Download Cytomegalovirus, Human Herpesvirus-6, and Human Herpesvirus

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Myelodysplastic syndrome wikipedia , lookup

Transcript
Cytomegalovirus, Human Herpesvirus-6, and Human
Herpesvirus-7 in Hematological Patients
Duncan A. Clark, Vincent C. Emery, and Paul D. Griffiths
The prototype member of the Betaherpesvirinae subfamily, cytomegalovirus (CMV), is the most important infectious
pathogen in transplant recipients, including those receiving bone marrow or stem cell grafts. Overt CMV disease such
as pneumonitis is notoriously difficult to treat. Antiviral prophylaxis, rapid diagnostic tests to identify CMV infection,
and preemptive antiviral chemotherapy are significant improvements in the management of CMV. As the kinetics of the
immune response to CMV become better defined, immunotherapeutic approaches should be introduced to complement
current management strategies. Two newly identified betaherpesviruses, human herpesvirus-6 (HHV-6) and human
herpesvirus-7 (HHV-7), are genetically more closely related to each other than to CMV. Both are highly prevalent in the
general population and infections post– bone marrow transplantation are common. These viruses are not as pathogenic
as CMV but HHV-6 at least can cause disease such as encephalitis, hepatitis, and bone marrow suppression. Both of
these newer herpesviruses are potentially susceptible to existing and licensed antiherpesvirus drugs.
Semin Hematol 40:154-162. © 2003 Elsevier Inc. All rights reserved.
T
HE BETAHERPESVIRUS subfamily includes cytomegalovirus (CMV) and the closely related
human herpesviruses-6 and -7 (HHV-6 and HHV-7).
CMV is well known as the most important infectious
pathogen in transplant recipients, and there is now
accumulating evidence that HHV-6 and HHV-7 also
exhibit enhanced pathogenicity in the immunocompromised host. This review will consider the relevance of infection with these viruses in hematology
patients.
Cytomegalovirus
CMV infects approximately 60% of adults in developed countries and almost all of those in developing
nations. The virus is usually acquired by close contact
between individuals, mainly by the transfer of saliva
but also through intrauterine or perinatal infection.
In addition, iatrogenic transmission can occur, as
from donated solid organs and through blood transfusion.78 Primary infections are usually asymptomatic although delayed infection in adulthood can
cause infectious mononucleosis–like illness.
CMV establishes life-long infection after primary
infection. Persistence likely includes both a latent
state with infectious virus only produced during episodes of reactivation, and chronic replication with
continuous or frequent but intermittent production
From the Department of Virology, Royal Free and University
College Medical School of UCL, London, UK.
Address correspondence to Duncan A. Clark, PhD, Department of
Virology, Royal Free and University College Medical School, Royal
Free Campus, Rowland Hill Street, London NW3 2PF, UK.
© 2003 Elsevier Inc. All rights reserved.
0037-1963/03/4002-0006$30.00/0
doi:10.1053/shem.2003.50015
154
of infectious virus. Monocyte/macrophages and endothelial cells are implicated as sites of CMV persistence and latency, and replication of the virus in these
two cell types is likely to be important in maintaining
life-long infection.42,82,87
CMV causes a variety of diseases in the immunocompromised host (Table 1), although the anatomical sites most commonly affected vary among patient
groups. In addition to these direct end-organ diseases, CMV has repeatedly been associated with rejection of solid organs or graft-versus-host disease
(GvHD) in bone marrow transplantation. Secondary
bacterial and fungal infections also are frequent after
CMV infection and, together with rejection and
GvHD, are considered indirect effects of the virus.79
CMV Infection Following Bone Marrow
Transplantation
Historically, CMV has been one of the most feared
infectious complications following bone marrow
transplantation. CMV disease in this setting can manifest as pneumonitis, gastrointestinal syndromes,
hepatitis, bone marrow suppression, and occasionally retinitis (Table 1). The lungs of the recipients
become a site for CMV replication, and CMV pneumonitis was one of the major diseases following
transplantation which, once established, led to high
morbidity and mortality. Prior to the advent of more
rapid diagnostic methods, the slow growth of CMV in
vitro allowed a diagnosis of active CMV infection
only late in the clinical course. Under these conditions, antiviral therapeutic intervention was of limited success.74 As discussed below, in more recent
years antiviral prophylaxis36 and/or improved diagnostic methods and correct deployment of antiviral
chemotherapy against CMV22 have dramatically re-
Seminars in Hematology, Vol 40, No 2 (April), 2003: pp 154-162
Betaherpesviruses in Hematological Patients
Table 1. CMV Diseases in Transplant Recipients
Symptoms
Solid Transplant
Bone Marrow Transplant
Fever/hepatitis
Gastrointestinal
Retinitis
Pneumonitis
Immunosuppression
Myelosuppression
Rejection, GvHD
⫹⫹
⫹
⫹
⫹
⫹
⫹
⫹
⫹
⫹⫹
⫹
⫹⫹
?
Adapted from Griffiths et al,34 by permission of Oxford University
Press.
duced the incidence of CMV pneumonitis in hematology patients and also led to improved outcomes in
infected patients.
CMV Replication and Viral Burden
CMV has the reputation of a slowly growing virus.
While true in conventional in vitro culture systems in
primary human fibroblasts, it is unlikely that such an
artificial system parallels in vivo complexity. The
recent application of dynamic mathematical modeling has illuminated our understanding of the replication rate of many human pathogens in vivo. Using
such analyses, we have shown that CMV replication
in vivo occurs rapidly, with doubling times of approximately 1 day.23 These findings have implications for the development of disease and the successful treatment of infection in patients with
hematological malignancies undergoing bone marrow transplantation.
