You are on page 1of 15

guideline

Management of cytomegalovirus infection in haemopoietic


stem cell transplantation

Vincent Emery,1 Mark Zuckerman,2 Graham Jackson,3 Celia Aitken,4 Husam Osman,5 Anthony Pagliuca,6 Mike Potter,7 Karl
Peggs,8 and Andrew Clark9 on behalf of the British Committee for Standards in Haematology, the British Society of Blood
and Marrow Transplantation and the UK Virology Network
1
Department of Virology, University College London School of Life and Medical Sciences, London, 2Department of Virology, Kings
College Hospital, London, 3Department of Haematology, Freeman Road Hospital, Newcastle, 4West of Scotland specialist virology
centre, Gartnavel General Hospital, Glasgow, 5Birmingham HPA Laboratory, Birmingham Heartlands Hospital, Birmingham,
6
Department of Haematology, Kings College Hospital, London, 7Section of Haemato-oncology, The Royal Marsden NHS Foundation
Trust, London, 8Department of Haematology, University College London Hospitals, London, and 9Blood and Marrow Transplant
Unit, Beatson Oncology Centre, Glasgow, UK

Keywords: stem cell, transplantation, bone marrow, periph- Primary prophylaxis with ganciclovir is not generally
eral blood stem cells, cytomegalovirus. recommended as toxicity outweighs efficacy in HSCT
patients (Grade 1B).
Primary prophylaxis with aciclovir or valaciclovir can
be deployed but only in conjunction with appropriate
List of Recommendations
monitoring of CMV in blood (Grade 1B).
Cytomegalovirus (CMV) infection and CMV disease Valaciclovir or valganciclovir are valid treatment
should be diagnosed according to established, interna- options for secondary prophylaxis with appropriate
tionally accepted, standardized criteria (Grade 1C). monitoring of CMV in blood (Grade 1C).
Risk-adapted patient assessment should inform clinical Intravenous immunoglobulin is not recommended for
management (Grade 1B). prophylaxis of CMV infection (Grade 1A).
All potential haemopoietic stem cell transplantation Real time quantitative polymerase chain reaction (PCR)
(HSCT) recipients should be tested for the presence of is the preferred choice for monitoring CMV DNA levels
CMV IgG antibody at diagnosis (Grade 1C). in HSCT patients (Grade 1B).
Once optimum human leucocyte antigen (HLA) match- All diagnostic laboratories should deploy the CMV
ing has been performed, a CMV IgG-negative donor international standard to allow viral loads to be
should be chosen for a CMV IgG-negative recipient and compared between centres (Grade 1C).
a CMV IgG-positive donor should be chosen for a CMV Monitoring of CMV load should be undertaken at
IgG-positive recipient when possible (Grade 1A). least weekly for the first 3 months post-HSCT (Grade
Donors or recipients who are initially found to be CMV 2C).
IgG-negative should be retested pre-transplant to CMV viral load monitoring should continue for 6-
exclude primary CMV infection (Grade 1C). 12 months if the patient has chronic graft-versus-host
Apparent CMV seroconversion in potential allograft disease (GvHD) or prolonged T-cell immunodeficiency
recipients who have received unscreened blood products (Grade 1B).
should be actively investigated to exclude passive acqui- Each transplant centre should have a risk-adapted policy
sition of antibody (Grade 1C) detailing threshold values for treatment of CMV infec-
Any CMV IgG-negative HSCT recipient transplanted tion, taking into account patient factors and PCR meth-
from a CMV IgG-negative donor who develops CMV odology (Grade 2C).
infection post-transplant must be reported to the Serious Ganciclovir is recommended as first line pre-emptive
Hazards of Transfusion (SHOT) scheme (Grade 1C). therapy for CMV in HSCT patients (Grade 1A).
Oral valganciclovir is a valid alternative when gastroin-
testinal absorption is normal or only minimally
Correspondence: Dr Andrew Clark, Bone Marrow Transplant Unit, impaired (Grade 1A).
Beatson Oncology Centre, West of Scotland Cancer Centre, 1053 Foscarnet is recommended as an alternative first-line
Great Western Road, Glasgow G12 0YN, UK. agent if neutropenia is present or for ganciclovir treat-
E-mail: andyclark@doctors.org.uk ment failure (Grade 1A).

2013 John Wiley & Sons Ltd First published online 6 May 2013
British Journal of Haematology, 2013, 162, 2539 doi:10.1111/bjh.12363
Guideline

Pre-emptive therapy with cidofovir can be considered as DENCE_LEVELS_AND_GRADES_OF_RECOMMENDA-


third-line in patients unresponsive to, or intolerant of, TION/43_GRADE.html).
both a ganciclovir preparation and foscarnet (Grade 2B). Initial review of manuscript, performed by the UK Clini-
In patients in whom CMV DNA loads in blood increase cal virology Network, British Society of Blood and Mar-
by 1 log10 over 2 weeks of pre-emptive therapy with a row Transplantation (BSBMT) executive committee and
first line drug, an alternative agent and drug resistance the British Committee for Standards in Haematology
profiling should be considered (Grade 2C). (BCSH) Haem-Onc Task Force.
Drug resistance should start to be suspected if CMV Final Review by sounding boards of the British Society for
loads in the blood fail to respond after 14 d of therapy, Haematology (BSH) and British Society of Blood and
especially in non-lymphopenic or multiply pre-treated Marrow Transplantation (BSBMT).
patients (Grade 2C).
Sequence analysis of the UL97 and UL54 genes is the
preferred option for resistance screening for currently Background
available drugs (grade 1B).
A multidisciplinary approach to management of CMV Clinical manifestations of CMV
disease is required (Grade 1C).
CMV is a herpes virus. Primary infection is followed by life-
De novo CMV disease should be treated with ganciclovir
long latency. Clinical manifestations vary widely and should
or foscarnet monotherapy and intravenous immuno-
be diagnosed according to established, internationally
globulin (Grade 1B).
accepted, standardized criteria (Ljungman et al, 2002a).
CMV disease that develops while on pre-emptive ther-
CMV infection is diagnosed when CMV is detected, most
apy or is clinically progressive requires drug resistance
commonly in the blood, using sensitive screening tools. It is
testing, increased drug doses and/or combination ther-
termed primary CMV infection when infection occurs in a
apy (Grade 1B).
CMV IgG-negative patient and CMV reactivation when the
Reduction in immunosuppression, especially reduction
patient, or donor, is known to be CMV antibody positive. In
in corticosteroid dose, is strongly recommended when
uncomplicated CMV infection, organ-specific signs and
possible (Grade 1B).
symptoms are absent, although non-specific symptoms, such
as fever and malaise, may occur (Ljungman et al, 2002a,
2011). The diagnosis of CMV disease requires the presence
Scope
of symptoms and signs compatible with end organ damage,
These evidence-based guidelines expand and adapt previous together with the detection of CMV by a validated method
guidance (Tomblyn et al, 2009; Andrews et al, 2011). While in an appropriate clinical specimen (Ljungman et al, 2002a).
specifically focusing on allogeneic haemopoietic stem cell If left untreated, asymptomatic CMV infection can progress
transplantation (HSCT), they are relevant to other areas of to CMV disease, most commonly affecting the lung, gastroin-
haematological oncology where there is an increased risk of testinal tract, eye, liver or central nervous system (CNS).
cytomegalovirus (CMV) infection, such as haematological CMV pneumonia is the most serious complication with
cancers where intense anti-T-cell therapy has been deployed a > 50% mortality (Ljungman, 1995). In addition to specific
(OBrien et al, 2006). organ damage, CMV also has profound, poorly characterized,
indirect immunosuppressive effects, leading to an increased
incidence of fungal and bacterial infection (Nichols et al,
Methodology 2002; Hakki et al, 2003; Blanquer et al, 2011) as well as
higher rates of both acute and chronic graft-versus-host
The production of these guidelines involved the following
disease (GvHD) (Soderberg et al, 1996; Larsson et al, 2004;
steps:
Wang et al, 2008). While some recent reports have suggested
Establishment of a working group comprising experts in
reduced relapse rates in selected patients who reactivate
the field of allogeneic transplantation and clinical virology
CMV (Behrendt et al, 2009; Elmaagacli et al, 2011), this
followed by literature review to 1 May 2012 including
finding remains controversial and should not hinder early
Medline, Pubmed and Cochrane reviews databases.
aggressive treatment of CMV infection.
Development of key recommendations based on ran-
domized, controlled trial evidence. Due to the paucity
of randomized studies, some recommendations are
Risk factors for CMV infection
based on literature review and a consensus of expert
opinion. Post HSCT, most patients will reactivate latent virus rather
The GRADE nomenclature was used to evaluate levels of than acquire primary infection. Mechanisms of latency are
evidence and to assess the strength of recommendations not completely understood but prevention of reactivation
(http://www.bcshguidelines.com/BCSH_PROCESS/EVI- appears to be primarily dependent upon a persistent, robust,

26 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

T-cell-mediated, immune response (Sylwester et al, 2005; complications and a CMV IgG-negative donor should be
Ljungman et al, 2011). Not surprisingly, the intensity of immu- chosen in these circumstances when possible. There is
nosuppression and the degree of T-cell depletion in transplant debate about the relative importance of CMV serostatus
protocols both critically affect the risk of reactivation (van Burik and HLA compatibility in donor selection. Specifically,
et al, 2007). Recipients of transplants from unrelated or human while HLA matching at HLA-A,B,C and DR remains the
leucocyte antigen (HLA)-mismatched donors, where immuno- most important factor in donor selection, the value of
suppression is increased and GvHD more likely, are particularly choosing a CMV IgG-negative donor over mismatches at
likely to reactivate CMV and these patients have a survival dis- other loci, such as HLA DQ or DP, or in protocols with
advantage. The deployment of T-cell depleting agents, such as aggressive T cell depletion remains unresolved (Boeckh &
alemtuzumab, or antithymocyte globulin (ATG) or increased/ Ljungman, 2009). It is also recognized that donor selection
prolonged courses of immunosuppression to treat GvHD can be a complex process and optimum CMV matching
significantly increases the risk of CMV infection (Schmidt-Hie- may not always be possible (Kollman et al, 2001; Spellman
ber et al, 2010). These risk factors can also increase the risk of et al, 2012). Provision of CMV-safe blood products is stan-
persistent, recurrent or late onset disease (Buyck et al, 2010), dard practice when both host and donor are negative, either
usually associated with impaired immune reconstitution (Ha- derived from CMV IgG-negative recipients or achieved by
kki et al, 2003; Zhou et al, 2009). Conversely, the reconstitution leucodepletion. Both techniques are effective, although rare
of specific anti-CMV cytotoxic T-lymphocytes (CTL) has been incidences of transfusion-transmitted infection have
shown to be protective (Lamba et al, 2005). CMV disease is as occurred using both approaches (Bowden et al, 1995; Ljung-
much of a problem following non-myeloablative transplanta- man et al, 2002b; Nichols et al, 2003). In the UK, universal
tion as it is in the myeloablative setting. Conflicting data have leucodepletion has largely replaced blood products from
been reported regarding the relative risk of bone marrow (BM) CMV-negative donors as of May 2012. It is important to be
compared to peripheral blood stem cells (PBSC) as a stem cell aware of the need to check the baseline CMV IgG status of
source (Nakamae et al, 2009; George et al, 2010; Pinana et al, potential transplant candidates at the time of initial diagno-
2010; Guerrero et al, 2012). Published incidence of CMV reacti- sis, to avoid confusion caused by passive transmission of
vation and CMV disease after umbilical cord blood transplanta- antibodies with transfusion of CMV IgG-positive blood
tion (UCBT) varies significantly. UCBT recipients have been products to negative recipients. Any transplant recipient
reported to be more susceptible to late complications of CMV, who converts from CMV IgG-negative to CMV IgG-positive
although this is not a universal finding and the relative role of T status pre-transplant will require careful assessment to sepa-
cell depletion in UCBT conditioning protocols requires further rate passive antibody from true seroconversion, as this has
investigation (Boeckh et al, 2004; Takami et al, 2005; Walker major implications for donor selection. In this setting, a
et al, 2007; Beck et al, 2010; Brown et al, 2010; Sauter et al, falling titre with time is suggestive of passively acquired
2011; Chiesa et al, 2012; Mikulska et al, 2012). antibody. Pre-allograft, it is recommended that samples for
IgM antibody and CMV polymerase chain reaction (PCR)
are sent as soon as a change in CMV status is suspected. If
Recommendations both are negative then this suggests the presence of pas-
sively acquired antibody. Blood products must be leucode-
CMV infection and CMV disease should be diagnosed
pleted in the blood bank facility (Ratko et al, 2001) and
according to established, internationally accepted, stan-
evidence of quality control for leucoreduction should be
dardized criteria (Grade 1C).
available to all transplant centres for JACIE [Joint Accredi-
Risk-adapted patient assessment should inform clinical
tation Committee International Society for Cellular Ther-
management (Grade 1B).
apy (Europe) & European Group for Blood and Marrow
Transplantation (EBMT)] accreditation purposes. R /D
transplants still require to be monitored for CMV infection.
Impact of host and donor CMV serostatus
Any cases of CMV infection occurring in this group of
patients should be reported to the Serious Hazards of
CMV screening
Transfusion (SHOT) scheme.
CMV screening should be performed using commercially
available CMV IgG assays. There is no gold standard assay
CMV IgG-positive donor or recipient
and it is important that laboratories running these tests par-
ticipate in external and internal quality assurance schemes. CMV seropositivity in either host or donor pre-transplant
continues to be associated with a poorer overall survival
post-allogeneic transplantation as a result of increased
CMV IgG-negative donor recipient pairs (R /D )
non-relapse mortality (NRM) (Broers et al, 2000; Kroger
CMV-negative recipients of grafts from CMV-negative et al, 2001; Albano et al, 2006; Tomonari et al, 2008). The
donors (R /D ) very rarely develop major CMV-related degree of this risk is determined primarily by the CMV IgG

