Seropositive umbilical cord blood transplant (UCBT) recipients are at increased risk for CMV complications. To reduce CMV complications, we adopted an intensive strategy that consisted of ganciclovir administered before transplantation (5 mg/kg intravenously daily from day −8 to day −2), high-dose acyclovir (2 g, 3 times daily) after transplanta-tion, and biweekly monitoring with a serum CMV PCR for preemptive therapy. Hazard rates and cumulative incidence of CMV complications along with days treated were compared in high-risk CMV-seropositive UCBT recipients who received the intensive strategy and a historical cohort who received a standard strategy. Of 72 seropositive patients, 29 (40%) received standard prophylaxis and 43 (60%) the new intensive approach. The hazard rate (HR) for CMV reactivation was lower for patients receiving the intensive strategy (HR 0.27, 95% confidence interval 0.15-0.48; P < .001) and led to fewer cases of CMV disease by 1 year (HR 0.11, 95% confidence interval 0.02-0.53; P = .006). In patients who reactivated, the intensive strategy also led to fewer days on CMV-specific antiviral therapy (median 42% [interquartile range 21-63] vs 70% [interquartile range 54-83], P < .001). Use of an intensive CMV prevention strategy in high-risk CMVseropositive UCBT recipients results in a significant decrease in CMV reactivation and disease.

CMV infection remains one of the most important infectious complications after hematopoietic stem cell transplantation (HSCT). CMV frequently reactivates in the posttransplantation period and can lead to life-threatening invasive disease, particularly in high-risk seropositive recipients.1  Additional negative effects, including increased rates of bacterial and fungal infections2,3  and graft failure,4  have also been shown to be associated with CMV reactivation in HSCT recipients. Current preemptive prevention strategies mitigate but have not eliminated life-threatening CMV disease, and the virus continues to be a cause of increased morbidity and mortality in multiple transplantation populations.3,5,6 

Umbilical cord blood transplant (UCBT) recipients in particular are at increased risk for CMV complications because of significant delay in immune reconstitution.7-10  Cord blood grafts are naive and have impaired functional recovery that may be more permissive to viral reactivation and less apt to control replication.11,12  Because high viral loads have been shown to be strong predictors for the development of CMV disease,13,14  UCBT recipients may also be at increased risk for the development of viral invasion. Incidence rates of CMV in UCBT vary, with reported rates of reactivation fluctuating from 21% to 100%15-17  and CMV disease between 6% and 21%,17-20  but different prevention methods and the inclusion of low-risk seronegative recipients limit comparisons.

Because of a concern for high rates of CMV complications in UCBT recipients at our institution, we instituted a new preemptive strategy that consists of the administration of ganciclovir before transplantation, primary prevention with high-dose acyclovir/valacyclovir after transplantation, and preemptive screening biweekly for CMV DNA. The authors of previous studies have demonstrated that the administration of both high-dose acyclovir and ganciclovir before transplantation are effective in reducing the rate of CMV reactivation and disease among allogeneic transplant recipients.21-23  To assess the safety and efficacy of this strategy on CMV outcomes, we compared a cohort of high-risk CMV-seropositive recipients of UCBT who received the standard institutional CMV prevention strategy and those who underwent this new intensive approach.

Patients

All patients who received a UCBT at the Fred Hutchinson Cancer Research Center between 2006 and 2010 and who were seropositive for CMV were eligible for inclusion in this study; only patients undergoing their first UCBT were included in these analyses. Patients were excluded if they died before day 14 after transplantation or had participated in primary CMV antiviral prevention trials. Patients were also excluded if they were receiving anti-CMV therapy at the time of transplantation for pretransplantation reactivation; all patients underwent pretransplantation CMV testing within 2 weeks before the start of conditioning.

Transplantation practices

Patients received a double cord blood transplantation if a suitable single-cord blood graft could not be found, as determined by institutional criteria. Selected cord blood units were required to be matched to the recipient at ≥ 4 of the 6 HLA loci on the basis of intermediate resolution typing at HLA-A and -B and allele-level for HLA-DRB1 typing; for recipients of 2 cord blood units, these units must be at least 3 of 6 HLA-matched to each other.

Decisions regarding the use of myeloablative or nonmyeloablative conditioning regimens were made by the primary transplantation team; CMV serostatus was not used as a criterion in selecting patient-specific conditioning regimens. Myeloablative conditioning typically consisted of cyclophosphamide 60 mg/kg intravenously daily for 2 days, total body irradiation (TBI) 1320 or 1200 cGy divided over 4 days, and fludarabine (Flu) 25 mg/m2 intravenously daily for 3 days. Other patients received Flu 30 mg/m2 intravenously daily for 5 days, treosulfan 14 g/m2 intravenously daily for 3 days, and a single fraction of TBI 200 cGy, or reduced-intensity conditioning consisting of Flu 40 mg/m2 intravenously daily for 5 days, a single dose of cyclophosphamide 50 mg/kg intravenously, and a single fraction of TBI 200 cGy. Patients who received either no previous chemotherapy or no chemotherapy in the 3 months preceding UCBT were given a greater dose of TBI at 300 cGy or had equine antithymocyte globulin at 15 mg/kg every 12 hours for 3 days added.

All patients received prophylactic immunosuppressive therapy for the prevention of GVHD consisting of cyclosporine A and mycophenolate mofetil. Acute GVHD was assessed by the use of standard criteria on the basis of organ involvement and categorized as acute GVHD grades 0-IV.24  The patient's underlying disease was categorized as standard or high risk on the basis of previously described criteria.25  All patients received standard prophylactic antimicrobial and antifungal agents during follow-up.26 

Antiviral prevention strategies during transplantation

UCBT patients in this study underwent 2 different prevention strategies. In the first historical cohort (“standard”), patients received our standard allogeneic regiment consisting of acyclovir 800 mg or valacyclovir 500 mg twice daily (and, during periods of mucositis, 250 mg/m2 intravenously acyclovir every 12 hours, adjusted for renal insufficiency) for varicella zoster virus and HSV prophylaxis. Patients were started on anti-CMV therapy if they developed ≥ 500 copies/mL or any antigenemia during weekly screening. A threshold for preemptive therapy of ≥ 100 copies/mL was used in patients receiving ≥ 1 mg/kg of steroids.26  After day 100, weekly PCR surveillance and preemptive therapy with valganciclovir (900 mg twice daily or appropriate dosing for pediatric patients) was started if patients had > 1000 copies/mL. A small number of patients underwent preemptive screening with pp65 antigenemia, and to compare prophylactic groups, these patients had their weekly clinical samples retrospectively retested for CMV DNA by PCR. These samples, which had been frozen at −20°C at the time of collection, were thawed and retested for CMV DNA by use of the same methods.27 

Because of observed rates of CMV-related complications in our UCBT recipients, an intensified strategy for CMV prophylaxis was implemented in June 2008; this strategy became standard for UCBT recipients in August 2008. In this second cohort (“intensive”), before transplantation CMV-seropositive patients received intravenous ganciclovir at 5 mg/kg daily from day −8 to day −2 during conditioning followed by high-dose acyclovir (2 g of valacyclovir every 8 hours or 500 mg/m2 acyclovir intravenously every 8 hours adjusted for renal insufficiency until tolerating oral medications) for the first 100 days. For patients < 40 kg and ≥ 20 kg, the dose of valacyclovir was 1 g every 8 hours; for those < 20 kg, the dose was 500 mg/m2 intravenously every 8 or 600 mg/m2 acyclovir every 6 hours. Patients in this cohort were tested biweekly by PCR, with a threshold for preemptive therapy at ≥ 25 copies/mL (limit of detection). After day 100, it was recommended that patients be placed on valganciclovir 900 mg once daily (dose adjusted for pediatric patients according standard guidelines) for 1 year; patients who could not tolerate valganciclovir had high-dose acyclovir continued.

