We performed a randomized trial comparing the use of quantitative DNAemia versus positive antigenemia for starting preemptive antihuman cytomegalovirus (HCMV) therapy in hematopoietic stem-cell transplantation (HSCT) recipients. In the DNAemia arm, antiviral therapy was initiated on reaching a DNAemia cut-off of 10 000 DNA copies/mL of whole blood, whereas in the antigenemia arm, therapy was started in the presence of a positive antigenemia. The aim of the study was to compare the number of patients treated in the 2 arms. On the whole, 178 patients (89 in each arm), receiving unmanipulated HSCT from either a relative or an unrelated donor, completed the study. Although the incidence of HCMV infection was comparable in DNAemia and antigenemia arms (34% vs 42%, respectively, P = .259), the number of patients treated was significantly lower in the DNAemia arm (18% vs 31%, P = .026). No patient developed HCMV disease. The use of a DNAemia cut-off avoids unnecessary antiviral treatment.

Preemptive antiviral therapy, based on the administration of drugs on detection of virus in blood, has been largely used to prevent the occurrence of human cytomegalovirus (HCMV) disease in patients given allogeneic hematopoietic stem-cell transplantation (HSCT).1  This strategy offers the advantage of treating only those patients experiencing HCMV infection and who are, thus, at risk of developing overt disease.2,4  Two assays have been mainly used during the last decade for early detection of HCMV in blood: antigenemia (detection of HCMV pp65 phosphoprotein in peripheral blood leukocytes)5,,8  and DNAemia (HCMV DNA detection in either leukocytes, plasma, or whole blood).3  In view of the high risk of fatal HCMV-related pneumonia in HSCT recipients, preemptive therapy for HSCT recipients is currently started on the first confirmed appearance of virus in blood, irrespective of the assay used.3,8,9 

We investigated, in a randomized study involving young patients, a different approach based on the comparison for preemptive therapy of a predefined cut-off for DNAemia with the first antigenemia positivity. The DNAemia cut-off was chosen based on a retrospective analysis of DNAemia levels in HSCT recipients undergoing antigenemia-guided HCMV surveillance.10,11 

From December 2002 to July 2006, a total of 184 patients were given an unmanipulated allogeneic HSCT from either a human leukocyte antigen (HLA)-identical sibling or an unrelated donor (UD) at the Oncoematologia Pediatrica, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy. Inclusion criteria for enrollment of patients in the study were as follows: serologic evidence of past HCMV infection in either donor (D) or recipient (R) or both, and either patient's or parents' written informed consent obtained in accordance with the Declaration of Helsinki. Overall, 178 patients (89 in each arm) were included in the study. Patients' characteristics are detailed in Table 1. The primary end point was the comparison of the number of treated patients in the 2 arms. Approval was obtained from the Fondazione IRCCS Policlinico San Matteo Institutional Review Board and ethics committee for these studies.