2013 John Wiley & Sons Ltd 27


British Journal of Haematology, 2013, 162, 2539
Guideline

status of the recipient. If a CMV IgG-negative recipient Adoptive immunotherapy


receives cells from a CMV IgG-positive donor (R /D+) then
There have been several small studies published using CD4
CMV infection occurs in 2030% of cases. CMV disease is
or CD8 T cells (Walter et al, 1995; Einsele et al, 2002; Peggs
unusual but NRM is increased, probably through the indirect
et al, 2003, 2009; Hanley et al, 2011; Sili et al, 2012), or
effects of CMV on immune status post-transplant (Nichols
CMV peptide-loaded dendritic cell vaccination (Grigoleit
et al, 2002; Ljungman et al, 2011; Pergam et al, 2012). There
et al, 2007) for treatment or prophylaxis of CMV infection,
is a greater risk of CMV reactivation and progression to
but too little evidence currently exists to make any recom-
CMV disease in CMV seropositive recipients, where 80% are
mendation at present although ongoing prospective studies
likely to reactivate CMV, irrespective of CMV status of
are addressing this issue.
donor (Ljungman et al, 2011). However, in most studies,
major complications were further increased when the donor
wass CMV IgG-negative (Ozdemir et al, 2007; Zhou et al,
Antiviral agents
2009; Ugarte-Torres et al, 2011).
Aciclovir prophylaxis has been extensively studied post-
HSCT. A large randomized trial of 310 patients initially
Recommendations
appeared to suggest a reduced incidence and delayed onset
All Potential HSCT recipients should be tested for the of CMV infection as well as a significant improvement in
presence of CMV IgG antibody at diagnosis (Grade 1C). survival. More mature follow up has shown no significant
Once optimum HLA matching has been performed, a difference in CMV reactivation between groups, although
CMV IgG-negative donor should be chosen for a CMV reactivation did occur later in the prophylactic group. A
IgG-negative recipient and a CMV IgG-positive donor modest survival benefit was still seen in the most aggressively
should be chosen for CMV IgG-positive recipient when treated patients, though it is difficult to attribute this to
possible (Grade 1A). anti-CMV activity alone (Prentice et al, 1994, 1997).
Donors or recipients who are initially found to be CMV Improved survival in allograft patients who received aciclovir
IgG-negative should be retested pre-transplant to prophylaxis post-engraftment was also shown in a meta-
exclude primary CMV infection (Grade 1C). analysis in which the vast majority of donor/recipient pairs
Apparent CMV seroconversion in potential allograft were CMV IgG-positive (Yahav et al, 2009). However, the
recipients who have received unscreened blood products impact of aciclovir on CMV reactivation/disease rates in the
should be actively investigated to exclude passively studies included was again minimal and survival advantage
acquired antibody (Grade 1C) was potentially mediated through anti-herpes simplex effects.
Any CMV IgG-negative HSCT recipient transplanted Subsequently, valaciclovir, 2 g four times a day, was
from a CMV IgG-negative donor who develops CMV compared with oral aciclovir at 800 mg four times a day, in
infection post-transplant must be reported to SHOT 727 patients following high dose intravenous (iv) aciclovir in
(Grade 1C). the immediate post-transplant period (Ljungman et al,
2002c). In this study, valaciclovir significantly reduced CMV
infection and disease rates (P < 00001). There was a 50%
Prevention of CMV disease occurrence
reduction in the use of pre-emptive therapy although there was
Prophylactic and pre-emptive strategies have both been used no difference in overall survival (Ljungman et al, 2002c). Similar
to reduce the incidence of CMV disease. Universal monitor- results were shown in a smaller case controlled study using valac-
ing of CMV levels in the blood is essential irrespective of iclovir at a dose of 1 g three times a day (Vusirikala et al, 2001).
whether prophylaxis is administered. The studies described above predominantly used myeloablative
conditioning and T-cell-replete BM as the stem cell source. Stud-
ies in PBSC recipients, though smaller, suggested that the benefi-
Primary prophylaxis for CMV
cial effects of high dose aciclovir prophylaxis appeared to be
maintained (Verma et al, 2003; Hazar et al, 2004). However, aci-
Intravenous immunoglobulin
clovir was shown to be significantly less effective in T-cell-
There is no evidence that intravenous immunoglobulin depleted BM transplant recipients, where 83% of T-cell-depleted
(IVIG), CMV-specific hyperimmune immunoglobulin or recipients still had a CMV reactivation compared to 41% of
anti-CMV monoclonal antibodies are useful alone or in com- unmanipulated stem cell sources (P < 00001) (Nakamura
bination with antiviral agents in primary prophylaxis against et al, 2002). As most of these studies predated widespread
CMV infection (Bowden et al, 1991; Ruutu et al, 1997; use of pre-emptive therapy based on quantitative PCR, their
Boeckh et al, 2001). A recent Cochrane review in solid organ significance is questionable in terms of current management
transplant did not recommend prophylactic immunoglobulin of CMV, though a compelling argument can be made for its
(Hodson et al, 2007). use for suppression of herpes simplex virus infection.

28 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

Ganciclovir prophylaxis significantly reduced the incidence with prolonged CMV viral screening. If prophylaxis is given,
of CMV infection and disease during the period of prophy- then oral valaciclovir 2 g three times a day or valganciclovir
laxis (Goodrich et al, 1993; Boeckh et al, 1996). However 900 mg daily is an option (Boeckh & Ljungman, 2009).
neutropenia occurred in up to 30% of cases treated (Salzber-
ger et al, 1997) and infective complications were increased
Recommendations
(Boeckh et al, 1996). Ganciclovir was less effective in T-cell-
depleted transplants and heavily immunosuppressed recipi- Primary prophylaxis with ganciclovir is not generally
ents (Maltezou et al, 1999). In a prospective randomized trial recommended as toxicity outweighs efficacy in HSCT
of ganciclovir versus aciclovir, cumulative rates of CMV patients (Grade 1B).
disease were equivalent, although more patients in the aciclo- Primary prophylaxis with aciclovir or valaciclovir can
vir group required pre-emptive therapy (P = 02) (Burns be deployed but only in conjunction with appropriate
et al, 2002). Post-prophylaxis, late onset CMV disease monitoring of CMV in the blood (Grade 1B).
remained a problem (Boeckh et al, 2003) and prolonged Valaciclovir or valganciclovir are valid treatment
exposure of CMV to ganciclovir, especially in the setting of options for secondary prophylaxis with appropriate
T-cell depletion may encourage resistance, as occurs in solid monitoring of CMV in the blood (Grade 1C).
organ transplantation (Eid et al, 2008). Valganciclovir pro- Intravenous immunoglobulins are not recommended for
phylaxis has been reported to reduce risk of CMV disease in prophylaxis of CMV infection (Grade 1A).
cord blood transplants (Montesinos et al, 2009).
More intensive prophylactic regimens involving pre-trans-
plant ganciclovir combined with high dose aciclovir prophy- Pre-emptive therapy
laxis or a combination of ganciclovir and foscarnet have
The current mainstay for managing CMV infection after
been employed in high-risk paediatric and cord blood trans-
HSCT is the rapid introduction of therapy, based on
plants where both have been reported to be successful in
evidence of CMV replication in blood (Goodrich et al,
reducing CMV infection and disease (Shereck et al, 2007;
1991). Success of pre-emptive therapy is dependent on the
Milano et al, 2011).
availability of a rapid, sensitive assay to allow early treatment
Maribavir, when given from engraftment, initially showed
at low levels of viral infection.
favourable results in phase II studies; but, at the dose chosen,
failed to show any effect on CMV disease or initiation of
CMV pre-emptive therapy compared to placebo in a larger
Diagnostic tests for early detection of CMV infection
phase III study. There was a small impact on CMV DNA
loads in plasma (Winston et al, 2008; Marty et al, 2011). Historically, the CMV antigenaemia assay provided a rapid
Letermovir (AIC246), a maturation inhibitor of CMV, has way to detect CMV infection and is still used as a cost-
been studied in phase 2 trials as anti-CMV prophylaxis in effective screening tool in many centres worldwide. However,
133 HSCT patients with potentially encouraging results it suffers from low sensitivity, is not accurately quantitative
(Goldner et al, 2011). Neither of these drugs can be recom- and, in HSCT patients, is limited by the leucopenia evident in
mended for prophylaxis at present. the early stages post-transplant. Here, CMV antigenaemia
In summary, post-HSCT, in contrast to solid organ trans- testing may be negative despite active viral replication
plantation, ganciclovir-induced myelosuppression limits its (Gondo et al, 1994; Koehler et al, 1995). The availability of
use for prophylaxis. Routine use of aciclovir or valaciclovir is real time quantitative PCR (RQ-PCR) approaches has revolu-
relatively non-toxic but will result in some patients being tionized the area of CMV DNA monitoring. RQ-PCR tech-
overtreated and the effect in T-cell-depleted transplants is niques provide rapid, high throughput platforms that are
small. However, the potential benefits of prophylaxis using significantly less affected by white cell counts (Einsele et al,
these drugs in selected patients include reducing the need for 1995; Emery et al, 2000; Fishman et al, 2007; Gimeno et al,
hospital admission, for iv pre-emptive therapy, reducing 2008). The choice of sample type for routine monitoring lies
indirect effects of CMV reactivation on immune status post- between plasma and whole blood and remains controversial.
transplant and delaying CMV reactivation until the patient The choice is often based on practical issues surrounding
has recovered from the toxicity associated with the transplant local sample handling and preparation. There are few pub-
and is no longer on immunosuppression. lished reports supporting the superiority of one sample type
over the other. In the past, plasma was regarded as an easier
sample to extract, but this is now less of an issue as there are
Secondary prophylaxis for CMV
a number of automated extraction platforms that can handle
In patients who have had previous CMV disease prior to whole blood. Several studies have shown that more CMV
transplant or with recurrent episodes of CMV infection, DNA can be extracted from whole blood samples than from
especially in the context of T-cell depletion or GvHD, sec- plasma (Garrigue et al, 2006; Koidl et al, 2008; Bravo et al,
ondary prophylaxis should be considered, in conjunction 2011a). Responses to therapy may also be more predictable in

