• The risk-based letermovir use strategy is effective for CMV infection prophylaxis and allows some patients at low risk not to use letermovir.

  • Among patients at low risk without letermovir, most had negative or transient positive CMV DNA without clinically significant CMV infection.

Letermovir is the first approved drug for cytomegalovirus (CMV) infection prophylaxis in adult patients who are CMV positive undergoing allogeneic hematopoietic cell transplantation (allo-HCT). Because CMV infection risk varies from patient to patient, we evaluated whether a risk-based strategy could be effective. In this single-center study, all consecutive adult patients who were CMV positive and underwent allo-HCT between 2015 and 2021 were included. During period 1 (2015-2017), letermovir was not used, whereas during period 2 (2018-2021), letermovir was used in patients at high risk but not in patients at low risk, except in those receiving corticosteroids. In patients at high risk, the incidence of clinically significant CMV infection (csCMVi) in period 2 was lower than that in period 1 (P < .001) by week 14 (10.5% vs 51.6%) and week 24 (16.9% vs 52.7%). In patients at low risk, although only 28.6% of patients received letermovir in period 2, csCMVi incidence was also significantly lower (P = .003) by week 14 (7.9% vs 29.0%) and week 24 (11.2% vs 33.3%). Among patients at low risk who did not receive letermovir (n = 45), 23 patients (51.1%) experienced transient positive CMV DNA without csCMVi, whereas 17 patients (37.8%) experienced negative results. In both risk groups, the 2 periods were comparable for CMV disease, overall survival, progression-free survival, relapse, and nonrelapse mortality. We concluded that a risk-based strategy for letermovir use is an effective strategy which maintains the high efficacy of letermovir in patients at high risk but allows some patients at low risk to not use letermovir.

Cytomegalovirus (CMV) infection, defined as detection of CMV nucleic acid in any body fluid or tissue specimen, has historically been common after allogeneic hematopoietic cell transplantation (allo-HCT), particularly in recipients who are CMV-positive, depending on the transplantation modalities and the characteristics of the donor/recipient match.1,2 CMV disease, which is the clinical expression of CMV infection, has significantly decreased with the use of a preemptive strategy based on regular monitoring of circulating CMV DNA and initiation of preemptive antiviral therapy at predefined thresholds.3,4 Recently, letermovir, a CMV DNA terminase complex inhibitor, has been approved for the prophylaxis of CMV infection and disease in adult recipients of allo-HCT who are CMV-positive, based on results of a clinical trial published by Marty et al5 in 2017. This large, randomized, placebo-controlled phase 3 study showed that letermovir significantly reduced the risk of clinically significant CMV infection (csCMVi; defined as CMV disease or CMV viremia leading to preemptive treatment) by week 24 after transplantation. Because results were consistent across risk groups, letermovir was considered as a universal prophylaxis through day 100 after allo-HCT in the CMV-positive population.6 Although previous retrospective studies linked CMV infection with an increased risk of overall mortality, thus allowing viral load to be used as a surrogate clinical end point in clinical trials, when compared with that in placebo recipients, all-cause mortality through week 48 after allo-HCT was not significantly lower in letermovir recipients, especially in patients at low risk for CMV infection.7,8 

In view of these conflicting results, it is quite possible that the efficiency of letermovir could be discussed in patients with low CMV infection risk.9 The recommended thresholds for initiating anti-CMV therapy in the phase 3 study were lower (>300 copies per mL for patients at low risk and >150 copies per mL for patients at high risk) than those used in practice, and we can assume that without treatment, these patients would have experienced only transient episodes of low detectable viremia without CMV disease.10 Furthermore, compared with solid transplantation in which valganciclovir prophylaxis has long been used without significant toxicity, recent studies in this area have shown that preemptive and prophylactic strategies are comparable in selected patients.11 CMV infection risk in the allo-HCT setting depends on several risk factors, as described in a number of retrospective studies.12-16 The high-risk group in the letermovir phase 3 study was defined as presenting one or more of the following defined criteria: related or unrelated donor with mismatch, haploidentical donor, use of cord blood, use of ex vivo T-cell depleted graft, and having graft-versus-host disease (GVHD).5 However, when considered separately, the predictive value of these factors is poor. Recently, we developed a simple weighted score (excluding haploidentical and second transplantations) from 0 to 20 points that evaluates, at the time of transplant, a patient’s risk of developing csCMVi (unrelated donor: +5; antithymocyte globulin: +4; CMV-negative donor: +3; mycophenolate mofetil: +3; total body irradiation: +3; and myeloablative conditioning: +2) and which classifies patients into 4 risk groups with distinct predicted rates of CMV infection.17 

The objective of this study was to evaluate whether a risk-based strategy could maintain the efficacy of letermovir against csCMVi without impairing related outcomes.

