiating antiviral treatment. Further

research is recommended to evaluate the benefit and applicability of this approach

(research need recommendation).

Numerous authors have reported the utility of sympathetic nerve blockade to reduce acute

pain in herpes zoster. Because greater acute pain severity increases the risk of PHN, any

intervention that reduces the duration or intensity of this pain has the potential to reduce

the severity or incidence of PHN. Thus, many of the studies of the efficacy of sympathetic

nerve blockade in acute zoster pain also examined the effect of this procedure on the

development of PHN.

In a retrospective analysis, Colding19 examined the effects of sympathetic nerve blocks

administered every second day in 243 patients with acute zoster-associated pain. The

patients were aged from 10 to 92 years, with 60% over 60 years. In all cases, the nerve

blocks were performed with a 1% solution of lidocaine and norepinephrine, and on

average, patients each received two doses. Improvement or complete relief from the acute

pain was observed in 206 patients, with the remaining 37 individuals reporting no change

in pain level. Seventy-one patients were followed up for 6 months, including 14 who had

reported no change in pain level following the nerve block. Five of the 71 patients

developed PHN but as the ages of the patients in the follow-up group were not specified,

it is not possible to determine how this observed rate compares with the expected rate.19

Page 67: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

66

A second retrospective analysis by Colding20 of 483 patients with acute herpes zoster,

using the same treatment regimen as described above, demonstrated improvement or

complete pain relief in 430 patients, with 53 not responding to treatment. Twelve-month

follow-up was obtained for 155 patients, of whom 5% developed PHN. The author

concluded that, based on a comparison with previously published data, nerve blocks

prevented PHN.20 However, given that different rates of PHN have been observed both in

clinical trials and in retrospective studies, this conclusion is questionable.21

A randomized, double-blind, placebo-controlled trial was conducted in 20 patients aged

over 50 years with herpes zoster of less than 6 weeks’ duration.22 The patients were

randomized to receive four daily injections of either bupivacaine or placebo.

A significantly higher proportion of the bupivacaine group were pain-free following the

four injections, compared with the placebo group (9/10 bupivacaine versus 2/10 placebo;

P<0.01).22 The remaining patient in the bupivacaine group and seven patients from the

placebo group who were still experiencing pain received a second series of four daily

injections of bupivacaine, irrespective of the group to which they were initially

randomized. The patient from the original bupivacaine group continued to experience

pain following the second series of injections. Four of the seven patients from the original

placebo group experienced complete pain relief following the second series of injections,

two reported persistent pain, but at approximately 20% of its original intensity, and one

continued to experience unmitigated persistent pain (Table 5).22 Over a 12-month follow-

up period, all subjects continued to report the same degree of pain relief that was noted

immediately following the treatment course. Based on these findings, the authors suggest

that nerve blocks may prevent the

development of PHN, although there

was no comparison with other studies.

A retrospective study of the effectiveness

of sympathetic nerve blocks in

preventing PHN when administered at

numerous intervals following the onset

of herpes zoster documented that nerve blocks initiated within 2 months were more

effective than treatment initiated after 2 months (n=122).23 Approximately 90% of patients

treated within 2 months of herpes zoster onset had complete pain resolution by 1 year,

compared with only 14% of patients whose treatment was initiated between 2 months and

1 year following onset of herpes zoster. This suggests, as expected, that earlier treatment,

before the establishment of chronic pain, is more effective. No relevant baseline data were

provided to allow comparison with expected outcome had no treatment been given.

Epidural administration of local anaesthetics with or without steroids was investigated in a

randomized, controlled study of 485 adults aged over 55 years with acute pain associated

with herpes zoster.24 Patients with rash of less than 7 days’ duration and severe pain were

randomized to receive either:

• Intravenous aciclovir 10 mg/kg three times daily for 9 days plus prednisolone (60 mg

daily with progressive reduction) for 21 days

or

• 6–12 ml bupivacaine (0.25%) every 6–8 or 12 hours plus methylprednisolone 40 mg

every 3–4 days by epidural catheter during a period ranging from 7 to 21 days.

TABLE 5: Number ofpatients experiencingpain due to herpes zosterfollowing treatment withsympathetic nerve block22

No pain Pain n (%) n

First series Bupivacaine 9 (90) 1 Electrolyte solution 2 (20) 8

Second series Bupivacaine-Bupivacaine 0 1 Electrolyte-Bupivacaine 4 3

Page 68: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

67

In this study, the incidence of pain at 12 months was 22.2% in patients receiving aciclovir

plus steroids, but only 1.6% after epidural analgesia plus steroids, supporting the suggestion

that earlier treatment, before the establishment of chronic pain, is more effective.

However, another study found that PHN developed in 14% (10/72) of patients given

sympathetic nerve blocks within 30 days of the onset of herpes zoster.25 The place of nerve

blocks in the management of acute pain is clearer than their use in prevention of PHN.

Sympathetic nerve blocks are not without hazard and would place a major demand on

healthcare resources if widely used. Further work may identify a group of patients where

benefit may justify increased risk and cost.

Whether or not the administration of sympathetic nerve blocks has the potential to prevent

PHN or reduce the likelihood of its development remains controversial.21 This is partly

because of methodological issues associated with the trials of this intervention (e.g.

patients selected for follow-up), but also due to the lack of proper comparison groups,

which makes it difficult to determine whether treatment resulted in fewer patients with

PHN than would be expected from natural history studies.

Gabapentin and opioids may also be beneficial for the treatment of acute pain in herpes

zoster. These agents should be titrated at similar doses as those given for the treatment of

chronic pain (see Chapter 9).

SummaryIn many individuals, herpes zoster comprises rash and pain of relatively short duration.

However in some individuals, especially the elderly, herpes zoster is associated with

complications and prolonged pain. The aims of treating herpes zoster are to minimize the

duration and severity of pain and prevent the associated complications.

Antiviral therapy for immunocompetent adults over 50 years of age with herpes zoster is

routinely recommended, particularly if it can be instituted within 72 hours of lesion onset.

Because of their improved pharmacokinetic profiles and simpler dosing regimens,

valaciclovir (1000 mg three times a day) or famciclovir (250 mg or 500 mg three times a

day) have replaced aciclovir (800 mg five times a day) as the preferred treatment. In

addition, it is recommended that physicians consider the use of steroids to reduce the

inflammation that may be contributing to zoster associated-pain.

There is some evidence that antiviral therapy should be considered for patients whose rash

has been present longer than 72 hours and in whom clinical examination reveals new

vesicle formation (indicative of ongoing virus replication). Similarly, if risk factors for

severe or protracted pain are evident at presentation, antiviral therapy should be initiated

irrespective of time since rash onset.

Tricyclic antidepressants, such as amitriptyline (25 mg once daily; 10 mg in the frail and

elderly, elevated every few days to a dose that is effective without severe side-effects),

particularly if initiated early in the clinical course of herpes zoster, can relieve the acute

pain associated with the disease and have the potential to prevent the development

of PHN.

Page 69: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

68

Sympathetic nerve blocks have been studied in several trials, most of which were

uncontrolled and lacked comparison groups. Thus, it is difficult to draw any conclusions

about the efficacy of this intervention in herpes zoster.

In general, patients with herpes zoster should receive antiviral therapy to limit viral

replication and, thereby, minimize the acute pain associated with herpes zoster, help

prevent PHN, prevent complications and speed the resolution of rash.

References

1. Cohen JI, Brunell PA, Straus SE et al. Recent advances in varicella-zoster virus infection. Ann Intern Med1999;130(11):922-932.

2. Wood MJ. Treatment of zoster. Rev Med Microbiol 1995;6:165-174.3. Wood MJ, Kay R, Dworkin RH et al. Oral acyclovir therapy accelerates pain resolution in patients with

herpes zoster: a meta-analysis of placebo-controlled trials. Clin Infect Dis 1996;22(2):341-347.4. Wood MJ, Shukla S, Fiddian AP et al. Treatment of acute herpes zoster: effect of early (< 48 h) versus late

(48-72 h) therapy with acyclovir and valaciclovir on prolonged pain. J Infect Dis 1998;178 Suppl 1:S81-84.5. Wood MJ, Kroon S. Management Strategies in Herpes: Reducing the burden of zoster-associated pain—

update. 2nd ed. Worthing: PPS Europe Ltd, 1995.6. Beutner KR, Friedman DJ, Forszpaniak C et al. Valaciclovir compared with acyclovir for improved therapy

for herpes zoster in immunocompetent adults. Antimicrob Agents Chemother 1995;39(7):1546-1553.7. Beutner KR. Valacyclovir: a review of its antiviral activity, pharmacokinetic properties, and clinical efficacy.

Antiviral Res 1995;28(4):281-290.8. Ormrod D, Goa K. Valaciclovir: a review of its use in the management of herpes zoster. Drugs

2000;59(6):1317-1340.9. Taylor-Wiedeman J, Yamashita K, Miyamura K et al. Varicella-zoster virus prevalence in Japan: no significant

change in a decade. Jpn J Med Sci Biol 1989;42(1):1-11.10. Niimura M, Honda M, Nishikawa T. Phase III study of valaciclovir hydrochloride tablets in patients with

herpes zoster: double-blind comparative study with aciclovir tablets [in Japanese]. Rinsho Iyaku1998;14(16):2867-2902.

11. Tyring S, Barbarash RA, Nahlik JE et al. Famciclovir for the treatment of acute herpes zoster: effects on acutedisease and postherpetic neuralgia. A randomized, double-blind, placebo-controlled trial. CollaborativeFamciclovir Herpes Zoster Study Group. Ann Intern Med 1995;123(2):89-96.

12. Tyring SK, Beutner KR, Tucker BA et al. Antiviral therapy for herpes zoster: randomized, controlled clinicaltrial of valacyclovir and famciclovir therapy in immunocompetent patients 50 years and older. Arch FamMed 2000;9(9):863-869.

13. Degreef H, Group FHZCS. Famciclovir, a new oral antiherpes drug: results of the first controlled clincialstudy demonstrating its efficacy and safety in the treatment of uncomplicated herpes zoster inimmunocompetent patients. Int J Antimicrob Agents 1994;4:241-246.

14. Wood MJ. Treatment of herpes zoster in the elderly. Herpes 1998;5:60-63.15. Decroix J, Partsch H, Gonzalez R et al. Factors influencing pain outcome in herpes zoster: an observational

study with valaciclovir. Valaciclovir International Zoster Assessment Group (VIZA). J Eur Acad DermatolVenereol 2000;14(1):23-33.

16. Whitley RJ, Weiss H, Gnann JW, Jr. et al. Acyclovir with and without prednisone for the treatment of herpeszoster. A randomized, placebo-controlled trial. The National Institute of Allergy and Infectious DiseasesCollaborative Antiviral Study Group. Ann Intern Med 1996;125(5):376-383.

17. Wood MJ, Johnson RW, McKendrick MW et al. A randomized trial of acyclovir for 7 days or 21 days withand without prednisolone for treatment of acute herpes zoster. N Engl J Med 1994;330(13):896-900.

18. Bowsher D. The effects of pre-emptive treatment of postherpetic neuralgia with amitriptyline: a randomized,double-blind, placebo-controlled trial. J Pain Symptom Manage 1997;13(6):327-331.

19. Colding A. The effect of regional sympathetic blocks in the treatment of herpes zoster. Acta AnaesthesiolScand 1969;13:133-141.

20. Colding A. Treatment of pain: organization of a pain clinic: treatment of acute herpes zoster. Proc R Soc Med1973;66(6):541-543.

21. Wu CL, Marsh A, Dworkin RH. The role of sympathetic nerve blocks in herpes zoster and postherpeticneuralgia. Pain 2000;87(2):121-129.

22. Tenicela R, Lovasik D, Eaglestein W. Treatment of herpes zoster with sympathetic blocks. Clin J Pain1985;1:63-67.

23. Winnie AP, Hartwell PW. Relationship between time of treatment of acute herpes zoster with sympatheticblockade and prevention of post-herpetic neuralgia: clinical support for a new theory of the mechanism bywhich sympathetic blockade provides therapeutic benefit. Reg Anesth 1993;18(5):277-282.

24. Pasqualucci A, Pasqualucci V, Galla F et al. Prevention of post-herpetic neuralgia: acyclovir andprednisolone versus epidural local anesthetic and methylprednisolone. Acta Anaesthesiol Scand2000;44(8):910-918.

25. Riopelle JM, Naraghi M, Grush KP. Chronic neuralgia incidence following local anesthetic therapy forherpes zoster. Arch Dermatol 1984;120(6):747-750.

Page 70: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Management RecommendationsCategory 2J To prevent ocular complications, antiviral therapy is recommended for all patients

with herpes zoster ophthalmicus who present within 1 week of onset of lesions.

Aciclovir (800 mg five times a day for 7 days), valaciclovir (1000 mg three times daily)

and famciclovir (500 mg three times daily) have been proven to reduce the ocular

complications of herpes zoster ophthalmicus. Valaciclovir and famciclovir have the

advantage of a simpler dosing schedule.

J Intravenous aciclovir (10 mg/kg every 8 hours for adults, 500 mg/m2 every 8 hours for

children) followed by the oral formulation is the treatment of choice for acute retinal

necrosis caused by varicella zoster virus (VZV). Valaciclovir (1000 mg three times

daily) and famciclovir (500 mg three times daily) should also be considered for use in

this situation.

Category 3J It is recommended that patients with herpes zoster ophthalmicus are treated with

systemic antiviral therapy, and that topical antivirals should only be administered in

addition to systemic medication during the acute phase of herpes zoster ophthalmicus.

J It is strongly recommended that patients with obvious ocular involvement should be

referred to an ophthalmologist.

J It is not recommended that corticosteroid therapy (either topical or systemic) be

prescribed except upon the advice of an ophthalmologist.

J It is recommended that antiviral treatment of herpes zoster ophthalmicus is initiated up

to 7 days following rash onset.

Research needJ It is recommended that a study is initiated to research the benefit of long-term antiviral

treatment on the outcome of herpes zoster ophthalmicus.

Herpes Zoster OphthalmicusHerpes zoster ophthalmicus occurs in 10–25% of patients with herpes zoster; this

dermatomal presentation is second in frequency only to thoracic herpes zoster.1-3 The scale

of the problem is demonstrated by the statistic that of the 1 million physician visits for

herpes zoster in the USA, one quarter involve the eye.4 Ocular complications occur in

50–90% of herpes zoster ophthalmicus patients and can lead to substantial visual

disability;5-8 10–15% of patients with herpes zoster ophthalmicus experience anatomical

or functional sequelae.9 69

VARICELLA ZOSTER VIRUS INFECTION OF THE EYE7

Page 71: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Herpes zoster ophthalmicus is due to reactivation of latent VZV in the first division of the

trigeminal nerve. The resultant dermal rash may extend from the nose and eye to the vertex

of the skull but rarely extends across the midline of the patient’s forehead.10 When the

nasociliary portion of the optic nerve is involved, which occurs in about 33% of patients

with herpes zoster ophthalmicus, there is a significant correlation with ocular

complications.8 Proliferation of VZV causes destruction of the ganglion but VZV is not

limited to the trigeminal nerve – it may travel to the central nervous system (CNS) where

it initiates necrosis in the corresponding sensory nuclei.11

Ocular complications of herpes zoster ophthalmicusJ It is strongly recommended that patients with obvious ocular involvement should be

referred to an ophthalmologist (category 3 recommendation).

The ocular complications of herpes zoster ophthalmicus may affect almost all structures of

the eye, including the orbit, conjunctiva, cornea, retina and optic nerve (Figure 1). The

complications are attributable to a number of mechanisms including virus replication and

dissemination, neuronal damage, ischaemic vasculitis and an inflammatory

granulomatous reaction.10 However, although much is known about manifestations of the

complications of herpes zoster ophthalmicus, less is known about the incidence and long-

term course of ocular involvement subsequent to it.

Conjunctiva. The most common manifestation is a non-specific conjunctivitis with a mixed

follicular and papillary reaction, which usually resolves within a week. Membranous

conjunctivitis can develop and lead to conjunctival scarring if inadequately treated.

Scleritis may occur during the acute phase of the

disease, but can also occur 2–3 months after the

rash has resolved.

Orbit. Complications may include proptosis,

chemosis and ocular motor palsies. The latter results

from localized spread of the virus within the orbit.10

Cornea. The complications are frequent and may be

both acute and chronic in nature. The cornea can

develop various signs of infection including an

epithelial or stromal keratitis. It develops shortly

after disease onset due to virus replication in the

corneal epithelium and occurs in 50% of patients with herpes zoster ophthalmicus. The

epithelial lesions can appear as small, slightly elevated, dendritic-shaped lesions that stain

slightly with fluorescein but vividly with rose Bengal. Because their shape resembles the

dendritic ulcerations seen in herpes simplex infections, they have been labelled

pseudodendrites. Later, corneal complications may result from host inflammatory

responses, and include anterior stromal infiltrates, sclerokeratitis, keratouveitis,

endotheleitis and disciform keratitis.

Uvea. The most frequently observed signal of intraocular involvement is mild-to-severe

anterior uveitis. It can occur independently of corneal or conjunctival disease and can

become chronic and recurrent. Patients with herpes zoster ophthalmicus-related iritis can

also develop elevated intraocular pressure due to inflammation or cellular damage.

70

FIGURE 1: Herpeszoster ophthalmicusinfection of theconjunctiva

Page 72: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Retina. Herpes zoster ophthalmicus can result in various retinal conditions, including

retinal haemorrhages, retinitis, ischaemic vasculitis and acute retinal necrosis syndrome.

The latter complication is discussed later in this chapter.

Optic nerve. The vasculitis associated with herpes zoster ophthalmicus may manifest in

the retina and optic nerve accompanied by central retinal vein occlusion, central retinal

artery occlusion, retinal vasculitis or ischaemic optic neuropathy.10 Herpes zoster

ophthalmicus-related optic neuropathy usually begins 1–4 weeks after the onset of rash

and can result in decreased visual acuity.

Therapy for herpes zoster ophthalmicusThe primary aim of treating herpes zoster ophthalmicus is to prevent the potentially severe

and sight-threatening ocular complications that can follow herpes zoster involving the

trigeminal nerve.

J To prevent ocular complications, antiviral therapy is recommended for all patients

with herpes zoster ophthalmicus who present within 1 week of onset of lesions.

Aciclovir (800 mg five times a day), valaciclovir (1000 mg three times daily for 7 days)

and famciclovir (500 mg three times daily) have been proven to reduce ocular

complications. Valaciclovir and famciclovir have the advantage of simpler dosing

schedules (category 2 recommendation).