2013 John Wiley & Sons Ltd 29


British Journal of Haematology, 2013, 162, 2539
Guideline

whole blood compared to plasma given the higher CMV Timing of CMV screening samples
DNA loads in the former (Atkinson & Emery, 2011). Other
There is no clinical trial evidence demonstrating how often
body fluids, where white cells may be scanty, such as urine,
CMV DNA load in blood samples should be determined
bronchoalveolar lavage, endotracheal aspirates, amniotic fluid
(Freyer et al, 2010). However, weekly assessment following
and cerebrospinal fluid can also be rapidly analysed using
allogeneic transplantation is advisable for the first 36 months
RQ- PCR. However, to date, RQ-PCR has not been validated
(Hebart et al, 1997). The duration of monitoring will depend
for diagnosis of CMV end-organ disease and no recommen-
on the level of immunosuppression, the degree of immune
dations can be made for the use of this technology in this
reconstitution, the presence of GvHD and response to antiviral
setting.
therapy. In these clinical settings, prolonged monitoring for
612 months may be required.
Methodological issues
A diverse range of in-house and commercially available
Recommendations
RT-PCR assays are used in many diagnostic laboratories
throughout the UK. The primers, probes and assay condi- Real-time quantitative PCR is the preferred option for
tions used in these tests vary. Laboratories running in- monitoring CMV DNA levels in HSCT patients (Grade
house assays will regularly make up their own new 1B).
reagents as part of the assay master mix, as well as a set All diagnostic laboratories should deploy the CMV
of log dilutions of a separately amplified quantification international standard to allow CMV DNA loads to be
standard, also known as a calibrator. This is used in order compared between centres (Grade 1C).
to produce a standard curve from which the number of Monitoring of CMV DNA load should be undertaken at
copies of CMV DNA per ml of sample can be reported. A least weekly for the first 3 months post-HSCT (Grade 2C).
number of controls must be used when running a diag- CMV viral load monitoring should continue to 6
nostic PCR assay including: 12 months if the patient has chronic GvHD or pro-
A positive control that is detected at the lower limit of longed T-cell immunodeficiency (Grade 1B).
the assay.
At least one negative control to monitor for assay con-
tamination. Treatment thresholds
An internal amplification control that is an unrelated target
There is no consensus concerning the CMV load at which
to monitor for any substances inhibitory to the assay.
pre-emptive treatment should be initiated (Boeckh & Ljung-
A run control to monitor inter- and intra-assay variation.
man, 2009; Ljungman et al, 2011). Some centres will start
Run controls are available commercially although some antiviral therapy once CMV DNA has been detected at any
laboratories will produce their own controls. Standardiza- level in two consecutive samples, while others have set
tion of nucleic acid extraction involves running an extrac- much higher thresholds on the basis of local assays and
tion control in each assay as well as an inhibition control clinical outcome data. Unfortunately, these thresholds will
for each sample. Recently, a World Health Organization remain difficult to compare between centres until PCR test-
standard for genome amplification of CMV from clinical ing is fully standardized. A randomized trial comparing
samples was produced by the National Institute for Biolog- antigenaemia with RQ-PCR has indicated that a CMV load
ical Standards and Control (Freyer et al, 2010; Bravo et al, >10 000 copies/ml may be an acceptable threshold for initi-
2011a). There are still significant differences in CMV DNA ation of therapy. (Gerna et al, 2008; Boeckh & Ljungman,
load values reported by different laboratories. These differ- 2009; Mullier et al, 2009). This allows some patients to gen-
ences have previously been attributed to the variety of in- erate effective immune responses and avoid toxic drug ther-
house methods used. However, it is likely that differences apy while still treating susceptible patients pre-emptively,
will still be seen using commercial assays as well. A recent before the onset of CMV disease. Demonstration of active
comparison showed substantial differences in the nucleic CMV-specific T-cell responses may help select patients who
acid extraction efficiency of a number of automated sys- do not require treatment (Cwynarski et al, 2001; Hebart
tems and differences in the commercial RQ PCR assays et al, 2002; Ozdemir et al, 2002). In addition to the peak
included in the study. This was most marked at the lower viral load there is evidence that the initial viral load and
CMV DNA loads (Bravo et al, 2011a). It is likely that the the rate of viral load increase are important predictors of
international standard will be increasingly used to calibrate development of CMV disease (Emery et al, 2000). Risk-
assays on a regular basis. This will allow CMV DNA levels adapted strategies are likely to be needed and early treat-
to be compared between different centres and aid multi- ment is advised in R+/D- patients, especially in those with
centre trials (Hirsch et al, 2013). Involvement with national severe lymphopenia, high initial viral loads (e.g. >34 log10
external quality assurance schemes is strongly advised. copies/ml), rapid viral load doubling times (e.g. >1 log10

30 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

copies/ml in 3 d) or those who are heavily immunocompro- bioavailability of active drug compared to iv ganciclovir
mised. 5 mg/kg bd and showed equivalent efficacy (Lim et al, 2009).
Good absorption was also shown in the context of intestinal
GvHD grade III (Einsele et al, 2006). Large prospective ran-
Recommendations
domized trials comparing valganciclovir and ganciclovir are
Each transplant centre should have a risk-adapted pol- lacking in HSCT patients. However, there are now several
icy detailing threshold values for treatment of CMV single centre randomized studies published, including over
infection, taking into account patient factors and local 150 patients, comparing the use of oral valganciclovir
PCR methodology (Grade 2C). 900 mg bd with 5 mg/kg bd iv ganciclovir. All these studies
showed no difference in any measure of efficacy although the
power of each individual study to exclude non-inferiority
Antiviral agents used in pre-emptive therapy was limited (Einsele et al, 2006; van der Heiden et al, 2006;
Lim et al, 2009; Chawla et al, 2011; Ruiz-Camps et al, 2011).
Ganciclovir. Ganciclovir requires phosphorylation by the UL
Further evidence of valganciclovir effectiveness was derived
97 viral kinase. It is virustatic. It inhibits viral replication by
from several single arm case series. These studies reported
competing with deoxyguanosine triphosphate as a substrate
response rates comparable to historic controls using ganciclo-
for viral DNA polymerase (UL 54) (Sia & Patel, 2000). Resis-
vir (Ayala et al, 2006; Rzepecki et al, 2008; Liu et al, 2010).
tance is mediated by mutations in genes encoding either of
CMV resistance rates appeared low following pre-emptive
these enzymes (Chou, 2008). Ganciclovir has been extensively
therapy with valganciclovir for CMV in HSCT (Allice et al,
used to treat CMV infection and disease (Goodrich et al,
2009). In solid organ transplants, where valganciclovir was
1991; Manteiga et al, 1998). It is still regarded as first choice
used extensively for prophylaxis it was shown to be non-infe-
pre-emptive therapy in most HSCT centres (Pollack et al,
rior to ganciclovir in preventing both CMV infection and
2011). The standard dose is 5 mg/kg intravenously twice
disease, with low rates of CMV resistance (Andrews et al,
daily for 14 d with maintenance of 5 mg/kg daily for a fur-
2011). After pre-emptive therapy in solid organ transplants
ther 714 d. Side effects and supportive therapies are listed
the kinetics of decrease in viral load were the same as for
in Table I.
intravenous ganciclovir (Mattes et al, 2005).
Valganciclovir. Valganciclovir is the valine ester of ganciclo-
Foscarnet. Foscarnet does not require phosphorylation and
vir, it is hydrolysed to ganciclovir after oral absorption. A
is virustatic through inhibition of viral DNA polymerase
small randomized trial in HSCT patients showed that oral
(Sia & Patel, 2000). Two randomized studies have compared
valganciclovir 900 mg twice daily led to higher effective
the outcomes of foscarnet versus ganciclovir pre-emptive
therapy of CMV infection post-allograft (Moretti et al,
Table I. Side effects of drugs used to treat CMV infection. 1998; Reusser et al, 2002). Foscarnet 90 mg/kg was com-
pared with ganciclovir 5 mg/kg, both administered
Drug Side effect Treatment/support
12 hourly iv, using CMV antigenaemia as a guide to com-
Ganciclovir Myelosuppression Granulocyte mencement of therapy (Moretti et al, 1998). Of 40 patients
colony-stimulating were randomly allocated to either treatment. There were no
factor statistically significant differences in rates of resolution of
Valganciclovir Haemolysis Toxicity-specific dose antigenaemia, treatment failure or progression to CMV
Nausea/vomiting reductions disease (510% in both groups). Both drugs required dose
Fever Regular twice daily
reductions of at least 20% in approximately half of all
Rash 5HT3 inhibitors
patients. A larger EBMT study compared 60 mg/kg foscar-
Mental state changes
net (n = 110) with ganciclovir 5 mg/kg (n = 103) both 12
Urinary symptoms
Foscarnet Electrolyte abnormalities Aggressive electrolyte hourly using either RQ-PCR or antigenaemia as a trigger to
Renal impairment replacement commence therapy (Reusser et al, 2002). No differences in
Nausea/vomiting Fluid support CMV disease occurrence, treatment-related mortality (TRM)
Genitourinary Regular twice daily or event-free survival were seen. Renal insufficiency was
Urinary symptoms 5HT3 inhibitors more common with foscarnet and myelosuppression with
Toxicity-specific dose ganciclovir. Further evidence of foscarnet efficacy comes
reductions from case series. In one, 313 patients were treated for CMV
Cidofovir Renal impairment Probenecid disease (n = 65), or were given pre-emptive therapy of
Nausea/vomiting Aggressive prehydration
CMV reactivation in related- donor transplants (n = 248)
Ocular Regular twice daily
(Asakura et al, 2010). Of 194 patients had failed previous
5HT3 inhibitors
ganciclovir due to lack of efficacy (n = 99) or myelosup-
Ophthalmology review
pression in (n = 95). Fifty two percent of patients with

2013 John Wiley & Sons Ltd 31


British Journal of Haematology, 2013, 162, 2539
Guideline

CMV disease and 90% of pre-emptively treated cases a possible reduction in TRM compared with historical con-
responded. More limited data were reported supporting trols (Bacigalupo et al, 1996b). However, not enough
foscarnet use in cord blood transplantation (Narimatsu evidence exists to make a recommendation regarding combi-
et al, 2007; Takami et al, 2007). nation therapy in this context.