Many risk factors for CMV disease have been defined in prospective studies spanning 20 years. Using
conventional cell culture methods and subsequently
rapid culture methods such as shell vial culture and
the detection of early antigen fluorescent foci
(DEAFF) test, active CMV replication in blood (viremia) was associated with an increased risk of development of CMV disease post– bone marrow transplantation.46,64,93 Subsequently, the detection of
virus using more sensitive methods such as antigenemia assays (where polymorphonuclear cells expressing the ppUL83 protein of CMV are identified
using specific monoclonal antibodies) and the polymerase chain reaction (PCR) have shown that both
CMV presence and the quantity of virus found
in the blood are risk factors for CMV disease.4,7,21,25,31,52,73,86
The traditional major predictor for a high risk of
CMV disease has been the transplantation of donor
marrow from CMV-seronegative individuals into a
recipient who is already CMV-seropositive. Under
such conditions, the donor marrow is naive to CMV,
and when endogenous CMV is reactivated the marrow-derived immune cells mount a primary immune
155
response to control replication. In contrast, donor
and recipient seropositivity for CMV is associated
with a lower risk of CMV disease, probably due
to the transfer of antiviral immunity with donor marrow.35 These qualitative effects of donorrecipient serostatus manifest themselves quantitatively through viral load, such that patients in the
D⫺R⫹ category exhibit higher viral loads (and hence
a higher probability of disease) post-transplantation
compared to patients who are in the D⫹R⫹ or D⫹R⫺
categories (Fig 1).31 Regression of the probability of
CMV disease with viral load showed an acute transition at critical quantities of virus, resulting in a large
increase in disease risk for relatively small increases
in viral load. As a consequence, viral load levels can
be defined that identify patients at risk of disease with
reasonable sensitivity and specificity. The advent of
real-time PCR monitoring of viral load is allowing
these concepts to be translated practically, to identify
patients at risk of future CMV disease and to initiate
therapy in a timely, cost-effective manner.60,67,73,86
Immune Control of CMV Following Bone
Marrow Transplantation
T cells are critical in the control of CMV replication.
Extensive research has shown that the cytotoxic T
lymphocyte (CTL) response to CMV is dominated by
activity against the pp65 (ppUL83) tegument protein, with contributions from T cells directed against
the immediate early 1 protein among others.95 In the
immunocompetent host, CD8 T cells specific for
ppUL83 can be visualized using human leukocyte
Figure 1. CMV peak virus load and its association with CMV
serostatus at the time of bone marrow transplantation. (F) Patients who experienced CMV disease; (E) asymptomatic patients.
Median viral load in the R⫹D⫺ group was significantly higher than
in the R⫹D⫹ group. Adapted with permission.31
156
Clark, Emery, and Griffiths
antigen (HLA) tetramer technology (⬃1%),29,37 and
in bone marrow transplant recipients undergoing
high-level CMV replication, frequencies as high as
20% of the total CD8⫹ T-cell population have been
observed.3,32 Using a series of phenotypic markers,
these CD8 T cells have a late effector phenotype
(CD45RO⫹, CD27⫺, CCR7⫺, or CD45RA⫹, CD27⫺,
CCR7⫺) and are therefore fully differentiated cytotoxic T cells.1,26 A large proportion of CMV-specific
CD8 T cells reside in the CD45RA population traditionally regarded as naive T cells. For CD4 T-cell
helper responses, there is a predominance in responses against glycoprotein B (gB, UL55; a major
target for neutralizing antibody response) and
ppUL83, although individual epitopes restricted by
HLA class II alleles have not been as extensively
mapped as have HLA class I–restricted peptides.5,53
Nevertheless, in immunocompetent individuals the
T-helper responses against total CMV antigens also
are relatively high and appear to be important to
maintain CMV-specific CD8 T cells.26
In the context of bone marrow transplant, the
engrafting marrow must cope with a range of infectious agents experienced by the stem cell recipient.
CD4 T-helper cells regenerate relatively slowly following allogeneic bone marrow transplantation, and
so appropriate help to CD8 T cells able to control
CMV replication may be limited. The importance of
the regeneration of T-cell immunity after bone marrow transplantation for the control of CMV replication has been demonstrated in studies of CMV-specific T cells and by adoptive transfer of CMV-specific
T cells.3,20,75 HLA tetramer technology50 has improved our understanding of the kinetics of regeneration of CD8-specific CTL responses against CMV
following transplantation and provided insight into
the level of CD8 T cells required to ensure that CMV
infection is maintained at very low levels. An absolute
CMV CD8 T-cell level of approximately 108 cells/L
was associated with total suppression of CMV replication, at least at the level of sensitivity of the PCR
assay employed (⬃200 genomes/mL blood).3 However, in patients who experienced a period of active
CMV infection (PCR positivity ⬎ 200 genomes/mL
blood), the median CMV CD8 T-cell frequency was
substantially lower, at approximately 107 cells/L,
whereas during periods where replication was suppressed, the median frequency increased to about 3 ⫻
107 cells/L. From these and other data it would appear that certain threshold levels of CMV CD8 T cells
may be required to effectively suppress CMV replication; when the absolute number of cells is below this
threshold, high levels of CMV replication ensue,
leading to a high risk of disease.
A variety of mechanisms might render the CMV
CD8 T-cell population suboptimal for control of viral
replication, including slow reconstitution of T-helper
Figure 2. CMV immunity in a patient following sibling donor
peripheral blood stem cell transplantation (CMV serostatus D⫺/
R⫹). In the top panel are simultaneously represented the levels of
CMV peptide AE42 tetramer-specific (Tet⫹) T cells expressed as an
absolute number per liter of blood (䢇——䢇, unbroken line) or as a
percentage of CD8⫹ T cells (} – – – }). The periods of prednisolone,
ganciclovir (G), and foscarnet (F) therapy are also shown. The CMV
viral load is shown in the lower panel (䢇——䢇, unbroken line).
Adapted with permission of the University of Chicago Press.3
responses in the post-transplant period and the effects of immunosuppressive drugs such as corticosteroids used to control GvHD. Prolonged exposure to
prednisolone is associated with relentless suppression of CMV specific CD8 T cells and a consequent
increase in viral load (Fig 2).3 The powerful effect of
CMV-specific T cells in limiting CMV replication was
dramatically demonstrated when an adoptive immunotherapeutic approach achieved suppression of
CMV replication in patients who had high viral loads
that were unresponsive to conventional antiviral chemotherapy.20 Studies are in progress to determine
whether a combination of viral load measurement
and CMV-specific CD8 T-cell determinations might
provide complementary information to identify patients at high risk of developing CMV infection and
disease following transplantation.