J It is recommended that antiviral treatment of herpes zoster ophthalmicus is initiated up

to 7 days following rash onset (category 3 recommendation).

J It is not recommended that corticosteroid therapy (either topical or systemic) be

prescribed except upon the advice of an ophthalmologist (category 3

recommendation).

J It is recommended that a study is initiated to research the benefit of long-term antiviral

treatment on the outcome of herpes zoster ophthalmicus (research need

recommendation).

Antiviral treatmentThe use of antiviral therapies in the management of herpes zoster ophthalmicus limits virus

replication and has been shown to have benefit in preventing its ocular complications.

Aciclovir has proved to be effective in the treatment of acute herpes zoster ophthalmicus

in two placebo-controlled trials. In the first of these, oral aciclovir (600 mg five times daily

for 10 days) was compared with placebo in the treatment of immunocompetent patients

with herpes zoster ophthalmicus.12 Aciclovir treatment resulted in quicker resolution of

dermatomal signs and symptoms, and reduced acute zoster-associated pain, particularly

when treatment was initiated within 72 hours of lesion onset.12 In addition, aciclovir

significantly reduced the incidence and severity of several frequent early ocular

complications (Table 1).12 There was no effect on late complications, but this may have

been due to the suboptimal dosage used.

In the second aciclovir trial, 42 patients with acute herpes zoster ophthalmicus of less than

72 hours’ duration received either aciclovir (800 mg five times daily for 10 days) or placebo.1371

Page 73: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

There was a significant reduction in eye disease persisting after 6 months with aciclovir. The

frequency and severity of complications during the early phase was also reduced but this

effect did not reach significance; it is possible that the failure to detect an effect was due to

the study not being of sufficient power.13

Although the results of the two studies

are somewhat in conflict, one showing

an early and the other a late effect,

aciclovir does appear to prevent the

complications of herpes zoster

ophthalmicus. These findings have been

followed by the investigation of both

valaciclovir and famciclovir in the

treatment of herpes zoster

ophthalmicus.

Valaciclovir has the potential to be as

effective as, or more effective than,

aciclovir for treating herpes zoster

ophthalmicus but with a simplified

dosing schedule. Valaciclovir results in

an aciclovir concentration in the

aqueous humour twice as high as that

after aciclovir administration.14

Moreover, corneal concentrations of

aciclovir correlate with plasma

concentrations.14 The potential for valaciclovir to reduce the complications of herpes

zoster ophthalmicus was suggested in a trial of 1141 patients with acute herpes zoster,

in which a small subset (119 patients) had herpes zoster ophthalmicus. There was a

trend to faster resolution of pain in herpes zoster ophthalmicus patients using

valaciclovir compared with aciclovir, but the sample size was too small to permit an

analysis of efficacy.15 Definitive evidence was obtained from a multicentre, randomized

trial comparing valaciclovir (1000 mg three times daily) with oral aciclovir (800 mg five

times daily for 7 days) in 110 immunocompetent patients with herpes zoster

ophthalmicus.14 The ocular complications were similar in the valaciclovir and aciclovir

groups, the most common being conjunctivitis affecting 54% and 52% of valaciclovir-

and aciclovir-treated patients, respectively (Table 2). The long-term outcomes of herpes

zoster ophthalmicus ocular complications were favourable and similar in the two

treatment groups, with only two patients (3.6%) in the valaciclovir group and one

patient (1.9%) in the aciclovir group experiencing ocular lesions that persisted for more

than 6 months. Thus, valaciclovir was as effective as aciclovir in preventing ocular

complications and the two treatments had similar tolerability.14

The safety and efficacy of famciclovir (500 mg three times daily) have also been compared

with oral aciclovir (800 mg five times daily for 7 days) in the treatment of herpes zoster

ophthalmicus.16 Famciclovir was well tolerated and demonstrated efficacy similar to

aciclovir in this study of 454 patients randomized to receive oral famciclovir 500 mg three

times daily or oral aciclovir 800 mg five times daily for 7 days and followed for up to

6 months. For the main outcome measure (loss of visual acuity), there was no significant72

TABLE 1: Ocularcomplications in herpeszoster ophthalmicuspatients receiving oralaciclovir (600 mg fivetimes daily) or placebo12

Aciclovir Placebo P-value ≤n (%) n (%)

Total patients (n) 36 35

Episcleritis 6 (17) 14 (40) Present at entry 1 (3) 7 (20) 0.0250 Developed after entry 5 (14) 7 (20) 0.2250

ScleritisAnterior 1 (3) 3 (9) Posterior 0 (0) 2(6)

Dendritiform keratopathy 12 (33) 21 (60) Present at entry 7 (19) 10 (29) 0.2670 Developed after entry 5 (14) 11 (31) 0.0319

Stromal keratitis (after entry) 9 (25) 19 (56) 0.0081 Mild 7 14 Moderate 2 5 Severe 0 1

Corneal scarring/vascularization 8 (22) 12 (34) 0.1934

Anterior uveitis (after entry) 6 (19) 18 (56) 0.0020 Mild 6 8 Moderate 0 8 Severe 0 2

Keratitic precipitates 7 (19) 17 (49) 0.0112 Mild 6 10 Moderate 0 5 Severe 1 2

Iris atrophy 1 4 (11) 0.1697

Page 74: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

difference between groups. The percentage of patients who experienced one or more ocular

manifestation was similar for famciclovir (142/245, 58.0%) and aciclovir (114/196, 58.2%)

recipients, with no significant difference between groups (odds ratio [OR] 0.99; 95%

confidence interval [CI] 0.68, 1.45). The percentage of patients experiencing severe and

non-severe manifestations was also similar between groups, with no significant difference,

and the incidence of individual ocular manifestations was comparable between groups.

73

TABLE 2: Incidence of ocular complications ofherpes zoster ophthalmicusin patients treated withvalaciclovir (1000 mgthree times daily) or oralaciclovir (800 mg fivetimes daily), and untreatedhistorical controls14

Valaciclovir (n=56) Aciclovir (n=54) Historical untreated controls Complication Incidence (%) 95% CI Incidence (%) 95% CI Incidence (%)

Conjunctivitis 30 (54) 40–67 28 (52) 38–66 86

Superficial keratitisPunctate keratitis 22 (39) 26–53 26 (48) 34–62 51 Dendritic keratitis 6 (11) 4–22 6 (11) 4–23 51–60

Dendritic ulcer 3 (5) 1–15 1 (2) 0–10 –

Stromal keratitis 7 (13) 5–25 7 (13) 5–24 41–56

Uveitis 7 (13) 5–24 9 (17) 8–29 34–60

Elevated intraocular pressure 8 (14) 6–26 7 (13) 5–25 –

Episcleritis 4 (7) 2–17 1 (2) 0–10 40

Thus, antiviral treatment of herpes zoster ophthalmicus is effective in reducing the

incidence and severity of ocular complications, with valaciclovir and famciclovir being as

effective as aciclovir but having simpler dosing schedules.

SteroidsAs discussed in Chapter 6, corticosteroids reduce the acute pain associated with herpes

zoster, but there is no proven benefit upon the development of post-herpetic neuralgia

(PHN) or the ocular complications of herpes zoster ophthalmicus.

The unanswered questions over the utility of steroids in the treatment of herpes zoster

ophthalmicus, and the severity of the complications that might warrant such therapy, lead

to the recommendation that corticosteroids, either systemic or topical, should not be

prescribed to patients with herpes zoster ophthalmicus without prior consultation with an

ophthalmologist, to ensure that patients are receiving the most appropriate therapy to help

prevent ocular complications, the most common of which is cataracts.

Sympathetic nerve blocksA small, randomized trial in patients with herpes zoster ophthalmicus demonstrated that

sympathetic nerve blocks given within 14 days of rash onset reduced the severity of acute

pain.17 Patients were administered a stellate ganglion block (bolus dose [9.5 ml]

comprising equal parts 1% lignocaine and 0.5% marcaine injected onto the C5/C6

prevertebral fascia) or were untreated. The treated patients reported significant reductions

in pain at 1 hour (P<0.001), 2 weeks to 1 month (P<0.01), 6 weeks to 2 months (P<0.05)

and 3 months (P<0.01). No significant reduction in the pain experienced by the untreated

control patients was observed until 3 months; the authors considered this reduction to be

due to resolution of pain over time in herpes zoster ophthalmicus.17 While sympathetic

nerve blockade may be considered for patients with severe acute pain, close liaison with

local pain clinics would be necessary to ensure that treatment is commenced within

14 days of rash onset, and not all pain clinics would accept that the value of this treatment

is proven.

Page 75: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Acute Retinal NecrosisVZV-associated acute retinal necrosis (ARN) has been described in both

immunocompetent and immunocompromised patients. Since the advent of AIDS, a more

aggressive variant of this disease (sometimes termed rapidly progressive herpetic retinal

necrosis [RPHRN]) has been identified in HIV-infected individuals, thought to be due to

their reduced level of cell-mediated immunity.18,19 Visual changes associated with ARN

usually occur weeks to months after the onset of herpes zoster. ARN can follow either

herpes zoster ophthalmicus or herpes zoster in a remote dermatome. Furthermore, retinal

involvement is bilateral in over half of cases, suggesting that VZV reaches the CNS via

haematogenous spread, possibly with extension along nerve pathways within the anterior

visual system.18

ARN presents with multifocal necrotizing lesions, often initially involving the peripheral

retina. The granular, non-haemorrhagic lesions rapidly extend and coalesce, accompanied

by relatively little intraocular inflammation.20-22 The typical signs and symptoms of

ARN are listed in the text box (right).

Recurrences of ARN are rare.23

Diagnosis of ARN is generally clinical.

Although PCR analysis of vitreous biopsy

samples can be performed to confirm

presence of VZV,24,25 the time taken for

biopsy results to be obtained may be

unacceptable.

Treatment of ARNJ Intravenous aciclovir (10 mg/kg

every 8 hours, 500 mg/m2 every

8 hours for children) followed by the oral formulation is the treatment of choice for

acute retinal necrosis caused by VZV (category 2 recommendation). Valaciclovir

(1000 mg three times daily) and famciclovir (500 mg three times daily) should also be

considered for use in this situation.

Antiviral treatmentThere have been no randomized trials of therapy in VZV-associated ARN. However,

intravenous aciclovir (10 mg/kg every 8 hours) for 7–10 days followed by 1–2 months of

oral aciclovir (400–800 mg five times a day) speeds the resolution of retinal lesions and

decreases the risk of disease in the other eye.23 Similarly, a clinical case report has

documented effective treatment of ARN with famciclovir (500 mg three times daily for

3 months) in an immunocompetent patient.26 Despite antiviral treatment, retinal

detachment occurs in most severe cases.

SummaryHerpes zoster ophthalmicus is observed in 1 in 5 patients with herpes zoster and up to

90% of these may experience ocular complications if left untreated. These complications

affect almost all of the structures of the eye and include conjunctivitis, scleritis, ocular

motor palsies, epithelial keratitis, stromal infiltrates, anterior uveitis and ARN.74

Signs and symptoms of acute retinal necrosis

• Pain

• Large white plaques in retinal periphery

• Photophobia

• Prominent vitritis

• Iritis

• Vascular sheathing

• Vitreous precipitates

• Necrotizing spreading retinal lesions

• Anterior uveitis

• 80% of cases become bilateral in 5–20 days

Page 76: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

The use of antiviral therapies in the management of herpes zoster ophthalmicus to limit

VZV replication has been shown to have benefit in preventing the ocular complications of

herpes zoster ophthalmicus. Valaciclovir (1000 mg three times daily) and famciclovir

(500 mg three times daily) are as effective as aciclovir in preventing the ocular

complications of herpes zoster ophthalmicus, but their simpler dosing schedules make

them more suitable for use.

The efficacy of steroids in herpes zoster ophthalmicus remains in doubt. It is not, therefore,

recommended that corticosteroids, either systemic or topical, are prescribed in herpes

zoster ophthalmicus without prior consultation with an ophthalmologist.

Sympathetic nerve blocks may play a role in limiting the pain associated with herpes

zoster ophthalmicus; however, the evidence for this is inconclusive.

Patients presenting with VZV-associated ARN should be treated with intravenous aciclovir

(10 mg/kg every 8 hours); however, anecdotal evidence suggests that valaciclovir

(1000 mg three times daily) and famciclovir (500 mg three times daily) may be beneficial

in these patients.

References1. Ragozzino MW, Melton LJ, 3rd, Kurland LT et al. Population-based study of herpes zoster and its sequelae.

Medicine (Baltimore) 1982;61:310-316.2. Burgoon CFJ, Burgoon JS, Bladridge GD. The natural history of herpes zoster. JAMA 1957;164:265-269.3. Mahalingam R, Wellish M, Lederer D et al. Quantitation of latent varicella-zoster virus DNA in human

trigeminal ganglia by polymerase chain reaction. J Virol 1993;67:2381-2384.4. Pavan-Langston D. Ophthalmic zoster. In: Varicella-zoster virus: virology and clinical management. (Arvin A,

Gershon A, eds). Cambridge: Cambridge University Press, 2000.5. Kestelyn P, Stevens AM, Bakkers E et al. Severe herpes zoster ophthalmicus in young African adults: a marker

for HTLV-III seropositivity. Br J Ophthalmol 1987;71:806-809.6. Marsh RJ, Cooper M. Ophthalmic herpes zoster. Eye 1993;7:350-370.7. Womack LW, Liesegang TJ. Complications of herpes zoster ophthalmicus. Arch Ophthalmol 1983;101:

42-45.8. Harding SP, Lipton JR, Wells JC. Natural history of herpes zoster ophthalmicus: predictors of postherpetic

neuralgia and ocular involvement. Br J Ophthalmol 1987;71:353-358.9. Liesegang TJ. Varicella-zoster virus eye disease. Cornea 1999;18:511-531.10. Liesegang TJ. Diagnosis and therapy of herpes zoster ophthalmicus. Ophthalmology 1991;98:1216-1229.11. Linnemann CC, Jr, Alvira MM. Pathogenesis of varicella-zoster angiitis in the CNS. Arch Neurol

1980;37:239-240.12. Cobo LM, Foulks GN, Liesegang T et al. Oral acyclovir in the treatment of acute herpes zoster ophthalmicus.

Ophthalmology 1986;93:763-770.13. Harding SP, Porter SM. Oral acyclovir in herpes zoster ophthalmicus. Curr Eye Res 1991;10:177-182.14. Colin J, Prisant O, Cochener B et al. Comparison of the efficacy and safety of valaciclovir and acyclovir for

the treatment of herpes zoster ophthalmicus. Ophthalmology 2000;107:1507-1511.15. Beutner KR, Friedman DJ, Forszpaniak C et al. Valaciclovir compared with acyclovir for improved therapy

for herpes zoster in immunocompetent adults. Antimicrob Agents Chemother 1995;39:1546-1553.16. Tyring SK. Famciclovir for ophthalmic zoster: A randomized acyclovir-controlled study. Br J Ophthalmol

2001;85:576-581.17. Harding SP, Lipton JR, Wells JC et al. Relief of acute pain in herpes zoster ophthalmicus by stellate ganglion

block. BMJ (Clin Res Ed) 1986;292:1428.18. Ormerod LD, Larkin JA, Margo CA et al. Rapidly progressive herpetic retinal necrosis: a blinding disease

characteristic of advanced AIDS. Clin Infect Dis 1998;26:34-45; discussion 46-37.19. Hellinger WC, Bolling JP, Smith TF et al. Varicella-zoster virus retinitis in a patient with AIDS-related

complex: case report and brief review of the acute retinal necrosis syndrome. Clin Infect Dis 1993;16:208-212.

20. Garthy DS, Spalton DJ, Hykin PG. Acute retinal necrosis syndrome. Br J Ophthalmol 1991May;75(5):292-297.

21. Margolis TP, Lowder CY, Holland GN et al. Varicella-zoster virus retinitis in patients with the acquiredimmunodeficiency syndrome. Am J Ophthalmol 1991;112:119-131.

22. Engstrom RE, Jr., Holland GN, Margolis TP et al. The progressive outer retinal necrosis syndrome. A variantof necrotizing herpetic retinopathy in patients with AIDS. Ophthalmology 1994;101:1488-1502.

23. Blumenkranz MS, Culbertson WW, Clarkson JG et al. Treatment of the acute retinal necrosis syndrome withintravenous acyclovir. Ophthalmology 1986;93:296-300.

24. Short GA, Margolis TP, Kuppermann BD et al. A polymerase chain reaction-based assay for diagnosing75

Page 77: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

varicella-zoster virus retinitis in patients with acquired immunodeficiency syndrome. Am J Ophthalmol1997;123:157-164.

25. Yamamoto S, Pavan-Langston D, Kinoshita S et al. Detecting herpesvirus DNA in uveitis using thepolymerase chain reaction. Br J Ophthalmol 1996;80:465-468.

26. Figueroa MS, Garabito I, Gutierrez C et al. Famciclovir for the treatment of acute retinal necrosis (ARN)syndrome. Am J Ophthalmol 1997;123:255-257.

76

Page 78: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Management RecommendationsCategory 1J Intravenous aciclovir therapy is the standard of care for immunocompromised patients

with disseminated varicella zoster virus (VZV) disease, including those with

complications such as varicella pneumonia.

The recommended doses are:

Immunocompromised adults with VZV infections: 10 mg/kg every 8 hours

UK: Immunocompromised children with VZV infections: 500 mg/m2 body surface area

every 8 hours. The dosage in children under 3 months of age is calculated on the basis

of body weight.

USA: Immunocompromised children with VZV infections: 20 mg/kg every 8 hours.

J Immunocompromised individuals should receive prophylaxis with varicella zoster

immune globulin (VZIG) as soon as possible but no later than 4 days following

exposure to varicella.

Category 2J In immunocompromised patients with suspected aciclovir-resistant VZV infection,

treatment with foscarnet (120–200 mg/kg/day, in two or three divided doses) should be

initiated unless renal failure is evident.

J It is recommended that suppressive antiviral therapy targeted at VZV should be

considered for transplant patients, especially in bone marrow transplant (BMT)

recipients and patients requiring immunosuppression for graft-versus-host disease

(GVHD). The recommended regimen is intravenous aciclovir (500 mg/m2 three times

a day) for 1 month followed by oral aciclovir (800 mg four times a day) for a further

6 months.