Cidofovir. Cidofovir is a nucleotide analogue that does not


Recommendations
require phosphorylation by viral UL97 kinase for activation.
The largest case series using cidofovir has been published by Ganciclovir is recommended as first line pre-emptive
the infectious disease working party of EBMT (Ljungman therapy for CMV in HSCT patients (Grade 1A).
et al, 2001). 82 patients were treated as primary (n = 24) or Oral valganciclovir is a useful alternative when gastroin-
secondary (n = 38) pre-emptive therapy for CMV infection testinal absorption is normal or minimally impaired
or for CMV disease (n = 20). Most patients were treated (Grade 1B).
with 5 mg/kg per week (range 15) for a median duration of Foscarnet is recommended as an alternative first line
3 weeks. Response rates were 50% for CMV disease, 66% for agent if neutropenia is present or for ganciclovir treat-
primary and 62% for secondary CMV infection. Other smal- ment failures (Grade 1A).
ler series demonstrated that cidofovir was effective but Pre-emptive therapy with cidofovir can be considered as
response rates varied from 50 to 90% with most series third line in patients unresponsive or intolerant of a
reporting significant failure rates (Chakrabarti et al, 2001; ganciclovir preparation or foscarnet (Grade 2B).
Platzbecker et al, 2001; Cesaro et al, 2005). Side effects are
shown in Table I. A more tolerable alternative dosing regi-
men of 1 mg/kg three times a week has been used to treat Switching pre-emptive therapy
patients with adenovirus infection (Lindemans et al, 2010)
The doubling time of CMV DNA load in untreated patients
but there is no information using this strategy for the treat-
is 12 d on average but may be even more rapid (Emery
ment of CMV. Modified hexadecyloxypropyl-cidofovir
et al, 1999; Buyck et al, 2010) At the start of pre-emptive
(HDP-CDV/CMX001), or other liposomal preparations of
therapy, before drugs can have a major impact, increases in
cidofovir have been developed but their use has only been
CMV DNA load occur in approximately one-third of
studied in small numbers of patients (Hostetler, 2010; Bravo
patients due to underlying immunosuppression and rapidity
et al, 2011b; Gokulgandhi et al, 2012) and no recommenda-
of replication (Buyck et al, 2010; Park et al, 2011). This
tions are possible.
should be appreciated and no changes in therapy instituted
in the first 14 d, in the absence of overt CMV disease or
Combination anti-viral drug therapy dramatic rises in viral PCR log values. It is difficult to be
dogmatic about triggers for changing pre-emptive antiviral
The theoretical advantages of in vivo combination therapy
drugs, as responses to treatment can be very slow in pro-
are reduced toxicity, and increased efficacy, both demon-
foundly lymphopenic patients. An increase in viral load by
strated in vitro (Manischewitz et al, 1990; Manion et al,
one log after 2 weeks of therapy is one option. Switching
1996): the primary disadvantage is generation of resistance,
therapy is seldom recommended earlier, and can often be
particularly in the T-cell-deficient, immunocompromised
delayed further, even in high-risk patients, unless disease
host, post-allograft. The combination of foscarnet and ganci-
has progressed clinically, or viral copy number is increasing
clovir (both at half dose) versus full dose ganciclovir was
rapidly.
evaluated in a small randomized study (n = 48) in transplant
patients, including HSCT recipients (Mattes et al, 2004).
There was less myelosuppression but overall increased toxic- Recommendations
ity, primarily renal, in the combination arm. Importantly
In patients where CMV DNA loads in blood increase by
though underpowered to show a clinical effect 71% of the
1 log10 over 2 weeks of pre-emptive therapy with a first
ganciclovir group reached the primary clinical endpoint of
line drug, an alternative agent and drug resistance profil-
viral clearance at 14 d compared to 50% in the combination
ing should be considered (Grade 2C).
group. At present, not enough evidence exists to support
treatment using these two drugs at sub-conventional doses.
Bacigalupo et al (1996a) have also reported combining
Antiviral drug resistance
foscarnet and ganciclovir at full dose, adjusted for organ
function, aiming for 180 mg/kg/d foscarnet and 10 mg/kg/d Drug resistance is relatively uncommon in the stem cell trans-
ganciclovir and delivering 5060% of these doses. Drugs were plant setting. Resistance to ganciclovir is estimated at 28% of
delivered together at full dose for 2 weeks followed by main- treated patients and generally occurs after 23 months of
tenance, where drugs were given at full dose but on alternate prolonged therapy (Marfori et al, 2007) but can occur after
days. This approach was effective and the authors suggested initial therapy (van der Beek et al, 2012; Kim et al, 2012).

32 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

Close liaison between transplant physicians and clinical virolo- net (or vice versa). Combination therapy is theoretically
gists is essential. Resistance is less common in drug-naive attractive but may prove difficult to deliver (Manion et al,
patients than in patients experiencing repeated episodes of late 1996). Foscarnet and ganciclovir can be combined, at full
onset CMV infection. Resistance should be suspected in dose (Bacigalupo et al, 1996a). Cidofovir is an alternative to
patients on antiviral drugs where the CMV DNA load has been ganciclovir, and is also synergistic with foscarnet in vitro, but
static or has increased for more than 2 weeks, although clini- should only be used if ganciclovir has failed or resistance has
cal resistance is still more likely in lymphopenic drug-nave been demonstrated (Manion et al, 1996) as both drugs are
patients. If drug resistance is suspected, samples should be sent nephrotoxic. Maribivir inhibits the enzyme that activates
for CMV UL97 and UL54 sequencing. ganciclovir so this combination should be avoided (Chou &
Marousek, 2006), although combining maribivir with foscar-
net or cidofovir is an option. Experimental therapies include
Recommendations
high dose maribivir, which has been successfully reported in
Drug resistance should be considered if the CMV DNA six patients (Avery et al, 2010a), though no firm recommen-
load in blood fails to respond after 14 d of therapy, dation can be made in the light of maribavir failing to reach
especially in non-lymphopenic or multiply pre-treated its target in the phase III study. Despite some reports of lack
patients (Grade 2C). of efficacy, IVIG in combination with antiviral therapy
Sequence analysis of the UL97 and UL54 genes is the remains the standard of care for CMV disease in many
preferred option for monitoring resistance to currently centres (Schmidt et al, 1988; Ljungman et al, 1992; Machado
available drugs (Grade 1B). et al, 2000). There appears to be no advantage to using
CMV-specific immunoglobulin (Ljungman et al, 1998). Every
attempt should be made to reduce or stop immunosuppres-
Management of CMV disease sion, especially corticosteroids. Switching a calcineurin inhib-
itor to sirolimus is a possible therapeutic intervention as
A multidisciplinary approach to the management of CMV
several studies have suggested a beneficial anti-CMV effect of
disease is important. Specific investigations and treatment
this immunosuppressant (Marty et al, 2007; Ozaki et al,
depends on which organs are affected and whether disease
2007; Demopoulos et al, 2008). The place of leflunamide
has developed de novo or from asymptomatic CMV infection
post-HSCT is still to be defined (Avery et al, 2010b; Chacko
while on first-line therapy. De novo CMV disease can be trea-
& John, 2012). Experimental adoptive immunotherapy with
ted with ganciclovir 5 mg/kg, or foscarnet 90 mg/kg twice
donor or third party anti-CMV-specific T cells may be avail-
daily for 2 weeks, followed by maintenance, although more
able within clinical trials but cannot be recommended as
prolonged treatment may be necessary. Careful ophthalmo-
standard at present.
logical review is mandatory as intraocular therapy may be
required (Song et al, 2008). Higher baseline CMV DNA
loads, faster kinetics of replication and a slower viral decay Recommendations
rate after starting therapy are associated with increased rates
A multidisciplinary approach to management of CMV
of treatment failure (Mattes et al, 2004) and mandate closer
disease is required (Grade 1C).
monitoring of these patients with lower thresholds for
De novo CMV disease should be treated with ganciclovir
diagnostic interventions. If CMV disease occurs during first
or foscarnet, plus intravenous immunoglobulin (Gra-
line pre-emptive therapy or if de novo CMV pneumonia is
de1B).
progressive at any time, then alternative strategies should be
CMV disease that develops while on pre-emptive ther-
considered. Failure to respond is more likely to be related to
apy or is clinically progressive requires drug resistance
the hosts inability to control the virus as a result of
testing, increased drug doses and/or a change of drug
impaired T cell immunity than to drug resistance, unless
(Grade 1B).
multiple courses of therapy have been given previously
Reduction in immunosuppression, especially corticoste-
(van der Beek et al, 2012). However, in critically unwell
roid dosage, is strongly recommended (Grade 1B).
patients, where available, drug resistance testing should be
performed early to optimize pharmacological management.
Alternative treatment approaches in this setting include
Disclaimer
increasing ganciclovir doses to 75 mg/kg twice a day (Bo-
eckh & Ljungman, 2009) or switching ganciclovir to foscar- The authors report no conflicts of interest.

References binstein, P. & Stevens, C.E. (2006) Umbilical Allice, T., Busca, A., Locatelli, F., Falda, M., Pitta-
cord blood transplantation and cytomegalovirus: luga, F. & Ghisetti, V. (2009) Valganciclovir as
Albano, M.S., Taylor, P., Pass, R.F., Scaradavou, posttransplantation infection and donor screen- pre-emptive therapy for cytomegalovirus infec-
A., Ciubotariu, R., Carrier, C., Dobrila, L., Ru- ing. Blood, 108, 42754282. tion post-allogenic stem cell transplantation:

2013 John Wiley & Sons Ltd 33


British Journal of Haematology, 2013, 162, 2539
Guideline

implications for the emergence of drug-resistant Halkes, C.J., Claas, E.C. & Kroes, A.C. (2012) (2011a) Comparative evaluation of three auto-
cytomegalovirus. Journal of Antimicrobial Che- Failure of pre-emptive treatment of cytomegalo- mated systems for DNA extraction in conjunc-
motherapy, 63, 600608. virus infections and antiviral resistance in stem tion with three commercially available real-time
Andrews, P.A., Emery, V.C. & Newstead, C. (2011) cell transplant recipients. Antiviral Therapy, 17, PCR assays for quantitation of plasma cytomeg-
Summary of the British transplantation society 4551. alovirus DNAemia in allogeneic stem cell trans-
guidelines for the prevention and management Behrendt, C.E., Rosenthal, J., Bolotin, E., Nakam- plant recipients. Journal of Clinical Microbiology,
of CMV disease after solid organ transplanta- ura, R., Zaia, J. & Forman, S.J. (2009) Donor 49, 28992904.
tion. Transplantation, 92, 11811187. and recipient CMV serostatus and outcome of Bravo, F.J., Bernstein, D.I., Beadle, J.R., Hostetler,
Asakura, M., Ikegame, K., Yoshihara, S., Tanigu- pediatric allogeneic HSCT for acute leukemia in K.Y. & Cardin, R.D. (2011b) Oral hexadecyloxy-
chi, S., Mori, T., Etoh, T., Takami, A., Yoshida, the era of CMV-preemptive therapy. Biology of propyl-cidofovir therapy in pregnant guinea pigs
T., Fukuda, T., Hatanaka, K., Kanamori, H., Blood and Marrow Transplantation, 15, 5460. improves outcome in the congenital model of
Yujiri, T., Atsuta, Y., Sakamaki, H., Suzuki, R. & Blanquer, J., Chilet, M., Benet, I., Aguilar, G., cytomegalovirus infection. Antimicrobial Agents
Ogawa, H. (2010) Use of foscarnet for cytomeg- Munoz-Cobo, B., Tellez, A., Costa, E., Bravo, D. and Chemotherapy, 55, 3541.
alovirus infection after allogeneic hematopoietic & Navarro, D. (2011) Immunological insights Broers, A.E., van Der Holt, R., van Esser, J.W.,
stem cell transplantation from a related donor. into the pathogenesis of active CMV infection Gratama, J.W., Henzen-Logmans, S., Kuenen-
International Journal of Hematology, 92, in non-immunosuppressed critically ill patients. Boumeester, V., Lowenberg, B. & Cornelissen,
351359. Journal of Medical Virology, 83, 19661971. J.J. (2000) Increased transplant-related mor-
Atkinson, C. & Emery, V.C. (2011) Cytomegalovi- Boeckh, M. & Ljungman, P. (2009) How we treat bidity and mortality in CMV-seropositive
rus quantification: where to next in optimising cytomegalovirus in hematopoietic cell transplant patients despite highly effective prevention of
patient management? Journal of Clinical Virol- recipients. Blood, 113, 57115719. CMV disease after allogeneic T-cell-depleted
ogy, 51, 223228. Boeckh, M., Gooley, T.A., Myerson, D., Cunning- stem cell transplantation. Blood, 95, 2240
Avery, R.K., Marty, F.M., Strasfeld, L., Lee, I., Arri- ham, T., Schoch, G. & Bowden, R.A. (1996) 2245.
eta, A., Chou, S., Tatarowicz, W. & Villano, S. Cytomegalovirus pp 65 antigenemia-guided early Brown, J.A., Stevenson, K., Kim, H.T., Cutler, C.,
(2010a) Oral maribavir for treatment of refrac- treatment with ganciclovir versus ganciclovir at Ballen, K., McDonough, S., Reynolds, C., Herre-
tory or resistant cytomegalovirus infections in engraftment after allogeneic marrow transplanta- ra, M., Liney, D., Ho, V., Kao, G., Armand, P.,
transplant recipients. Transplant Infectious tion: a randomized double-blind study. Blood, Koreth, J., Alyea, E., McAfee, S., Attar, E., Dey,
Disease, 12, 489496. 88, 40634071. B., Spitzer, T., Soiffer, R., Ritz, J., Antin, J.H. &
Avery, R.K., Mossad, S.B., Poggio, E., Lard, M., Boeckh, M., Bowden, R.A., Storer, B., Chao, N.J., Boussiotis, V.A. (2010) Clearance of CMV vire-
Budev, M., Bolwell, B., Waldman, W.J., Braun, Spielberger, R., Tierney, D.K., Gallez-Hawkins, mia and survival after double umbilical cord
W., Mawhorter, S.D., Fatica, R., Krishnamurthi, G., Cunningham, T., Blume, K.G., Levitt, D. & blood transplantation in adults depends on
V., Young, J.B., Shrestha, R., Stephany, B., Zaia, J.A. (2001) Randomized, placebo- reconstitution of thymopoiesis. Blood, 115,
Lurain, N. & Yen-Lieberman, B. (2010b) Utility controlled, double-blind study of a cytomegalo- 41114119.
of leflunomide in the treatment of complex virus-specific monoclonal antibody (MSL-109) van Burik, J.A., Carter, S.L., Freifeld, A.G., High,
cytomegalovirus syndromes. Transplantation, 90, for prevention of cytomegalovirus infection after K.P., Godder, K.T., Papanicolaou, G.A., Mendi-
419426. allogeneic hematopoietic stem cell transplanta- zabal, A.M., Wagner, J.E., Yanovich, S. &
Ayala, E., Greene, J., Sandin, R., Perkins, J., Field, tion. Biology of Blood and Marrow Transplanta- Kernan, N.A. (2007) Higher risk of cytomegalo-
T., Tate, C., Fields, K.K. & Goldstein, S. (2006) tion, 7, 343351. virus and aspergillus infections in recipients of
Valganciclovir is safe and effective as pre-emp- Boeckh, M., Leisenring, W., Riddell, S.R., Bowden, T cell-depleted unrelated bone marrow: analysis
tive therapy for CMV infection in allogeneic R.A., Huang, M.L., Myerson, D., Stevens-Ayers, of infectious complications in patients treated
hematopoietic stem cell transplantation. Bone T., Flowers, M.E., Cunningham, T. & Corey, L. with T cell depletion versus immunosuppressive
Marrow Transplantation, 37, 851856. (2003) Late cytomegalovirus disease and mortal- therapy to prevent graft-versus-host disease.
Bacigalupo, A., Bregante, S., Tedone, E., Isaza, A., ity in recipients of allogeneic hematopoietic Biology of Blood and Marrow Transplantation,
Van Lint, M.T., Moro, F., Trespi, G., Occhini, stem cell transplants: importance of viral load 13, 14871498.
D., Gualandi, F., Lamparelli, T. & Marmont, and T-cell immunity. Blood, 101, 407414. Burns, L.J., Miller, W., Kandaswamy, C., DeFor,
A.M. (1996a) Combined foscarnet -ganciclovir Boeckh, M., Fries, B. & Nichols, W.G. (2004) T.E., MacMillan, M.L., Van Burik, J.A. & Weis-
treatment for cytomegalovirus infections after Recent advances in the prevention of CMV dorf, D.J. (2002) Randomized clinical trial of
allogeneic hemopoietic stem cell transplantation infection and disease after hematopoietic stem ganciclovir vs acyclovir for prevention of cyto-
(Hsct). Bone Marrow Transplantation, 18 (Suppl. cell transplantation. Pediatric Transplantation, 8 megalovirus antigenemia after allogeneic trans-
2), 110114. (Suppl. 5), 1927. plantation. Bone Marrow Transplantation, 30,
Bacigalupo, A., Bregante, S., Tedone, E., Isaza, A., Bowden, R.A., Fisher, L.D., Rogers, K., Cays, M. & 945951.
Van Lint, M.T., Trespi, G., Occhini, D., Gua- Meyers, J.D. (1991) Cytomegalovirus (CMV)- Buyck, H.C., Prentice, H.G., Griffiths, P.D. &
landi, F., Lamparelli, T. & Marmont, A.M. specific intravenous immunoglobulin for the Emery, V.C. (2010) The risk of early and late
(1996b) Combined foscarnet-ganciclovir treat- prevention of primary CMV infection and dis- CMV DNAemia associated with Campath use in
ment for cytomegalovirus infections after alloge- ease after marrow transplant. Journal of Infec- stem cell transplant recipients. Bone Marrow
neic hemopoietic stem cell transplantation. tious Diseases, 164, 483487. Transplantation, 45, 12121219.
Transplantation, 62, 376380. Bowden, R.A., Slichter, S.J., Sayers, M., Weisdorf, Cesaro, S., Zhou, X., Manzardo, C., Buonfrate,
Beck, J.C., Wagner, J.E., DeFor, T.E., Brunstein, D., Cays, M., Schoch, G., Banaji, M., Haake, R., D., Cusinato, R., Tridello, G., Mengoli, C.,
C.G., Schleiss, M.R., Young, J.A., Weisdorf, Welk, K., Fisher, L., McCullough, J. & Miller, Palu, G. & Messina, C. (2005) Cidofovir for
D.H., Cooley, S., Miller, J.S. & Verneris, M.R. W. (1995) A comparison of filtered leukocyte- cytomegalovirus reactivation in pediatric
(2010) Impact of cytomegalovirus (CMV) reacti- reduced and cytomegalovirus (CMV) seronega- patients after hematopoietic stem cell trans-
vation after umbilical cord blood transplanta- tive blood products for the prevention of trans- plantation. Journal of Clinical Virology, 34,
tion. Biology of Blood and Marrow fusion-associated CMV infection after marrow 129132.
Transplantation, 16, 215222. transplant. Blood, 86, 35983603. Chacko, B. & John, G.T. (2012) Leflunomide for
van der Beek, M.T., Marijt, E.W., Vossen, A.C., Bravo, D., Clari, M.A., Costa, E., Munoz-Cobo, B., cytomegalovirus: bench to bedside. Transplant
van der Blij-de Brouwer, C.S., Wolterbeek, R., Solano, C., Jose Remigia, M. & Navarro, D. Infectious Disease, 14, 111120.

34 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

Chakrabarti, S., Collingham, K.E., Osman, H., Fe- Einsele, H., Reusser, P., Bornhauser, M., Kalhs, P., transplant recipients. Journal of Clinical Microbi-
gan, C.D. & Milligan, D.W. (2001) Cidofovir as Ehninger, G., Hebart, H., Chalandon, Y., Kro- ology, 46, 33113318.
primary pre-emptive therapy for post-transplant ger, N., Hertenstein, B. & Rohde, F. (2006) Oral Gokulgandhi, M.R., Barot, M., Bagui, M., Pal, D.
cytomegalovirus infections. Bone Marrow Trans- valganciclovir leads to higher exposure to ganci- & Mitra, A.K. (2012) Transporter-targeted lipid
plantation, 28, 879881. clovir than intravenous ganciclovir in patients prodrugs of cyclic cidofovir: a potential
Chawla, J.S., Ghobadi, A., Mosley, J. 3rd, Ver- following allogeneic stem cell transplantation. approach for the treatment of cytomegalovirus
kruyse, L., Trinkaus, K., Abboud, C.N., Cashen, Blood, 107, 30023008. retinitis. Journal of Pharmaceutical Sciences, 101,
A.F., Stockerl-Goldstein, K.E., Uy, G.L., Wester- Elmaagacli, A.H., Steckel, N.K., Koldehoff, M., 32493263.
velt, P., Dipersio, J.F. & Vij, R. (2011) Oral Hegerfeldt, Y., Trenschel, R., Ditschkowski, M., Goldner, T., Hewlett, G., Ettischer, N., Ruebsa-
valganciclovir versus ganciclovir as delayed pre- Christoph, S., Gromke, T., Kordelas, L., Ottinger, men-Schaeff, H., Zimmermann, H. & Lischka,
emptive therapy for patients after allogeneic H.D., Ross, R.S., Horn, P.A., Schnittger, S. & P. (2011) The novel anticytomegalovirus com-
hematopoietic stem cell transplant: a pilot trial Beelen, D.W. (2011) Early human cytomegalovi- pound AIC246 (Letermovir) inhibits human
(040274) and review of the literature. Trans- rus replication after transplantation is associated cytomegalovirus replication through a specific
plant Infectious Disease, 14, 259267. with a decreased relapse risk: evidence for a puta- antiviral mechanism that involves the viral ter-
Chiesa, R., Gilmour, K., Qasim, W., Adams, S., tive virus-versus-leukemia effect in acute myeloid minase. Journal of Virology, 85, 1088410893.
Worth, A.J., Zhan, H., Montiel-Equihua, C.A., leukemia patients. Blood, 118, 14021412. Gondo, H., Minematsu, T., Harada, M., Akashi,
Derniame, S., Cale, C., Rao, K., Hiwarkar, P., Emery, V.C., Cope, A.V., Bowen, E.F., Gor, D. & K., Hayashi, S., Taniguchi, S., Yamasaki, K.,
Hough, R., Saudemont, A., Fahrenkrog, C.S., Griffiths, P.D. (1999) The dynamics of human Shibuya, T., Takamatsu, Y., Teshima, T.
Goulden, N., Amrolia, P.J. & Veys, P. (2012) cytomegalovirus replication in vivo. Journal of (1994) Cytomegalovirus (CMV) antigenaemia
Omission of in vivo T-cell depletion promotes Experimental Medicine, 190, 177182. for rapid diagnosis and monitoring of CMV-
rapid expansion of naive CD4 + cord blood lym- Emery, V.C., Sabin, C.A., Cope, A.V., Gor, D., associated disease after bone marrow trans-
phocytes and restores adaptive immunity within Hassan-Walker, A.F. & Griffiths, P.D. (2000) plantation. British Journal of Haematology, 86,
2 months after unrelated cord blood transplant. Application of viral-load kinetics to identify 130137.
British Journal of Haematology, 156, 656666. patients who develop cytomegalovirus disease Goodrich, J.M., Mori, M., Gleaves, C.A., Du
Chou, S. (2008) Cytomegalovirus UL97 mutations after transplantation. Lancet, 355, 20322036. Mond, C., Cays, M., Ebeling, D.F., Buhles,
in the era of ganciclovir and maribavir. Reviews Fishman, J.A., Emery, V., Freeman, R., Pascual, W.C., DeArmond, B. & Meyers, J.D. (1991)
in Medical Virology, 18, 233246. M., Rostaing, L., Schlitt, H.J., Sgarabotto, D., Early treatment with ganciclovir to prevent cyto-
Chou, S. & Marousek, G.I. (2006) Maribavir Torre-Cisneros, J. & Uknis, M.E. (2007) Cyto- megalovirus disease after allogeneic bone mar-
antagonizes the antiviral action of ganciclovir on megalovirus in transplantation challenging the row transplantation. New England Journal of
human cytomegalovirus. Antimicrobial Agents status quo. Clinical Transplantation, 21, 149 Medicine, 325, 16011607.
and Chemotherapy, 50, 34703472. 158. Goodrich, J.M., Bowden, R.A., Fisher, L., Keller,
Cwynarski, K., Ainsworth, J., Cobbold, M., Freyer, H.A., Anderson, R. & Minor, P.D. (2010) C., Schoch, G. & Meyers, J.D. (1993) Ganciclo-
Wagner, S., Mahendra, P., Apperley, J., Gold- Collaborative study to evaluate the proposed 1st vir prophylaxis to prevent cytomegalovirus dis-
man, J., Craddock, C. & Moss, P.A. (2001) WHO international standard for human cyto- ease after allogeneic marrow transplant. Annals
Direct visualization of cytomegalovirus-specific megalovirus for nucleic acid amplification-based of Internal Medicine, 118, 173178.
T-cell reconstitution after allogeneic stem cell assays. WHO/BS/10.2138, pp. 140. Grigoleit, G.U., Kapp, M., Hebart, H., Fick, K.,
transplantation. Blood, 97, 12321240. Garrigue, I., Boucher, S., Couzi, L., Caumont, A., Beck, R., Jahn, G. & Einsele, H. (2007) Den-
Demopoulos, L., Polinsky, M., Steele, G., Mines, Dromer, C., Neau-Cransac, M., Tabrizi, R., dritic cell vaccination in allogeneic stem cell
D., Blum, M., Caulfield, M., Adamkovic, A., Schrive, M.H., Fleury, H. & Lafon, M.E. (2006) recipients: induction of human cytomegalovirus
Liu, Q., Harler, M.B., Hahn, C. & Singh, A. Whole blood real-time quantitative PCR for (HCMV)-specific cytotoxic T lymphocyte
(2008) Reduced risk of cytomegalovirus infec- cytomegalovirus infection follow-up in trans- responses even in patients receiving a transplant
tion in solid organ transplant recipients treated plant recipients. Journal of Clinical Virology, 36, from an HCMV-seronegative donor. Journal of
with sirolimus: a pooled analysis of clinical tri- 7275. Infectious Diseases, 196, 699704.
als. Transplant Proc, 40, 14071410. George, B., Kerridge, I., Gilroy, N., Huang, G., Guerrero, A., Riddell, S.R., Storek, J., Stevens-
Eid, A.J., Arthurs, S.K., Deziel, P.J., Wilhelm, M.P. Hertzberg, M., Gottlieb, D. & Bradstock, K. Ayers, T., Storer, B., Zaia, J.A., Forman, S.,
& Razonable, R.R. (2008) Emergence of drug- (2010) Fludarabine-based reduced intensity con- Negrin, R.S., Chauncey, T., Bensinger, W. & Bo-
resistant cytomegalovirus in the era of valganci- ditioning transplants have a higher incidence of eckh, M. (2012) Cytomegalovirus viral load and
clovir prophylaxis: therapeutic implications and cytomegalovirus reactivation compared with virus-specific immune reconstitution after
outcomes. Clinical Transplantation, 22, 162170. myeloablative transplants. Bone Marrow Trans- peripheral blood stem cell versus bone marrow
Einsele, H., Ehninger, G., Hebart, H., Wittkowski, plantation, 45, 849855. transplantation. Biology of Blood and Marrow
K.M., Schuler, U., Jahn, G., Mackes, P., Herter, Gerna, G., Lilleri, D., Caldera, D., Furione, M., Transplantation, 18, 6675.
M., Klingebiel, T., Loffler, J., Wagner, S. & Mul- Zenone Bragotti, L. & Alessandrino, E.P. (2008) Hakki, M., Riddell, S.R., Storek, J., Carter, R.A.,
ler, C.A. (1995) Polymerase chain reaction mon- Validation of a DNAemia cutoff for preemptive Stevens-Ayers, T., Sudour, P., White, K., Corey,
itoring reduces the incidence of cytomegalovirus therapy of cytomegalovirus infection in adult L. & Boeckh, M. (2003) Immune reconstitution
disease and the duration and side effects of hematopoietic stem cell transplant recipients. to cytomegalovirus after allogeneic hematopoi-
antiviral therapy after bone marrow transplanta- Bone Marrow Transplantation, 41, 873879. etic stem cell transplantation: impact of host
tion. Blood, 86, 28152820. Gimeno, C., Solano, C., Latorre, J.C., Hernandez- factors, drug therapy, and subclinical reactiva-
Einsele, H., Roosnek, E., Rufer, N., Sinzger, C., Boluda, J.C., Clari, M.A., Remigia, M.J., Furio, tion. Blood, 102, 30603067.
Riegler, S., Loffler, J., Grigoleit, U., Moris, A., S., Calabuig, M., Tormo, N. & Navarro, D. Hanley, P.J., Shaffer, D.R., Cruz, C.R., Ku, S.,
Rammensee, H.G., Kanz, L., Kleihauer, A., (2008) Quantification of DNA in plasma by an Tzou, B., Liu, H., Demmler-Harrison, G.,
Frank, F., Jahn, G. & Hebart, H. (2002) Infusion automated real-time PCR assay (cytomegalovi- Heslop, H.E., Rooney, C.M., Gottschalk, S. &
of cytomegalovirus (CMV)-specific T cells for rus PCR kit) for surveillance of active cytomega- Bollard, C.M. (2011) Expansion of T cells tar-
the treatment of CMV infection not responding lovirus infection and guidance of preemptive geting multiple antigens of cytomegalovirus,
to antiviral chemotherapy. Blood, 99, 39163922. therapy for allogeneic hematopoietic stem cell Epstein-Barr virus and adenovirus to provide