Table 1

Characteristics of the 178 patients completing follow-up

ParameterRandomization arm
DNAemiaAntigenemia
Median age, y (range) 9 (0-25) 9 (0-24) 
Sex, no. male patients/no. female patients 43/46 55/34 
Diagnosis, no. patients (%)   
    Acute myeloid leukemia 8 (9) 13 (14) 
    Acute lymphoblastic leukemia 24 (27) 26 (29) 
    Chronic myeloid leukemia 1 (1) 0 (0) 
    Myelodysplastic syndrome 10 (11) 4 (5) 
    Non-Hodgkin lymphoma 4 (5) 1 (1) 
    Hemophagocytic lymphohistiocytosis 1 (1) 3 (3) 
    Hemoglobinopathies 24 (27) 30 (34) 
    Bone marrow failure 11 (12) 5 (6) 
    Inborn metabolism disorders 4 (5) 4 (5) 
    Other* 2 (2) 3 (3) 
HCMV serology, no. patients (%)   
    R+/D 27 (30) 30 (34) 
    R+/D+ 50 (56) 39 (44) 
    R/D+ 12 (14) 20 (22) 
Donor type, no. patients (%)   
    Sibling 41 (46) 39 (44) 
    Unrelated 48 (54) 50 (56) 
Source of stem-cell graft, no. patients (%)   
    Bone marrow 80 (90) 74 (84) 
    Peripheral blood 2 (2) 3 (3) 
    Cord blood 3 (3) 9 (10) 
    Bone marrow + cord blood 4 (5) 3 (3) 
Conditioning regimen, no. patients (%)   
    Chemotherapy based 61 (68) 60 (67) 
    TBI based 28 (32) 29 (33) 
    ATG administration 45 (51) 46 (52) 
GvHD prophylaxis, no. patients (%)   
    Cyclosporine-A 22 (25) 25 (28) 
    Cyclosporine-A + methotrexate 67 (75) 64 (72) 
Days to PMN recovery, median (range) 20 (8-39) 20 (9-45) 
Days to PLT recovery, median (range) 23 (8-95) 25 (11-195) 
Grade II-IV acute GvHD 24 (27) 26 (29) 
Chronic GvHD 7 (8) 6 (7) 
ParameterRandomization arm
DNAemiaAntigenemia
Median age, y (range) 9 (0-25) 9 (0-24) 
Sex, no. male patients/no. female patients 43/46 55/34 
Diagnosis, no. patients (%)   
    Acute myeloid leukemia 8 (9) 13 (14) 
    Acute lymphoblastic leukemia 24 (27) 26 (29) 
    Chronic myeloid leukemia 1 (1) 0 (0) 
    Myelodysplastic syndrome 10 (11) 4 (5) 
    Non-Hodgkin lymphoma 4 (5) 1 (1) 
    Hemophagocytic lymphohistiocytosis 1 (1) 3 (3) 
    Hemoglobinopathies 24 (27) 30 (34) 
    Bone marrow failure 11 (12) 5 (6) 
    Inborn metabolism disorders 4 (5) 4 (5) 
    Other* 2 (2) 3 (3) 
HCMV serology, no. patients (%)   
    R+/D 27 (30) 30 (34) 
    R+/D+ 50 (56) 39 (44) 
    R/D+ 12 (14) 20 (22) 
Donor type, no. patients (%)   
    Sibling 41 (46) 39 (44) 
    Unrelated 48 (54) 50 (56) 
Source of stem-cell graft, no. patients (%)   
    Bone marrow 80 (90) 74 (84) 
    Peripheral blood 2 (2) 3 (3) 
    Cord blood 3 (3) 9 (10) 
    Bone marrow + cord blood 4 (5) 3 (3) 
Conditioning regimen, no. patients (%)   
    Chemotherapy based 61 (68) 60 (67) 
    TBI based 28 (32) 29 (33) 
    ATG administration 45 (51) 46 (52) 
GvHD prophylaxis, no. patients (%)   
    Cyclosporine-A 22 (25) 25 (28) 
    Cyclosporine-A + methotrexate 67 (75) 64 (72) 
Days to PMN recovery, median (range) 20 (8-39) 20 (9-45) 
Days to PLT recovery, median (range) 23 (8-95) 25 (11-195) 
Grade II-IV acute GvHD 24 (27) 26 (29) 
Chronic GvHD 7 (8) 6 (7) 

All P values were nonsignificant.

R indicates recipient; D, donor; TBI, total body irradiation; ATG, anti-thymocyte globulin; GvHD, graft-versus-host disease; PMN, polymorphonuclear; PLT, platelet; ns; not significant.

*

Other includes 1 congenital sideroblastic anemia, 1 metastatic renal cell carcinoma, 1 Chediak-Higashi syndrome, 1 IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked), 1 Kostmann syndrome.

Defined as time needed to reach an absolute neutrophil count equal to or greater than 0.5 × 109/L.

Defined as time needed to reach an unsupported platelet count equal to or greater than 50 ×109/L.

The number of patients to be enrolled was calculated by a sample size evaluation method based on the χ2 test. The minimum number of patients to be randomized was 84 per arm, based on a significance level of 0.05, a study power of 0.80, and the hypothesized need for treatment in 40% patients allocated to the antigenemia arm9  and 20% patients allocated to the DNAemia arm.

Patients were randomized using a block random design on the basis of donor type (namely HLA-identical sibling vs UD). In the antigenemia arm, patients were treated upon detection of either 2 or more pp65-positive leukocytes (see below) or first confirmed positivity. In the DNAemia arm, patients were treated upon reaching a DNA level of 10 000 copies/mL of blood, a threshold chosen in view of previously obtained results.10,11 

Intravenous ganciclovir (5 mg/kg twice a day) was administered as preemptive therapy in both arms and stopped upon virus clearance from blood. In the case of drug toxicity, foscarnet (90 mg/kg twice a day) was given as replacement for ganciclovir.