J It is recommended that the use of prophylactic varicella vaccination be considered in

selected groups of immunocompromised persons, such as those with leukaemia that

is in remission or who are undergoing haematopoietic stem cell transplantation.

However, varicella vaccine is not recommended in these patients for post-exposure

prophylaxis because of the risk of vaccine-induced varicella.

Category 3J Anecdotal evidence suggests that oral antiviral therapy may be appropriate for the

treatment of varicella in some immunocompromised individuals. In this setting, the

newer antivirals, valaciclovir (1000 mg three times daily for adults) and famciclovir

(500 mg three times daily for adults), are likely to be at least as effective as aciclovir

(800 mg four times daily). The minimum duration of therapy should be 7 days.77

MANAGEMENT OF VARICELLA ZOSTER VIRUS

INFECTIONS IN THE IMMUNOCOMPROMISED HOST8

Page 79: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

J For some immunocompromised individuals, oral antiviral therapy may be appropriate

for the treatment of herpes zoster – in this setting, the newer antivirals, valaciclovir

(1000 mg three times daily for adults) and famciclovir (500 mg three times daily for

adults), are likely to be at least as effective as aciclovir (800 mg five times daily).

J Clinicians should be aware that in immunocompromised individuals, herpes zoster

might initially present as unexplained abdominal pain.

J Combined intravenous foscarnet (60 mg/kg three times per week) plus intravitreal

ganciclovir (400 mg twice per week) therapy is the best option for rapidly progressive

herpetic retinal necrosis. However, the dose of foscarnet to be used has not been

established in the clinical trial setting, and further dose-comparative studies should be

undertaken.

J It is recommended that VZV isolates obtained from hyperkeratotic papules and

ecthymatous lesions from HIV-positive individuals be submitted so that antiviral

susceptibility testing may be undertaken if clinically necessary.

Research needJ Studies are recommended comparing oral valaciclovir and famciclovir with

intravenous aciclovir for the management of immunocompromised individuals with

VZV infections.

J There is a need to define the patients for whom oral antiviral therapy might be used in

place of intravenous therapy.

IntroductionIn individuals who have defective or compromised cell-mediated immunity, VZV may

cause extensive cutaneous disease and invade visceral organs, especially the lungs, liver,

central nervous system (CNS) and bone marrow. The complications that may arise in

immunocompromised individuals can be severe and sometimes life-threatening, e.g.

pneumonitis and encephalitis.

Immunocompromised individuals at greater risk of developing severe varicella include

patients with lymphoproliferative malignancies (especially T-cell dysplasias), individuals

who have received either a BMT or solid organ transplant within the previous 12 months,

HIV-positive individuals with a significantly reduced CD4 cell count (<200 CD4

cells/mm3) and those receiving high doses of corticosteroids or cytotoxic chemotherapy.

Immunocompromised individuals who may be at lower risk of severe varicella include

those receiving short-term corticosteroid therapy, patients receiving long-term aspirin

therapy and those with chronic cutaneous, cardiac or pulmonary disease.

The potential severity of varicella and its attendant complications in the

immunocompromised patient emphasize the need for effective prevention and treatment

of disease, particularly in these populations. This chapter considers post-exposure

prophylaxis and treatment of varicella in this vulnerable patient group. Vaccination of

immunocompromised individuals is reviewed in Chapter 3.78

Page 80: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

HIV-infected children who develop varicella are at increased risk of developing herpes

zoster soon after the initial varicella infection. These patients may also develop recurrent

disease – defined as new episodes of disseminated skin lesions in the absence of exposure

occurring at least 1 month after a previous attack.1 Such patients have modest decreases

in CD4 cell count, whereas low CD4 cell counts are associated with progressive varicella,

in which lesions continue to appear for at least 1 month.1

While many cases of herpes zoster in individuals with HIV/AIDS are similar to disease in

the otherwise healthy host, unusual presentations of herpes zoster may occur. Infections in

the immunocompromised host may have atypical features – large rash area, long-term

persistence, or recurrent outbreaks.2 Pain in these individuals may also be complicated by

AIDS neuropathy. Furthermore, the growing use of chemotherapy may result in an increase

in the reactivation of VZV, which is more likely to be severe or complicated than in the

otherwise healthy individual if untreated. The widespread use of potent antiretroviral

therapy has prevented the incidence of herpes zoster in individuals with HIV increasing

compared with that in the general population.

Other immunocompromised populations at greater risk of herpes zoster include those

receiving immunosuppressant medication, steroids or chemotherapy (transplant recipients,

patients with malignancies and patients with immune-mediated diseases such as

rheumatoid arthritis and systemic lupus erythematosus [SLE]). These groups of patients are

most likely to benefit from antiviral treatment.

Epidemiology of VZV Infections in the Immunocompromised VaricellaHIV-infected individualsThe prevalence and clinical presentation of varicella in children with HIV infection in

developed countries is similar to that in immunocompetent individuals, with respect to

likelihood of infection and proportion of severe cases and complications.3 In the European

Collaborative Study, approximately 130 HIV-infected children and 750 uninfected

children born to HIV-positive mothers were followed. There were no reports of severe

varicella in any of the children who were infected during the study period.4,5 However, in

children with AIDS, the proportion of severe cases and complications is greater.2

Since 95% of adults in developed countries are VZV-seropositive, most HIV-positive adults

will already have had varicella earlier in life. However, for those who are VZV-seronegative

and HIV-positive, the consequences of a primary VZV infection in adulthood are likely to

be serious. This situation could occur more frequently in rural areas of tropical countries,

which may have a larger susceptible adult population. However, there is a lack of data on

the prevalence of varicella in HIV-infected adults in those parts of the world.

Prevalence of infection in BMT recipientsBMT recipients have been reported as being at high risk of developing varicella

irrespective of the VZV serostatus of either the recipient or the donor.6-8 The prevention of

varicella in this population is discussed later in this chapter.

Herpes zoster Herpes zoster is more common in immunocompromised than in immunocompetent

individuals. Studies of HIV-positive individuals conducted in the late 1980s and early

79

Page 81: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

1990s document that the incidence of herpes zoster varied from 2900–5100 per 100 000

patient-years.9-12 This compares with 130–480 new cases per 100 000 people per year in

the overall UK and USA populations.13-15 Thus the incidence is 15–25 times greater than

the rate in the general population and 3–7 times greater than that in the elderly.9 The

cumulative incidence of herpes zoster in HIV-infected individuals is estimated to be

30–40% over approximately 10 years of follow-up.9 Patients who present with herpes

zoster in areas of high HIV prevalence may have underlying HIV infection; therefore,

zoster may suggest HIV infection in susceptible individuals.16,17

Patients with malignancies are also an important population at increased risk of herpes zoster.

The strongest evidence of this comes from a study conducted from 1972–1980 in a cancer

care centre in Ontario, Canada. Patients were followed up for a minimum of

5 years.18 There were over 7000 new cancer cases per year, from which 811 cases of herpes

zoster were identified in 785 individuals with a median age of 57 years. The cumulative

incidence rate in this population 5 years after diagnosis was 625 per 100 000 patient-years,

some five times higher than in the general population. The study also found that risk for herpes

zoster is greater for patients with haematological malignancies than for those with solid

tumours, and is greatest for Hodgkin’s disease compared with all other malignancies.19,20

Bone marrow and solid organ transplant recipients are likely to be at increased risk of

herpes zoster but data on its incidence often come from small studies or retrospective

reviews. Despite this, it appears that BMT recipients are particularly at risk, with the

cumulative frequency of herpes zoster reported to range from 13% to 50% in the

28 months after BMT.8 The majority of cases occur within 12 months of transplantation,

with the highest incidence between 3 and 6 months after transplantation.21 Two

randomized, controlled studies have compared 6 months of aciclovir prophylaxis with

placebo.22,23 Aciclovir was effective in reducing the risk of herpes zoster in both studies

during the 6-month treatment period, but there were no differences between the groups

12 months post-transplant. This suggests that a long duration of antiviral prophylaxis is

needed to prevent herpes zoster in these patients.

The data on solid organ transplants are limited to surveys conducted in the 1960s and

1970s, in which different immunosuppressive regimens were employed. These studies

report that the frequency of herpes zoster was 7–14% in renal transplant recipients within

2–5 years of transplantation, and 22% after 6 years of follow-up in cardiac transplant

patients.9 Valaciclovir has not been studied for VZV prophylaxis, but when valaciclovir was

given for cytomegalovirus (CMV) prophylaxis in renal transplant recipients,24 no cases of

VZV were reported in the treatment arm.

Presentation and Complications of VZV Infections in theImmunocompromisedVaricellaThe reduced cellular immunity observed in the immunocompromised host results in

enhanced susceptibility to severe varicella and its associated complications compared

with immunocompetent individuals. Those more likely to be at high risk of developing

varicella and associated complications include:

• Individuals with AIDS

• Patients with malignancies

• BMT recipients80

Page 82: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

• Solid organ transplant recipients

• Patients receiving high-dose corticosteroids

• Children with leukaemia.

Pneumonitis is the major complication of varicella among immunocompromised hosts

and is associated with appreciable mortality. The risk of it developing is inversely related

to lymphocyte count, and children with leukaemia appear to be particularly susceptible.25

Among these patients, varicella pneumonitis has an incidence of 32% and a mortality rate

of 31% in the absence of treatment.25

HIV-infected individualsThere are no published population-based studies, but a few case studies of varicella in

HIV-infected children have been reported.26-30 Complications and severity of varicella are

probably related to CD4 cell count; varicella-associated pneumonia and bacterial skin

infections are more severe in children with reduced CD4 cell counts (<200 CD4

lymphocytes per mm3 at presentation).27,28 In a study involving nine HIV-infected children,

aged between 9 months and 8 years, eight of the patients developed visceral dissemination

involving the lungs, two had liver involvement and one patient had brain and pancreas

involvement in addition to lungs and liver.27 During the initial episode of herpes zoster

infection, five were treated with intravenous aciclovir, three with oral aciclovir and one

patient did not receive aciclovir. Four of the nine patients developed secondary bacterial

infections, four developed chronic or recurrent varicella and one patient died following

progressive pneumonia. Despite this patient having received intravenous aciclovir, autopsy

findings also showed disseminated varicella zoster infection involving lung, liver, brain

and pancreas.27

When varicella occurs in HIV-infected adults and those with AIDS, it can result in

significant morbidity, including encephalitis and hepatitis.31 In a study of five HIV-positive

men who contracted varicella, two had complications characterized by severe headache

and meningismus, and one of these patients also exhibited hepatitis and

thrombocytopenia.31 All five responded to aciclovir therapy.

Although uncommon, the potentially serious nature of varicella and associated

complications in immunocompromised patients demonstrates the importance of

effectively treating those cases that do occur.

Herpes zoster Immunocompromised patients (e.g. BMT recipients) have an increased probability of

cutaneous and visceral dissemination of VZV compared with immunocompetent

individuals.32 Commonly reported complications in the immunocompromised include

VZV pneumonia, encephalitis, and hepatitis. The increased risk of complications in this

group is illustrated by observations of untreated herpes zoster. A study in 76 adults with

herpes zoster who had either lymphoma, a lymphoproliferative malignancy, or who had

undergone BMT or solid organ transplant, demonstrated that cutaneous dissemination

occurred in 32%, visceral dissemination in 9.5%, and two of these individuals died.33,34

In most instances, the clinical features of herpes zoster in HIV-positive individuals are

similar to those seen in the immunocompetent. However, a unique feature of herpes zoster

in those who are HIV-positive is the higher frequency of recurrence.10 In studies, 20–30%

81

Page 83: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

of HIV-infected patients developed one or more subsequent episodes of herpes zoster,

which may have involved the same or different dermatomes.10,12 It is not clear whether these

patients were receiving aciclovir therapy. The probability of a recurrence within 1 year of

the index case is estimated to be 12% in these patients;35 this compares with a rate of second

recurrences in immunocompetent individuals of less than 5%.1 However, recent data from

an analysis of 1071 elderly subjects in the UK suggest a higher figure, of around 14%.36

Although studies have shown a variety of delayed-onset neurological complications,37

visceral dissemination of VZV as reported in other immunocompromised populations is

uncommon in HIV-infected individuals.

Evidence from some studies which show an apparent link between immunosuppression

and a higher incidence of post-herpetic neuralgia (PHN) is insubstantial. Immuno-

compromised and immunosuppressed individuals may be more at risk from other

complications (i.e. dissemination) than immunocompetent individuals.

Atypical generalized zosterAn unusual presentation of herpes zoster in the immunocompromised host is atypical

generalized zoster. Sufferers present with diffuse varicella-like skin lesions with limited

primary dermatomal involvement.38 Patients presenting with this are VZV seropositive

prior to rash development,38 thus the syndrome is due to reactivation of VZV, not primary

varicella. Herpes zoster begins with a limited area of involvement in the primary

dermatome, quickly followed by generalized cutaneous dissemination. Risk factors for

development of atypical generalized zoster appear to be the same as those for classic

herpes zoster, i.e. increasing age and reduced cell-mediated immunity.

Abdominal herpes zosterJ Clinicians should be aware that in some immunocompromised individuals, herpes zoster

might initially present as unexplained abdominal pain (category 3 recommendation).

A serious manifestation of herpes zoster in the immunocompromised individual is

abdominal herpes zoster. Patients present with severe, unexplained abdominal pain that

may precede the appearance of the cutaneous rash by hours or days.39,40 The diagnosis of

herpes zoster is usually not considered until the typical skin vesicles begin to appear,

usually in a thoracic dermatome. Abdominal herpes zoster is associated with a high

mortality rate, even when appropriate antiviral therapy is administered.

Autopsy studies have revealed a high frequency of abdominal visceral involvement in

patients with abdominal herpes zoster. Polymerase chain reaction (PCR) detection of VZV

in peripheral blood mononuclear cells and whole blood has been used in a few cases to

make the diagnosis of visceral VZV before the appearance of the rash.40

Chronic VZV encephalitisChronic VZV encephalitis is seen almost exclusively in patients with conditions involving

depressed cellular immune responses. In one study, two of 47 BMT patients with herpes

zoster developed encephalitis.41 Encephalitis may occur months after an episode of herpes

zoster, making a diagnosis problematic.42 The clinical presentation comprises headache,

fever, altered mental status, seizures, and focal neurological defects (including aphasia,

hemiplegia, and reduced visual field).43-45

82

Page 84: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Pathology studies reveal multifocal lesions in the white matter of the brain, near the grey-

white junction, with small-vessel vasculitis and nerve demyelination being observed.46,47

Magnetic resonance imaging (MRI) studies demonstrate plaque-like lesions in the white

matter, consistent with demyelination, and late development of ischaemic or

haemorrhagic infarcts of cortical and subcortical grey and white matter (Figure 1).48,49

Examination of CSF reveals mild mononuclear pleocytosis. VZV DNA has been amplified

from the CSF of encephalitis patients by PCR.50,51

The clinical course of chronic VZV encephalitis is

often deterioration and death in up to half of cases,41

although anecdotal reports have suggested benefit

from therapy with high-dose intravenous aciclovir.

Atypical cutaneous lesions J It is recommended that VZV isolates obtained

from hyperkeratotic papules and ecthymatous

lesions from HIV-positive individuals be

submitted for antiviral susceptibility testing

(category 3 recommendation).

VZV can cause a variety of atypical cutaneous lesions

in individuals with HIV infection and low CD4 cell

counts. One example is multiple hyperkeratotic

papules, measuring 3–20 mm in diameter, which follow no dermatomal pattern (Figure

2).52,53 These lesions may be chronic, persisting for months or years, and are sometimes

associated with aciclovir-resistant strains of VZV.54 A second type of dermatological

manifestation observed in HIV-positive individuals is ecthymatous VZV lesions (Figure 3).

Patients present with multiple, large (10–30 mm), punched-out ulcerations with a central

black eschar and a peripheral rim of vesicles.55,56

These atypical cutaneous lesions may be caused by antiviral-resistant

strains of VZV, and, thus, isolates obtained from these papules or lesions

should routinely be submitted for antiviral susceptibility testing.

Pre-Exposure Vaccination in the Immunocompromised J It is recommended that the use of prophylactic varicella

vaccination be considered in selected groups of

immunocompromised persons, such as those with leukaemia that

is in remission or who are undergoing haematopoietic stem cell

transplantation. However, varicella vaccine is not recommended

in these patients for post-exposure prophylaxis because of the risk

of vaccine-induced varicella (category 2 recommendation).

Clinical trials of the varicella vaccine were initially performed in

immunocompromised children because of the likely positive risk:benefit ratio. The vaccine

had been shown to be safe in trials in Japanese children and it was recognized that

protection against varicella would be of significant benefit to immunocompromised

children.57 A number of trials have documented seroconversion and protection following

varicella vaccination.22-2983

FIGURE 2:Hyperkeratotic papules of VZV lesions in anHIV-positive individual

FIGURE 1: MRI of thebrain or an HIV-positiveindividual with VZVencephalitis. Multifocalareas of infarction areshown: the long solidarrow shows a superficialwedge-shaped lesion.Open arrows indicatedeep ovoid lesions in thewhite matter. Short solidarrows show smallerlesions at junctions ofgrey and white matter

© 2

002.

Rep

rodu

ced

with

per

mis

sion

, Gild

en D

H, K

lein

schm

idt-

DeM

aste

rs B

K, L

aGua

rdia

JJet

al.

Neu

rolo

gic

com

plic

atio

ns o

fth

e re

activ

atio

n of

var

icel

la-z

oste

r vi

rus.

N E

ngl J

Med

2000

;342

(9):6

35-6

45. M

assa

chus

etts

Med

ical

Soc

iety

Rep

rodu

ced

with

per

mis

sion

, Pro

fess

or Jo

hn G

nann

.

Page 85: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Leukaemic childrenVaricella vaccination has been shown to be safe and effective in selected groups of

immunocompromised children (including those with acute leukaemia, lymphoma and

malignancies). In studies conducted in Japan

between 1975 and 1983, a total of 330 children

with leukaemia were vaccinated with one dose of

vaccine.58-61 Two hundred and fifty-one children

were vaccinated during temporary suspension of

chemotherapy (i.e. for 1 week before and after

vaccination). Of these children, 18% had a mild,

varicella-associated rash and seroconversion

occurred in 91%. Of the remaining 79 children who

were immunized without suspension of

chemotherapy, 47% had a rash and seroconversion

occurred in 96%. Similar trials in Europe produced

comparable results.62-65 The vaccine-associated rash

observed in these trials is likely due to these patients’ reduced cell-mediated immunity.