For the purposes of preemptive therapy, patients were started on intravenous ganciclovir or foscarnet. Patients who were pre-engraftment or had intolerance to ganciclovir were given foscarnet. All patients received either ganciclovir 5 mg/kg intravenously or foscarnet 90 mg/kg twice daily 7-14 days as induction therapy, followed by maintenance therapy with once-daily dosing until routine surveillance testing was negative. Patients who did not respond after the second week of induction therapy continued on twice-daily dosing until CMV PCR levels began to decrease. Patients who rapidly cleared their CMV received at least 1 week of induction and 1 week of maintenance therapy. Resistance testing and decisions to change to alternate therapy (ie, foscarnet from ganciclovir) were at the discretion of the primary team and the infectious diseases consult service. Appropriate dose adjustments were made for patients with renal dysfunction.

Definitions

CMV reactivation was defined as any detection of CMV DNA in serum, and CMV disease was defined by standardized criteria.28  The initial CMV PCR level was defined as the CMV DNA copies/mL in serum at first detection, and maximum CMV PCR was highest recorded level during the first 100 days; total days of CMV were considered cumulative. For the purposes of analyses, a binary outcome for high-viral load defined as any CMV DNAemia level > 1000 copies/mL. Total days of CMV-specific antiviral use (ganciclovir and/or foscarnet) were calculated from start date to final dose administered during the first 100 days; days of multiple episodes of reactivation were summed cumulatively. Induction therapy was considered to be the period during which patients received the equivalent of twice-daily dosing of anti-CMV therapy. Acute kidney injury was assessed up to 100 days and was classified as a serum creatinine concentration that was 2 or 3 times as high as the baseline value.29 

Statistical methods

Patient and transplantation characteristics were compared by use of the Fisher exact test and Wilcoxon rank-sum test where applicable. We estimated the probability of CMV reactivation and disease for each treatment cohort by using cumulative incidence methods, with death considered a competing risk in analyses; similar cumulative incidence methods were used to estimate the rate of engraftment and acute GVHD. Statistical differences in cumulative incidence curves between groups were assessed by use of the Gray test.30  A multivariable Cox proportional hazards model was used to evaluate the impact of the prevention strategy on CMV reactivation and disease; separate hazard ratios (HRs) were determined for high-viral load and for early/preengraftment or late/postengraftment reactivation.

For the purposes of multivariable analyses, we defined 2 separate periods during follow-up: early/pre-engraftment (≤ 30 days after transplantation) and late/postengraftment (day > 30 to day 100); patients who reactivated during early/pre-engraftment were excluded for late/postengraftment analyses. Factors identified a priori for inclusion in the multivariate model for CMV reactivation were myeloablative versus nonmyeloablative conditioning, donor number (1 vs 2 cord blood grafts), and acute GVHD (grade > 2) as a time-dependent covariate.

To compare the amount of exposure to antiviral therapy, we determined the percentage of time on CMV antiviral therapy between treatment groups in the first 100 days by dividing the number of days on anti-CMV treatment by the total survival days in the first 100 days. The percentage of days that patients were exposed to anti-CMV therapy was compared between the 2 prevention strategies by use of the Wilcoxon-rank sum test. All P values were 2-sided and considered significant at the α = 0.05 level. All study activities were approved by the Fred Hutchinson Cancer Research Center Institutional Review Board, and all participants provided written informed consent according the principles of the Declaration of Helsinki.

Patient characteristics

Of 135 who underwent UCBT, 78 patients (58%) were CMV seropositive (Figure 1). Of these 78, a total of 6 patients were excluded because they died before day 14 after transplantation (n = 2), were on antiviral therapy at time of transplantation (n = 3), or were enrolled in a CMV prevention trial (n = 1). Of the remaining 72 patients, 29 (40%) received standard prophylaxis, and 43 (60%) received the intensive prevention strategy. Patient, transplantation, and graft characteristics stratified for the 2 cohorts are summarized in Table 1.25  The major difference between the 2 groups was that those who received the more intensive prophylactic strategy had greater risk of disease (P = .02). The 2 groups were otherwise similar with respect to HLA disparity, intensity of conditioning regimen, sex, total nucleated cells infused, and diagnosis, although there was a trend toward an increased age among those who received the more intensive prophylaxis (P = .10).

Figure 1

Schema of seropositive UCBT study population.

Figure 1

Schema of seropositive UCBT study population.

Close modal
Table 1

Characteristics of CMV-seropositive recipients undergoing UCBT (n = 72)

CharacteristicIntensive strategy, n = 43, n (%)Standard strategy, n = 29, n (%)P
Median age, y (IQR) 31.7 (16-57) 21.4 (10.1-41.9) .10 
Sex   .74 
    Female 22 (51) 16 (55)  
    Male 21 (49) 13 (45)  
No. of donors   .50 
    1 5 (12) 5 (17)  
    2 38 (88) 24 (83)  
HLA disparity*   .14 
    4/6 25 (58) 15 (52)  
    5/6 14 (32) 14 (48)  
    6/6 4 (10)  
Transplantation type   .98 
    Myeloablative 34 (79) 23 (79)  
    Nonmyeloablative 9 (21) 6 (21)  
Total nucleated dose (× 107/kg)   .82 
    median (IQR) 3.9 (3.1-5.1) 4.2 (2.5-6.0)  
Diagnosis   .60 
    Acute lymphoblastic leukemia 12 (28) 7 (24)  
    Acute myeloid leukemia 21 (49) 17 (59)  
    CML 3 (7) 3 (9)  
    Other 7 (16) 2 (6)  
Disease risk   .02 
    Standard risk 29 (67) 26 (90)  
    High risk 14 (33) 3 (10)  
CharacteristicIntensive strategy, n = 43, n (%)Standard strategy, n = 29, n (%)P
Median age, y (IQR) 31.7 (16-57) 21.4 (10.1-41.9) .10 
Sex   .74 
    Female 22 (51) 16 (55)  
    Male 21 (49) 13 (45)  
No. of donors   .50 
    1 5 (12) 5 (17)  
    2 38 (88) 24 (83)  
HLA disparity*   .14 
    4/6 25 (58) 15 (52)  
    5/6 14 (32) 14 (48)  
    6/6 4 (10)  
Transplantation type   .98 
    Myeloablative 34 (79) 23 (79)  
    Nonmyeloablative 9 (21) 6 (21)  
Total nucleated dose (× 107/kg)   .82 
    median (IQR) 3.9 (3.1-5.1) 4.2 (2.5-6.0)  
Diagnosis   .60 
    Acute lymphoblastic leukemia 12 (28) 7 (24)  
    Acute myeloid leukemia 21 (49) 17 (59)  
    CML 3 (7) 3 (9)  
    Other 7 (16) 2 (6)  
Disease risk   .02 
    Standard risk 29 (67) 26 (90)  
    High risk 14 (33) 3 (10)  

CML indicates chronic myelogenous leukemia; IQR, interquartile range; and UCBT, umbilical cord blood transplantation.