All patients were monitored for HCMV infection until 3 months after HSCT by collecting blood samples twice weekly until day + 45 and weekly thereafter. Subsequently, HCMV infection was monitored in all patients at least at scheduled controls at 6, 9, and 12 months and in the presence of clinical signs or symptoms.

Quantitation of viral DNA in whole blood for patients allocated to the DNAemia arm was performed using real-time polymerase chain reaction (PCR).12  The antigenemia assay was evaluated by counting the number of leukocytes positive for pp65/2 × 105 cells examined, according to a previously described procedure13  and more recently standardized procedure.14  In a subgroup of 48 patients, HCMV-specific CD4+ and CD8+ T-cell reconstitution was monitored as reported.15 

Data were analyzed as of March 1, 2007. Differences between medians were compared using the Mann-Whitney U test. Differences in percentages were tested using the Pearson χ2 test. All tests were two-tailed. The probabilities of HCMV infection and need for preemptive therapy were calculated and expressed as cumulative incidence, taking into account competing risks.16,17 

The median follow-up of surviving patients enrolled into this study was 23 months (range, 7-49 months). For at least 1 year after allograft, 129 patients were observed; they represented 88% of patients still alive. Thirty of the 89 HSCT recipients (34%) enrolled in the DNAemia arm and 37 of 89 patients (42%) enrolled in the antigenemia arm developed HCMV infection (P = not significant). The cumulative incidence curves (Figure 1A) showed a probability of HCMV infection at day + 360 of 34% and 42% in the DNAemia and antigenemia arms, respectively. Median time to first HCMV detection was comparable in the 2 arms (43 days, range, 11-118, in the DNAemia arm; and 43 days, range, 19-85, in the antigenemia arm).

Figure 1

Cumulative incidence of HCMV infection and preemptive treatment in the 2 randomization arms. (A) HCMV infection. (B) Preemptive treatment. Numbers in parentheses indicate the actual number of infected (A) or treated (B) patients and the relevant probability at day + 360 with 95% confidence intervals (CI).

Figure 1

Cumulative incidence of HCMV infection and preemptive treatment in the 2 randomization arms. (A) HCMV infection. (B) Preemptive treatment. Numbers in parentheses indicate the actual number of infected (A) or treated (B) patients and the relevant probability at day + 360 with 95% confidence intervals (CI).

Close modal

In multivariate analysis, the only factor significantly associated with the occurrence of HCMV infection in the whole population was recipient HCMV seropositivity (data not shown).

Twenty-three patients who had DNAemia levels consistently below the cut-off level in the relevant arm, or negative test on a second control after a first positive result in the antigenemia arm (ie, patients with self-resolving infection), were not treated. The remaining 44 patients were treated according to criteria already detailed. As shown in Table 2, the number of patients treated was significantly lower in the DNAemia arm (P = .037). The cumulative incidence curves (Figure 1B) of the probability of starting preemptive treatment at day + 360 was 18% in the DNAemia arm and 31% in the antigenemia arm (P = .026). In addition, treatment onset was significantly delayed in the DNAemia arm compared with antigenemia arm (Table 2). All of the other parameters investigated were comparable in the 2 arms, as well as 100-day cumulative incidence of grade II-IV acute graft-versus-host disease (GvHD) (27% in the DNAemia and 29% in the antigenemia arm) and 1-year transplantation-related mortality (3% in the DNAemia and 7% in the antigenemia arm). No cases of HCMV disease were observed in either arm during the study period.