A large study of vaccination of leukaemic children has been conducted in the USA and

Canada.66 Five hundred and seventy-five children with leukaemia were vaccinated (Figure 4).

Most received the VZVoka vaccine produced by Merck & Co Inc, although a few received

SmithKline Beecham’s (now GlaxoSmithKline) VZVoka-RIT vaccine. Chemotherapy was

suspended for 1 week before and 1 week after vaccination in 511 of the children; a further

64 had already completed therapy at the time of vaccination. To be eligible for the study,

the children had to be in remission for at least 1 year and have an absolute lymphocyte

count of at least 700 cells/mm3. Initially, vaccinees received a single dose of vaccine, but

18% of them failed to seroconvert. The schedule was subsequently changed to give two

doses of vaccine, 3 months apart, with chemotherapy suspended only for the first dose.

Following two doses, the seroconversion rate was 95%. Vaccine-associated rash occurred

2–6 weeks after vaccination; the incidence of this was lower in those no longer receiving

chemotherapy (5%) than in children whose chemotherapy had been suspended (50%).

Only 10% of vaccinees developed rash after the second dose. Although the frequency of

the rash was high, it was much less severe than natural varicella.67 To minimize the severity

of the rash, vaccinees with 50 lesions or more were offered high-dose oral aciclovir

(800 mg five times daily). The rate and severity of vaccine-associated rash were higher in the

US studies than in the Japanese. This may be attributable to the more immunosuppressive

chemotherapy regimens used for leukaemia treatment in the USA and Canada.66 A similar

explanation may account for the need for two vaccine doses in the US and Canadian study.

In addition to documenting seroconversion, the study also provided evidence for vaccine

efficacy. Of 123 leukaemic vaccinees who had a household exposure to varicella, 86%

were completely protected and 14% had mild (median 100 lesions) breakthrough disease.

Over an 11-year follow-up period, 87% of vaccinated individuals remained seropositive

and there was no evidence that the incidence or severity of breakthrough varicella

increased with time.68

84

FIGURE 3:Echthymatous VZVlesions in an HIV-positiveindividual54

© R

epro

duce

d w

ith p

erm

issi

on, J

acob

son

MA

, Ber

ger T

G, F

ikrig

Set

al.A

cycl

ovir-

resi

stan

t var

icel

la z

oste

r vi

rus

infe

ctio

n af

ter

chro

nic

oral

acyl

ovir

ther

apy

in p

atie

nts

with

the

acqu

ired

imm

unod

efic

ienc

ysy

ndro

me

(AID

S). A

nn In

tern

Med

1990

;112

:187

-191

.Am

eric

anC

olle

ge o

f Phy

sici

ans.

Page 86: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Children with solid organ transplantsVaccination of children prior to or after solid organ transplantation results in

seroconversion in the majority of individuals. In a small study, 34 children (17 in renal

failure and 17 post-renal transplant) were immunized with one dose of the VZVoka-RIT

vaccine produced by SmithKline Beecham.69 The overall seroconversion rate was 85%, the

rate being equal in both groups. One child

developed a mild, vaccine-associated rash and three

developed mild varicella within 2–4 years. In 68

children with renal or hepatic failure, vaccination

with one dose of the SmithKline Beecham VZVoka-RIT

vaccine resulted in a 50% seroconversion rate and a

second dose gave an 85% seroconversion rate.70

There were no cases of varicella during the 6-year

follow-up period.70

Children with BMTVaccination also results in seroconversion after BMT. Seroconversion was seen in eight of

nine susceptible children immunized 12–23 months after they received a BMT.71

Seroconversion in immunocompromised versus immunocompetent childrenSeroconversion rates in immunocompromised children vaccinated with the varicella

vaccine are similar to the rate observed in immunocompetent children. In a study

conducted in Mexico, 67 immunocompromised and 33 immunocompetent children with

no clinical history of varicella or herpes zoster were vaccinated with live attenuated

varicella vaccine.72 The immunocompromised group comprised children with leukaemia,

solid tumours, chronic renal failure and cirrhosis. Serum IgG antibodies against VZV were

measured by ELISA at baseline, 3-months post-vaccination and 6-months post-

vaccination. Thirty-six (53.7%) of the immunocompromised and 22 (66%) of the

immunocompetent children had positive VZV-ELISA

results at baseline. Among VZV-seronegative

children, seroconversion at 6-months post-

vaccination was demonstrated in 90.3% of the

immunocompromised and 100% of the

immunocompetent children (Figure 5). Three of the

immunocompromised children had a mild rash

symptomatic of varicella following vaccination.72

Vaccination in immunocompromised adultsWhile the potential benefit of varicella vaccination

has been demonstrated in selected groups of

immunocompromised children, there is little data on the efficacy and safety of varicella

vaccine in immunocompromised adults.

One randomized study has indicated that the risk of developing zoster is significantly

reduced when inactivated varicella vaccine is administered prior to haematopoietic stem

cell transplantation and for the first 90 days thereafter.73 In this study, patients received the

vaccine 30 days before transplantation, and again at Days 30, 60 and 90 after

transplantation. Zoster subsequently developed in 7 of 53 vaccinated and 19 of 58

unvaccinated patients (13% versus 33%; P=0.001). The mean stimulation index (defined

85

FIGURE 4: Vaccinationof leukaemic children66

Patients

Single dose of vaccine

Vaccine-associated rash

Second dose of vaccine

Vaccine-associated rash

Chemotherapy suspended (n=511)Chemotherapy completed (n=64)

18% did not seroconvert

50% in those whose chemotherapy had been suspended

Seroconversion in 95%

Incidence of rash 10%

5% in those no longer receiving chemotherapy

FIGURE 5: Antibodytitres measured by ELISAat baseline, 3 months and6 months after varicellavaccination in healthy andimmunocompromisedchildren who were VZVseronegative at baseline72

43.5

32.5

2

1.51

0.5

00 3

ELIS

A u

nits

6

HealthyImmunocompromised

Months

© 2

002

Rep

rodu

ced

with

per

mis

sion

from

Els

evie

r Sc

ienc

eM

oral

es-C

astil

lo M

E, A

lvar

ex-M

unoz

MT,

Sol

orza

no-S

anto

s F

etal

.Liv

e va

ricel

la v

acci

ne in

bot

h im

mun

ocom

prom

ised

and

heal

thy

child

ren.

Arc

h M

ed R

es20

00;3

1:85

-87.

Page 87: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

as the in vitro CD4 T-cell proliferation response to VZV) was significantly higher in patients

who did, rather than did not, receive the vaccine at 90 days, 120 days and 12 months.

Increases in the stimulation index above 1.6 correlated with a reduced risk of zoster.73

Post-Exposure Prophylaxis in the ImmunocompromisedJ Immunocompromised individuals should receive prophylaxis with VZIG as soon as

possible but no later than 4 days following exposure to varicella (category 1

recommendation).

J Post-exposure prophylaxis with varicella vaccine is not suitable for immuno-

compromised individuals because of the risk of vaccine-induced varicella (category 2

recommendation).

J Extended antiviral suppression for VZV infections should be considered in transplant

patients, especially BMT recipients and patients requiring immunosuppression for

GVHD. The recommended regimen is intravenous aciclovir (500 mg/m2 three times a

day) for 1 month followed by oral aciclovir (800 mg four times a day) for a further

6 months (category 2 recommendation).

Transmission of VZV to susceptible individuals is difficult to prevent as those infected are

contagious 24–48 hours prior to clinical signs of disease. Thus, it is necessary to undertake

post-exposure prophylaxis in immunocompromised individuals who are at risk of severe

varicella and have had significant exposure to VZV.

VZIG post-exposure prophylaxis The only product licensed for post-exposure prophylaxis of varicella is VZIG, which

continues to be the mainstay of prophylaxis in immunocompromised individuals. Studies

performed in the 1960s and 1970s proved the efficacy of VZIG in modifying or preventing

VZV infections if administered soon after exposure (within 96 hours).

Although VZIG is widely used as post-exposure prophylaxis against varicella in

immunocompromised individuals, there is no evidence that VZIG is better than natural

immunity in these patients. If administered within 96 hours of exposure, VZIG reduces the

incidence of disease complications, lessens the severity of the disease and increases the

proportion of immunocompromised patients who experience subclinical varicella.74 In

some instances, VZIG appears to prolong the incubation period of varicella with the result

that a proportion of treated children develop varicella up to 28 days after its

administration. VZIG may be given to immunocompromised individuals who do not have

a clear history of varicella and are exposed to VZV, although VZIG has no antiviral effect

when given after the onset of rash.

The usual route of administration is intramuscular injection. In the UK, VZIG

administration is targeted only at those who are VZV-seronegative. Serum immune

globulin may also be administered intravenously (IVIG) to those with bleeding disorders.

Peak and trough VZV titres are comparable after administration of either VZIG or IVIG.75

If an individual receiving VZIG does not develop clinical varicella, subsequent VZV

exposures should also prompt VZIG prophylaxis unless active immunity is documented by

serological testing, as immunity derived from a subclinical infection is not known to be

86

Page 88: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

protective. Between 25% and 50% of all immunocompromised VZIG recipients may

remain susceptible to varicella after its administration.74

VZIG has been used successfully as post-exposure prophylaxis in a variety of different

immunocompromised populations

(mostly children), including renal and

liver transplant recipients and those

with leukaemia or solid tumours.74,76,77

In one study, VZIG was administered to

2412 immunocompromised children

(<16 years of age) with a range of

underlying illnesses (including

leukaemia, non-Hodgkin’s lymphoma

and Hodgkin’s disease) within 4 days of

exposure to VZV.74 Varicella subsequently

occurred in 561 children (454 clinical

varicella, 107 subclinical). However, in

these immunocompromised children, VZIG reduced the incidence of pneumonia,

encephalitis and death from varicella 10-fold, and was associated with milder disease and

a higher incidence of subclinical varicella compared with historical controls (Table 1).74,78,79

Immunocompromised individuals should receive post-exposure prophylaxis with VZIG as

soon as possible, but no later than 4 days following exposure to varicella. If varicella develops,

the patient should be treated with intravenous antiviral compounds at least initially.

In settings where VZIG is not available, other modalities of prophylaxis, such as antiviral

compounds, should be considered. Recent studies have suggested that post-exposure

prophylaxis using antiviral compounds may also be of use in immunocompromised

patients, particularly when used in conjunction with VZIG.2,80 Oral aciclovir was a safe

and effective adjunctive adjunct to VZIG for post-exposure prophylaxis following VZV

exposure in 12 children receiving corticosteroid treatment for chronic renal disease.80 In

this small study, eight patients (10 separate VZV exposures) received oral aciclovir

(40 mg/kg/day) in conjunction with VZIG, and four patients (six separate VZV exposures)

received VZIG alone. None of the eight patients who received aciclovir plus VZIG

developed varicella, and one developed humoral immunity to VZV despite absence of

clinical disease. In comparison, one of the patients who received VZIG alone developed

varicella. No adverse reactions were seen with aciclovir prophylaxis. These data suggest

that aciclovir co-administered with VZIG may be more protective than VZIG alone.

Aciclovir prophylaxis BMT recipients are likely to be at high risk of primary (varicella) and recurrent (herpes

zoster) VZV infection irrespective of the VZV serostatus of the patient or donor.6,7 In an

observational study, low-dose aciclovir and ganciclovir were shown to be effective at

delaying VZV infection following allogeneic BMT.

One hundred and fifty-one BMT recipients received low-dose aciclovir or ganciclovir for

prophylaxis against herpes simplex virus (HSV) or CMV disease. Patients who were HSV

seropositive received either oral aciclovir (400 mg three times daily) or intravenous

87

TABLE 1: Varicellafollowing VZIGadministration to exposedimmunocompromisedchildren versus historicalcontrols74,78,79

Disease category VZIG Historic control (no VZIG) n (% of evaluable)* Expected n (%)†

Clinical varicella 1–10 pox 56 (13) 0 (0) 11–50 pox 132 (31) 23 (5) 51–100 pox 59 (13) 41 (9) >100 pox 178 (42) 390 (86)

Unknown 29 (1.2) Subclinical varicella 107 (4.4) 14 (~3)

Total 561 (23) 468

Pneumonitis 13 (2.9)†† 113 (25)††

Encephalitis 2 (0.4)†† 15 (3.3)††

Death 3 (0.6)†† 32 (7)††

*Adapted from Levin et al.74 Total number receiving VZIG = 2412†Data from Feldman et al.78 for pox number and subclinical varicella, and from Ross79 for pneumonitis, encephalitisand death

††Per cent of varicella cases

Page 89: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

aciclovir (250 mg three times a day) up to the day of engraftment, followed by oral

aciclovir (200 mg three times a day) for at least 6 months following transplantation. CMV

seropositive patients received intravenous ganciclovir (5 mg/kg three times weekly) from

engraftment to Day 84.8 None of the patients developed VZV infection whilst receiving

either aciclovir or ganciclovir (P<0.0001 compared with historic controls). However,

when antiviral prophylaxis ended, 21 of 121 (17%)

patients developed VZV infection, five of which

occurred within 5 weeks of the end of therapy. The

cumulative incidence of VZV infection was 13% at

12 months, 32% at 24 months and 38% at 28

months following transplantation (Figure 6).8

The investigators concluded that 3–6 months, low-

dose aciclovir or ganciclovir was effective at

delaying VZV infection following BMT, even though

the therapies did not appear to affect the overall

incidence of VZV infection. This delay may allow

the patient time to develop sufficient immunity to

overcome varicella infection after cessation of

prophylaxis, which may reduce the incidence of

associated complications. Therefore, antiviral

suppression of varicella may be warranted in certain

high-risk patients, including those undergoing BMT.

More studies are needed to investigate whether

longer periods of prophylaxis than those analysed in

this study would further reduce or prevent

susceptibility to VZV reactivation.

Prophylaxis with aciclovir has been documented to

prevent herpes zoster in allogeneic BMT recipients in

two placebo-controlled trials. In the first trial,

intravenous aciclovir (250 mg/m2 three times per day)

starting 5 days before transplant and continued for

5 weeks, was followed by oral aciclovir (400 mg

three times daily) for 6 months.81 Herpes zoster

developed in 5 of 22 placebo recipients but there

were no cases in the 20 aciclovir recipients.

In the second study,82 in which aciclovir was given primarily for the prevention of CMV

disease, a variety of regimens were used:

• Intravenous aciclovir (500 mg/m2, three times a day for 1 month) followed by oral

aciclovir (800 mg four times a day for a further 6 months)

• Intravenous aciclovir (500 mg/m2, three times a day) followed by oral placebo

• Low-dose oral aciclovir (200 or 400 mg, four times a day) followed by placebo.

The probability of VZV disease by Day 210 of the trial was lower with the use of

intravenous aciclovir followed by oral aciclovir (3%) than with the other two regimens

(both 9%).82 While these results are encouraging, an earlier, placebo-controlled study did

not document an overall reduction in the incidence of herpes zoster in allogeneic BMT88

FIGURE 6: Cumulativeincidence of VZVinfection (a) following BMT and (b) followingcessation ofaciclovir/ganciclovirprophylaxis8

500.940

0.730

0.520

0.310

0.10Es

timat

ed p

erce

ntag

e w

ith V

ZV

0 2 3 76541

Years following cessation of aciclovir/ganciclovir

121 15 11 017825

Number at risk

500.940

0.730

0.520

0.310

0.10Es

timat

ed p

erce

ntag

e w

ith V

ZV

0 2 3 76541

Years following allogeneic BMT

151 43 27 03121678

Number at risk

b)

a)

© R

epro

duce

d w

ith p

erm

issi

on, S

teer

CB,

Sze

r J,

Sasa

deus

z Je

t al

.Var

icel

la-z

oste

r in

fect

ion

afte

r al

loge

neic

bon

e m

arro

w tr

ansp

lant

atio

n: in

cide

nce,

ris

k fa

ctor

s an

d pr

even

tion

with

low

-dos

e ac

iclo

vir

and

ganc

iclo

vir.

Bon

e M

arro

w T

rans

plan

t20

00;2

5:65

7-66

4. N

atur

e Pu

blis

hing

Gro

up.

Page 90: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

recipients given intravenous aciclovir (5 mg/kg three times daily) for 23 days followed by

oral aciclovir (800 mg four times daily) for 6 months.23 However, the balance of evidence

suggests that aciclovir prophylaxis can prevent herpes zoster in BMT recipients.

Similarly, data from a series of trials of aciclovir for the prevention of CMV infection and

disease in liver transplant recipients suggest that it has the potential to minimize the risk

of herpes zoster. In these trials, intravenous and oral aciclovir prevented VZV infection and

reactivation.8,83,84

One study has advocated treatment of HIV-positive individuals who develop recurrent or

progressive herpes zoster with intravenous aciclovir followed by lifelong oral

prophylaxis.85 However, potent antiretroviral therapy may preserve or restore immune

function so that continual prophylaxis of opportunistic infections, including herpes zoster,

will no longer be necessary.

An alternative approach for the prophylaxis of herpes zoster is varicella vaccination. In 14

VZV-seropositive patients undergoing BMT, the use of a varicella vaccine during the early

post-transplantation period (administered in three doses at 1, 2 and 3 months) boosted

VZV-specific cellular immunity and decreased the severity of herpes zoster.86 A trial of a

modified approach, in which one dose of vaccine is given prior to BMT and three doses

are given after transplantation, is underway.

Despite efforts to prevent varicella in immunocompromised patients, passive prophylaxis

can sometimes fail, and unrecognized exposures to VZV can occur. Consequently, antiviral

treatment after the onset of clinical disease is a key component of the management of

varicella in immunocompromised patients. Antiviral therapy requires continual

reassessment, regardless of how the patient is immunocompromised.

Aciclovir has proven useful and has become the drug of choice in this setting. Early treatment

with it stops VZV viraemia and virus replication in skin, shortens the duration of virus shedding

and new lesion formation, and speeds lesion healing.2,87 The intervention appears to

compensate for the delayed acquisition of virus-specific immunity in patients in whom the

cell-mediated immune responses that are necessary to limit primary VZV infection are altered.