Therapeutic Interventions Against CMV
Following Bone Marrow Transplantation
Two anti-CMV agents have been the mainstay for the
treatment of established disease in the hematology
patient: ganciclovir (an acyclic guanosine analogue)
and foscarnet (an analogue of inorganic pyrophosphate). Both block activity of CMV DNA polymerase
(UL54 gene), ganciclovir by acting as a competitive
inhibitor of the natural substrate and thus causing
chain termination, and foscarnet as a product inhibitor of the DNA synthetic reaction catalyzed by the
Betaherpesviruses in Hematological Patients
viral-specific DNA polymerase. For ganciclovir to be
effective it must be phosphorylated to the triphosphate moiety; the first stage of phosphorylation is
effected by a viral specific protein kinase encoded by
the UL97 gene, while subsequent phosphorylation
steps are catalyzed by cellular kinases. Ganciclovir
therapy (5 mg/kg twice daily) has an antiviral efficacy
of approximately 91.5% at controlling CMV replication.23 Similar estimates for the efficacy of foscarnet
are not available, but trials comparing ganciclovir
with foscarnet indicate much similarity.76 The effectiveness of preemptive therapy triggered by antigenemia positivity or PCR positivity has been shown in
two trials6,19; in both the treatment was initiated far
earlier than using conventional cell culture methods
and patients required less overall ganciclovir therapy
with the benefits of minimizing the associated neutropenia. Consequently, many centers now use antigenemia- or PCR-triggered preemptive therapy (usually 14 days) for patient management.
An alternative approach to reduce the probability
of CMV infection/disease has been the prophylactic
deployment of agents such as aciclovir and/or ganciclovir. Placebo-controlled clinical trials showed that
high-dose aciclovir could reduce CMV disease and
also improve survival.65,72 More recently, the availability of the pro-drug of aciclovir, valaciclovir,
which produces increased oral bioavailability, has
been shown to reduce the incidence of CMV disease
in bone marrow transplant recipients54 and also in
other immunocompromised hosts.56 Intravenous, or
more recently oral ganciclovir (1 g three times per
day), have produced clinical benefit in controlled
trials.30,96 Therapy up to 100 days following bone
marrow engraftment is effective at controlling CMV
infection and its consequent disease. However, there
are a variety of ramifications to these strategies. First,
since CMV replication is substantially inhibited by
oral ganciclovir (this formulation has an antiviral
efficacy of approximately 46.5%),23 the regenerating
immune system is not exposed to sufficient levels of
viral replication to elicit a T-cell–mediated immune
response, and so patients remain unprimed to CMV
following the cessation of prophylaxis.16 Second, because the drug concentration is inadequate to fully
eradicate replication during the period of prophylaxis, coupled with the lack of regeneration of an
early immune response against CMV, viremia can
appear following the cessation of prophylaxis (usually between 3 and 4 weeks) especially in the highrisk D⫺R⫹ group. Unfortunately, late CMV infection
has been associated with a high incidence of late
CMV disease,68 which has been difficult to manage,
stimulating many centers to pursue aggressive preemptive therapy triggered by antigenemia or PCR
positivity.
Although drug resistance is an emerging question
157
in the context of CMV,24,51 a recent report has shown
that antigenemia-driven preemptive therapy with
ganciclovir did not produce ganciclovir-resistant viruses: there was no selection of viruses with mutations in UL97, despite recurrence of antigenemia.28 A
major risk factor for the recurrent viremia appears to
be the slope of decline in viral load during initial
therapy, so that a tardy viral load response to intervention increases the likelihood of subsequent viremic episodes.38
Intravenous and oral formulations of ganciclovir
have been used extensively, but the prodrug of ganciclovir, valganciclovir is now available.62,71 Although there are no results from controlled trials of
this compound in bone marrow transplant recipients,
the prodrug should provide levels of ganciclovir in
the blood that are comparable to intravenous ganciclovir, but attention to poor absorption may be necessary in the presence of gut GvHD. Valganciclovir
likely will replace intravenous ganciclovir. Since the
effective concentration of ganciclovir has been increased over oral ganciclovir, the antiviral efficacy
achieved should be also be greater. How use of valganciclovir in treatment and prophylaxis will affect
late infection and generation of ganciclovir-resistant
viruses remains to be defined, but analysis of samples
obtained from a clinical trial of valganciclovir versus
intravenous ganciclovir in HIV-infected individuals
showed that the frequency of ganciclovir-resistant
strains of CMV were comparable between the two
treatment groups.8
Human Herpevirus-6
HHV-6 is highly prevalent worldwide and infection
usually occurs in early childhood. In young children,
HHV-6 is a causative agent of febrile illness including
exanthem subitum (ES).97 Two variants of HHV-6
have been defined (termed A and B), and they are
increasingly viewed as closely related although distinct species.18
HHV-6 is tropic for T lymphocytes and neural cells
but has the ability to infect a wide variety of cell types
in vitro.13 HHV-6 utilizes CD46 as a cellular receptor
(CD46, also termed membrane cofactor protein, is a
member of a family of complement regulators).80
Activated CD4⫹ T cells and monocytes/macrophages
appear to be the preferential targets for replication in
vivo.48,83
Similar to CMV, HHV-6 remains in the host lifelong following primary infection. The virus, predominantly variant B, can be detected in the peripheral
blood of healthy individuals by nested PCR when
sufficient quantities of DNA are tested.14 The actual
site of latency has yet to be established, although
candidates include monocytes47 and early bone marrow progenitor cells.58 An uncommon form of
158
Clark, Emery, and Griffiths
Table 2. Prospective Studies of HHV-6 Infection Following Bone Marrow or Stem Cell Transplantation
Study
Method of Detection
Observed Disease
Wilborn et al94
Appleton et al2
Kadakia et al43
Wang et al91
Chan et al11
Cone et al15
Maeda et al61
Ljungman et al55
Imbert-Marcille et al39
Yoshikawa et al99
PCR
PCR⫹IHC
VI
PCR
PCR
PCR
PCR
QCPCR
PCR
VI
GvHD*
GvHD* (PCR in skin/rectal biopsies)
None
Delayed granulocyte and platelet engraftment*
None
Rash*
Delayed platelet engraftment
Viral load correlated with delayed platelet engraftment
Myelosuppression and fever
Rash*
* Analysis on subgroup of patient population
Abbreviations: PCR, polymerase chain reaction; QCPCR, quantitative competitive PCR, VI, virus isolation; IHC, immunohistochemistry.