Table 2

Preemptive therapy in the 2 randomization arms

ParameterWhole patient population
P
DNAemia armAntigenemia arm
No. of patients 89 89 na 
No. (%) of HCMV infected patients 30 (34) 37 (42) ns 
No. (%) of treated patients 16 (18) 28 (31) .037 
Median days (range) to start of antiviral therapy 56 (17-144) 45 (22-88) .032 
Median days (range) of first course of antiviral therapy 15 (6-35) 14 (5-39) ns 
No. (%) of relapsing HCMV infections 8/16 (50) 15/28 (54) ns 
No. (%) of treated relapses 3/16 (19) 10/28 (36) ns 
Median days (range) of relapse treatment 11 (7-52) 14 (7-49) ns 
Median days (range) of total antiviral therapy 17 (6-69) 17 (5-69) ns 
Median days (range) of total antiviral therapy:    
    in patients with grade 0-I acute GvHD 12 (6-20) 14 (7-66) ns 
    in patients with grade II-IV acute GvHD 20 (6-69) 23 (5-69) ns 
        P grade 0-I vs grade II-IV (0.08) (ns) na 
ParameterWhole patient population
P
DNAemia armAntigenemia arm
No. of patients 89 89 na 
No. (%) of HCMV infected patients 30 (34) 37 (42) ns 
No. (%) of treated patients 16 (18) 28 (31) .037 
Median days (range) to start of antiviral therapy 56 (17-144) 45 (22-88) .032 
Median days (range) of first course of antiviral therapy 15 (6-35) 14 (5-39) ns 
No. (%) of relapsing HCMV infections 8/16 (50) 15/28 (54) ns 
No. (%) of treated relapses 3/16 (19) 10/28 (36) ns 
Median days (range) of relapse treatment 11 (7-52) 14 (7-49) ns 
Median days (range) of total antiviral therapy 17 (6-69) 17 (5-69) ns 
Median days (range) of total antiviral therapy:    
    in patients with grade 0-I acute GvHD 12 (6-20) 14 (7-66) ns 
    in patients with grade II-IV acute GvHD 20 (6-69) 23 (5-69) ns 
        P grade 0-I vs grade II-IV (0.08) (ns) na 

No patients from either arm contracted HCMV disease.

na indicates not applicable; ns, not significant.

Upon initiation of treatment, patients allocated to the DNAemia arm had median levels of both viral DNAemia (17 900, range 10 100-62 800 copies/mL of blood) and antigenemia (48, range 0-300 pp65-positive leukocytes) significantly higher (P < .001) than those in the antigenemia arm: 0 (range 0-32800) DNA copies/mL of blood and 3 (range 1-75) pp65-positive leukocytes.

HCMV-specific CD4+ and CD8+ T-cell reconstitution was available for 48 patients,15  equally distributed between the 2 randomization arms. Sixteen were HCMV-seronegative before transplantation and were transplanted from a seropositive donor. None of them developed HCMV infection; HCMV-specific T-cells appeared within the first 3 months in 4 of them. All 32 HCMV-seropositive patients (25 of whom developed HCMV infection) reconstituted HCMV-specific T-cell immunity after a median time of 36 (range 27-153) days for the CD8+ T-cell compartment and 47 (range 27-153) days for the CD4+ T-cell compartment. No difference in time to specific immune recovery was found between the 2 arms.

Results of our study indicate that (i) a DNAemia cut-off can be safely used to guide preemptive therapy for HCMV infections in HSCT recipients, as no case of HCMV disease was observed in the study population; (ii) adoption of a cut-off of 10 000 copies/mL of blood significantly decreases the number of patients candidate to preemptive treatment with respect to antigenemia with advantages in terms of toxic effect and drug cost reduction and no disadvantages in terms of GvHD occurrence and transplantation-related mortality; and (iii) treatment inception was significantly delayed in the DNAemia arm, thus in a situation where hematologic and immunologic recovery is expected to be more advanced.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank the technical staff of the Servizio di Virologia for performing the assays, Daniela Sartori for preparing the manuscript, and Laurene Kelly for revision of the English in this manuscript.

This work was supported in part by grants from the Ministero della Salute-Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo (Ricerca Corrente grant 80 541 and Ricerca Finalizzata 2003 grant 89 269) and Fondazione Cassa di Risparmio delle Provincie Lombarde (CARIPLO) (grant 93 005) (G. Gerna) and by grants from Associazione Italiana Ricerca sul Cancro (AIRC), Consiglio Nazionale delle Ricerche (CNR), Ministero dell'Università e della Ricerca Scientifica e Tecnologica (MURST), the European Union (FP6 program ALLOSTEM), and Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo (F.L.).

Contribution: D.L., G. Gerna, and F.L. designed and performed the research, collected and analyzed the data, and wrote the paper; M.F., M.E.B., and S.T. collected the data; G. Giorgiani performed the research; F.B. designed the research.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Giuseppe Gerna, Servizio di Virologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, via Taramelli 5, 27100 Pavia, Italy; e-mail:g.gerna@smatteo.pv.it.