There is a limited window of opportunity for antiviral therapy to affect the outcome of

varicella. In the immunocompetent host, most virus replication has ceased by 72 hours

after rash onset. In immunocompromised patients, however, duration of virus replication

and virus shedding is extended. This limited chance to affect disease outcome means that

aggressive management of disease in these patients is justifiable and necessary. This has

been emphasized by studies investigating the impact of untreated varicella. In a study of

127 children with leukaemia and varicella who were not treated with antivirals, 28% of

the patients developed varicella-associated pneumonia, with a mortality rate of 7%.88

Treatment of VZV Infections in the Immunocompromised Antiviral therapy In immunocompetent individuals, post-exposure administration of antiviral therapy with

aciclovir is effective in reducing the severity of varicella and herpes zoster. Antiviral

therapy suppresses virus replication, and in immunocompetent hosts allows immune

responses to develop. Consequently, protective immunity may develop without the

89

Page 91: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

associated severe clinical symptoms. However, in immunocompromised individuals, there

are few specific data available on the efficacy of antiviral post-exposure prophylaxis. In

these patients, immunity to VZV may not develop during administration of aciclovir, and

residual virus infection may still cause severe disease after prophylaxis is stopped. Despite

the lack of data, some physicians believe that aciclovir is effective in immunocompromised

patients and recent studies support this.87 The primary reason to treat herpes zoster in

immunocompromised hosts is to prevent life-threatening visceral dissemination. Alleviation

of acute pain and prevention or amelioration of post-herpetic neuralgia (PHN) are also

desirable therapeutic goals, as they are for immunocompetent patients.

J Intravenous aciclovir therapy is the standard of care for immunocompromised patients

with disseminated VZV disease, including those with complications such as varicella

pneumonia (category 1 recommendation).

Intravenous aciclovir treatment in the immunocompromisedVaricellaIntravenous aciclovir (10 mg/kg for adults and 500 mg/m2 or 20 mg/kg for children every

8 hours) remains the standard of care for the treatment of varicella in the immunocompromised,

including those within 9–12 months of BMT. Such therapy prevents virus dissemination,

reduces visceral complications,89 and in severe cases of varicella, reduces the duration of

prolonged attacks.27,13 Early treatment (within 24 hours of rash onset) with intravenous

aciclovir (500 mg/m2 every 8 hours for 5–7 days) significantly lowers the probability of

pneumonitis and death in immunocompromised children with varicella.25,88 In a very small

study of 20 paediatric cancer patients with varicella,25 a higher number of placebo recipients

(five of 11) developed pneumonia compared with the aciclovir recipients (none of seven).25

Intravenous aciclovir should be administered to transplant recipients and HIV-positive

individuals with low CD4 cell counts with varicella whether or not it is initiated within

24 hours of rash onset.

Early treatment is important to limit the chance of visceral dissemination; patients with

evidence of visceral dissemination before starting aciclovir therapy have a poorer

prognosis than those with none.90

Herpes zosterIntravenous aciclovir is used consistently for the highest-risk patients with localized or

disseminated herpes zoster, based on evidence from one placebo-controlled trial33 and

three comparative studies with vidarabine.91-93

In the placebo-controlled study, immunocompromised patients (those with

lymphoproliferative or other cancers, BMT or renal transplant recipients, or those with

primary immune deficiency) were enrolled into the trial up to 45 days after the onset of

localized herpes zoster provided they had new vesicles forming.33 Patients in the aciclovir

arm received a 7-day intravenous course of aciclovir (1500 mg/m2/day). There was no

significant effect of aciclovir on rash healing or on the resolution of acute pain, and this

was still the case when the analysis was confined to those patients randomized within

72 hours of rash onset, although it should be noted that the number of patients in this

subset was small. However, aciclovir did prevent further potentially life-threatening

cutaneous and visceral dissemination.33

90

Page 92: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

In a study of aciclovir (10 mg/kg at 8-hour intervals for 7 days) and vidarabine (continuous

12-hour infusion of 10 mg/kg/day for 7 days) in 73 cancer patients with disseminated

herpes zoster, there were no differences between the treatment groups in rates of

cutaneous healing, resolution of acute neuritis or frequency of PHN,94 although aciclovir-

treated patients were discharged from hospital earlier. These data suggest that both

intravenous aciclovir and intravenous vidarabine are effective for treatment of

disseminated herpes zoster in immunocompromised individuals.

In the comparative studies of aciclovir and vidarabine,91-93 there was no discernible

advantage of vidarabine; in one trial, aciclovir was more effective. Moreover, aciclovir was

associated with fewer side-effects and was easier to administer.93 Consequently,

intravenous aciclovir remains the standard therapy for the treatment of recurrent VZV

infection in the immunocompromised host. There are no controlled trials in the peer-

reviewed literature comparing intravenous aciclovir with oral aciclovir, famciclovir or

valaciclovir for the treatment of herpes zoster in the immunocompromised host.

The place of oral antiviralsJ Studies are recommended comparing valaciclovir and famciclovir with intravenous

aciclovir for the management of immunocompromised individuals with VZV infections

(research need recommendation).

J There is a need to define the patients for whom oral antiviral therapy might be used in

place of intravenous therapy (research need recommendation).

Oral antiviral therapy in varicellaJ Anecdotal evidence suggests that oral antiviral therapy may be appropriate for the

treatment of varicella in some immunocompromised individuals. In this setting, the newer

antivirals, valaciclovir (1000 mg three times daily for adults) and famciclovir (500 mg three

times daily for adults), are likely to be at least as effective as aciclovir (800 mg four times

daily). The minimum duration of therapy should be 7 days (category 3 recommendation).

Although standard management of immunocompromised patients with varicella involves

intravenous aciclovir, this approach is expensive, has a negative effect on patient quality

of life, and is inconvenient to the recipient and their family.95 There is also a risk of

intravenous catheter-related complications such as phlebitis and secondary infection.

Consequently, the role of oral antiviral therapies for the treatment of varicella in less

immunocompromised individuals has gained interest.

To date, there have been no controlled trials in the peer-reviewed literature of oral

aciclovir, valaciclovir or famciclovir for the treatment of varicella in the

immunocompromised host. There are, however, anecdotal reports and small studies that

suggest such therapy may be adequate in some patients.16

In such circumstances, the newer oral antiviral therapies – valaciclovir and famciclovir –

are likely to be as effective as or more effective than oral aciclovir. Indeed, oral valaciclovir

(1000 mg three times a day) provides comparable systemic exposure, but reduced peak

aciclovir concentrations, compared with intravenous aciclovir (5 mg/kg three times a day)

in immunocompromised cancer patients, without adverse effects.96 In fact, Cohen et al1

recommended that these newer oral antiviral compounds should be considered alongside

91

Page 93: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

oral aciclovir for treatment of varicella in some immunocompromised individuals. Oral

valaciclovir, famciclovir or aciclovir for 7 days was recommended for those taking

continuous or intermittent high-dose corticosteroids and those taking low-dose daily

cytotoxic chemotherapy.1 However, there are no paediatric formulations of valaciclovir

and famciclovir currently available.

Sequential intravenous–oral antiviral therapyConsidering the potential benefit of oral antiviral therapy in immunocompromised patients,

an approach using sequential intravenous–oral antiviral therapy may also prove effective.

A study was conducted in 26 immunocompromised children to assess the efficacy of

sequential intravenous and oral aciclovir.95 Intravenous aciclovir treatment was initiated at

1500 mg/m2/day in three equal doses. Concurrent management included reducing

immunosuppressive therapies (mercaptopurine, methotrexate, corticosteroids, cyclosporin)

by 50%, or stopping administration of these therapies. Sixteen of the patients had a

documented exposure to VZV. Eleven of these 16 (69%) received VZIG in addition to

intravenous aciclovir. Patients receiving 48 hours of intravenous therapy were deemed eligible

to switch to oral aciclovir (20 mg/kg four times daily) if they were afebrile, had no new lesion

formation within the previous 24 hours, had no internal organ involvement, and were able

to tolerate oral medication. Upon switching to oral aciclovir, patients were observed in

hospital for a further 24 hours; if their condition did not deteriorate, they were discharged.

Twenty five of the 26 patients successfully switched from intravenous to oral therapy after

4 days (exact 4.1±1.2 [mean±SD] days). Varicella resolved in all 25 children who switched

to oral aciclovir, and none of the patients required resumption of intravenous therapy.95

Considering that sequential intravenous–oral antiviral therapy for immunocompromised children

with severe varicella may decrease the duration of hospitalization and hence reduce the

associated inconvenience and high cost, this multi-modal therapy merits further investigation.

Oral antiviral therapy for herpes zosterJ For some immunocompromised individuals, oral antiviral therapy may be appropriate for

the treatment of herpes zoster – in this setting, the newer antivirals, valaciclovir (1000 mg

three times daily for adults) and famciclovir (500 mg three times daily for adults), are

likely to be at least as effective as aciclovir (800 mg five times daily) (category 3

recommendation).

J Studies are recommended comparing valaciclovir and famciclovir with intravenous

aciclovir in the management of immunocompromised individuals with herpes zoster

(research need recommendation).

The disadvantages of intravenous therapy (e.g. cost, risk of infection) have driven the

search for effective alternatives. Evidence is now emerging that some HIV-infected patients

(with normal CD4 cell counts) and other immunocompromised patients with herpes zoster

respond well to oral antiviral therapy.87,97 Patients who may be eligible for oral antiviral

therapy include HIV-infected individuals and those receiving:

• High-dose corticosteroids (intermittent or continuous)

• Low-dose cytotoxic chemotherapy (daily cyclophosphamide, methotrexate, 6-MP and

azathioprine).92

Page 94: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

A double-blind study compared famciclovir (500 mg three times daily) with oral aciclovir

(800 mg five times daily) for 10 days for treatment of localized dermatomal herpes zoster in

immunocompromised patients.87 Patients were randomized following BMT, solid organ

transplantation, or cancer chemotherapy. Famciclovir and aciclovir were equivalent with

respect to the proportion of patients experiencing new lesion formation whilst receiving

treatment (77% versus 73% for famciclovir and aciclovir groups, respectively). In addition,

there were no significant differences

observed between the treatment groups

with respect to time to cessation of new

lesion formation, time to full crusting

and time to loss of acute pain (Table 2).87

Both oral antiviral therapies appeared to

reduce the risk of cutaneous or visceral

dissemination of herpes zoster. The

number of patients developing

disseminated zoster was low in both

treatment groups (famciclovir 3% [two

patients] aciclovir 8% [six patients]).

These results are similar to historical data

reported for intravenous aciclovir in

immuncompromised patients.33

The two oral agents were well tolerated.

This safety profile, combined with the

efficacy results, suggests that oral antiviral therapy offers a convenient alternative to

intravenous therapy. Valaciclovir (1000 mg three times daily) will probably be at least as

effective as aciclovir in this patient group. In neutropenic patients, valaciclovir (1000 mg

8-hourly) produced systemic aciclovir exposure comparable with that produced by

5 mg/kg aciclovir administered by 1-hour intravenous infusion every 8 hours (Figure 7).96

The efficacy of valaciclovir in this and other immunocompromised patient groups should

be established in clinical trials.

In a study of sorivudine (40 mg daily for 10 days)

versus aciclovir (800 mg five times daily for 10 days) in

170 HIV-positive patients presenting with herpes

zoster, sorivudine was effective and well tolerated.97 In

this study, the median CD4 cell count was 171

cells/mm3; 34% of the population had a CD4 cell

count of <100 cells/mm3 and 45% had CD4 cell

counts of >200 cells/mm3. Sorivudine was significantly

superior to aciclovir for several parameters, including

time to cessation of new vesicle formation and time to

total lesion crusting. Both aciclovir and sorivudine

were well tolerated and not associated with serious

clinical adverse events, and adverse event profiles

were not different between recipients. However, a

number of fatal adverse reactions have been reported

in Japan associated with the use of sorivudine in

patients already receiving the anticancer drug

5-fluorouracil.

93

Famciclovir Aciclovir500 mg 3 x/day 800 mg 5 x/day

n=71 n=77

Time to full crusting Patients in analysis 71 77 Patients with event 60 60 Median (days) 8 9 Hazard ratio 1.26 95% confidence interval (0.88, 1.82)*

Time to complete healing Patients in analysis 71 77 Patients with event 56 57 Median (days) 20 21 Hazard ratio 0.9895% confidence interval (0.67, 1.42)*

Time to loss of acute phase pain Patients in analysis 64 69 Patients with event 39 36 Median (days) 14 17 Hazard ratio 1.1195% confidence Interval (0.71, 1.75)*

*No significant difference

FIGURE 7: Meansteady-state plasmaaciclovir concentrationsover time followingadministration of oralvalaciclovir (1000 mgthree times daily) orintravenous aciclovir toneutropenic patients(5 mg/kg every 8 hours)96

25

20

15

10

5

00 1 3 4 5 6 7

Aci

clov

ir c

once

ntra

tion

(µM

)

121110982

Time (hours)

35

30

IV aciclovir (5 mg/kg)Oral valaciclovir (1000 mg)

TABLE 2: Lesion healingand resolution of acutepain in immuno-compromised patientsreceiving either oralaciclovir (800 mg fivetimes daily) or famciclovir(500 mg three timesdaily)87

© R

epro

duce

d w

ith p

erm

issi

on,H

oglu

nd M

, Lju

ngm

an P

, Wel

ler S

. Com

para

ble

acic

lovi

r exp

osur

espr

oduc

ed b

y or

al v

alac

iclo

vir a

nd in

trave

nous

aci

clov

ir in

imm

unoc

ompr

omis

ed c

ance

r pat

ient

s.

J Ant

imic

rob

Che

mot

her 2

002;

47:8

55-8

56.O

xfor

d U

nive

rsity

Pre

ss.

Page 95: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

VZV Ocular Infections in HIV-Infected IndividualsJ Combined intravenous foscarnet (60 mg/kg three times per week) plus intravitreal

ganciclovir (400 mg twice per week) therapy is the best option for rapidly progressive

herpetic retinal necrosis (category 3 recommendation). However, the dose of foscarnet

to be used has not been established in the clinical trial setting, and further dose-

comparative studies should be undertaken.

The reduced level of cell-mediated immunity in herpes zoster patients who are HIV-positive

may lead to a higher level of unchecked virus replication and hence more frequent and serious

complications. VZV infection, and particularly herpes zoster ophthalmicus, is often the first

manifestation of HIV infection. Sellitti et al98 conducted a retrospective review of 112 herpes

zoster ophthalmicus patients to determine the incidence of HIV infection in patients

presenting with the complication. Twenty-nine of the 112 herpes zoster ophthalmicus

patients (26%) were HIV-positive. All 29 patients were younger than 50 years at the time of

herpes zoster ophthalmicus diagnosis. Acute retinal necrosis following herpes zoster

ophthalmicus was observed in five of the 29 immunocompromised patients, and none of

the remaining 83 patients.98 The authors recommended that all patients younger than

50 years presenting with herpes zoster ophthalmicus be tested for HIV serostatus, and that

HIV-infected patients should be monitored for acute retinal necrosis.98

Margolis et al99 reported a retrospective study on the incidence of ocular complications in

48 HIV-positive individuals with herpes zoster ophthalmicus. All patients had received

either oral or intravenous aciclovir. The median CD4 lymphocyte count for the study

population was 48 cells/mm3 (range 2–490 cells/mm3).99 The prevalence of ocular

complications is shown in Table 3. In addition to these complications, two patients (4%)

developed PHN, and two (4%) zoster-associated CNS disease.99 The authors reported that

although the overall level of complications was lower than expected for the study

population, those caused by active virus replication – chronic infectious pseudodendritic

keratitis and CNS disease – were

particularly devastating to the patients

involved and difficult to manage.99 This

highlights the need for effective

treatment to limit the VZV replication in

patients with HIV infection.

Rapidly progressive herpetic retinal necrosisRapidly progressive herpetic retinal necrosis (RPHRN) is a syndrome caused by VZV

infection, and is observed almost exclusively in patients with HIV infection (CD4 cell

count <100/mm3). Presentation is with multifocal necrotizing lesions involving the

peripheral retina. The granular, non-haemorrhagic lesions rapidly extend and coalesce,

and progress to full thickness retinal necrosis (Figure 8). In HIV-positive patients, bilateral

blindness ensues within days or weeks.100-110

For RPHRN in individuals with AIDS, combination therapy with intravenous ganciclovir (5 mg/kg

twice per day) plus systemic foscarnet may improve the outcome, and prevent involvement of

the other eye.101-105 However, visual acuity may still be reduced.101 More encouraging results

were reported in a single case report on the use of cidofovir (5 mg/kg weekly for 2 weeks and

subsequently once every 2 weeks as maintenance dose); the patient’s visual acuity improved and

stabilized (for at least 16 months) following five treatment cycles (10 weeks).106

94

TABLE 3: Prevalenceand nature of ocularcomplications of herpeszoster ophthalmicus inHIV-positive patientsreceiving aciclovir99

Ocular complication Prevalence (%)

Mild or no ocular involvement 31 Stromal keratitis (usually mild) 35 Chronic infectious pseudodendritic keratitis 4 Iritis 50 Raised intraocular pressure 6 Progressive outer retinal necrosis 4

Page 96: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Antiviral-resistant VZV infectionsJ In immunocompromised patients with suspected

aciclovir-resistant VZV infection, treatment with foscarnet

(120–200 mg/kg/day, in two or three divided doses)

should be initiated unless renal failure is evident

(category 2 recommendation).

VZV resistant to aciclovir has been reported as a clinical

problem only in HIV-positive individuals with long-term,

chronic VZV infection.54,107 These individuals frequently

experience persistent or recurrent VZV infections, which may

be atypical in presentation (e.g. ecthymatous VZV

lesions).55,56 The resistant isolates are often cross-resistant to

penciclovir (the prodrug of famciclovir). This occurs as most

drug-resistant isolates have an altered or deleted thymidine

kinase gene. Aciclovir and penciclovir require activation by

virus thymidine kinase prior to inhibiting the virus polymerase. Therefore, alternative drugs

that act to directly inhibit the DNA polymerase, such as foscarnet or cidofovir, are often

effective as second-line treatments.

The efficacy of foscarnet in the setting of aciclovir-resistant VZV was shown in a recent

review of 18 HIV-positive individuals with chronic VZV lesions.108 Thirteen of these

patients received intravenous foscarnet (120–200 mg/kg/day, in two or three divided

doses). Ten patients (77%) experienced initial complete healing of their lesions, although

herpes zoster reoccurred after a mean of 110 days in five.

While foscarnet can be effective, there have been rare instances of foscarnet resistance in

VZV.109 Cidofovir may be a suitable alternative for infections caused by foscarnet and

nucleoside analogue-resistant strains.110

SummaryImmunocompromised individuals who have impaired cell-mediated immunity are at great

risk of developing life-threatening varicella and its associated complications. Consequently,

both post-exposure prophylaxis and aggressive treatment of varicella, if it does occur, are

warranted in this group.