Updated and adapted with permission.13 Copyright 2000. © John Wiley & Sons Ltd.
HHV-6 persistence is characterized by integration of
apparently whole-genome length HHV-6 sequences
into host-cell chromosomes,89 but the clinical relevance of this unusual biology, if any, is not clear.
Integration sites have been mapped to chromosome
17p13.3, chromosome 22q13, and chromosome
1q44 in different individuals. The inheritance of
chromosomally integrated HHV-6 also has been reported.17
HHV-6 and Lymphoproliferative Disease
The involvement of HHV-6 in a range of hematological malignancies has been investigated with largely
negative findings.13 In situ detection of HHV-6 has
been demonstrated in the abnormal cells of T-cell
chronic lymphoproliferative disease9 and sinus histiocytosis with massive lymphadenopathy (RosaiDorfman disease).57 Further studies are necessary to
determine if HHV-6 is causally associated with either
of these disorders.
HHV-6 Infection Following Bone Marrow
Transplantation
Infection with HHV-6 following bone marrow transplantation is common and, considering the high rate
of seroprevalence of the population, is likely to be
due almost always to reactivation of recipient’s virus
or reinfection from the donor. The virus has been
found in 28% of bone marrow samples from healthy
individuals, suggesting that it can be transmitted
from the donor to the recipient,27 and cases of primary infection have been reported.49 HHV-6A and
especially HHV-6B infections have been detected in
the post-transplant period; most HHV-6 infections
occur within the first 4 weeks.39,61,99
Case reports and selective studies have associated
HHV-6 with a range of clinical diseases following
bone marrow transplantation, including encephalitis, idiopathic interstitial pneumonitis, hepatitis,
early and late graft failure, and bone marrow suppression.12,13 In vitro, both HHV-6 variants are able to
suppress the maturation and growth of normal bone
marrow precursors, including granulocyte/macrophage, erythroid, and megakaryocytic lineages.40,41
The detection of HHV-6 in cerebrospinal fluid by
PCR also has been associated with a range of CNS
disease in bone marrow transplant recipients.90,92,100
Prospective studies have been conducted to measure the medical impact of HHV-6 following stem cell
transplantation (Table 2). Some have reported associations between HHV-6 and delayed engraftment,
myelosuppression, and fever.39,61 HHV-6 viral load
also significantly correlated with delayed platelet engraftment following stem cell transplantation.55
However, other analyses have failed to show a clear
association between HHV-6 and disease when the
whole study population was analyzed2,11,15,43,91,94;
subgroup analysis sometimes has linked the virus to
GvHD,94 delayed engraftment,91 and rash.15,99
Contradictory findings in prospective studies are
likely to represent variations in the demographics of
transplant populations, their medical care, and, importantly, the methods used in detection of virus
infection (although the majority have employed
PCR). Most authors have found a low incidence of
the disease types previously identified by case reports
when large numbes of bone marrow transplant recipients were examined.
Human Herpesvirus-7
As with HHV-6, HHV-7 also is highly prevalent
worldwide and infection usually occurs in early
childhood, perhaps slightly delayed in comparison to
HHV-6. Similar to HHV-6, HHV-7 can cause febrile
159
Betaherpesviruses in Hematological Patients
Table 3. Prospective Studies of HHV-7 Infection Following
Transplantation
Study
Transplant
Type
Method of
Detection
Wang et al91
Chan et al11
Maeda et al61
Osman et al70
Kidd et al45
Bone marrow
Bone marrow
Bone marrow
Renal
Renal
PCR
PCR
PCR
PCR
PCR
Tong et al88
Griffiths et al33
Mendez et al63
Renal
Liver
Liver
PCR
PCR
PCR
Observed Disease
None
Delayed engraftment*
None
CMV disease
CMV disease
Graft rejection*
CMV disease
None
CMV disease
* Analysis on subgroup of patient population.
illness in young children.85 Analysis of the HHV-7
genome shows that it is most closely related to HHV6.69
HHV-7 utilizes CD4 as a cellular receptor to infect
T cells59 and although CXCR4 was initially reported
to be downregulated in infected cells,81 more recent
studies suggest that this molecule is not a co-receptor.102 The virus has also been shown to productively
infect macrophages in vitro.101
Similar to HHV-6, HHV-7 can be detected in the
peripheral blood of healthy individuals by nested
PCR,44 although the actual site of latency has yet to be
identified. Monocyte/macrophages harbor virus in a
nonreplicating form following infection in vitro.101
HHV-7 Following Transplantation
Fewer studies have examined the role of HHV-7 infection post-transplantation (Table 3). Similar to
HHV-6, active HHV-7 infection does occur, likely
due to reactivation of recipient’s virus or reinfection,
given the high seroprevalence in the population.
HHV-7 DNA has been detected by PCR in 50% of
bone marrow samples from healthy persons,27 including CD34⫹ and CD34⫺ cell fractions,66 suggesting that donor material may be a potential source of
virus.