1
Boeckh
M
Ljungman
P
Paya
C
Cytomegalovirus infection after bone marrow transplantation.
Transplant infections
1998
Philadelphia, PA
Lippincott-Raven
215
227
2
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood
1996
88
4063
4071
3
Einsele
H
Ehninger
G
Hebart
H
et al
Polymerase chain reaction monitoring reduces the incidence of cytomegalovirus disease and the duration and side effects of antiviral therapy after bone marrow transplantation.
Blood
1995
86
2815
2820
4
Ljungman
P
Aschan
J
Lewensohn-Fuchs
I
et al
Results of different strategies for reducing cytomegalovirus-associated mortality in allogeneic stem cell transplant recipients.
Transplantation
1998
66
1330
1334
5
Van der Bij
W
Schirm
J
Torensma
R
Van Son
WJ
Tegzess
AM
The
TH
Comparison between viremia and antigenemia for detection of cytomegalovirus in blood.
J Clin Microbiol
1988
26
2531
2535
6
Revello
MG
Zavattoni
M
Percivalle
E
Grossi
P
Gerna
G
Correlation between immunofluorescent detection of human cytomegalovirus immediate early antigens in polymorphonuclear leukocytes and viremia.
J Infect Dis
1989
160
159
160
7
Boeckh
M
Bowden
RA
Goodrich
JM
Pettinger
M
Meyers
JD
Cytomegalovirus antigen detection in peripheral blood leukocytes after allogeneic marrow transplantation.
Blood
1992
80
1358
1364
8
Locatelli
F
Percivalle
E
Comoli
P
et al
Human cytomegalovirus (HCMV) infection in paediatric patients given allogeneic bone marrow transplantation: role of early antiviral treatment for HCMV antigenaemia on patients' outcome.
Br J Hematol
1994
88
64
71
9
Gerna
G
Lilleri
D
Baldanti
F
et al
Human cytomegalovirus immediate-early mRNAemia versus pp65 antigenemia for guiding pre-emptive therapy in children and young adults undergoing hematopoietic stem cell transplantation: a prospective, randomized, open-label trial.
Blood
2003
101
5053
5060
10
Lilleri
D
Baldanti
F
Gatti
M
et al
Clinically-based determination of safe DNAemia cutoff levels for preemptive therapy of human cytomegalovirus infections in solid organ and hematopoietic stem cell transplant recipients.
J Med Virol
2004
73
412
418
11
Gerna
G
Lilleri
D
Monitoring transplant patients for human cytomegalovirus: diagnostic update.
Herpes
2006
13
4
11
12
Gerna
G
Vitulo
P
Rovida
F
et al
Impact of human metapneumovirus and human cyto-megalovirus versus other respiratory viruses on the lower respiratory tract infections of lung transplant recipients.
J Med Virol
2006
78
408
416
13
Gerna
G
Revello
MG
Percivalle
E
Morini
F
Comparison of different immunostaining techniques and monoclonal antibodies to the lower matrix phosphoprotein (pp65) for optimal quantitation of human cytomegalovirus antigenemia.
J Clin Microbiol
1992
30
1232
1237
14
Gerna
G
Percivalle
E
Torsellini
M
Revello
MG
Standardization of the human cytomegalovirus antigenemia assay by means of in vitro generated pp65-positive peripheral blood polymorphonuclear leukocytes.
J Clin Microbiol
1998
36
3585
3589
15
Lilleri
D
Gerna
G
Fornara
C
Lozza
L
Maccario
R
Locatelli
F
Prospective simultaneous quantification of human cytomegalovirus-specific CD4+ and CD8+ T-cell reconstitution in young recipients of allogeneic hematopoietic stem cell transplants.
Blood
2006
108
1406
1412
16
Gooley
TA
Leisenring
W
Crowley
J
Storer
BE
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators.
Stat Med
1999
18
695
706
17
Klein
JP
Rizzo
JD
Zhang
M-J
Keiding
N
Statistical methods for analysis and presentation of the results of bone marrow transplants. Part I: unadjusted analysis.
Bone Marrow Transplant
2001
28
909
915
Sign in via your Institution