The efficacy and safety of VZIG as prophylaxis for varicella in immunocompromised

populations have been well documented and, as such, remains the mainstay of post-

exposure prophylaxis in this population. Recent studies have indicated that post-exposure

prophylaxis with aciclovir is effective, particularly if used in combination with VZIG, and

it is recommended that this combination is used in immunocompromised patients to

improve the outcome following exposure to varicella.

Long-term oral antiviral therapy for suppression of VZV may be of value in organ transplant

patients, especially BMT recipients and patients requiring immunosuppression for GVHD.

It should be noted that post-exposure prophylaxis with varicella vaccine is not suitable for

the immunocompromised. 95

FIGURE 8: RPHRN inan HIV-positive individual

© R

epro

duce

d w

ith p

erm

issi

on,O

rmer

od L

D, L

arki

n JA

, Mar

go C

Aet

al.

Rap

idly

prog

ress

ive

herp

etic

ret

inal

nec

rosi

s: a

blin

ding

dis

ease

cha

ract

eris

tic o

f adv

ance

d A

IDS.

Clin

Infe

ct D

is19

98;2

6:34

-45;

dis

cuss

ion

46-3

7.U

nive

rsity

of C

hica

go.

Page 97: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

When prophylaxis fails or unwitting exposure occurs, aggressive treatment of varicella

with antiviral compounds is necessary and has been well documented. Intravenous

antiviral therapy with aciclovir (10 mg/kg for adults and 500 mg/m2 or 20 mg/kg for

children every 8 hours) remains the standard of care for immunocompromised patients

and those presenting with complications such as varicella-associated pneumonia. In the

profoundly immunocompromised with suspected aciclovir resistance, therapy with

foscarnet (120–200 mg/kg/day, in two or three divided doses) is recommended in the first

instance.

Studies suggest that oral antiviral therapy may be appropriate for the treatment of varicella in

some immunocompromised individuals, which may help alleviate the cost, inconvenience

and negative impact on patients’ quality of life associated with intravenous therapy. As is the

case for varicella in the immunocompetent, valaciclovir (1000 mg three times daily) and

famciclovir (500 mg three times daily) in adults will probably be at least as effective as

aciclovir (800 mg five times daily).

In immunocompromised hosts, the primary reason to treat herpes zoster is to prevent visceral

dissemination, which can be fatal. Alleviation of acute pain and prevention or amelioration

of PHN are also desirable therapeutic goals, as they are for immunocompetent patients.

Dissemination of herpes zoster is more likely to occur in immunocompromised individuals

than in the immunocompetent of the same age. However, despite the greater impact of

herpes zoster, there have been fewer studies conducted in these populations than in

immunocompetent groups.

Prophylactic treatment with intravenous or oral antivirals, potentially administered

sequentially, is recommended in transplant patients to prevent the development of herpes

zoster in the period immediately following transplantation when they are at their most

immunocompromised and, therefore, most at risk of severe herpes zoster and its associated

complications.

Intravenous aciclovir (10 mg/kg every 8 hours) should be considered the standard of care for

immunocompromised patients with herpes zoster. In those with suspected aciclovir

resistance, therapy with foscarnet (120–200 mg/kg/day, in two or three divided doses) should

be initiated, unless renal failure is evident. In rare situations where foscarnet-resistant VZV is

encountered, cidofovir may be a suitable therapeutic alternative.

Preliminary evidence suggests that oral antiviral therapy is appropriate for the treatment of

herpes zoster in some immunocompromised individuals. Famciclovir (500 mg three times

daily for adults) is as effective as aciclovir (800 mg five times daily) in the treatment of herpes

zoster in the immunocompromised. Valaciclovir (1000 mg three times daily for adults) may

well be at least as effective as oral aciclovir and famciclovir but this hypothesis has yet to be

tested in a clinical trial.

References1. Cohen JI, Brunell PA, Straus SE et al. Recent advances in varicella-zoster virus infection. Ann Intern Med

1999;130:922-932.2. Vafai A, Berger M. Zoster in patients infected with HIV: a review. Am J Med Sci 2001;321:372-380.3. Jue S, Whitley RJ. Herpesvirus Infections in Children with Human Immunodeficiency Virus. 2nd ed.

Baltimore: Williams & Wilkins, 1994.96

Page 98: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

4. Newell M. Personal communication.5. Conant M, Wood MJ. Management Strategies in Herpes: Herpesvirus and HIV Infection – Co-factors and

Opportunistic Infections. Worthing: PPS Europe Ltd, 1995.6. Wilkins EG, Leen CL, McKendrick MW et al. Management of chickenpox in the adult. A review prepared

for the UK Advisory Group on Chickenpox on behalf of the British Society for the Study of Infection. J InfectDis 1998;36 (Suppl 1):49-58.

7. Webster A, Grint P, Brenner MK et al. Titration of IgG antibodies against varicella zoster virus before bonemarrow transplantation is not predictive of future zoster. J Med Virol 1989;27:117-119.

8. Steer CB, Szer J, Sasadeusz J et al. Varicella-zoster infection after allogeneic bone marrow transplantation:incidence, risk factors and prevention with low-dose aciclovir and ganciclovir. Bone Marrow Transplant2000;25:657-664.

9. Schmader K. Epidemiology of herpes zoster. In: Varicella-Zoster Virus: Virology and Clinical Management.(Arvin A, Gershon A, eds). Cambridge: Cambridge University Press, 2000: 220-245.

10. Buchbinder SP, Katz MH, Hessol NA et al. Herpes zoster and human immunodeficiency virus infection. J Infect Dis 1992;166:1153-1156.

11. Rogues AM, Dupon M, Ladner J et al. Herpes zoster and human immunodeficiency virus infection: a cohortstudy of 101 co-infected patients. J Infect Dis 1993;168:245.

12. Glesby MJ, Moore RD, Chaisson RE. Clinical spectrum of herpes zoster in adults infected with humanimmunodeficiency virus. Clin Infect Dis 1995;21:370-375.

13. McGregor RM. Herpes zoster, chicken pox and cancer in general practice. BMJ 1957;1:84-87.14. Hope-Simpson RE. The nature of herpes zoster: a long-term study and a new hypothesis. Proc R Soc Med

1965;58:9-20.15. Ragozzino MW, Melton LJ, 3rd, Kurland LT et al. Population-based study of herpes zoster and its sequelae.

Medicine (Baltimore) 1982;61:310-316.16. Liesegang T. Varicella zoster viral disease. Mayo Clin Proc 1999;74:983-998.17. Friedman-Kien AE, Lafleur FL, Gendler E et al. Herpes zoster: a possible early clinical sign for development

of acquired immunodeficiency syndrome in high-risk individuals. J Am Acad Dermatol 1986;14:1023-1028.18. Rusthoven JJ, Ahlgren P, Elhakim T et al. Varicella-zoster infection in adult cancer patients. A population

study. Arch Intern Med 1988;148:1561-1566.19. Rusthoven JJ. The risk of varicella-zoster infections in different patient populations: a critical review. Transfus

Med Rev 1994;8:96-116.20. Rusthoven JJ, Ahlgren P, Elhakim T et al. Risk factors for varicella zoster disseminated infection among adult

cancer patients with localized zoster. Cancer 1988;62:1641-1646.21. Ljungman P. Prophylaxis against herpesvirus infections in transplant recipients. Drugs 2001;61:187-196.22. Ljungman P, Wilczek H, Gahrton G et al. Long-term acyclovir prophylaxis in bone marrow transplant

recipients and lymphocyte proliferation responses to herpes virus antigens in vitro. Bone Marrow Transplant1986;1:185-192.

23. Selby PJ, Powles RL, Easton D et al. The prophylactic role of intravenous and long-term oral acyclovir afterallogeneic bone marrow transplantation. Br J Cancer 1989;59:434-438.

24. Lowance D, Neumayer HH, Legendre CM et al. Valacyclovir for the prevention of cytomegalovirus diseaseafter renal transplantation. International Valacyclovir Cytomegalovirus Prophylaxis Transplantation StudyGroup. N Engl J Med 1999;340:1462-1470.

25. Prober CG, Kirk LE, Keeney RE. Acyclovir therapy of chickenpox in immunosuppressed children—acollaborative study. J Pediatr 1982;101:622-625.

26. Pahwa S, Biron KK, Lim W. Continuous varicella-zoster virus infection associated with acyclovir resistancein a child with AIDS. JAMA 1988;260:2879-2882.

27. Jura E, Chadwick EG, Josephs SH. Varicella-zoster virus infections in children infected with humanimmunodeficiency virus. Pediatr Infect Dis J 1989;8:586-590.

28. Srugo I, Israele V, Wittek AE et al. Clinical manifestations of varicella-zoster virus infections in humanimmunodeficiency virus-infected children. Am J Dis Child 1993;147:742-745.

29. Lieibovitz E, Cooper DK, Giurguitin D. Varicella zoster virus infection in Romanian children infected withthe human immunodeficiency virus. Pediatrics 1993;92:838-842.

30. Kelley R, Mancao M, Lee F. Varicella in children with perinatally acquired human immunodeficiency virusinfection. J Pediatr 1994;124:271-273.

31. Wallace MR, Hooper DG, Pyne JM et al. Varicella immunity and clinical disease in HIV-infected adults.South Med J 1994;87:74-76.

32. Gnann JW, Whitley RJ. Natural history and treatment of varicella-zoster in high-risk populations. J HospInfect 1991;18:317-329.

33. Balfour HH, Jr., Bean B, Laskin OL et al. Acyclovir halts progression of herpes zoster inimmunocompromised patients. N Engl J Med 1983;308:1448-1453.

34. Balfour HH, Jr., McMonigal KA, Bean B. Acyclovir therapy of varicella-zoster virus infections inimmunocompromised patients. J Antimicrob Chemother 1983;12 (Suppl B):S169-S179.

35. Glesby MJ, Moore RD, Chaisson RE. Herpes zoster in patients with advanced human immunodeficiencyvirus infection treated with zidovudine. J Infect Dis 1993;168:1264-1268.

36. Bowsher D. The lifetime occurrence of herpes zoster and prevalence of post-herpetic neuralgia: aretrospective survey in an elderly population. Eur J Pain 1999;3:335-342.

37. De La Blanchardiere A, Rozenberg F, Caumes E. Neurological complications of varicella-zoster virusinfection in adults with human immunodeficiency virus-infected patients: results from a randomized,controlled trial. Antimicrob Agents Chemother 2000;42:1139-1145.

38. Locksley RM, Flournoy N, Sullivan KM et al. Infection with varicella-zoster virus after marrow transplantation. J Infect Dis 1985;152.

39. Stemmer SM, Kinsman K, Tellschow S et al. Fatal noncutaneous visceral infection with varicella-zoster virus ina patient with lymphoma after autologous bone marrow transplant. Clin Infect Dis 1993;16:497-499.

40. Rogers SY, Irving W, Harris A et al. Visceral varicella zoster infection after bone marrow transplantation withoutskin involvement and the use of PCR for diagnosis. Bone Marrow Transplant 1995;15:805-807.

97

Page 99: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

41. Koc Y, Miller KB, Schenkein DP et al. Varicella zoster virus infections following allogeneic bone marrowtransplantation: frequency, risk factors, and clinical outcome. Biol Blood Marrow Transplant 2000;6:44-49.

42. Horten B, Price RW, Jimenez D. Multifocal varicella-zoster virus leukoencephalitis temporally remote fromherpes zoster. Ann Neurol 1981;9:251-266.

43. Gray F, Mohr M, Rozenberg F. Varicella-zoster virus encephalitis in acquired immunodeficiency syndrome: areport of four cases. Neuropathol Appl Neurobiol 1992;18:502-514.

44. Janssen RS, Saykin AJ, Kaplan JE. Nuerological complications of human immunodeficiency virus infection inpatients with lymphadenopathy syndrome. Ann Neurol 1988;23:49-55.

45. Gray F, Belec L, Lescs MC. Varicella-zoster virus infection of the central nervous system in the acquired immunedeficiency syndrome. Brain 1994;117:987-999.

46. Kleinschmidt-DeMasters BK, Amlie-Lefond C, Gilden DH. The patterns of varicella zoster virus encephalitis. Hum Pathol 1996;27:927-938.

47. Morgello S, Block GA, Price RW et al. Varicella-zoster virus leukoencephalitis and cerbral vasculopathy. ArchPathol Lab Med 1988;112:173-177.

48. Lentz D, Jordan JE, Pike GB et al. MRI in varicella zoster virus leukoencephalitis in the immunocompromisedhost. J Comput Assist Tomogr 1993;17:313-316.

49. Aygun N, Finelli DA, Rodgers MS et al. Multifocal varicella-zoster virus leukoencephalitis in a patient withAIDS: MR findings. Am J Neuroradiol 1998;19:1897-1899.

50. Gilden DH, Kleinschmidt-DeMasters BK, LaGuardia JJ et al. Neurologic complications of the reactivation ofvaricella-zoster virus. N Engl J Med 2000;342:635-645.

51. Iten A, Chatelard P, Vuadens P et al. Impact of cerebrospinal fluid PCR on the management of HIV-infectedpatients with varicella-zoster virus infection of the central nervous system. J Neurovirol 1999;5:172-180.

52. Tronnier M, Plettenberg A, Meigel WN et al. Recurrent verrucous herpes zoster in an HIV patient.Demonstration of the virus by immunofluorescence and electron microscopy. Eur J Dermatol 1994;4:604-607.

53. Vaughan-Jones SA, McGibbon DH, Bradbeer CS. Chronic verrucous varicella-zoster infection in a patient withAIDS. Clin Exp Dermatol 1994;19:327-329.

54. Jacobson MA, Berger TG, Fikrig S et al. Acyclovir-resistant varicella zoster virus infection after chronic oralacylovir therapy in patients with the acquired immunodeficiency syndrome (AIDS). Ann Intern Med1990;112:187-191.

55. Gilson IH, Barnett JH, Conant M et al. Disseminated ecthymatous herpes varicella zoster virus infection inpatients with acquire immunodeficiency syndrome. J Am Acad Dermatol 1989;20:637-642.

56. Alessi E, Cusini M, Zerboni R. Unusual varicella zoster virus infection in patients with the acquiredimmunodeficiency syndrome. Arch Dermatol 1988;124:1011-1013.

57. Annunziato PW, Gershon AA. Primary immunization against varicella. In: Varicella-Zoster Virus: Virology andClinical Management. (Arvin A, Gershon AA, eds). Cambridge: Cambridge University Press, 2000: 460-476.

58. Takahashi M, Gershon AA. Varicella vaccine. In: Vaccines. 2nd edn. (Plotkin SA, Mortomer EA, eds).Philadelphia PA: WB Saunders, 1994: 387-417.

59. Sato Y, Miyano T, Kawauchi K et al. Use of live varicella vaccine in children with acute leukemia and malignantlymphoma. Biken J 1984;27:111-113.

60. Nunoue T. Clinical observations on varicella-zoster vaccinees treated with immunosuppressants for amalignancy. Biken J 1984;27:115-118.

61. Konno T, Yamaguchi Y, Minegishi M et al. A clinical trial of live attenuated varicella vaccine (Biken) in childrenwith malignant diseases. Biken J 1984;27:73-75.

62. Ninane J, Latinne D, Heremans-Bracke MT et al. Live varicella vaccine in severely immunodepressed children.Postgrad Med J 1985;61:97-102.

63. Haas RJ, Belohradsky B, Dickerhoff R et al. Active immunization against varicella of children with acuteleukaemia or other malignancies on maintenance chemotherapy. Postgrad Med J 1985;61:69-72.

64. Heath RB, Malpas JS. Experience with the live Oka-strain varicella vaccine in children with solid tumours.Postgrad Med J 1985;61:107-111.

65. Heller L, Berglund G, Ahstrom L et al. Early results of a trial of the Oka-strain varicella vaccine in children withleukaemia or other malignancies in Sweden. Postgrad Med J 1985;61:79-83.

66. LaRussa P. Experience with live-attenuated varicella-zoster vaccines. Contrib Microbiol 1999;3:173-192.67. Gershon AA, LaRussa P, Steinberg S. The varicella vaccine. Clinical trials in immunocompromised individuals.

Infect Dis Clin North Am 1996;10:583-594.68. Gershon AA. Varicella-zoster virus: prospects for control. Adv Pediatr Infect Dis 1995;10:93-124.69. Zamora I, Simon JM, Da Silva ME et al. Attenuated varicella virus vaccine in children with renal transplants.

Pediatr Nephrol 1994;8:190-192.70. Giacchino R, Marcellini M, Timitilli A et al. Varicella vaccine in children requiring renal or hepatic

transplantation. Transplantation 1995;60:1055-1056.71. Sauerbrei A, Prager J, Hengst U et al. Varicella vaccination in children after bone marrow transplantation. Bone

Marrow Transplant 1997;20:381-383.72. Morales-Castillo ME, Alvarex-Munoz MT, Solorzano-Santos F et al. Live varicella vaccine in both

immunocompromised and healthy children. Arch Med Res 2000;31:85-87.73. Hata A, Asanuma H, Rinki M et al. Use of an inactivated varicella vaccine in recipients of hematopoietic-cell

transplants. N Engl J Med 2002;347:26-34.74. Levin MJ, Nelson WL, Preblud SR. Clinical trials with varicella-zoster virus immunoglobulins. London:

Academic Press, 1986.75. Paryani SG, Arvin AM, Koropchak CM et al. Comparison of varicella zoster antibody titers in patients given

intravenous immune serum globulin or varicella zoster immune globulin. J Pediatr 1984;105:200-205.76. Lynfield R, Herrin JT, Rubin RH. Varicella in pediatric renal transplant patients. Pediatrics 1992;90:216-220.77. McGregor RS, Zitelli BJ, Urbach AH. Varicella in pediatric orthotopic liver tranplant recipients. Pediatrics

1989;82:256-261.78. Feldman S, Hughes WT, Daniel CB. Varicella in children with cancer: Seventy-seven cases. Pediatrics 1975;56:

388-397.

98

Page 100: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

79. Ross H. Modification of chickenpox in family contacts by administration of gamma globulin. N Engl J Med1962;267:369-376.

80. Goldstein SL, Somers MJ, Lande MB et al. Acyclovir prophylaxis of varicella in children with renal disease receivingsteroids. Pediatr Nephrol 2000;14:305-308.