*

For recipients of 2 UCB units, the HLA matching reflects the worse matched of the 2 units.

Disease risk: standard refers to aplastic anemia, chronic myeloid leukemia in chronic phase, myelodysplastic syndromes without excess blasts, and leukemia and lymphoma in remission. High refers to all other hematologic malignancies.25  The Fisher exact test and Wilcoxon rank-sum analyses were used to calculate categorical and continuous variables, respectively.

Incidence and timing of CMV reactivation

As part of the intervention, patients in the intensive strategy had more frequent CMV testing in the first 100 days during follow-up (intensive, total 948 tests [median 24 tests per patient {IQR 17-28}] vs standard, total 559 tests [median 18 tests {IQR 16-25}], P = .049). In patients receiving the intensive strategy, first reactivation occurred at a median of 27 days (range, 3-77 days), compared with a median of 17 days (range, 3-65 days) to first reactivation in those treated with our standard strategy (P = .29), and the mean duration of serum CMV PCR detection was significantly shorter among patients who received the intensive approach (16.7 days [range, 2-95 days] vs 46.7 days [range, 4-91 days]; P < .001).

The cumulative incidence estimate of CMV reactivation was lower in those who received the more intensive approach compared with the standard group (26/43 [60%] vs 29/29 [100%], P < .001; Figure 2).30  The intensive strategy was also associated with a significant reduction in CMV reactivation in time-to-event analyses (HR 0.27; 95% confidence interval [CI] 0.15-0.48; P < .001; Table 2). Interestingly, a total of 15 of 29 (52%) in the standard cohort and 8 of 43 (19%) in the intensive cohort developed CMV reactivation before engraftment (P = .003). The hazards of early/preengraftment CMV reactivation were less in those receiving the more intensive strategy (HR 0.25; 95% CI 0.13-0.49; P < .001), but the risk was no different during the late/postengraftment period (HR 0.39; 95% CI 0.11-1.35; P = .14).

Figure 2

Cumulative incidence of CMV reactivation to day +100 by prevention strategy in seropositive UCBT recipients (n = 72). Competing risk for CMV reactivation considered death or retransplantation; P value determined by the Gray test.30 

Figure 2

Cumulative incidence of CMV reactivation to day +100 by prevention strategy in seropositive UCBT recipients (n = 72). Competing risk for CMV reactivation considered death or retransplantation; P value determined by the Gray test.30 

Close modal
Table 2

CMV outcomes in seropositive recipients undergoing UCBT by prevention strategy (n = 72)

OutcomeIntensive, n = 43 n (%)Standard, n = 29 n (%)Unadjusted HR (95% CI)PAdjusted HR (95% CI)*P
CMV reactivation 26 (60) 29 (100) 0.27 (0.15-0.47) < .001 0.27 (0.15-0.48) < .001 
    Timing       
        Early/pre-engraftment 15 (58) 25 (86) 0.26 (0.14-0.50) < .001 0.25 (0.13-0.49) < .001 
        Late/postengraftment 11 (42) 4 (14) 0.29 (0.09-0.93) .038 0.39 (0.11-1.35) .14 
    Complications       
        High-level viremia 3 (7) 24 (83) 0.05 (0.01-0.16) < .001 0.04 (0.01-0.15) < .001 
        Resistant CMV 1 (2) 3 (10)   – .30§ 
CMV disease 2 (5) 8 (28) 0.19 (0.04-0.91) .038 0.11 (0.02-0.53) .006 
CMV-associated death 0 (0) 3 (10)   – .06§ 
OutcomeIntensive, n = 43 n (%)Standard, n = 29 n (%)Unadjusted HR (95% CI)PAdjusted HR (95% CI)*P
CMV reactivation 26 (60) 29 (100) 0.27 (0.15-0.47) < .001 0.27 (0.15-0.48) < .001 
    Timing       
        Early/pre-engraftment 15 (58) 25 (86) 0.26 (0.14-0.50) < .001 0.25 (0.13-0.49) < .001 
        Late/postengraftment 11 (42) 4 (14) 0.29 (0.09-0.93) .038 0.39 (0.11-1.35) .14 
    Complications       
        High-level viremia 3 (7) 24 (83) 0.05 (0.01-0.16) < .001 0.04 (0.01-0.15) < .001 
        Resistant CMV 1 (2) 3 (10)   – .30§ 
CMV disease 2 (5) 8 (28) 0.19 (0.04-0.91) .038 0.11 (0.02-0.53) .006 
CMV-associated death 0 (0) 3 (10)   – .06§ 

CI indicates confidence interval; HR, hazard ratio; and UCBT, umbilical cord blood transplantation.

*

Multivariable model includes myeloablative vs nonmyeloablative conditioning, number of donor grafts, and GVHD (grade ≥ II, and time-dependent covariate) except where indicated.

Only patients who reactivated, early ≤ 30 days after transplantation and late > 30-100 days after transplantation.

High-level viremia is a PCR value ≥ 1000 copies at any point of time in the first 100 days after transplantation.

§

Calculated by the use of the Fisher exact probability test.

Adjusted only for acute GHVD in the multivariable model.

All but 6 of 72 patients (8%) were tested weekly or biweekly with CMV PCR. These 6 patients were all in the standard cohort, and 3 were tested with a mix of PCR and antigenemia testing whereas the 3 others had testing for the entire after transplantation period by antigenemia only. All 6 of these patients were documented to have developed CMV reactivation before retrospective testing and were treated with standard CMV preemptive therapy. PCR testing on frozen blood collected at the time of antigenemia determination demonstrated similar positive and negative results on retesting except in 2 patients. These 2 patients were tested after transplantation by antigenemia only and were found on retrospective PCR testing to be positive 7 and 11 days before their first positive antigenemia test.

During the first 100 days after transplantation, the mean PCR viral load in the intensive strategy cohort was significantly less than in the standard cohort at every week, except for the first (Figure 3). When we compared viral loads in those who developed CMV reactivation, we found that the initial and the maximum levels of PCR viral load were significantly lower for patients who received intensive prophylaxis compared with those who received the standard prevention; median initial viral load: 88 copies/mL (IQR, 67-100) versus 210 (IQR 63-649, P = .01) and median maximum viral load: 170 copies/mL (IQR 88-310) versus 3200 (IQR 1400-11 000, P < .001). The hazards of developing a viral load of ≥ 1000 copies was significantly less in the intensive strategy (HR 0.04; 95% CI 0.01-0.15; P < .001; Table 2).