2013 John Wiley & Sons Ltd 35


British Journal of Haematology, 2013, 162, 2539
Guideline

broad antiviral specificity after stem cell trans- Kollman, C., Howe, C.W., Anasetti, C., Antin, Bone Marrow Transplant Group. Clinical Infec-
plantation. Cytotherapy, 13, 976986. J.H., Davies, S.M., Filipovich, A.H., Hegland, J., tious Diseases, 14, 831835.
Hazar, V., Kansoy, S., Kupesiz, A., Aksoylar, S., Kamani, N., Kernan, N.A., King, R., Ratanatha- Ljungman, P., Cordonnier, C., Einsele, H., Bender-
Kantar, M. & Yesilipek, A. (2004) High-dose rathorn, V., Weisdorf, D. & Confer, D.L. (2001) Gotze, C., Bosi, A., Dekker, A., De la Camara,
acyclovir and pre-emptive ganciclovir in preven- Donor characteristics as risk factors in recipients R., Gmur, J., Newland, A.C., Prentice, H.G.,
tion of cytomegalovirus disease in pediatric after transplantation of bone marrow from Robinson, A.J., Rovira, M., Rosler, W. & Veil,
patients following peripheral blood stem cell unrelated donors: the effect of donor age. Blood, D. (1998) Use of intravenous immune globulin
transplantation. Bone Marrow Transplantation, 98, 20432051. in addition to antiviral therapy in the treatment
33, 931935. Kroger, N., Zabelina, T., Kruger, W., Renges, H., of CMV gastrointestinal disease in allogeneic
Hebart, H., Schroder, A., Loffler, J., Klingebiel, T., Stute, N., Schrum, J., Kabisch, H., Schafhausen, bone marrow transplant patients: a report from
Martin, H., Wassmann, B., Gerneth, F., Rabe- P., Jaburg, N., Loliger, C., Schafer, P., Hinke, A. the European Group for Blood and Marrow
nau, H., Jahn, G., Kanz, L., Muller, C.A. & Ein- & Zander, A.R. (2001) Patient cytomegalovirus Transplantation (EBMT). Infectious Diseases
sele, H. (1997) Cytomegalovirus monitoring by seropositivity with or without reactivation is the Working Party of the EBMT. Bone Marrow
polymerase chain reaction of whole blood sam- most important prognostic factor for survival Transplantation, 21, 473476.
ples from patients undergoing autologous bone and treatment-related mortality in stem cell Ljungman, P., Deliliers, G.L., Platzbecker, U.,
marrow or peripheral blood progenitor cell transplantation from unrelated donors using Matthes-Martin, S., Bacigalupo, A., Einsele, H.,
transplantation. Journal of Infectious Diseases, pretransplant in vivo T-cell depletion with anti- Ullmann, J., Musso, M., Trenschel, R., Ribaud,
175, 14901493. thymocyte globulin. British Journal of Haematol- P., Bornhauser, M., Cesaro, S., Crooks, B.,
Hebart, H., Daginik, S., Stevanovic, S., Grigoleit, ogy, 113, 10601071. Dekker, A., Gratecos, N., Klingebiel, T., Tagliaf-
U., Dobler, A., Baur, M., Rauser, G., Sinzger, C., Lamba, R., Carrum, G., Myers, G.D., Bollard, erri, E., Ullmann, A.J., Wacker, P. & Cordon-
Jahn, G., Loeffler, J., Kanz, L., Rammensee, H.G. C.M., Krance, R.A., Heslop, H.E., Brenner, M.K. nier, C. (2001) Cidofovir for cytomegalovirus
& Einsele, H. (2002) Sensitive detection of & Popat, U. (2005) Cytomegalovirus (CMV) infection and disease in allogeneic stem cell
human cytomegalovirus peptide-specific cyto- infections and CMV-specific cellular immune transplant recipients. The Infectious Diseases
toxic T-lymphocyte responses by interferon- reconstitution following reduced intensity condi- Working Party of the European Group for
gamma-enzyme-linked immunospot assay and tioning allogeneic stem cell transplantation with Blood and Marrow Transplantation. Blood, 97,
flow cytometry in healthy individuals and in Alemtuzumab. Bone Marrow Transplantation, 388392.
patients after allogeneic stem cell transplanta- 36, 797802. Ljungman, P., Griffiths, P. & Paya, C. (2002a) Def-
tion. Blood, 99, 38303837. Larsson, K., Aschan, J., Remberger, M., Ringden, initions of cytomegalovirus infection and disease
van der Heiden, P.L., Kalpoe, J.S., Barge, R.M., O., Winiarski, J. & Ljungman, P. (2004) in transplant recipients. Clinical Infectious Dis-
Willemze, R., Kroes, A.C. & Schippers, E.F. Reduced risk for extensive chronic graft-versus- eases, 34, 10941097.
(2006) Oral valganciclovir as pre-emptive ther- host disease in patients receiving transplants Ljungman, P., Larsson, K., Kumlien, G., Aschan,
apy has similar efficacy on cytomegalovirus with human leukocyte antigen-identical sibling J., Barkholt, L., Gustafsson-Jernberg, A., Lewen-
DNA load reduction as intravenous ganciclovir donors given polymerase chain reaction-based sohn-Fuchs, I. & Ringden, O. (2002b) Leukocyte
in allogeneic stem cell transplantation recipients. preemptive therapy against cytomegalovirus. depleted, unscreened blood products give a low
Bone Marrow Transplantation, 37, 693698. Transplantation, 77, 526531. risk for CMV infection and disease in CMV
Hirsch, H.H., Lautenschlager, I., Pinsky, B.A., Car- Lim, Z.Y., Cook, G., Johnson, P.R., Parker, A., seronegative allogeneic stem cell transplant
denoso, L., Aslam, S., Cobb, B., Vilchez, R.A. & Zuckerman, M., Marks, D., Wiltshire, H., Mufti, recipients with seronegative stem cell donors.
Valsamakis, A. (2013) An international multicen- G.J. & Pagliuca, A. (2009) Results of a phase I/II Scandinavian Journal of Infectious Diseases, 34,
ter performance analysis of cytomegalovirus load British Society of Bone Marrow Transplantation 347350.
tests. Clinical Infectious Diseases, 56, 367373. study on PCR-based pre-emptive therapy with Ljungman, P., de La Camara, R., Milpied, N.,
Hodson, E.M., Jones, C.A., Strippoli, G.F., Web- valganciclovir or ganciclovir for active CMV Volin, L., Russell, C.A., Crisp, A. & Webster,
ster, A.C. & Craig, J.C. (2007) Immunoglobu- infection following alemtuzumab-based reduced A. Valacyclovir International Bone Marrow
lins, vaccines or interferon for preventing intensity allogeneic stem cell transplantation. Transplant Study Group. (2002c) Randomized
cytomegalovirus disease in solid organ trans- Leukemia Research, 33, 244249. study of valacyclovir as prophylaxis against
plant recipients. Cochrane Database Systematic Lindemans, C.A., Leen, A.M. & Boelens, J.J. (2010) cytomegalovirus reactivation in recipients of
Review, 2, CD005129. How I treat adenovirus in hematopoietic stem allogeneic bone marrow transplants. Blood, 99,
Hostetler, K.Y. (2010) Synthesis and early develop- cell transplant recipients. Blood, 116, 54765485. 30503056.
ment of hexadecyloxypropylcidofovir: an oral Liu, K.Y., Wang, Y., Han, M.Z., Huang, H., Chen, Ljungman, P., Hakki, M. & Boeckh, M. (2011)
antipoxvirus nucleoside phosphonate. Viruses, 2, H., Liu, Q.F., Wang, J.M., Liu, T., Song, Y.P., Cytomegalovirus in hematopoietic stem cell
22132225. Ma, J., Wu, D.P., Zou, P. & Huang, X.J. (2010) transplant recipients. Hematology/oncology Clin-
Kim, Y.J., Boeckh, M., Cook, L., Stempel, H., Jer- Valganciclovir for pre-emptive therapy of cyto- ics of North America, 25, 151169.
ome, K.R., Boucek, R. Jr, Burroughs, L. & Engl- megalovirus viraemia after hematopoietic stem Machado, C.M., Dulley, F.L., Boas, L.S., Castelli,
und, J.A. (2012) Cytomegalovirus infection and cell transplantation: a prospective multi-center J.B., Macedo, M.C., Silva, R.L., Pallota, R.,
ganciclovir resistance caused by UL97 mutations trial. Chinese Medical Journal (English Edition), Saboya, R.S. & Pannuti, C.S. (2000) CMV
in pediatric transplant recipients. Transplant 123, 21992205. pneumonia in allogeneic BMT recipients
Infectious Disease, 14, 611617. Ljungman, P. (1995) Cytomegalovirus pneumonia: undergoing early treatment of pre-emptive
Koehler, M., St George, K. & Rinaldo, C. Jr (1995) presentation, diagnosis, and treatment. Seminars ganciclovir therapy. Bone Marrow Transplanta-
The evaluation of results of CMV antigenaemia in Respiratory Infections, 10, 209215. tion, 26, 413417.
assays in bone marrow transplant recipients. Ljungman, P., Engelhard, D., Link, H., Biron, P., Maltezou, H., Whimbey, E., Abi-Said, D., Przepi-
British Journal of Haematology, 89, 427. Brandt, L., Brunet, S., Cordonnier, C., Debus- orka, D., Champlin, R. & Goodrich, J. (1999)
Koidl, C., Bozic, M., Marth, E. & Kessler, H.H. scher, L., de Laurenzi, A. & Kolb, H.J. (1992) Cytomegalovirus disease in adult marrow trans-
(2008) Detection of CMV DNA: is EDTA whole Treatment of interstitial pneumonitis due to plant recipients receiving ganciclovir prophy-
blood superior to EDTA plasma? Journal of cytomegalovirus with ganciclovir and intrave- laxis: a retrospective study. Bone Marrow
Virological Methods, 154, 210212. nous immune globulin: experience of European Transplantation, 24, 665669.