Prospective studies in bone marrow transplant recipients have not found an obvious correlation between HHV-7 infection and a range of clinical endpoints including GvHD and engraftment61,91 (Table
3). The detection of HHV-7 in peripheral blood by
PCR during the early post-transplantation period was
associated with a longer time to neutrophil engraftment.11 In vitro HHV-7 was not suppressive of granulocyte/macrophage, erythroid, and megakaryocyte
colony formation in some experiments,40,41 but inoculation of cord blood CD34⫹ cells in vitro with
HHV-7 significantly decreased the number of pluripotent (granulocyte/monocyte/erythroid/megakaryocyte)
progenitors in semisolid assays with a more modest
effect on committed (granulocyte/macrophage and
erythroid) progenitors.66 In addition, HHV-7 appeared to hasten differentiation of cord blood CD34⫹
cells to myeloid but not erythroid cells.66 Thus, there
are in vitro data to suggest the ability of HHV-7 to
perturb the maturation of haematopoietic progenitor
cells. Two cases of HHV-7 encephalitis, one fatal,
following bone marrow or stem cell transplantation
have been reported.10,11
A number of prospective studies have followed
solid organ transplant patients (Table 3).33,45,63,70,88
In renal recipients, concomitant CMV and HHV-7
infection was associated with a greater risk of developing CMV disease70 (Table 3). Others have reported
a similar association between HHV-7 and an increased risk of CMV disease,45,63,88 but putative
mechanisms are unknown.
HHV-6 and HHV-7 Antiviral Susceptibility
Ganciclovir, foscarnet, and cidofovir have been reported to be inhibitors of HHV-6 and HHV-7 replication in vitro,84 although not consistently.77,98 There
have been no controlled trials of antiviral therapy
against HHV-6 or HHV-7 infection, but individual
published cases suggested a clinical response to treatment of HHV-6 disease (encephalitis/CNS disease
and bone marrow suppression) after bone marrow
transplantation, using either ganciclovir or foscarnet
or both.12 Currently licensed antiherpetic compounds may be effective against HHV-6 and HHV-7,
but treatment strategies need to be formulated
through appropriate clinical protocols.
Conclusion
CMV remains an important pathogen in hematological patients, specifically bone marrow/stem cell transplant recipients. However, important advances have
been made in the monitoring and treatment of CMV
infection in these patients. The future use of immunotherapeutic approaches may complement the current management strategies. The clinical relevance of
HHV-6 and HHV-7 in hematological patients is certainly more restricted than CMV, although accumulating evidence highlights the ability of HHV-6 to
cause disease post bone marrow/stem cell transplantation.
References
1. Appay V, Dunbar PR, Callan M, et al: Memory CD8⫹ T cells
vary in differentiation phenotype in different persistent virus infections. Nat Med 8:379-385, 2002
2. Appleton AL, Sviland L, Peiris JS, et al: Human herpesvirus-6 infection in marrow graft recipients: Role in pathogenesis of graft-versus-host disease. Bone Marrow Transplant
16:777-782, 1995
160
Clark, Emery, and Griffiths
3. Aubert G, Hassan-Walker AF, Madrigal JA, et al: Cytomegalovirus-specific cellular immune responses and viremia in
recipients of allogeneic stem cell transplants. J Infect Dis
184:955-963, 2001
4. Baldanti F, Revello MG, Percivalle E, et al: Use of the human
cytomegalovirus (HCMV) antigenemia assay for diagnosis
and monitoring of HCMV infections and detection of antiviral drug resistance in the immunocompromised. J Clin
Virol 11:51-60, 1998
5. Beninga J, Kropff B, Mach M: Comparative analysis of fourteen individual human cytomegalovirus proteins for helper
T cell response. J Gen Virol 76:153-160, 1995
6. Boeckh M, Gooley TA, Myerson D, et al: Cytomegalovirus
pp65 antigenemia-guided early treatment with ganciclovir
versus ganciclovir at engraftment after allogeneic marrow
transplantation: A randomized double-blind study. Blood
88:4063-4071, 1996
7. Boivin G, Belanger R, Delage R, et al: Quantitative analysis of
cytomegalovirus (CMV) viremia using the pp65 antigenemia assay and the Cobas Amplicor CMV Monitor PCR test
after blood and marrow allogeneic transplantation. J Clin
Microbiol 38:4356-4360, 2000
8. Boivin G, Gilbert C, Gaudreau A, et al: Rate of emergence of
cytomegalovirus (CMV) mutations in leukocytes of patients
with acquired immunodeficiency syndrome who are receiving valganciclovir as induction and maintenance therapy for
CMV retinitis. J Infect Dis 184:1598-1602, 2001
9. Braun DK, Pellett PE, Hanson CA: Presence and expression
of human herpesvirus 6 in peripheral blood mononuclear
cells of S100-positive, T cell chronic lymphoproliferative
disease. J Infect Dis 171:1351-1355, 1995
10. Chan PK, Chik KW, To KF, et al: Case report: human
herpesvirus 7 associated fatal encephalitis in a peripheral
blood stem cell transplant recipient. J Med Virol 66:493496, 2002
11. Chan PK, Peiris JS, Yuen KY, et al: Human herpesvirus-6 and
human herpesvirus-7 infections in bone marrow transplant
recipients. J Med Virol 53:295-305, 1997
12. Clark DA, Griffiths PD: Human herpesvirus 6: Relevance of
infection in the immunocompromised host. Br J Haematol
(in press)
13. Clark DA: Human herpesvirus 6. Rev Med Virol 10:155-173,
2000
14. Clark DA, Ait-Khaled M, Wheeler AC, et al: Quantification
of human herpesvirus 6 in immunocompetent persons and
post-mortem tissues from AIDS patients by PCR. J Gen Virol
77:2271-2275, 1996
15. Cone RW, Huang ML, Corey L, et al: Human herpesvirus 6
infections after bone marrow transplantation: Clinical and
virologic manifestations. J Infect Dis 179:311-318, 1999
16. Cwynarski K, Ainsworth J, Cobbold M, et al: Direct visualization of cytomegalovirus-specific T-cell reconstitution after allogeneic stem cell transplantation. Blood 97:12321240, 2001
17. Daibata M, Taguchi T, Nemoto Y, et al: Inheritance of
chromosomally integrated human herpesvirus 6 DNA.