Figure 3

Mean observed CMV viral load in UCBT recipients during the first 100 days after transplantation by type of prevention strategy (n = 72). Whiskers equal 95% CIs for weekly mean value.

Figure 3

Mean observed CMV viral load in UCBT recipients during the first 100 days after transplantation by type of prevention strategy (n = 72). Whiskers equal 95% CIs for weekly mean value.

Close modal

CMV disease

CMV disease was documented in a total of 8 patients during the first 100 days, 2 in the intensive group and 6 in the standard group (P = .054; Table 3). Two other patients in the standard cohort developed CMV disease after day 100 (both pneumonia, days 165, 191). The overall cumulative incidence of CMV disease at 1 year was 4.7% for patients treated with the new strategy and 27.6% for those treated with the standard strategy. When we evaluated CMV disease in time-to-event analyses, we found that the aggressive strategy was associated with a significant reduction in CMV disease (HR 0.11; 95% CI 0.02-0.53; P = .006; Table 2).

Table 3

Characteristics and outcomes of UCBT recipients who developed CMV disease (n = 10)

Age, yProphylaxisDiagnosisConditioning regimenNo. donorsaGVHD grade, daysTime to CMV reactivation, daysViral load at first reactivation, copies/mLTime to CMV disease, daysSites of CMV diseaseOut-come
Early disease (days 0-100)           
    42 Standard AML Cy + Flu + TBI (1320 cGy) 2 (21) 57 3354 92 GI Alive 
    64 Standard AML Cy + Flu + TBI (1320 cGy) 2 (34) 18 22 000 34 Lung Dead 
    23 Standard ALL Cy + Flu + ATG + TBI (200 cGy) 3 (15) 17* 35 17* Lung Alive 
    28 Standard AML CY + Flu + TBI (1320 cGy) NE 21* 63 33 Disseminated Dead 
    21 Standard ALL Cy + Flu + TBI (1320 cGy) NE 8* 6000 11* GI Dead 
    42 Standard ALL Cy + Flu + TBI (1320 cGy) 2 (35) 3* 100 66 GI Alive 
    1 Intensive Hystiocytosis Campath + Mel + Flu 3* 47 11* Lung Alive 
    53 Intensive AML Treo + Flu + TBI (200 cGy) NE 42 Lung Dead 
Late disease (days 101-365)           
    54 Standard AML Cy + Flu + ATG, TBI (200 cGy) 21 1053 191 Lung Dead 
    2 Standard AML Cy + Flu + TBI (1320 cGy) 3 (10) 8* 3200 165 Lung Alive 
Age, yProphylaxisDiagnosisConditioning regimenNo. donorsaGVHD grade, daysTime to CMV reactivation, daysViral load at first reactivation, copies/mLTime to CMV disease, daysSites of CMV diseaseOut-come
Early disease (days 0-100)           
    42 Standard AML Cy + Flu + TBI (1320 cGy) 2 (21) 57 3354 92 GI Alive 
    64 Standard AML Cy + Flu + TBI (1320 cGy) 2 (34) 18 22 000 34 Lung Dead 
    23 Standard ALL Cy + Flu + ATG + TBI (200 cGy) 3 (15) 17* 35 17* Lung Alive 
    28 Standard AML CY + Flu + TBI (1320 cGy) NE 21* 63 33 Disseminated Dead 
    21 Standard ALL Cy + Flu + TBI (1320 cGy) NE 8* 6000 11* GI Dead 
    42 Standard ALL Cy + Flu + TBI (1320 cGy) 2 (35) 3* 100 66 GI Alive 
    1 Intensive Hystiocytosis Campath + Mel + Flu 3* 47 11* Lung Alive 
    53 Intensive AML Treo + Flu + TBI (200 cGy) NE 42 Lung Dead 
Late disease (days 101-365)           
    54 Standard AML Cy + Flu + ATG, TBI (200 cGy) 21 1053 191 Lung Dead 
    2 Standard AML Cy + Flu + TBI (1320 cGy) 3 (10) 8* 3200 165 Lung Alive 

ALL indicates acute lymphoblastic leukemia; AML, acute myelogenous leukemia; ATG, antithymocyte globulin; Cy, cyclophosphamide; Flu, fludarabine; GI, gastrointestinal; Intensive, intensive prophylactic strategy; Mel, melphalan, NE, not evaluable; Standard, standard prophylactic strategy; TBI, total body irradiation; and UCBT, umbilical cord blood transplantation.

*

Complication developed preengraftment.

Developed early relapse.

When considering all 8 occurrences of CMV disease (both early and late CMV disease) in the standard cohort, we found that 4 developed pneumonia, 3 gastrointestinal, and 1 disseminated disease (Table 3). The median time-to-early CMV disease (≤ 100 days after transplantation, n = 6) was 33 days (range, 11-92 days), 2 of whom (33%) died secondary to CMV disease. One of the 2 patients in the standard group who developed disease during the late period (day > 100 to 1 year) also died from CMV pneumonia.

In the intensive cohort, 2 patients developed CMV disease (Table 3). The first patient was a pediatric patient who underwent transplantation because of Langerhans cell histiocytosis. While on intravenous acyclovir, the patient reactivated at day 3, and a bronchoalveolar lavage was positive for CMV by shell vial and PCR at day 11 after transplantation. He was treated with foscarnet and had a full recovery. In the second patient CMV was isolated in BAL by shell vial at day 42, at which time he had several other coinfections (vancomycin-resistant Enterococcus, gram negative bacteremia); CMV was never detected in serum. The patient died of multiorgan failure in the setting of relapse at day 47.

CMV-specific antiviral therapy

Patients who had documented reactivation in the intensive cohort had a smaller percentage of days in the first 100 days after transplantation on active anti-CMV therapy (median 42% [IQR 21-63] vs 70% [IQR 54-83], P < .001) and fewer days on induction dosing (median 16% [IQR 8-24] vs 29% [IQR 18-42], P < .001) compared with those who reactivated in the standard cohort (Figure 4). Of those who reactivated, a total of 4 patients developed CMV resistance during follow-up. In total 3 of 29 (10.3%) in the standard group developed UL97 mutations associated with ganciclovir resistance (day 30, 36, 113), whereas only 1 of 26 (3.8%) developed a UL97 mutation (day 246; P = .61). No patient developed a UL54 mutation during follow-up.

Figure 4

Use of anti-CMV antiviral therapy by type of prevention strategy. Anti-CMV therapy was considered ganciclovir and foscarnet; induction therapy was the period during which patients received the equivalent of twice-daily dosing of anti-CMV therapy. Only patients with proven CMV reactivation included. Total percentage was determined by days on anti-CMV therapy divided by total days alive during the first 100 days after transplantation. Whiskers equal 10th to 90th percentile, and solid dots equal outliers.