36 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

Manion, D.J., Vibhagool, A., Chou, T.C., Kaplan, serostatus. Biology of Blood and Marrow Trans- cytomegalovirus infection after receipt of leuko-
J., Caliendo, A. & Hirsch, M.S. (1996) Suscepti- plantation, 18, 9299. reduced blood products. Blood, 101, 41954200.
bility of human cytomegalovirus to two-drug Milano, F., Pergam, S.A., Xie, H., Leisenring, OBrien, S.M., Keating, M.J. & Mocarski, E.S.
combinations in vitro. Antiviral Therapy, 1, W.M., Gutman, J.A., Riffkin, I., Chow, V., (2006) Updated guidelines on the management
237245. Boeckh, M.J. & Delaney, C. (2011) Intensive of cytomegalovirus reactivation in patients with
Manischewitz, J.F., Quinnan, G.V. Jr, Lane, H.C. & strategy to prevent CMV disease in seropositive chronic lymphocytic leukemia treated with ale-
Wittek, A.E. (1990) Synergistic effect of ganci- umbilical cord blood transplant recipients. mtuzumab. Clinical Lymphoma and Myeloma, 7,
clovir and foscarnet on cytomegalovirus replica- Blood, 118, 56895696. 125130.
tion in vitro. Antimicrobial Agents and Montesinos, P., Sanz, J., Cantero, S., Lorenzo, I., Ozaki, K.S., Camara, N.O., Nogueira, E., Pereira,
Chemotherapy, 34, 373375. Martin, G., Saavedra, S., Palau, J., Romero, M., M.G., Granato, C., Melaragno, C., Camargo,
Manteiga, R., Martino, R., Sureda, A., Labeaga, R., Montava, A., Senent, L., Martinez, J., Jarque, I., L.F. & Pacheco-Silva, A. (2007) The use of sirol-
Brunet, S., Sierra, J. & Rabella, N. (1998) Cyto- Salavert, M., Cordoba, J., Gomez, L., Weiss, S., imus in ganciclovir-resistant cytomegalovirus
megalovirus pp 65 antigenemia-guided pre-emp- Moscardo, F., de la Rubia, J., Larrea, L., Sanz, infections in renal transplant recipients. Clinical
tive treatment with ganciclovir after allogeneic M.A. & Sanz, G.F. (2009) Incidence, risk factors, Transplantation, 21, 675680.
stem transplantation: a single-center experience. and outcome of cytomegalovirus infection and Ozdemir, E., St John, L.S., Gillespie, G., Rowland-
Bone Marrow Transplantation, 22, 899904. disease in patients receiving prophylaxis with Jones, S., Champlin, R.E., Molldrem, J.J. &
Marfori, J.E., Exner, M.M., Marousek, G.I., Chou, oral valganciclovir or intravenous ganciclovir Komanduri, K.V. (2002) Cytomegalovirus reacti-
S. & Drew, W.L. (2007) Development of new after umbilical cord blood transplantation. Biol- vation following allogeneic stem cell transplanta-
cytomegalovirus UL97 and DNA polymerase ogy of Blood and Marrow Transplantation, 15, tion is associated with the presence of
mutations conferring drug resistance after val- 730740. dysfunctional antigen-specific CD8 + T cells.
ganciclovir therapy in allogeneic stem cell Moretti, S., Zikos, P., Van Lint, M.T., Tedone, E., Blood, 100, 36903697.
recipients. Journal of Clinical Virology, 38, Occhini, D., Gualandi, F., Lamparelli, T., Mordi- Ozdemir, E., Saliba, R.M., Champlin, R.E., Couriel,
120125. ni, N., Berisso, G., Bregante, S., Bruno, B. & D.R., Giralt, S.A., de Lima, M., Khouri, I.F.,
Marty, F.M., Bryar, J., Browne, S.K., Schwarzberg, Bacigalupo, A. (1998) Forscarnet vs ganciclovir Hosing, C., Kornblau, S.M., Anderlini, P.,
T., Ho, V.T., Bassett, I.V., Koreth, J., Alyea, for cytomegalovirus (CMV) antigenemia after Shpall, E.J., Qazilbash, M.H., Molldrem, J.J.,
E.P., Soiffer, R.J., Cutler, C.S., Antin, J.H. & allogeneic hemopoietic stem cell transplantation Chemaly, R.F. & Komanduri, K.V. (2007) Risk
Baden, L.R. (2007) Sirolimus-based graft-versus- (HSCT): a randomised study. Bone Marrow factors associated with late cytomegalovirus
host disease prophylaxis protects against cyto- Transplantation, 22, 175180. reactivation after allogeneic stem cell transplan-
megalovirus reactivation after allogeneic hema- Mullier, F., Kabamba-Mukadi, B., Bodeus, M. & tation for hematological malignancies. Bone
topoietic stem cell transplantation: a cohort Goubau, P. (2009) Definition of clinical thresh- Marrow Transplantation, 40, 125136.
analysis. Blood, 110, 490500. old for CMV real-time PCR after comparison Park, S.Y., Lee, S.O., Choi, S.H., Kim, Y.S., Woo,
Marty, F.M., Ljungman, P., Papanicolaou, G.A., with PP65 antigenaemia and clinical data. Acta J.H., Sung, H., Kim, M.N., Kim, D.Y., Lee, J.H.,
Winston, D.J., Chemaly, R.F., Strasfeld, L., Clinica Belgica, 64, 477482. Lee, J.H., Lee, K.H. & Kim, S.H. (2011) Para-
Young, J.A., Rodriguez, T., Maertens, J., Nakamae, H., Kirby, K.A., Sandmaier, B.M., Nora- doxical rising cytomegalovirus antigenemia dur-
Schmitt, M., Einsele, H., Ferrant, A., Lipton, setthada, L., Maloney, D.G., Maris, M.B., Davis, ing preemptive ganciclovir therapy in
J.H., Villano, S.A., Chen, H. & Boeckh, M.; C., Corey, L., Storb, R. & Boeckh, M. (2009) hematopoietic stem cell transplant recipients:
Maribavir -300 Clinical Study Group. (2011) Effect of conditioning regimen intensity on incidence, risk factors, and clinical outcomes.
Maribavir prophylaxis for prevention of cyto- CMV infection in allogeneic hematopoietic cell Journal of Clinical Microbiology, 49, 41794184.
megalovirus disease in recipients of allogeneic transplantation. Biology of Blood and Marrow Peggs, K.S., Verfuerth, S., Pizzey, A., Khan, N.,
stem-cell transplants: a phase 3, double-blind, Transplantation, 15, 694703. Guiver, M., Moss, P.A. & Mackinnon, S. (2003)
placebo-controlled, randomised trial. The Lancet Nakamura, R., Cortez, K., Solomon, S., Battiwalla, Adoptive cellular therapy for early cytomegalovi-
Infectious Diseases, 11, 284292. M., Gill, V.J., Hensel, N., Childs, R. & Barrett, rus infection after allogeneic stem-cell transplan-
Mattes, F.M., Hainsworth, E.G., Geretti, A.M., A.J. (2002) High-dose acyclovir and pre-emptive tation with virus-specific T-cell lines. Lancet,
Nebbia, G., Prentice, G., Potter, M., Burroughs, ganciclovir to prevent cytomegalovirus disease 362, 13751377.
A.K., Sweny, P., Hassan-Walker, A.F., Okwuadi, in myeloablative and non-myeloablative alloge- Peggs, K.S., Verfuerth, S., Pizzey, A., Chow, S.L.,
S., Sabin, C., Amooty, G., Brown, V.S., Grace, neic stem cell transplantation. Bone Marrow Thomson, K. & Mackinnon, S. (2009) Cytomeg-
S.C., Emery, V.C. & Griffiths, P.D. (2004) A Transplantation, 30, 235242. alovirus-specific T cell immunotherapy pro-
randomized, controlled trial comparing ganci- Narimatsu, H., Kami, M., Kato, D., Matsumura, motes restoration of durable functional antiviral
clovir to ganciclovir plus foscarnet (each at half T., Murashige, N., Kusumi, E., Yuji, K., Hori, immunity following allogeneic stem cell trans-
dose) for preemptive therapy of cytomegalovirus A., Shibata, T., Masuoka, K., Wake, A., Miyako- plantation. Clinical Infectious Diseases, 49, 1851
infection in transplant recipients. Journal of shi, S., Morinaga, S. & Taniguchi, S. (2007) 1860.
Infectious Diseases, 189, 13551361. Reduced dose of foscarnet as preemptive therapy Pergam, S.A., Xie, H., Sandhu, R., Pollack, M.,
Mattes, F.M., Hainsworth, E.G., Hassan-Walker, for cytomegalovirus infection following reduced- Smith, J., Stevens-Ayers, T., Ilieva, V., Kimball,
A.F., Burroughs, A.K., Sweny, P., Griffiths, P.D. intensity cord blood transplantation. Transplant L.E., Huang, M.L., Hayes, T.S., Corey, L. &
& Emery, V.C. (2005) Kinetics of cytomegalovi- Infectious Disease, 9, 1115. Boeckh, M.J. (2012) Efficiency and risk factors
rus load decrease in solid-organ transplant Nichols, W.G., Corey, L., Gooley, T., Davis, C. for CMV transmission in seronegative hemato-
recipients after preemptive therapy with & Boeckh, M. (2002) High risk of death due poietic stem cell recipients. Biology of Blood and
valganciclovir. Journal of Infectious Diseases, 191, to bacterial and fungal infection among cyto- Marrow Transplantation, 18, 13911400.
8992. megalovirus (CMV)-seronegative recipients of Pinana, J.L., Martino, R., Barba, P., Margall, N.,
Mikulska, M., Raiola, A.M., Bruzzi, P., Varaldo, stem cell transplants from seropositive donors: Roig, M.C., Valcarcel, D., Sierra, J. & Rabella,
R., Annunziata, S., Lamparelli, T., Frassoni, F., evidence for indirect effects of primary CMV N. (2010) Cytomegalovirus infection and disease
Tedone, E., Galano, B., Bacigalupo, A. & Viscoli, infection. Journal of Infectious Diseases, 185, after reduced intensity conditioning allogeneic
C. (2012) CMV infection after transplant from 273282. stem cell transplantation: single-centre experi-
cord blood compared to other alternative Nichols, W.G., Price, T.H., Gooley, T., Corey, L. & ence. Bone Marrow Transplantation, 45, 534
donors: the importance of donor-negative CMV Boeckh, M. (2003) Transfusion-transmitted 542.