Blood 94:1545-1549, 1999
18. Dominguez G, Dambaugh TR, Stamey FR, et al: Human
herpesvirus 6B genome sequence: Coding content and comparison with human herpesvirus 6A. J Virol 73:8040-8052,
1999
19. Einsele H, Ehninger G, Hebart H, et al: Polymerase chain
reaction monitoring reduces the incidence of cytomegalovirus disease and the duration and side effects of antiviral
therapy after bone marrow transplantation. Blood 86:28152820, 1995
20. Einsele H, Roosnek E, Rufer N, et al: Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV
infection not responding to antiviral chemotherapy. Blood
99:3916-3922, 2002
21. Einsele H, Steidle M, Vallbracht A, et al: Early occurrence of
human cytomegalovirus infection after bone marrow transplantation as demonstrated by the polymerase chain reaction technique. Blood 77:1104-1110, 1991
22. Emery VC: Prophylaxis for CMV should not now replace
pre-emptive therapy in solid organ transplantation. Rev Med
Virol 11:83-86, 2001
23. Emery VC, Cope AV, Bowen EF, et al: The dynamics of
human cytomegalovirus replication in vivo. J Exp Med 190:
177-182, 1999
24. Emery VC, Griffiths PD: Prediction of cytomegalovirus load
and resistance patterns after antiviral chemotherapy. Proc
Natl Acad Sci USA 97:8039-8044, 2000
25. Emery VC, Sabin CA, Cope AV, et al: Application of viralload kinetics to identify patients who develop cytomegalovirus disease after transplantation. Lancet 355:2032-2036,
2000
26. Gamadia LE, Rentenaar RJ, Baars PA, et al: Differentiation of
cytomegalovirus-specific CD8(⫹) T cells in healthy and
immunosuppressed virus carriers. Blood 98:754-761, 2001
27. Gautheret-Dejean A, Dejean O, Vastel L, et al: Human herpesvirus-6 and human herpesvirus-7 in the bone marrow
from healthy subjects. Transplantation 69:1722-1723, 2000
28. Gilbert C, Roy J, Belanger R, et al: Lack of emergence of
cytomegalovirus UL97 mutations conferring ganciclovir
(GCV) resistance following preemptive GCV therapy in
allogeneic stem cell transplant recipients. Antimicrob
Agents Chemother 45:3669-3671, 2001
29. Gillespie GM, Wills MR, Appay V, et al: Functional heterogeneity and high frequencies of cytomegalovirus- specific
CD8(⫹) T lymphocytes in healthy seropositive donors. J Virol 74:8140-8150, 2000
30. Goodrich JM, Bowden RA, Fisher L, et al: Ganciclovir prophylaxis to prevent cytomegalovirus disease after allogeneic
marrow transplant. Ann Intern Med 118:173-178, 1993
31. Gor D, Sabin C, Prentice HG, et al: Longitudinal fluctuations
in cytomegalovirus load in bone marrow transplant patients:
relationship between peak virus load, donor/recipient
serostatus, acute GVHD and CMV disease. Bone Marrow
Transplant 21:597-605, 1998
32. Gratama JW, van Esser JW, Lamers CH, et al: Tetramerbased quantification of cytomegalovirus (CMV)-specific
CD8⫹ T lymphocytes in T-cell-depleted stem cell grafts and
after transplantation may identify patients at risk for progressive CMV infection. Blood 98:1358-1364, 2001
33. Griffiths PD, Ait-Khaled M, Bearcroft CP, et al: Human
herpesvirus 6 and 7 as potential pathogens after liver transplant: Prospective comparison with the effect of cytomegalovirus. J Med Virol 59:496-501, 1999
34. Griffiths PD, Clark DA, Emery VC: Betaherpesviruses in
transplant recipients. J Antimicrob Chemother 45:29-34,
2000 (suppl T3)
35. Grob JP, Grundy JE, Prentice HG, et al: Immune donors can
protect marrow-transplant recipients from severe cytomegalovirus infections. Lancet 1:774-776, 1987
36. Hart GD, Paya CV: Prophylaxis for CMV should now replace
pre-emptive therapy in solid organ transplantation. Rev Med
Virol 11:73-81, 2001
37. Hassan-Walker AF, Vargas Cuero AL, Mattes FM, et al:
CD8⫹ cytotoxic lymphocyte responses against cytomegalovirus after liver transplantation: correlation with time from
Betaherpesviruses in Hematological Patients
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
transplant to receipt of tacrolimus. J Infect Dis 183:835-843,
2001
Humar A, Kumar D, Boivin G, et al: Cytomegalovirus
(CMV) virus load kinetics to predict recurrent disease in
solid-organ transplant patients with CMV disease. J Infect
Dis 186:829-833, 2002
Imbert-Marcille BM, Tang XW, Lepelletier D, et al: Human
herpesvirus 6 infection after autologous or allogeneic stem
cell transplantation: A single-center prospective longitudinal study of 92 patients. Clin Infect Dis 31:881-886, 2000
Isomura H, Yamada M, Yoshida M, et al: Suppressive effects
of human herpesvirus 6 on in vitro colony formation of
hematopoietic progenitor cells. J Med Virol 52:406-412,
1997
Isomura H, Yoshida M, Namba H, et al: Suppressive effects
of human herpesvirus-6 on thrombopoietin-inducible
megakaryocytic colony formation in vitro. J Gen Virol 81:
663-673, 2000
Jarvis MA, Nelson JA: Human cytomegalovirus persistence
and latency in endothelial cells and macrophages. Curr Opin
Microbiol 5:403-407, 2002
Kadakia MP, Rybka WB, Stewart JA, et al: Human herpesvirus 6: Infection and disease following autologous and allogeneic bone marrow transplantation. Blood 87:5341-5354,
1996
Kidd IM, Clark DA, Ait-Khaled M, et al: Measurement of
human herpesvirus 7 load in peripheral blood and saliva of
healthy subjects by quantitative polymerase chain reaction.