Figure 4

Use of anti-CMV antiviral therapy by type of prevention strategy. Anti-CMV therapy was considered ganciclovir and foscarnet; induction therapy was the period during which patients received the equivalent of twice-daily dosing of anti-CMV therapy. Only patients with proven CMV reactivation included. Total percentage was determined by days on anti-CMV therapy divided by total days alive during the first 100 days after transplantation. Whiskers equal 10th to 90th percentile, and solid dots equal outliers.

Close modal

Other transplantation outcomes

The time-to-engraftment and platelet recovery between those who received the intensive and standard strategies was similar (Table 4).29  Two patients in each cohort developed graft failure; one patient with secondary graft failure was observed in the standard cohort. In time-to-event analyses, the cumulative incidences of engraftment and GVHD (grade III-IV) were not significantly different (P = .20 and P = .07, respectively). In the first 100 days nonrelapse mortality was similar between the 2 groups (P = .30, log-rank) and at 1 year did not significantly differ between the 2 treatments groups (P = .63, log-rank). Importantly, high-dose acyclovir/valacyclovir did not appear to lead to additional renal toxicity during the first 100 days after transplantation (Table 4).

Table 4

Other transplantation-related outcomes in UCBT recipients by prevention strategy (n = 72)

OutcomesIntensive, n = 43 n (%)Standard, n = 29 n (%)P*
Time to engraftment 20 20 .49§ 
    Median days (IQR) (17-28) (14-29)  
Time to platelets ≥ 20 000 36 34 .37 
    Median days (IQR) (31-50) (29-45)  
Acute GVHD    
    Grade II-IV 28 (74) 25 (86) .21 
    Grade III-IV 8 (21) 10 (34) .22§ 
Renal function    
    Mean max creatinine (SD) 1.8 (1.2) 1.6 (1.0) .47 
    Acute kidney injury    
        × 2 baseline 28 (65) 18 (62) .81 
        × 3 baseline 16 (37) 7 (24) .31 
Nonrelapse mortality    
    First 100 days 8 (19) 2 (7) .30 
    1 year 11 (26) 6 (21) .63 
OutcomesIntensive, n = 43 n (%)Standard, n = 29 n (%)P*
Time to engraftment 20 20 .49§ 
    Median days (IQR) (17-28) (14-29)  
Time to platelets ≥ 20 000 36 34 .37 
    Median days (IQR) (31-50) (29-45)  
Acute GVHD    
    Grade II-IV 28 (74) 25 (86) .21 
    Grade III-IV 8 (21) 10 (34) .22§ 
Renal function    
    Mean max creatinine (SD) 1.8 (1.2) 1.6 (1.0) .47 
    Acute kidney injury    
        × 2 baseline 28 (65) 18 (62) .81 
        × 3 baseline 16 (37) 7 (24) .31 
Nonrelapse mortality    
    First 100 days 8 (19) 2 (7) .30 
    1 year 11 (26) 6 (21) .63 

IQR indicates interquartile range; UCBT, umbilical cord blood transplantation.

*

P values calculated with the Fisher exact probability test for categorical variables and Wilcoxon-rank sum for continuous variables, unless otherwise specified.

Only in patients who engrafted.

When calculated by time-to-event analysis P = .07 (Gray test).

§

When calculated by time to event analysis P = .20 (Gray test).

Acute kidney injury as defined by ≥ 2 times baseline creatinine or ≥ 3 times baseline, during the first 100 days.29 

Calculated by log-rank.

In this study, an intensive strategy of ganciclovir before transplantation followed by primary prophylaxis with high-dose acyclovir and frequent preemptive screening was highly effective in preventing CMV reactivation and disease in a high-risk cohort of CMV-seropositive UCBT recipients. Compared with a standard prophylaxis with a moderate dose of acyclovir that is used at our center, this new strategy was associated with fewer episodes of both CMV reactivation and invasive disease as well as lower levels of viral replication. In addition, this strategy led to fewer days on CMV specific antiviral therapy, fewer cases of drug resistance, and was not associated with kidney dysfunction, delayed engraftment, or other transplantation-related outcomes.

The most important finding in our study was that use of this intensive approach decreased a patient's risk of developing CMV disease in the first 100 days to 4.6%, a figure similar to rates observed when conventional BM and peripheral blood stem cell sources are used.31  The outcomes from this intensive strategy were likely a cumulative effect of different interventions aimed at CMV prevention, one of which was high-dose acyclovir/valacyclovir prophylaxis. Acyclovir's low side effect profile makes it an attractive option for prevention, and because high-dose acyclovir/valacyclovir has been shown to decrease CMV reactivation in other HSCT populations,21,32-34  some centers use this method of prevention as a standard in their UCBT recipients.35  However, high-dose acyclovir alone may have limited ability to decrease the risk of CMV disease in UCBT and other HSCT recipients.17,21,32,35  Other antiviral options may offer protection from early CMV disease19,36  but are known to cause additional toxicities.37  Primary prophylaxis has also been shown to be associated with a greater rate of late CMV disease in part because of the delayed recovery of CMV-specific T-cell immunity,13,19 

In combination with high-dose acyclovir, the application of twice-weekly CMV PCR testing in this strategy allowed for improved identification of early CMV reactivation and intervention at lower viral loads during episodes of reactivation. CMV replicates with a doubling time of approximately 1 day in HCT recipients,38  suggesting that more frequent testing may have the advantage of detecting low levels of CMV DNA before the development of rapid logarithmic growth. CMV viral load predicts the development of CMV disease14,39 ; therefore, interventions at a lower viral load threshold could also partially explain our decreased rate of CMV disease.

Patients also received ganciclovir before transplantation and late valganciclovir as part of our intensive prevention strategy. Ganciclovir administered before transplantation has been shown to decrease CMV complications in other HSCT populations23,34,40,41  and is hypothesized to decrease the risk of early posttransplantion CMV reactivation.41  Perhaps because of this intervention, we found that patients in our intensive strategy were significantly less likely to have preengraftment CMV. Most importantly, early reactivation appeared to have a negative effect on the rates of CMV disease in our study, and therefore the addition of ganciclovir before transplantation may have contributed to lower rates of invasive disease. In addition, although there are too few cases to evaluate in this study, this intervention before transplantation may also provide some protection against preengraftment disease, which is known to be associated with increased mortality.42  Because the authors of other studies have shown safety and efficacy of greater dosing before transplantation,34  an increase to treatment levels (5 mg/kg twice daily) before transplantation may have provided additional benefits. The use of valganciclovir likely led to less late CMV disease events, but because of limited late disease events in either cohort, we were not able to demonstrate a statistically significant benefit to its use.