81. Lundgren G, Wilczek H, Lonnqvist B et al. Acyclovir prophylaxis in bone marrow transplant recipients. Scand JInfect Dis Suppl 1985;47:137-144.

82. Prentice HG, Gluckman E, Powles RL et al. Impact of long-term acyclovir on cytomegalovirus infection and survivalafter allogeneic bone marrow transplantation. European Acyclovir for CMV Prophylaxis Study Group. Lancet1994;343:749-753.

83. Singh N, Yu VL, Mieles L et al. High-dose acyclovir compared with short-course preemptive ganciclovir therapy toprevent cytomegalovirus disease in liver transplant recipients. A randomized trial. Ann Intern Med 1994;120:375-381.

84. Winston DJ, Wirin D, Shaked A et al. Randomised comparison of ganciclovir and high-dose acyclovir for long- termcytomegalovirus prophylaxis in liver-transplant recipients. Lancet 1995;346:69-74.

85. Wood MJ. Antivirals in the context of HIV disease. J Antimicrob Chemother 1996;37 (Suppl B):S97-S112.86. Redman RL, Nader S, Zerboni L et al. Early reconstitution of immunity and decreased severity of herpes zoster in

bone marrow transplant recipients immunized with inactivated varicella vaccine. J Infect Dis 1997;176:578-585.87. Tyring S, Belanger R, Bezwoda W et al. A randomized, double-blind trial of famciclovir versus acyclovir for the

treatment of localized dermatomal herpes zoster in immunocompromised patients. Cancer Invest 2001;19:13-22.88. Feldman S, Lott L. Varicella in children with cancer: impact of antiviral therapy and prophylaxis. Pediatrics

1987;80:465-472.89. Lekstrom-Himes JA, Straus SE. Varicella-zoster virus infections in the normal and immunocompromised host. In:

Clinical Management of Herpes Viruses. (Sacks SL, Straus SE, Whitley RJ et al, eds). Amsterdam: IOS Press, 1995:175-192.

90. Balfour HH, Jr. Intravenous acyclovir therapy for varicella in immunocompromised children. J Pediatr1984;104:134-136.

91. Shepp DH, Dandliker PS, Meyers JD. Treatment of varicella-zoster virus infection in severely immunocompromisedpatients. A randomized comparison of acyclovir and vidarabine. N Engl J Med 1986;314:208-212.

92. Vilde JL, Bricaire F, Leport C et al. Comparative trial of acyclovir and vidarabine in disseminated varicella-zostervirus infections in immunocompromised patients. J Med Virol 1986;20:127-134.

93. Whitley RJ, Gnann JW, Jr, Hinthorn D et al. Disseminated herpes zoster in the immunocompromised host: acomparative trial of acyclovir and vidarabine. The NIAID Collaborative Antiviral Study Group. J Infect Dis1992;165:450-455.

94. Whitley RJ. Therapeutic approaches to varicella-zoster virus infections. J Infect Dis 1992;166 (Suppl B):S51-S57.95. Carcao MD, Lau RC, Gupta A et al. Sequential use of intravenous and oral acyclovir in the therapy of varicella in

immunocompromised children. Pediatr Infect Dis J 1998;17:626-631.96. Hoglund M, Ljungman P, Weller S. Comparable aciclovir exposures produced by oral valaciclovir and intravenous

aciclovir in immunocompromised cancer patients. J Antimicrob Chemother 2002;47:855-856.97. Gnann JW, Jr., Crumpacker CS, Lalezari JP et al. Sorivudine versus acyclovir for treatment of dermatomal herpes

zoster in human immunodeficiency virus-infected patients: results from a randomized, controlled clinical trial.Collaborative Antiviral Study Group/AIDS Clinical Trials Group, Herpes Zoster Study Group. Antimicrob AgentsChemother 1998;42:1139-1145.

98. Sellitti TP, Huang AJ, Schiffman J et al. Association of herpes zoster ophthalmicus with acquired immunodeficiencysyndrome and acute retinal necrosis. Am J Ophthalmol 1993;116:297-301.

99. Margolis TP, Milner MS, Shama A et al. Herpes zoster ophthalmicus in patients with human immunodeficiency virusinfection. Am J Ophthalmol 1998;125:285-291.

100. Austin RB. Progressive outer retinal necrosis syndrome: a comprehensive review of its clinical presentation,relationship to immune system status, and management. Clin Eye Vis Care 2000;12:119-129.

101. Moorthy RS, Weinberg DV, Teich SA et al. Management of varicella zoster virus retinitis in AIDS. Br J Ophthalmol1997;81:189-194.

102. Meffert SA, Kertes PJ, Lim PL et al. Successful treatment of progressive outer retinal necrosis using high-doseintravitreal ganciclovir. Retina 1997;17:560-562.

103. Yang CM, Wang WW, Lin CP. Progressive outer retinal necrosis syndrome as an early manifestation of humanimmunodeficiency virus. J Formos Med Assoc 1999;98:141-144.

104. Ciulla TA, Rutledge BK, Morley MG et al. The progressive outer retinal necrosis syndrome: successful treatment withcombination antiviral therapy. Ophthalmic Surg Lasers 1998;29:198-206.

105. Perez-Blazquez E, Traspas R, Mendez Marin I et al. Intravitreal ganciclovir treatment in progressive outer retinalnecrosis. Am J Ophthalmol 1997;124:418-421.

106. Schliefer K, Gumbel HO, Rockstroh JK et al. Management of progressive outer retinal necrosis with cidofovir in ahuman immunodeficiency virus-infected patient. Clin Infect Dis 1999;29:684-685.

107. Snoeck R, Gerard M, Sadzot-Delvaux C et al. Meningoradiculoneuritis due to acyclovir-resistant varicella-zostervirus in a patient with AIDS. J Infect Dis 1993;168:1330-1331.

108. Breton G, Fillet AM, Katlama C et al. Acyclovir-resistant herpes zoster in human immunodeficiency virus-infectedpatients: results of foscarnet therapy. Clin Infect Dis 1998;27:1525-1527.

109. Visse B, Dumont B, Huraux JM et al. Single amino acid change in DNA polymerase is associated with foscarnetresistance in a varicella-zoster virus strain recovered from a patient with AIDS. J Infect Dis 1998;178 (Suppl 1):S55-S57.

110. Andrei G, Snoeck R, Reymen D et al. Comparative activity of selected antiviral compounds against clinical isolatesof varicella-zoster virus. Eur J Clin Microbiol Infect Dis 1995;14:318-329

99

Page 101: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Management RecommendationsFirst-line treatment of established post-herpetic neuralgiaCategory 1J Tricyclic antidepressants, such as amitriptyline or nortriptyline (initiated at 10–25 mg

and increasing weekly by 25 mg [10 mg if patient >65 years or frail]), relieve pain in

some patients. It is recommended that these treatments be initiated at a low dose for

acute herpes zoster-associated pain. If post-herpetic neuralgia (PHN) develops, the

dose should be increased gradually until pain control is achieved or side-effects

become intolerable.

J Gabapentin is recommended as an effective treatment for PHN and can be titrated

from 300–900 mg/day up to 3600 mg/day (provided there is unimpaired renal

function).

J Administration of a topical local anaesthetic (e.g. lidocaine patch 5%) is

recommended.

Category 3J Encouragement, counselling and education on quality-of-life matters should be given

to patients with established PHN, as should cold packs and advice on clothing and

lifestyle.

J Efforts should be made to encourage treatment compliance in patients with established

pain.

J Analgesia using paracetamol with or without a weak opioid is effective and

recommended in established pain, but there is no evidence to support the use of non-

steroidal anti-inflammatory drugs (NSAIDs) in the treatment of PHN.

Second-line treatment of established PHNCategory 1J Topical capsaicin (0.025% or 0.075%) may be effective in some patients and they

should receive advice on its use and side-effects to achieve compliance.

Category 2J Stronger opioids (including oxycodone, morphine and methadone) may be considered

for therapy of PHN, but, as they are usually administered in a pain clinic, cannot be

considered as first-line therapy.

J If a first-line tricyclic antidepressant fails, any of the available non-tricyclics may be

tried, such as venlafaxine or maprotiline. Selective serotonin reuptake inhibitors

(SSRIs) such as fluoxetine, paroxetine or sertraline may also be effective in some

patients.100

MANAGING ESTABLISHED PAIN IN HERPES ZOSTER9

Page 102: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Category 3J Transcutaneous electrical nerve stimulation (TENS) may be effective but evidence to

support this is scarce.

J With persistent pain, topical preparations, tricyclic antidepressants, gabapentin and

opioids may be effective if used in combination. It is recommended that this approach

be based upon the physician’s experience and the condition of the individual patient.

J Additional approaches to consider in established PHN include nerve blocks, and

topical aspirin in ether. Some patients may wish to consider complementary or

alternative medicine, such as homeopathy, faith healing or acupuncture, although

there is no evidence to support these approaches.

J Patients who fail to respond to several different treatment approaches despite

monitored compliance should be referred to a pain management clinic.

Research needsJ Cost-benefit analysis of different treatment modalities and management techniques

utilized in established PHN would be beneficial to assess the clinical impact and cost

of therapeutic alternatives.

J The development of effective pain management programmes for PHN patients,

especially elderly patients, is of utmost importance.

IntroductionJ Cost-benefit analysis of different treatment modalities and management techniques

utilized in established PHN would be beneficial to assess the clinical impact and cost

of therapeutic alternatives (research need recommendation).

J The development of effective pain management programmes for PHN patients,

especially elderly patients, is of utmost importance (research need recommendation).

PHN, the ongoing pain that follows the resolution of herpes zoster, can result in significant

morbidity that interferes with the activities of daily living.1 It can manifest as one or more

of:

• Spontaneous aching or burning or paroxysmal shooting pains

• Allodynia (pain evoked by application of a mild, normally non-noxious, stimulus – this

may be static or dynamic)

• Hyperalgesia (severe pain evoked by application of a normally mildly painful stimulus).

In addition to being severe, PHN is often prolonged. Approximately 22% of patients with

herpes zoster still have spontaneous and/or evoked pain 3 months after rash appearance.2

At 1 year, about 5–10% still have PHN and by this stage further spontaneous resolution is

limited.2 Such is the frequency of herpes zoster that at any one time, the estimated

prevalence of PHN in the UK is 200 000 cases.2 Thus, many patients will suffer moderate-

to-severe pain which can last for many years and in some cases for life.

Aciclovir and, by inference, other antivirals cannot treat established PHN.3,4 The ongoing

discomfort and distress caused to patients by established PHN highlight the need for

effective treatment and management. Work published on tricyclic antidepressants,

101

Page 103: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

opioids,5,6 anticonvulsants including gabapentin,7 topical lidocaine,8 and topical

capsaicin6 documents their varying degrees of effectiveness for established PHN. With the

increasing number of therapy options available, successful PHN treatment is likely to be

be multimodal.

First-Line Treatment of Established PHNPhysical measuresJ Encouragement, counselling and education on quality-of-life matters should be given

to patients with established PHN, as should cold packs and advice on clothing and

lifestyle (category 3 recommendation).

J Efforts should be made to encourage treatment compliance in patients with established

pain (category 3 recommendation).

Patients with established PHN should receive encouragement, counselling and education

on quality-of-life matters. When discussing treatments for PHN, the importance of

compliance should be emphasized.

Physical measures can offer benefit to the patient with PHN.9 Cold packs can give

temporary relief of symptoms in patients who do not have cold allodynia. They must be

applied frequently and with a layer such as a cotton towel between the pack and the skin.

A protective layer, such as plastic ‘artificial skin’ sprays between the skin and clothing may

reduce tactile allodynia from the rubbing of clothes.

Analgesic therapy for established PHNJ Analgesia using paracetamol, with or without a weak opioid, is effective and

recommended in established pain, but there is no evidence to support the use of

NSAIDs in PHN (category 3 recommendation).

Paracetamol is well tolerated and is a suitable first-line analgesic in PHN.10 When pain

relief is insufficient with paracetamol alone, codeine may be added as their combination

produces a significant increase in analgesia.11

Although it has been argued for a number of years that neuropathic pain is unresponsive

to opiates, non-blinded studies suggest that patients with PHN may obtain significant pain

relief from opioid analgesics, leading to the wider use of these agents in the treatment of

PHN.5,12,13 The higher potency opioid analgesics (e.g. morphine) are probably best

administered in a multidisciplinary pain clinic, as monitoring and dose titration against

pain relief are essential. If opioids do not provide significant analgesia, they should be

discontinued to prevent dependence.

Oxycodone may provide significant relief from PHN. A cross-over trial compared

controlled-release oxycodone with placebo in 50 patients with PHN of at least moderate

intensity for an average of 31 months. The initial dose of oxycodone was 10 mg every

12 hours increasing to a maximum of 30 mg every 12 hours over 4 weeks. The average

dose in the last week was 45 mg per day. Patients treated with oxycodone exhibited

significantly greater pain relief (P<0.0001), and reduction of allodynia (P=0.0004)

compared with those receiving placebo.6 This study provides justification for opioid

administration in patients with established PHN. However, the side-effects of opioids,102

Page 104: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

which include respiratory depression, constipation, sedation, nausea and vomiting, may

be problematic. A further side-effect of concern is delirium which may be a barrier to the

use of this class of drug.

Another analgesic, tramadol, a weak opioid m-receptor agonist and monoamine re-uptake

inhibitor, has shown potential in the treatment of established PHN. In a pilot study, nine

out of 10 patients reported tramadol as providing pain relief described as satisfactory or

very satisfactory.14,15 However, tramadol should not be co-administered with

antidepressants because of the risk of serotonin syndrome.

Tricyclic antidepressants in PHNJ Tricyclic antidepressants, such as amitriptyline or nortriptyline (initiated at 10–25 mg

and increasing weekly by 25 mg [10 mg if patient >65 years or frail]), relieve pain in

some patients. The dose should be increased gradually until pain control is achieved

or side-effects become intolerable (category 1 recommendation).

The place of tricyclic antidepressants in

the first-line treatment of PHN has been

established in a number of clinical

trials.16-22 These demonstrate an

analgesic effect on chronic neuropathic

pain that is independent of their

antidepressant action (Table 1), and an

effect on sodium channels in primary

afferent fibres. In reviews of treatments

for PHN, Volmink et al23 and McQuay

et al24 concluded that both amitriptyline

and desipramine are effective at relieving pain in PHN.13,23-25 The starting dose of

amitriptyline should be 25 mg (10 mg in frail patients) as a single night-time dose.26 The

dose should be increased by 25 mg at weekly intervals until either it achieves pain relief or

adverse effects become problematic. The maximum dose is 150 mg. Patients should be

warned to expect a dry mouth and drowsiness.

Although amitriptyline has become the most widely used antidepressant for the treatment

of PHN, it is associated with a large number of adverse effects, especially in the elderly

(e.g. anticholinergic side-effects and orthostatic hypotension).13,27 To address this, Watson

and colleagues22 performed a double-blind crossover trial comparing amitriptyline with

nortriptyline in 31 patients with PHN. There were no differences between the two

antidepressants with regard to relief of skin pain, mood, disability, satisfaction, or

preference between the two drugs. However, intolerable side-effects were more common

with amitriptyline. Thus, nortriptyline is a suitable alternative, especially for those who

may experience side-effects with the latter.

Anticonvulsants in PHNJ Gabapentin is recommended as an effective treatment for PHN and can be titrated

from 300–900 mg/day up to 3600 mg/day (provided there is unimpaired renal

function) (category 1 recommendation).103

TABLE 1: Summary ofcontrolled trials ofantidepressant therapiesin PHN6,17–22

Treatment Patients (n) Positive pain relief reported (%)

Amitriptyline17 24 67 Placebo 5 Amitriptyline18 34 47 Placebo 8 Desipramine19 19 63 Placebo 11 11 Amitriptyline20 15 60 Zimelidine 7 Amitriptyline21 32 44 Maprotiline 18 Amitriptyline22 31 58 Nortriptyline 55

Page 105: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

Evidence for the benefit of anticonvulsants in PHN was lacking until the publication of a

report by Rowbotham and colleagues5 of a large-scale clinical trial evaluating the use of

gabapentin in this indication. This randomized, double-blind study comparing gabapentin

(titrated to a maximum dose of 3600 mg daily) with placebo in 229 patients with PHN7 had

a primary efficacy outcome of change in average daily pain score, based upon an

11-point Likert pain scale (0=no pain, 10=worst possible pain). Patients receiving

gabapentin exhibited a significant reduction in average daily pain score from 6.3 to 4.2

compared with 6.5 to 6.0 for the placebo group (P<0.001).7 Gabapentin was found to be

effective not only in pain control but also in the treatment of sleep interference associated

with PHN, which in turn improved mood and quality of life. Gabapentin was associated with

somnolence, dizziness, ataxia, peripheral oedema, and infection. However, the number of

withdrawals due to side-effects was similar to that seen with placebo. Similar results were

seen in another multicentre, randomized, double-blind, placebo-controlled trial involving

334 patients with PHN.28 Patients who received gabapentin (either 1800 or 2400 mg/day

in three divided doses) showed significantly greater improvements in pain scores and

measures of sleep quality than those who received placebo.28

Gabapentin has not been directly compared with other treatments for PHN. However, its

efficacy has been compared with tricyclic antidepressants using the ‘number needed to

treat‘ (NNT) method.24 The comparison used data from the gabapentin trials and from a

review of tricyclic antidepressants. The analysis indicated that gabapentin was as effective

and safe as tricyclic antidepressants but with fewer contraindications to its use. The

placebo-controlled study, therefore, supports the use of gabapentin as a first-line therapy

for the treatment of the pain of established PHN. The FDA in the USA has also approved

gabapentin for use in the treatment of PHN.

Other anticonvulsants have been less well studied in PHN. For example, lamotrigine

reduced hyperalgesia and allodynia in experimental studies,29 and anecdotal reports

indicate that lamotrigine was effective in neuropathic pain, including central pain, which

is notoriously difficult to treat.29 However, this effect occurred at comparatively high doses

(up to 600 mg/day). Side-effects of lamotrigine are similar to those of other

anticonvulsants, but dermatological complications may be more common and more

severe.30 The risk of developing adverse effects can be reduced by prescribing a low initial

dose (25–50 mg/day) and increasing the dose slowly. Carbamazepine was ineffective at

relieving continuous pain in an uncontrolled study.31

Lidocaine patchJ Administration of a topical local anaesthetic (e.g. lidocaine patch 5%) is

recommended (category 1 recommendation).

Topical lidocaine has been shown to provide effective pain relief in established PHN.