2013 John Wiley & Sons Ltd 37


British Journal of Haematology, 2013, 162, 2539
Guideline

Platzbecker, U., Bandt, D., Thiede, C., Helwig, A., Group. Bone Marrow Transplantation, 19, poietic stem cell transplantation with ale-
Freiberg-Richter, J., Schuler, U., Plettig, R., 233236. mtuzumab. Ophthalmology, 115, 17661770.
Geissler, G., Rethwilm, A., Ehninger, G. & Bor- Rzepecki, P., Barzal, J., Sarosiek, T., Oborska, S. & Spellman, S.R., Eapen, M., Logan, B.R., Mueller,
nhauser, M. (2001) Successful preemptive ci- Szczylik, C. (2008) Prevention of cytomegalovi- C., Rubinstein, P., Setterholm, M.I., Woolfrey,
dofovir treatment for CMV antigenemia after rus reactivaction after allogenic hematopoietic A.E., Horowitz, M.M., Confer, D.L. & Hurley,
dose-reduced conditioning and allogeneic blood stem cell transplantation with valganciclovir: C.K.; National Marrow Donor, P., Center for
stem cell transplantation. Transplantation, 71, single center experience. Journal of Chemother- International Blood and Marrow Transplant
880885. apy, 20, 140142. Research. (2012) A perspective on the selection
Pollack, M., Heugel, J., Xie, H., Leisenring, W., Sto- Salzberger, B., Bowden, R.A., Hackman, R.C., of unrelated donors and cord blood units for
rek, J., Young, J.A., Kukreja, M., Gress, R., Davis, C. & Boeckh, M. (1997) Neutropenia in transplantation. Blood, 120, 259265.
Tomblyn, M. & Boeckh, M. (2011) An interna- allogeneic marrow transplant recipients receiving Sylwester, A.W., Mitchell, B.L., Edgar, J.B., Taor-
tional comparison of current strategies to prevent ganciclovir for prevention of cytomegalovirus mina, C., Pelte, C., Ruchti, F., Sleath, P.R.,
herpesvirus and fungal infections in hematopoi- disease: risk factors and outcome. Blood, 90, Grabstein, K.H., Hosken, N.A., Kern, F., Nelson,
etic cell transplant recipients. Biology of Blood 25022508. J.A. & Picker, L.J. (2005) Broadly targeted
and Marrow Transplantation, 17, 664673. Sauter, C., Abboud, M., Jia, X., Heller, G., human cytomegalovirus-specific CD4 + and
Prentice, H.G., Gluckman, E., Powles, R.L., Ljung- Gonzales, A.M., Lubin, M., Hawke, R., Perales, CD8 + T cells dominate the memory compart-
man, P., Milpied, N., Fernandez Ranada, J.M., M.A., van den Brink, M.R., Giralt, S., Papani- ments of exposed subjects. Journal of Experimen-
Mandelli, F., Kho, P., Kennedy, L. & Bell, A.R. colaou, G., Scaradavou, A., Small, T.N. & Bar- tal Medicine, 202, 673685.
(1994) Impact of long-term acyclovir on cyto- ker, J.N. (2011) Serious infection risk and Takami, A., Mochizuki, K., Asakura, H., Yamazaki,
megalovirus infection and survival after alloge- immune recovery after double-unit cord blood H., Okumura, H. & Nakao, S. (2005) High inci-
neic bone marrow transplantation. European transplantation without antithymocyte globulin. dence of cytomegalovirus reactivation in adult
Acyclovir for CMV Prophylaxis Study Group. Biology of Blood and Marrow Transplantation, recipients of an unrelated cord blood transplant.
Lancet, 343, 749753. 17, 14601471. Haematologica, 90, 12901292.
Prentice, H.G., Gluckman, E., Powles, R.L., Ljung- Schmidt, G.M., Kovacs, A., Zaia, J.A., Horak, D.A., Takami, A., Mochizuki, K., Ito, S., Sugimori, C.,
man, P., Milpied, N.J., Camara, R., Mandelli, F., Blume, K.G., Nademanee, A.P., ODonnell, Yamashita, T., Asakura, H., Okumura, H. &
Kho, P., Kennedy, L. & Bell, A.R. (1997) Long- M.R., Snyder, D.S. & Forman, S.J. (1988) Ganci- Nakao, S. (2007) Safety and efficacy of foscarnet
term survival in allogeneic bone marrow trans- clovir/immunoglobulin combination therapy for for preemptive therapy against cytomegalovirus
plant recipients following acyclovir prophylaxis the treatment of human cytomegalovirus-associ- reactivation after unrelated cord blood trans-
for CMV infection. The European Acyclovir for ated interstitial pneumonia in bone marrow plantation. Transplant Proc, 39, 237239.
CMV Prophylaxis Study Group. Bone Marrow allograft recipients. Transplantation, 46, Tomblyn, M., Chiller, T., Einsele, H., Gress, R.,
Transplantation, 19, 129133. 905907. Sepkowitz, K., Storek, J., Wingard, J.R., Young,
Ratko, T.A., Cummings, J.P., Oberman, H.A., Schmidt-Hieber, M., Schwarck, S., Stroux, A., J.A. & Boeckh, M.J. (2009) Guidelines for pre-
Crookston, K.P., DeChristopher, P.J., Eastlund, Ganepola, S., Reinke, P., Thiel, E., Uharek, L. & venting infectious complications among hemato-
D.T., Godwin, J.E., Sacher, R.A., Yawn, D.H. & Blau, I.W. (2010) Immune reconstitution and poietic cell transplantation recipients: a global
Matuszewski, K.A.; University Health System, C. cytomegalovirus infection after allogeneic stem perspective. Biology of Blood and Marrow Trans-
(2001) Evidence-based recommendations for the cell transplantation: the important impact of in plantation, 15, 11431238.
use of WBC-reduced cellular blood components. vivo T cell depletion. International Journal of Tomonari, A., Takahashi, S., Ooi, J., Tsukada, N.,
Transfusion, 41, 13101319. Hematology, 91, 877885. Konuma, T., Kato, S., Kasahara, S., Iseki, T.,
Reusser, P., Einsele, H., Lee, J., Volin, L., Rovira, Shereck, E.B., Cooney, E., van de Ven, C., Della- Yamaguchi, T., Tojo, A. & Asano, S. (2008)
M., Engelhard, D., Finke, J., Cordonnier, C., Lotta, P. & Cairo, M.S. (2007) A pilot phase II Impact of cytomegalovirus serostatus on out-
Link, H., Ljungman, P., Infectious Diseases study of alternate day ganciclovir and foscarnet come of unrelated cord blood transplantation
Working Party of the European Group for in preventing cytomegalovirus (CMV) infections for adults: a single-institute experience in
Blood and Marrow Transplantation. (2002) in at-risk pediatric and adolescent allogeneic Japan. European Journal of Haematology, 80,
Randomized multicenter trial of foscarnet versus stem cell transplant recipients. Pediatric Blood & 251257.
ganciclovir for preemptive therapy of cytomega- Cancer, 49, 306312. Ugarte-Torres, A., Hoegh-Petersen, M., Liu, Y.,
lovirus infection after allogeneic stem cell trans- Sia, I.G. & Patel, R. (2000) New strategies for pre- Zhou, F., Williamson, T.S., Quinlan, D., Sy, S.,
plantation. Blood, 99, 11591164. vention and therapy of cytomegalovirus infec- Roa, L., Khan, F., Fonseca, K., Russell, J.A. &
Ruiz-Camps, I., Len, O., de la Camara, R., Gur- tion and disease in solid-organ transplant Storek, J. (2011) Donor serostatus has an impact
gui, M., Martino, R., Jarque, I., Barrenetxea, C., recipients. Clinical Microbiology Reviews, 13, on cytomegalovirus-specific immunity, cytom-
Diaz de Heredia, C., Batlle, M., Rovira, M., de 83121. egaloviral disease incidence, and survival in
la Torre, J., Torres, A., Aguilar, M., Espigado, Sili, U., Leen, A.M., Vera, J.F., Gee, A.P., Huls, H., seropositive hematopoietic cell transplant
I., Martin-Davila, P., Bou, G., Borrell, N. & Heslop, H.E., Bollard, C.M. & Rooney, C.M. recipients. Biology of Blood and Marrow Trans-
Aguado, J.M.; Spanish Network for Research on (2012) Production of good manufacturing prac- plantation, 17, 574585.
Infection in Transplantation. (2011) Valganci- tice-grade cytotoxic T lymphocytes specific for Verma, A., Devine, S., Morrow, M., Chen, Y.H.,
clovir as pre-emptive therapy for cytomegalovi- Epstein-Barr virus, cytomegalovirus and adeno- Mihalov, M., Peace, D., Stock, W., Pursell, K.,
rus infection in allogeneic haematopoietic stem virus to prevent or treat viral infections post- Wickrema, A., Yassine, M., Jessop, E. & van Be-
cell transplant recipients. Antiviral Therapy, 16, allogeneic hematopoietic stem cell transplant. sien, K. (2003) Low incidence of CMV viremia
951957. Cytotherapy, 14, 711. and disease after allogeneic peripheral blood
Ruutu, T., Ljungman, P., Brinch, L., Lenhoff, S., Soderberg, C., Larsson, S., Rozell, B.L., Sumitran- stem cell transplantation. Role of pretransplant
Lonnqvist, B., Ringden, O., Ruutu, P., Volin, L., Karuppan, S., Ljungman, P. & Moller, E. (1996) ganciclovir and post-transplant acyclovir. Bone
Albrechtsen, D., Sallerfors, B., Ebeling, F. & My- Cytomegalovirus-induced CD13-specific autoim- Marrow Transplantation, 31, 813816.
llyla, G. (1997) No prevention of cytomegalovi- munitya possible cause of chronic graft-vs-host Vusirikala, M., Wolff, S.N., Stein, R.S., Brandt, S.J.,
rus infection by anti-cytomegalovirus disease. Transplantation, 61, 600609. Morgan, D.S., Greer, J.P., Schuening, F.G., Dum-
hyperimmune globulin in seronegative bone Song, W.K., Min, Y.H., Kim, Y.R. & Lee, S.C. mer, J.S. & Goodman, S.A. (2001) Valacyclovir
marrow transplant recipients. The Nordic BMT (2008) Cytomegalovirus retinitis after hemato- for the prevention of cytomegalovirus infection

38 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 2539
Guideline

after allogeneic stem cell transplantation: a single Wang, L.R., Dong, L.J., Zhang, M.J. & Lu, D.P. Yahav, D., Gafter-Gvili, A., Muchtar, E., Skalsky,
institution retrospective cohort analysis. Bone (2008) Correlations of human herpesvirus 6B K., Kariv, G., Yeshurun, M., Leibovici, L. &
Marrow Transplantation, 28, 265270. and CMV infection with acute GVHD in recipi- Paul, M. (2009) Antiviral prophylaxis in
Walker, C.M., van Burik, J.A., De For, T.E. & ents of allogeneic haematopoietic stem cell haematological patients: systematic review and
Weisdorf, D.J. (2007) Cytomegalovirus infection transplantation. Bone Marrow Transplantation, meta-analysis. European Journal of Cancer, 45,
after allogeneic transplantation: comparison of 42, 673677. 31313148.
cord blood with peripheral blood and marrow Winston, D.J., Young, J.A., Pullarkat, V., Papa- Zhou, W., Longmate, J., Lacey, S.F., Palmer, J.M.,
graft sources. Biology of Blood and Marrow nicolaou, G.A., Vij, R., Vance, E., Alangaden, Gallez-Hawkins, G., Thao, L., Spielberger, R.,
Transplantation, 13, 11061115. G.J., Chemaly, R.F., Petersen, F., Chao, N., Nakamura, R., Forman, S.J., Zaia, J.A. & Dia-
Walter, E.A., Greenberg, P.D., Gilbert, M.J., Finch, Klein, J., Sprague, K., Villano, S.A. & Bo- mond, D.J. (2009) Impact of donor CMV
R.J., Watanabe, K.S., Thomas, E.D. & Riddell, eckh, M. (2008) Maribavir prophylaxis for status on viral infection and reconstitution of
S.R. (1995) Reconstitution of cellular immunity prevention of cytomegalovirus infection in multifunction CMV-specific T cells in CMV-
against cytomegalovirus in recipients of alloge- allogeneic stem cell transplant recipients: a positive transplant recipients. Blood, 113, 6465
neic bone marrow by transfer of T-cell clones multicenter, randomized, double-blind, pla- 6476.
from the donor. New England Journal of Medi- cebo-controlled, dose-ranging study. Blood,
cine, 333, 10381044. 111, 54035410.

2013 John Wiley & Sons Ltd 39


British Journal of Haematology, 2013, 162, 2539

You might also like