The incidence of CMV reactivation in our standard cohort is consistent with previous studies in which seropositive UCBT recipients not receiving high-dose acyclovir or anti-CMV antiviral prophylaxis had reactivation rates reported to be between 70% and 100%.15,16,43  However, reactivation rates in our intensive strategy were slightly greater than those reported in CMV-seropositive UCBT recipients who received high dose of acyclovir prophylaxis.35  The use of CMV PCR as the screening method for preemptive therapy may have provided additional advantages in the UCBT population and may help explain differences in rates of CMV reactivation between studies. The presence of antigenemia detects fewer cases of CMV reactivation,44,45  may necessitate greater viral loads for detection,46  and positive results are more likely to be delayed until after the presence of symptoms of disease compared with patients screened by PCR.47  In fact, even when given identical prophylactic regimens, UCBT recipients tested by PCR for preemptive therapy developed fewer episodes of invasive disease compared with those who were screened with an antigenemia-based strategy.19 

In both cohorts CMV was detected by a highly sensitive quantitative double-primer PCR assay that has been shown to be superior to pp65 antigenemia with regard to sensitivity, specificity, and predictive values for CMV detection in serum specimens.27  Interestingly, 2 patients who were tested by antigenemia only developed preengraftment CMV, where diagnosis was delayed by more than a week compared with retrospective PCR testing. This increased sensitivity may have improved early detection in our study and allowed for prompt intervention. Combined with early detection, our lower threshold also enhanced the use of early CMV-specific antiviral preemptive therapy.

The increased rate of identification and the use of lower thresholds in our intensive cohort had the potential to increase the use of CMV specific antiviral therapy. The intensified strategy, however, did not lead to an increased use of antiviral therapy. In fact, numbers of days on ganciclovir or foscarnet were decreased significantly in patients who had documented reactivation (Figure 4). In addition, patients in the intensive cohort needed fewer days of induction therapy. The use of less CMV-specific antiviral therapy also likely contributed to fewer cases of CMV resistance in this cohort. Finally, because standard anti-CMV drugs used in preemptive therapy have toxicities that can in lead to increased mortality,37  the significant reduction observed in our intensive strategy likely provided additional benefits.

The exposure to high-dose acyclovir/valacyclovir also had the potential to increase the rate of drug-specific side effects because acyclovir has been shown to be associated with nephrotoxicty48  and neurologic complications.49  During study follow-up, there were no difference in renal outcomes between the 2 study cohorts, and no reports of drug-associated neurologic complications were noted in those treated with high-dose acyclovir. In addition, there appeared to be no effect on engraftment or nonrelapse mortality comparing the 2 cohorts. Perhaps most importantly, the reductions in intravenous ganciclovir/foscarnet use and CMV disease observed when using this intensive strategy likely outweigh any excess costs and or potential drug side effects from the increased use of these agents as primary prophylaxis.

As with any retrospective study, there are limits that are imposed by our data. We acknowledge that our 2 populations were not entirely comparable because patients in our more intensive prophylactic strategy were greater-risk transplantation recipients because of age and pretransplantation risk stratification. The most important limitation to our study is the small sample size of our study cohort. The majority of studies in which the authors assess CMV risk focus on entire cohorts of UCBT recipients and often include very low-risk CMV-seronegative patients.17,18,35,50  Because recipient seropositivity remains the most important risk factor for CMV in HSCT and because others have demonstrated increased rates of disease and reactivation in CMV-seropositive UCBT recipients,16,19  we limited our analyses to this high-risk population. Although it was a smaller cohort size, it allowed us to assess a greater incidence of adverse CMV end points and demonstrate significant differences between our 2 strategies.

Finally, by implementing multiple components in this intensive approach, we found it was not possible to unravel the benefit of each specific intervention. For example, the protection from early reactivation (< day 30) could be because of pretransplantation ganciclovir, high-dose acyclovir, or a combination of both components. On the basis of these data, we can only recommend this strategy as a combination of therapies, but future prospective randomized trials could better clarify the importance of each respective intervention.

In conclusion, our data show that an intensive approach to CMV prevention in seropositive UCBT recipients leads to decreased rates of CMV complications. Through the use of pre- and posttransplantation antiviral prophylaxis, increased frequency of preemptive screening, and lowered thresholds for the institution of preemptive therapy, we were able to demonstrate additional protection against CMV disease and the development of preengraftment CMV reactivation. This intensive approach was well tolerated and led to a significant reduction in the use of preemptive antiviral therapy. Until the development of less-toxic antiviral prophylaxis for CMV prevention, this aggressive approach may be used to provide enhanced protection from CMV in high risk UCBT recipients and could be considered in other populations that are at increased risk of CMV complications.

Presented in part as an oral presentation at the 52nd Annual Meeting of the American Society of Hematology, Orlando, FL, December 4, 2010 and the 9th Annual International Umbilical Cord Blood Transplantation Symposium, San Francisco, CA, June 24, 2011.

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.

The authors Terry Stevens-Ayers, Tracy Santo, and Meei-Li Huang for their assistance with late CMV PCR testing. They also thank Denise Ziegler and Mary Joy Lopez for their assistance in care of the patients.

This work was supported by National Institutes of Health grants K23HL077446, R24HL74445, 1RC2HL101844, K23HL096831, 1K24HL093294, CA18029, and CA15704. S.P. is also supported by an ASBMT/Viropharma New Investigator Award. V.C. was supported by the Infectious Diseases Society of America's Medical Scholars Program. C.D. is a Damon Runyon Clinical Investigator supported in part by the Damon Runyon Cancer Research Foundation (CI# 35-07).

National Institutes of Health

Contribution: F.M. and S.A.P. participated in research design, data collection, performed statistical analyses, and wrote the manuscript; H.X. performed statistical analyses under direction of W.M.L., and both contributed to the research design, writing, and review of the manuscript; V.C. participated in data collection and contributed to the writing and review of the manuscript; and J.A.G., I.R., M.J.B., and C.D. participated in research design and contributed to the writing and review of the manuscript.

Conflict-of-interest disclosure: S.A.P. has received research support from Chimerix Inc and Viropharma Inc and has received consulting fees from Chimerix, Inc. J.A.G. has received research support and consulting fees from Chimerix Inc. M.J.B. has received research support from Chimerix Inc, GlaxoSmithKline, Roche Laboratories, Vical Inc, and Viropharma Inc; he has received consulting fees from Astellas, Boehringer Ingelheim, Chimerix Inc, Novartis, Roche/Genentech, Vical Inc, and Viropharma Inc. The remaining authors declare no competing financial interests.

Correspondence: Steven Pergam, MD, MPH, 1100 Fairview Ave, N D3-100, Seattle, WA 98109; e-mail: spergam@fhcrc.org.