J Infect Dis 174:396-401, 1996
Kidd IM, Clark DA, Sabin CA, et al: Prospective study of
human betaherpesviruses after renal transplantation: association of human herpesvirus 7 and cytomegalovirus coinfection with cytomegalovirus disease and increased rejection. Transplantation 69:2400-2404, 2000
Kidd IM, Fox JC, Pillay D, et al: Provision of prognostic
information in immunocompromised patients by routine
application of the polymerase chain reaction for cytomegalovirus. Transplantation 56:867-871, 1993
Kondo K, Kondo T, Okuno T, et al: Latent human herpesvirus 6 infection of human monocytes/macrophages. J Gen
Virol 72:1401-1408, 1991
Kondo K, Kondo T, Shimada K, et al: Strong interaction
between human herpesvirus 6 and peripheral blood monocytes/macrophages during acute infection. J Med Virol 67:
364-369, 2002
Lau YL, Peiris M, Chan GC, et al: Primary human herpes
virus 6 infection transmitted from donor to recipient
through bone marrow infusion. Bone Marrow Transplant
21:1063-1066, 1998
Lechner F, Cuero AL, Kantzanou M, et al: Studies of human
antiviral CD8⫹ lymphocytes using class I peptide tetramers.
Rev Med Virol 11:11-22, 2001
Limaye AP, Corey L, Koelle DM, et al: Emergence of ganciclovir-resistant cytomegalovirus disease among recipients of
solid-organ transplants. Lancet 356:645-649, 2000
Limaye AP, Huang ML, Leisenring W, et al: Cytomegalovirus (CMV) DNA load in plasma for the diagnosis of CMV
disease before engraftment in hematopoietic stem-cell transplant recipients. J Infect Dis 183:377-382, 2001
Liu YN, Curtsinger J, Donahue PR, et al: Molecular analysis
of the immune response to human cytomegalovirus glycoprotein B. I. Mapping of HLA-restricted helper T cell
epitopes on gp93. J Gen Virol 74:2207-2214, 1993
Ljungman P, de La CR, Milpied N, et al: Randomized study
of valacyclovir as prophylaxis against cytomegalovirus reac-
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
161
tivation in recipients of allogeneic bone marrow transplants.
Blood 99:3050-3056, 2002
Ljungman P, Wang FZ, Clark DA, et al: High levels of
human herpesvirus 6 DNA in peripheral blood leucocytes
are correlated to platelet engraftment and disease in allogeneic stem cell transplant patients. Br J Haematol 111:774781, 2000
Lowance D, Neumayer HH, Legendre CM, et al: Valacyclovir
for the prevention of cytomegalovirus disease after renal
transplantation. N Engl J Med 340:1462-1470, 1999
Luppi M, Barozzi P, Garber R, et al: Expression of human
herpesvirus-6 antigens in benign and malignant lymphoproliferative diseases. Am J Pathol 153:815-823, 1998
Luppi M, Barozzi P, Morris C, et al: Human herpesvirus 6
latently infects early bone marrow progenitors in vivo. J Virol 73:754-759, 1999
Lusso P, Secchiero P, Crowley RW, et al: CD4 is a critical
component of the receptor for human herpesvirus 7: Interference with human immunodeficiency virus. Proc Natl
Acad Sci USA 91:3872-3876, 1994
Machida U, Kami M, Fukui T, et al: Real-time automated
PCR for early diagnosis and monitoring of cytomegalovirus
infection after bone marrow transplantation. J Clin Microbiol 38:2536-2542, 2000
Maeda Y, Teshima T, Yamada M, et al: Monitoring of human
herpesviruses after allogeneic peripheral blood stem cell
transplantation and bone marrow transplantation. Br J
Haematol 105:295-302, 1999
Martin DF, Sierra-Madero J, Walmsley S, et al: A controlled
trial of valganciclovir as induction therapy for cytomegalovirus retinitis. N Engl J Med 346:1119-1126, 2002
Mendez JC, Dockrell DH, Espy MJ, et al: Human betaherpesvirus interactions in solid organ transplant recipients.
J Infect Dis 183:179-184, 2001
Meyers JD, Ljungman P, Fisher LD: Cytomegalovirus excretion as a predictor of cytomegalovirus disease after marrow
transplantation: Importance of cytomegalovirus viremia.
J Infect Dis 162:373-380, 1990
Meyers JD, Reed EC, Shepp DH, et al: Acyclovir for prevention of cytomegalovirus infection and disease after allogeneic marrow transplantation. N Engl J Med 318:70-75, 1988
Mirandola P, Secchiero P, Pierpaoli S, et al: Infection of
CD34(⫹) hematopoietic progenitor cells by human herpesvirus 7 (HHV-7). Blood 96:126-131, 2000
Mori T, Okamoto S, Watanabe R, et al: Dose-adjusted preemptive therapy for cytomegalovirus disease based on realtime polymerase chain reaction after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 29:
777-782, 2002
Nguyen Q, Champlin R, Giralt S, et al: Late cytomegalovirus
pneumonia in adult allogeneic blood and marrow transplant
recipients. Clin Infect Dis 28:618-623, 1999
Nicholas J: Determination and analysis of the complete
nucleotide sequence of human herpesvirus. J Virol 70:59755989, 1996
Osman HK, Peiris JS, Taylor CE, et al: “Cytomegalovirus
disease” in renal allograft recipients: Is human herpesvirus
7 a co-factor for disease progression? J Med Virol 48:295301, 1996
Pescovitz MD, Rabkin J, Merion RM, et al: Valganciclovir
results in improved oral absorption of ganciclovir in liver
transplant recipients. Antimicrob Agents Chemother 44:
2811-2815, 2000
Prentice HG, Gluckman E, Powles RL, et al: Impact of
long-term acyclovir on cytomegalovirus infection and survival after allogeneic bone marrow transplantation. Euro-
162
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
Clark, Emery, and Griffiths
pean Acyclovir for CMV Prophylaxis Study Group. Lancet