There have been a number of double-blind, vehicle-controlled studies on the efficacy of

topical lidocaine in the treatment of PHN.8,32,33 In these studies, 5% lidocaine, either as a

gel or as a non-woven patch, provided moderate or better pain relief in more than half of

patients compared with vehicle controls.8,32,33 In the most recent of these studies, the

lidocaine patch gave pain relief in patients with established PHN. In the trial,33 the

lidocaine patch provided greater pain relief than the vehicle control, with patients

receiving the lidocaine patch remaining on treatment 3.7-fold longer than on the vehicle104

Page 106: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

patch. All of the patients in the study had previously been successfully treated with topical

lidocaine. Upon completion of the study, 78.1% (25/32) of the lidocaine-patch group

preferred the treatment patch to previous treatment they had received, compared with

9.4% (3/32) of the control group (P<0.001).32

There is minimal systemic uptake and no need for dose escalation with use of the patch

which, combined with the rapid onset of relief, makes the lidocaine patch a useful option

for relieving the pain of PHN. Lidocaine patches and gabapentin are the only treatments

with a specific approval for the treatment of PHN by the FDA in the USA.

In countries where lidocaine is not available, EMLA cream under an occlusive dressing

may be effective.34

Second-Line Treatments for Established PHNAnalgesic therapyJ Stronger opioids (including oxycodone, morphine and methadone) may be considered

for therapy of PHN, but, as they are usually administered in a pain clinic, cannot be

considered as first-line therapy (category 2 recommendation).

Alternative tricyclic antidepressantsJ If a first-line tricyclic antidepressant fails, any of the available non-tricyclics may be

tried, such as venlafaxine or maprotiline. SSRIs such as fluoxetine, paroxetine or

sertraline may also be effective in some patients (category 2 recommendation).

Amitriptyline may cause significant anticholinergic side-effects in the elderly or it may fail

to offer sufficient pain relief. Similarly, patients may experience unacceptable sedation

from nortriptyline. Thus, an alternative tricyclic antidepressant, such as desipramine, may

be considered.35 In a randomized, double blind, placebo-controlled trial, 6 weeks of

desipramine treatment (mean dose 167 mg/day) resulted in significant improvements in

PHN.17 Furthermore, desipramine has a less toxic side-effect profile than amitriptyline,

especially with respect to anticholinergic effects.27

Maprotiline is also thought to have fewer side-effects than amitriptyline;36 both have been

compared in the treatment of established PHN in a double-blind, randomized trial.21

Maprotiline relieved PHN (steady pain, brief pain and pain on skin contact) but was not

as effective as amitryptiline. Moreover, more patients receiving maprotiline experienced

side-effects, many of which were more severe, than those in the amitryptiline group.

Therefore, it is recommended that amitryptiline should be the first choice of tricyclic

antidepressant to treat PHN, but, if it fails agents such as maprotiline should be used.29

If a patient tolerates a full dose of the first tricyclic antidepressant over a trial period of

2–4 weeks, there is little justification for trying a second tricyclic antidepressant, except as

third-line therapy. If a first-line tricyclic antidepressant is not tolerated, any of the available

non-tricyclics may be tried, such as venlafaxine or maprotiline. SSRIs such as fluoxetine,

paroxetine or sertraline may also be effective in some patients and can be considered as

second-line treatments.

105

Page 107: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

CapsaicinJ Topical capsaicin (0.025% or 0.075%) may be effective in some patients and they

should receive advice on its use and side-effects to achieve compliance (category 1

recommendation).

The place of topical capsaicin is still not completely resolved. Capsaicin (N-Vanillyl-8-

methyl-6-(E)-noneamide) is a pungent derivative of hot chilli peppers. Its application to

sensory nerve tissue first stimulates then inhibits (by causing depletion of the

neurotransmitter substance P) activity in nociceptive C fibres. Topical applications of

concentrations varying from 0.025% to 0.075% have been studied in a number of painful

conditions including osteoarthritis of the knee and PHN. A 0.075% preparation of

capsaicin has been reported to provide a significant benefit in the treatment of PHN.23 In

contrast, McQuay and Moore25 reviewed its use and concluded that there was no

evidence of significant improvement in pain relief following capsaicin treatment. These

disparate results are likely to be due to the problem of placebo-blinding in controlled

studies of capsaicin, because of the burning sensation associated with the active treatment.

Topical capsaicin is not a first-line choice for the treatment of patients with PHN. For

many, there is a lack of compliance due to the burning sensation experienced following

application.13

Other Treatment Approaches for Established PHNJ With persistent pain, topical preparations, tricyclic antidepressants, gabapentin and

opioids may be effective if used in combination. It is recommended that this approach

be based upon the physician’s experience and the condition of the individual patient

(category 3 recommendation).

J Patients who fail to respond to several different treatment approaches despite

monitored compliance should be referred to a pain management clinic (category 3

recommendation).

J Additional approaches to consider in established PHN include nerve blocks, and

topical aspirin in ether. Some patients may wish to consider complementary or

alternative medicine, such as homeopathy, faith healing or acupuncture, although

there is no evidence for these approaches (category 3 recommendation).

Sympathetic nerve blocksAlthough sympathetic nerve blocks may be effective in relieving acute zoster-associated

pain, they do not appear to provide prolonged pain relief in patients with established

PHN.7,37 Studies by Colding38 and Nurmikko et al39 have demonstrated little or no benefit

from the use of sympathetic nerve blocks in the treatment of PHN. This lack of efficacy may

be explained in part by evidence which suggests only a limited role for the sympathetic

nervous system in the pain and allodynia experienced in PHN.39,40

In contrast, intrathecal administration of methylprednisolone was found to be effective for

the treatment of PHN in a study of 270 patients who had had PHN for at least 1 year; the

population was restricted to those patients with long-lasting pain that was resistant to

conventional treatments.41 Patients were randomized to one of three groups:106

Page 108: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

• Methylprednisolone and lidocaine (3 ml of 3% lidocaine with 60 mg methylprenisolone)

• Lidocaine alone

• No treatment.

Treatment was once weekly for 4 weeks. Pain was evaluated at the end of the 4-week study

period, then at 4 weeks, 1 year and 2 years later. At the end of the study, patients in the

methylprednisolone–lidocaine group had a decrease in the pain area and intensity, and

their use of diclofenac was 70% lower than at baseline, suggesting that this treatment may

be of benefit in PHN.41 However, this study has received much criticism because of the

perceived serious risk of intrathecal methylprednisolone administration. Until

confirmation from properly-conducted studies is available, this treatment cannot be

advised.

TENSJ TENS may be effective but evidence to support its efficacy is scarce (category 3

recommendation).

Evidence for the efficacy of TENS is scarce but it may be beneficial in some patients and

has little risk.42,43 However, TENS should be part of a larger management programme that

includes other modalities.44

N-methyl-D-aspartate (NMDA) receptor antagonistsThere have been no controlled trials of the N-methyl-D-aspartate (NMDA) receptor

antagonist ketamine in PHN but evidence that it may be effective for reducing pain comes

from a non-comparative trial in five patients.45 Continuous subcutaneous infusion of

ketamine was effective in relieving the spontaneous pain and allodynia of PHN. However,

itching and painful indurations at the injection site were common and other side-effects

included nausea, fatigue and dizziness. Another NMDA antagonist, dextramethorphan

(mean dose 439 mg/day), did not reduce pain in 13 patients with established PHN when

compared with placebo, although the antagonist was effective in diabetic neuropathy in

the same trial.46

Topical aspirin in etherThe evidence that other treatments can ameliorate established PHN is often poor. There

has been a limited number of studies on aspirin suspended in ether, chloroform or

acetone.47-49 Although these therapies can help some patients, there are doubts about the

extent of clinical benefit, especially as the trials have been of short duration. There are also

concerns about the safety of the mixtures.

Alternative and complementary medicineAcupuncture was shown to have no effect on pain resolution in a placebo-controlled trial

involving 62 individuals; patients in both treatment groups experienced similar levels of

pain relief.50 Faith healing may offer benefit to some patients but this non-invasive

approach has not been tested in formal studies.

SummaryPHN can result in significant morbidity and interfere with patients’ activities of daily living.

It can manifest as spontaneous pain (aching or burning in nature), allodynia or 107

Page 109: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

hyperalgesia. Not only is PHN potentially severe, it is often prolonged with some 5–10%

of patients still experiencing pain at 1 year after rash appearance. This ongoing discomfort

and distress emphasize the need for effective management and treatment.

First-line therapy for PHN may include analgesics, such as paracetamol or a weak opioid,

and tricyclic antidepressants, such as amitriptyline or nortriptyline, initiated at 10–25 mg

and increasing weekly by 25 mg. Gabapentin (up to 3600 mg per day provided there is

unimpaired renal function) is also recommended as an effective first-line treatment for

PHN. However, other anticonvulsants have been less well studied for the treatment of

PHN. Physical measures such as cold packs have been shown to offer pain relief to some

patients. In addition, patients with established PHN should receive encouragement,

counselling, and education on quality-of-life matters and the importance of treatment

compliance.

If a first-line tricyclic antidepressant fails to provide pain relief, or the patient experiences

unacceptable side-effects, treatment with any of the other available tricyclics should be

considered. Other first-line treatments that have been shown to be of use in PHN include

topical lidocaine patches or capsaicin and stronger opioid analgesics such as oxycodone

or tramadol.

Alternative approaches to consider in the second-line treatment of PHN that does not

respond to the therapies described above include nerve blocks, homeopathy, topical

aspirin in ether, faith healing, acupuncture, TENS and referral to a pain management clinic.

In summary, effective treatment of established PHN is likely to be multimodal and may involve:

• Tricyclic antidepressants (amitriptyline initiated at 10–25 mg)

• Anticonvulsants (gabapentin titrated from 300–900 mg daily up to 3600 mg every

24 hours provided there is unimpaired renal function)

• Opioids (oxycodone, morphine and methadone)

• Topical agents (lidocaine or capsaicin).

References1. Choo PW, Galil K, Donahue JG et al. Risk factors for postherpetic neuralgia. Arch Intern Med

1997;157:1217-1224.2. Bowsher D. The lifetime occurrence of herpes zoster and prevalence of post-herpetic neuralgia:

A retrospective survey in an elderly population. Eur J Pain 1999;3:335-342.3. Acosta EP, Balfour HH, Jr. Acyclovir for treatment of postherpetic neuralgia: efficacy and pharmacokinetics.

Antimicrob Agents Chemother 2001;45:2771-2774.4. Surman OS, Flynn T, Schooley RT et al. A double-blind, placebo-controlled study of oral acyclovir in

postherpetic neuralgia. Psychosomatics 1990;31:287-292.5. Rowbotham MC, Reisner-Keller LA, Fields HL. Both intravenous lidocaine and morphine reduce the pain of

postherpetic neuralgia. Neurology 1991;41:1024-1028.6. Watson CP, Babul N. Efficacy of oxycodone in neuropathic pain: a randomized trial in postherpetic

neuralgia. Neurology 1998;50:1837-1841.7. Rowbotham M, Harden N, Stacey B et al. Gabapentin for the treatment of postherpetic neuralgia: a

randomized controlled trial. JAMA 1998;280:1837-1842.8. Rowbotham MC, Davies PS, Verkempinck C et al. Lidocaine patch: double-blind controlled study of a new

treatment method for post-herpetic neuralgia. Pain 1996;65:39-44.9. Johnson RW. Herpes zoster and postherpetic neuralgia. Optimal treatment. Drugs Aging 1997;10:80-94.10. Lancaster T, Silagy C, Gray S. Primary care management of acute herpes zoster: systematic review of

evidence from randomized controlled trials. Br J Gen Pract 1995;45:39-45.11. de Craen AJ, Di Giulio G, Lampe-Schoenmaeckers JE et al. Analgesic efficacy and safety of paracetamol-

codeine combinations versus paracetamol alone: a systematic review. BMJ 1996;313:321-325.12. Pappagallo M, Campbell JN. Chronic opioid therapy as alternative treatment for post-herpetic neuralgia.

Ann Neurol 1994;35:S54-56.

108

Page 110: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

13. Kanazi GE, Johnson RW, Dworkin RH. Treatment of postherpetic neuralgia: an update. Drugs 2000;59:1113-1126.

14. Raffa RB, Friderichs E, Reimann W et al. Opioid and nonopioid components independently contribute to themechanism of action of tramadol, an ‘atypical’ opioid analgesic. J Pharmacol Exp Ther 1992;260:275-285.

15. Gobel H, Stadler T. Treatment of post-herpes zoster pain with tramadol. Results of an open pilot study versusclomipramine with or without levomepromazine. Drugs 1997;53 (Suppl 2):S34-S39.

16. Max MB. Thirteen consecutive well-designed randomized trials show that antidepressants reduce pain indiabetic neuropathy and post-herpetic neuralgia. Pain Forum 1995;4:248-253.

17. Watson CPN, Evans RJ, Reed K. Amitriptyline versus placebo in postherpetic neuralgia. Neurology1982;32:671-673.

18. Max MB, Schafer SC, Culnane M. Amitriptyline, but not Iorazepam, relives postherpetic neuralgia.Neurology 1988;38:1427-1432.

19. Kishore-Kumar R, Max MB, Schafer SC. Desipramine relieves postherpetic neuralgia. Clin Pharmacol Ther1990;47:305-312.

20. Watson CPN, Evans RJ. A comparative trial of amitriptyline and zimelidine in post-herpetic neuralgia. Pain1985;23:387-394.

21. Watson CPN, Chipman M, Reed K. Amitriptyline versus maprotiline in postherpetic neuralgia: Arandomized, double-blind, crossover trial. Pain 1992;48:29-36.

22. Watson CP, Verusch L, Chapman M et al. Nortriptyline versus amitriptyline in post herpetic neuralgia: arandomised trial. Neurology 1998;51(4):1166-1171.

23. Volmink J, Lancaster T, Gray S et al. Treatments for postherpetic neuralgia – a systematic review ofrandomized controlled trials. Fam Pract 1996;13:84-91.

24. McQuay HJ, Tramer M, Nye BA et al. A systematic review of antidepressants in neuropathic pain. Pain1998;75:160-161.

25. McQuay HJ, Moore RA. An evidence-based resource for pain relief. Oxford: Oxford University Press, 1998.26. McQuay HJ, Moore RA. Antidepressants and chronic pain. BMJ 1997;314:763-764.27. McCue RE. Using tricyclic antidepressants in the elderly. Clin Geriatr Med 1992;8:323-334.28. Rice AS, Maton S et al. Gabapentin in postherpetic neuralgia: a randomised, double-blind, placebo

controlled study. Pain 2001;94:215-224.29. Nakamura-Craig M, Follenfant RL. Effect of lamotrigine in the acute and chronic hyperalgesia induced by

PGE2 and in the chronic hyperalgesia in rats with streptozotocin-induced diabetes. Pain 1995;63:33-37.30. Nurmikko TJ, Nash TP, Wiles JR. Recent advances: control of chronic pain. BMJ 1998;317:1438-1441.31. Killian JM, Fromm GH. Carbamazepine in the treatment of neuralgia. Use of side effects. Arch Neurol

1968;19:129-136.32. Rowbotham MC, Davies PS, Fields HL. Topical lidocaine gel relieves postherpetic neuralgia. Ann Neurol

1995;37:246-253.33. Galer BS, Rowbotham MC, Perander J et al. Topical lidocaine patch relieves postherpetic neuralgia more

effectively than a vehicle topical patch: results of an enriched enrollment study. Pain 1999;80:533-538.34. Stow PJ, Glynn CJ, Minor B. EMLA cream in the treatment of post-herpetic neuralgia. Efficacy and

pharmacokinetic profile. Pain 1989;39:301-305.35. Schmader K. Postherpetic neuralgia in immunocompetent elderly people. Vaccine 1998;16:1768-1770.36. Coccaro EF, Siever LJ. Second generation antidepressants: a comparative review. J Clin Pharmacol

1985;25:241-260.37. Dworkin RH. Prevention of postherpetic neuralgia. Lancet 1999;353:1636-1637.38. Colding A. The effect of regional sympathetic blocks in the treatment of herpes zoster. Acta Anaesthesiol

Scand 1969;13:133-141.39. Nurmikko T, Wells C, Bowsher D. Pain and allodynia in postherpetic neuralgia: role of somatic and

sympathetic nervous systems. Acta Neurol Scand 1991;84:146-152.40. Rowbotham MC, Fields HL. The relationship of pain, allodynia and thermal sensation in post-herpetic

neuralgia. Brain 1996;119:347-354.41. Kotani N, Kushikata T, Hashimoto H et al. Intrathecal methylprednisolone for intractable postherpetic

neuralgia. N Engl J Med 2000;343:1514-1519.42. Nathan PW, Wall PD. Treatment of post-herpetic neuralgia by prolonged electric stimulation. BMJ

1974;3:645-647.43. Yamashiro H, Hara K, Gotoh Y. [Relief of intractable post-herpetic neuralgia with gasserian ganglion block

using methyl prednisolone acetate and with TENS]. Masui 1990;39:1239-1244.44. Thorsteinsson G. Chronic pain: use of TENS in the elderly. Geriatrics 1987;42:75-77, 81-72.45. Eide K, Stubhaug A, Oye I et al. Continuous subcutaneous administration of the N-methyl-D-aspartic acid

(NMDA) receptor antagonist ketamine in the treatment of post-herpetic neuralgia. Pain 1995;61:221-228.46. Nelson KA, Park KM, Robinovitz E et al. High-dose oral dextromethorphan versus placebo in painful

diabetic neuropathy and postherpetic neuralgia. Neurology 1997;48:1212-1218.47. King RB. Topical aspirin in chloroform and the relief of pain due to herpes zoster and postherpetic neuralgia.

Arch Neurol 1993;50:1046-1053.48. King RB. Concerning the management of pain associated with herpes zoster and of postherpetic neuralgia.

Pain 1988;33:73-78.49. De Benedittis G, Lorenzetti A. Topical aspirin/diethyl ether mixture versus indomethacin and

diclofenac/diethyl ether mixtures for acute herpetic neuralgia and postherpetic neuralgia: a double-blindcrossover placebo-controlled study. Pain 1996;65:45-51.

50. Lewith GT, Field J, Machin D. Acupuncture compared with placebo in post-herpetic pain. Pain1983;17:361-368.

109

Page 111: IHMF 2002 - Improving the Management of Varicella, Herpes Zoster & Zoster-Associated Pain

ISBN 0 904052 74 5HE6251/05.00/11.0/PAREXEL MMS