1
Boeckh
 
M
Nichols
 
WG
Papanicolaou
 
G
Rubin
 
R
Wingard
 
JR
Zaia
 
J
Cytomegalovirus in hematopoietic stem cell transplant recipients: current status, known challenges, and future strategies.
Biol Blood Marrow Transplant
2003
, vol. 
9
 
9
(pg. 
543
-
558
)
2
Marr
 
KA
Carter
 
RA
Boeckh
 
M
Martin
 
P
Corey
 
L
Invasive aspergillosis in allogeneic stem cell transplant recipients: changes in epidemiology and risk factors.
Blood
2002
, vol. 
100
 
13
(pg. 
4358
-
4366
)
3
Nichols
 
WG
Corey
 
L
Gooley
 
T
Davis
 
D
Boeckh
 
M
High risk of death due to bacterial and fungal infection among cytomegalovirus (CMV)-seronegative recipients of stem cell transplants from seropositive donors: evidence for indirect effects of primary CMV infection.
J Infect Dis
2002
, vol. 
185
 
3
(pg. 
273
-
282
)
4
Steffens
 
HP
Podlech
 
J
Kurz
 
S
Angele
 
P
Dreis
 
D
Reddehase
 
MJ
Cytomegalovirus inhibits the engraftment of donor bone marrow cells by downregulation of hemopoietin gene expression in recipient stroma.
J Virol
1998
, vol. 
72
 
6
(pg. 
5006
-
5015
)
5
Broers
 
AE
van Der Holt
 
R
van Esser
 
JW
, et al. 
Increased transplant-related morbidity and mortality in CMV-seropositive patients despite highly effective prevention of CMV disease after allogeneic T-cell–depleted stem cell transplantation.
Blood
2000
, vol. 
95
 
7
(pg. 
2240
-
2245
)
6
Humar
 
A
Wood
 
S
Lipton
 
J
, et al. 
Effect of cytomegalovirus infection on 1-year mortality rates among recipients of allogeneic bone marrow transplants.
Clin Infect Dis
1998
, vol. 
26
 
3
(pg. 
606
-
610
)
7
Brunstein
 
CG
Barker
 
JN
Weisdorf
 
DJ
, et al. 
Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease.
Blood
2007
, vol. 
110
 
8
(pg. 
3064
-
3070
)
8
Cornetta
 
K
Laughlin
 
M
Carter
 
S
, et al. 
Umbilical cord blood transplantation in adults: results of the prospective Cord Blood Transplantation (COBLT).
Biol Blood Marrow Transplant
2005
, vol. 
11
 
2
(pg. 
149
-
160
)
9
Long
 
GD
Laughlin
 
M
Madan
 
B
, et al. 
Unrelated umbilical cord blood transplantation in adult patients.
Biol Blood Marrow Transplant
2003
, vol. 
9
 
12
(pg. 
772
-
780
)
10
Brown
 
JA
Boussiotis
 
VA
Umbilical cord blood transplantation: basic biology and clinical challenges to immune reconstitution.
Clin Immunol
2008
, vol. 
127
 
3
(pg. 
286
-
297
)
11
D'Arena
 
G
Musto
 
P
Cascavilla
 
N
, et al. 
Flow cytometric characterization of human umbilical cord blood lymphocytes: immunophenotypic features.
Haematologica
1998
, vol. 
83
 
3
(pg. 
197
-
203
)
12
Komanduri
 
KV
St John
 
LS
de Lima
 
M
, et al. 
Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T-cell skewing.
Blood
2007
, vol. 
110
 
13
(pg. 
4543
-
4551
)
13
Boeckh
 
M
Leisenring
 
W
Riddell
 
SR
, et al. 
Late cytomegalovirus disease and mortality in recipients of allogeneic hematopoietic stem cell transplants: importance of viral load and T-cell immunity.
Blood
2003
, vol. 
101
 
2
(pg. 
407
-
414
)
14
Emery
 
VC
Sabin
 
CA
Cope
 
AV
Gor
 
D
Hassan-Walker
 
AF
Griffiths
 
PD
Application of viral-load kinetics to identify patients who develop cytomegalovirus disease after transplantation.
Lancet
2000
, vol. 
355
 
9220
(pg. 
2032
-
2036
)
15
Takami
 
A
Mochizuki
 
K
Asakura
 
H
Yamazaki
 
H
Okumura
 
H
Nakao
 
S
High incidence of cytomegalovirus reactivation in adult recipients of an unrelated cord blood transplant.
Haematologica
2005
, vol. 
90
 
9
(pg. 
1290
-
1292
)
16
Tomonari
 
A
Iseki
 
T
Ooi
 
J
, et al. 
Cytomegalovirus infection following unrelated cord blood transplantation for adult patients: a single institute experience in Japan.
Br J Haematol
2003
, vol. 
121
 
2
(pg. 
304
-
311
)
17
Walker
 
CM
van Burik
 
JA
De For
 
TE
Weisdorf
 
DJ
Cytomegalovirus infection after allogeneic transplantation: comparison of cord blood with peripheral blood and marrow graft sources.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
9
(pg. 
1106
-
1115
)
18
Matsumura
 
T
Narimatsu
 
H
Kami
 
M
, et al. 
Cytomegalovirus infections following umbilical cord blood transplantation using reduced intensity conditioning regimens for adult patients.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
5
(pg. 
577
-
583
)
19
Montesinos
 
P
Sanz
 
J
Cantero
 
S
, et al. 
Incidence, risk factors, and outcome of cytomegalovirus infection and disease in patients receiving prophylaxis with oral valganciclovir or intravenous ganciclovir after umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
6
(pg. 
730
-
740
)
20
Yoo
 
KH
Lee
 
SH
Sung
 
KW
, et al. 
Current status of pediatric umbilical cord blood transplantation in Korea: a multicenter retrospective analysis of 236 cases.
Am J Hematol
2011
, vol. 
86
 
1
(pg. 
12
-
17
)
21
Ljungman
 
P
de La Camara
 
R
Milpied
 
N
, et al. 
Randomized study of valacyclovir as prophylaxis against cytomegalovirus reactivation in recipients of allogeneic bone marrow transplants.
Blood
2002
, vol. 
99
 
8
(pg. 
3050
-
3056
)
22
Meyers
 
JD
Reed
 
EC
Shepp
 
DH
, et al. 
Acyclovir for prevention of cytomegalovirus infection and disease after allogeneic marrow transplantation.
N Engl J Med
1988
, vol. 
318
 
2
(pg. 
70
-
75
)
23
Winston
 
DJ
Ho
 
WG
Bartoni
 
K
, et al. 
Ganciclovir prophylaxis of cytomegalovirus infection and disease in allogeneic bone marrow transplant recipients. Results of a placebo-controlled, double-blind trial.
Ann Intern Med
1993
, vol. 
118
 
3
(pg. 
179
-
184
)
24
Przepiorka
 
D
Smith
 
TL
Folloder
 
J
, et al. 
Risk factors for acute graft-versus-host disease after allogeneic blood stem cell transplantation.
Blood
1999
, vol. 
94
 
4
(pg. 
1465
-
1470
)
25
Mielcarek
 
M
Storer
 
BE
Boeckh
 
M
, et al. 
Initial therapy of acute graft-versus-host disease with low-dose prednisone does not compromise patient outcomes.
Blood
2009
, vol. 
113
 
13
(pg. 
2888
-
2894
)
26
Nakamae
 
H
Kirby
 
KA
Sandmaier
 
BM
, et al. 
Effect of conditioning regimen intensity on CMV infection in allogeneic hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
6
(pg. 
694
-
703
)
27
Boeckh
 