343:749-753, 1994
Qamruddin AO, Oppenheim BA, Guiver M, et al: Screening
for cytomegalovirus (CMV) infection in allogeneic bone
marrow transplantation using a quantitative whole blood
polymerase chain reaction (PCR) method: Analysis of potential risk factors for CMV infection. Bone Marrow Transplant 27:301-306, 2001
Reed EC, Shepp DH, Dandliker PS, et al: Ganciclovir treatment of cytomegalovirus infection of the gastrointestinal
tract after marrow transplantation. Bone Marrow Transplant
3:199-206, 1988
Reusser P, Attenhofer R, Hebart H, et al: Cytomegalovirusspecific T-cell immunity in recipients of autologous peripheral blood stem cell or bone marrow transplants. Blood
89:3873-3879, 1997
Reusser P, Einsele H, Lee J, et al: Randomized multicenter
trial of foscarnet versus ganciclovir for preemptive therapy
of cytomegalovirus infection after allogeneic stem cell transplantation. Blood 99:1159-1164, 2002
Reymen D, Naesens L, Balzarini J, et al: Antiviral activity of
selected acyclic nucleoside analogues against human herpesvirus 6. Antiviral Res 28:343-357, 1995
Roback JD: CMV and blood transfusions. Rev Med Virol
12:211-219, 2002
Rubin RH: The indirect effects of cytomegalovirus infection
on the outcome of organ transplantation. JAMA 261:36073609, 1989
Santoro F, Kennedy PE, Locatelli G, et al: CD46 is a cellular
receptor for human herpesvirus 6. Cell 99:817-827, 1999
Secchiero P, Zella D, Barabitskaja O, et al: Progressive and
persistent downregulation of surface CXCR4 in CD4(⫹) T
cells infected with human herpesvirus 7. Blood 92:45214528, 1998
Soderberg-Naucler C, Fish KN, Nelson JA: Reactivation of
latent human cytomegalovirus by allogeneic stimulation of
blood cells from healthy donors. Cell 91:119-126, 1997
Takahashi K, Sonoda S, Higashi K, et al: Predominant CD4
T-lymphocyte tropism of human herpesvirus 6-related virus. J Virol 63:3161-3163, 1989
Takahashi K, Suzuki M, Iwata Y, et al: Selective activity of
various nucleoside and nucleotide analogues against herpesvirus 6 and 7. Antiviral Chem Chemother 8:24-31, 1997
Tanaka K, Kondo T, Torigoe S, et al: Human herpesvirus 7:
Another causal agent for roseola (exanthem subitum). J Pediatr 125:1-5, 1994
Tanaka Y, Kanda Y, Kami M, et al: Monitoring cytomegalovirus infection by antigenemia assay and two distinct plasma
real-time PCR methods after hematopoietic stem cell transplantation. Bone Marrow Transplant 30:315-319, 2002
Taylor-Wiedeman J, Sissons JG, Borysiewicz LK, et al:
Monocytes are a major site of persistence of human cytomegalovirus in peripheral blood mononuclear cells. J Gen
Virol 72:2059-2064, 1991
88. Tong CY, Bakran A, Williams H, et al: Association of human
herpesvirus 7 with cytomegalovirus disease in renal transplant recipients. Transplantation 70:213-216, 2000
89. Torelli G, Barozzi P, Marasca R, et al: Targeted integration of
human herpesvirus 6 in the p arm of chromosome 17 of
human peripheral blood mononuclear cells in vivo. J Med
Virol 46:178-188, 1995
90. Wainwright MS, Martin PL, Morse RP, et al: Human herpesvirus 6 limbic encephalitis after stem cell transplantation.
Ann Neurol 50:612-619, 2001
91. Wang FZ, Dahl H, Linde A, et al: Lymphotropic herpesviruses in allogeneic bone marrow transplantation. Blood 88:
3615-3620, 1996
92. Wang FZ, Linde A, Hagglund H, et al: Human herpesvirus 6
DNA in cerebrospinal fluid specimens from allogeneic bone
marrow transplant patients: Does it have clinical significance? Clin Infect Dis 28:562-568, 1999
93. Webster A, Blizzard B, Pillay D, et al: Value of routine
surveillance cultures for detection of CMV pneumonitis
following bone marrow transplantation. Bone Marrow
Transplant 12:477-481, 1993
94. Wilborn F, Brinkmann V, Schmidt CA, et al: Herpesvirus
type 6 in patients undergoing bone marrow transplantation:
Serologic features and detection by polymerase chain reaction. Blood 83:3052-3058, 1994
95. Wills MR, Carmichael AJ, Mynard K, et al: The human
cytotoxic T-lymphocyte (CTL) response to cytomegalovirus
is dominated by structural protein pp65: Frequency, specificity, and T- cell receptor usage of pp65-specific CTL.
J Virol 70:7569-7579, 1996
96. Winston DJ, Ho WG, Bartoni K, et al: Ganciclovir prophylaxis of cytomegalovirus infection and disease in allogeneic
bone marrow transplant recipients. Results of a placebocontrolled, double-blind trial. Ann Intern Med 118:179-184,
1993
97. Yamanishi K, Okuno T, Shiraki K, et al: Identification of
human herpesvirus-6 as a causal agent for exanthem subitum. Lancet 1:1065-1067, 1988
98. Yoshida M, Yamada M, Tsukazaki T, et al: Comparison of
antiviral compounds against human herpesvirus 6 and 7.
Antiviral Res 40:73-84, 1998
99. Yoshikawa T, Asano Y, Ihira M, et al: Human herpesvirus 6
viremia in bone marrow transplant recipients: clinical features and risk factors. J Infect Dis 185:847-853, 2002
100. Zerr DM, Gupta D, Huang ML, et al: Effect of antivirals on
human herpesvirus 6 replication in hematopoietic stem cell
transplant recipients. Clin Infect Dis 34:309-317, 2002
101. Zhang Y, de Bolle L, Aquaro S, et al: Productive infection of
primary macrophages with human herpesvirus 7. J Virol
75:10511-10514, 2001
102. Zhang Y, Hatse S, De Clercq E, et al: CXC-chemokine
receptor 4 is not a coreceptor for human herpesvirus 7 entry
into CD4(⫹) T cells. J Virol 74:2011-2016, 2000