M
Huang
 
M
Ferrenberg
 
J
, et al. 
Optimization of quantitative detection of cytomegalovirus DNA in plasma by real-time PCR.
J Clin Microbiol
2004
, vol. 
42
 
3
(pg. 
1142
-
1148
)
28
Ljungman
 
P
Griffiths
 
P
Paya
 
C
Definitions of cytomegalovirus infection and disease in transplant recipients.
Clin Infect Dis
2002
, vol. 
34
 
8
(pg. 
1094
-
1097
)
29
Hingorani
 
SR
Guthrie
 
K
Batchelder
 
A
, et al. 
Acute renal failure after myeloablative hematopoietic cell transplant: incidence and risk factors.
Kidney Int
2005
, vol. 
67
 
1
(pg. 
272
-
277
)
30
Gray
 
RJ
A Class of K-sample tests for comparing the cumulative incidence of a competing risk.
Ann Stat
1998
, vol. 
16
 
3
(pg. 
1141
-
1154
)
31
Gooley
 
TA
Chien
 
JW
Pergam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
32
Prentice
 
HG
Gluckman
 
E
Powles
 
RL
, et al. 
Impact of long-term acyclovir on cytomegalovirus infection and survival after allogeneic bone marrow transplantation. European Acyclovir for CMV Prophylaxis Study Group.
Lancet
1994
, vol. 
343
 
8900
(pg. 
749
-
753
)
33
Hazar
 
V
Kansoy
 
S
Kupesiz
 
A
Aksoylar
 
S
Kantar
 
M
Yesilipek
 
A
High-dose acyclovir and pre-emptive ganciclovir in prevention of cytomegalovirus disease in pediatric patients following peripheral blood stem cell transplantation.
Bone Marrow Transplant
2004
, vol. 
33
 
9
(pg. 
931
-
935
)
34
Kline
 
J
Pollyea
 
DA
Stock
 
W
, et al. 
Pre-transplant ganciclovir and post transplant high-dose valacyclovir reduce CMV infections after alemtuzumab-based conditioning.
Bone Marrow Transplant
2006
, vol. 
37
 
3
(pg. 
307
-
310
)
35
Beck
 
JC
Wagner
 
JE
DeFor
 
TE
, et al. 
Impact of cytomegalovirus (CMV) reactivation after umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
2
(pg. 
215
-
222
)
36
Narimatsu
 
H
Kami
 
M
Kato
 
D
, et al. 
Reduced dose of foscarnet as preemptive therapy for cytomegalovirus infection following reduced-intensity cord blood transplantation.
Transplant Infectious Dis
2007
, vol. 
9
 
1
(pg. 
11
-
15
)
37
Salzberger
 
B
Bowden
 
RA
Hackman
 
RC
Davis
 
C
Boeckh
 
M
Neutropenia in allogeneic marrow transplant recipients receiving ganciclovir for prevention of cytomegalovirus disease: risk factors and outcome.
Blood
1997
, vol. 
90
 
6
(pg. 
2502
-
2508
)
38
Emery
 
VC
Cope
 
AV
Bowen
 
EF
Gor
 
D
Griffiths
 
PD
The dynamics of human cytomegalovirus replication in vivo.
J Exp Med
1999
, vol. 
190
 
2
(pg. 
177
-
182
)
39
Ljungman
 
P
Perez-Bercoff
 
L
Jonsson
 
J
, et al. 
Risk factors for the development of cytomegalovirus disease after allogeneic stem cell transplantation.
Haematologica
2006
, vol. 
91
 
1
(pg. 
78
-
83
)
40
Atkinson
 
K
Downs
 
K
Golenia
 
M
, et al. 
Prophylactic use of ganciclovir in allogeneic bone marrow transplantation: absence of clinical cytomegalovirus infection.
Br J Haematol
1991
, vol. 
79
 
1
(pg. 
57
-
62
)
41
Verma
 
A
Devine
 
S
Morrow
 
M
, et al. 
Low incidence of CMV viremia and disease after allogeneic peripheral blood stem cell transplantation. Role of pretransplant ganciclovir and post-transplant acyclovir.
Bone Marrow Transplant
2003
, vol. 
31
 
9
(pg. 
813
-
816
)
42
Limaye
 
AP
Bowden
 
RA
Myerson
 
D
Boeckh
 
M
Cytomegalovirus disease occurring before engraftment in marrow transplant recipients.
Clin Infect Dis
1997
, vol. 
24
 
5
(pg. 
830
-
835
)
43
Parody
 
R
Martino
 
R
Rovira
 
M
, et al. 
Severe infections after unrelated donor allogeneic hematopoietic stem cell transplantation in adults: comparison of cord blood transplantation with peripheral blood and bone marrow transplantation.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
7
(pg. 
734
-
748
)
44
Halfon
 
P
Berger
 
P
Khiri
 
H
, et al. 
Algorithm based on CMV kinetics DNA viral load for preemptive therapy initiation after hematopoietic cell transplantation.
J Med Virol
2011
, vol. 
83
 
3
(pg. 
490
-
495
)
45
Cortez
 
KJ
Fischer
 
SH
Fahle
 
GA
, et al. 
Clinical trial of quantitative real-time polymerase chain reaction for detection of cytomegalovirus in peripheral blood of allogeneic hematopoietic stem-cell transplant recipients.
J Infect Dis
2003
, vol. 
188
 
7
(pg. 
967
-
972
)
46
de la Cruz-Vicente
 
F
Perez-Romero
 
P
Aguilar-Guisado
 
M
Cisneros-Herreros
 
JM
Urbano-Ispizua
 
A
Espigado
 
I
Differences in cytomegalovirus replication quantified using quantitative polymerase chain reaction and antigenemia after allogeneic stem cell transplantation.
Transplant Proc
2010
, vol. 
42
 
8
(pg. 
3230
-
3231
)
47
Mori
 
T
Aisa
 
Y
Shimizu
 
T
, et al. 
Prevention of cytomegalovirus infection by valaciclovir after allogeneic bone marrow transplantation from an unrelated donor.
Int J Hematol
2006
, vol. 
83
 
3
(pg. 
266
-
270
)
48
Perazella
 
MA
Drug-induced renal failure: update on new medications and unique mechanisms of nephrotoxicity.
Am J Med Sci
2003
, vol. 
325
 
6
(pg. 
349
-
362
)
49
Izzedine
 
H
Mercadal
 
L
Aymard
 
G
, et al. 
Neurotoxicity of valacyclovir in peritoneal dialysis: a pharmacokinetic study.
Am J Nephrol
2001
, vol. 
21
 
2
(pg. 
162
-
164
)
50
Sauter
 
C
Abboud
 
M
Jia
 
X
, et al. 
Serious infection risk and immune recovery after double-unit cord blood transplantation without antithymocyte globulin.
Biol Blood Marrow Transplant
2011
, vol. 
17
 (pg. 
1460
-
1471
)

Author notes

*

F.M. and S.A.P. contributed equally to this work.

Sign in via your Institution