Patients

All consecutive adult patients who were CMV positive and underwent allo-HCT from 1 January 2015 to 31 July 2021 at the Lille University Hospital, France were included. Patients were divided into 2 periods based on the date of transplantation: a first period (period 1) without any use of letermovir between 1 January 2015 and 31 December 2017, and a second period (period 2) with the use of letermovir based on the risk of CMV infection between 1 January 2018 and 31 July 2021. Inside of each period, patients were identified as being low-risk (CMV risk score low [0-4 points] or intermediate-low [5-10 points]) or as high-risk for csCMVi (CMV risk score intermediate-high [11-14 points] or high [15-20 points], haploidentical transplantation and second transplantation). Prospectively collected data came from Project Manager Internet Server (ProMISe), the international database coordinated by European Society for Bone Marrow Transplantation, and on-site individual patient files were used. The follow-up was censored as of 31 December 2021. The study was led according to the Declaration of Helsinki and registered with the French Data Protection Authority (CNIL). All patients and donors provided informed consent concerning the retrospective use of their clinical data.

Letermovir use strategy

In period 1, letermovir was not available and was therefore not used. In period 2, letermovir was administered in oral form to all patients at high risk on day 8 posttransplant and continued until day 100 (week 14) after transplantation according to approval. In contrast, letermovir was not systematically administered to patients at low risk during period 2, but because of the change in CMV infection risk over time, letermovir was initiated and continued until day 100 if corticosteroids were used before day 100 after transplantation. In both risk groups, in case of csCMVi, letermovir was discontinued and not resumed. CMV DNA quantification was performed weekly on EDTA-anticoagulated whole blood samples by quantitative real-time polymerase chain reaction using the kPCR PLX CMV DNA Assay on the Versant kPCR Molecular System (Siemens, Erlangen, Germany) until 2018 and, from 2019, the AltoStar CMV PCR Kit 1.5, on the AltoStar Automation System (Altona Diagnostics, Hamburg, Germany). The threshold for use of preemptive therapy in CMV viremia was determined in a consensual manner at 3.5 log10 IU/mL.3,18-20 

Statistical analysis

Categorical variables were expressed as frequency (percentage). Age was reported as a median with interquartile range. The primary end point was the cumulative incidence of csCMVi defined as the use of preemptive therapy for CMV viremia or CMV disease. The secondary endpoints were the following: cumulative incidence of CMV disease, overall survival (OS) defined as the time from allo-HCT to death, progression-free survival (PFS) defined as the time from allo-HCT to disease progression or death, cumulative incidence of relapse (CIR) defined as the time from allo-HCT to disease progression, and nonrelapse mortality (NRM) defined as the time from allo-HCT to death without evidence of underlying disease relapse.

Comparisons of patient characteristics between the 2 periods were performed using χ2 test or Fisher exact test for categorical variables and Mann–Whitney U test for the age at allo-HCT. The cumulative incidences of csCMVi (with landmark analysis for late csCMVi), CMV disease, relapse, and NRM with 95% confidence interval (CI) were estimated using the Kalbfleisch and Prentice method by considering death as a competing event (except for NRM: relapse) and was compared between the 2 periods (period 1 vs period 2) in each risk group (low- and high-risk) by using the Fine and Gray competing risk regression model. OS and PFS were estimated using the Kaplan–Meier method and compared between the 2 periods using the Cox regression model. All analyses were censored at 52 weeks after transplantation. The 14-week cumulative incidence of csCMVi was estimated in subgroups of patients. Heterogeneity tests were performed to assess whether the effect of period on the risk of csCMVi differed based on patient characteristics.

Statistical testing was conducted at the two-tailed α-level of 0.05. Statistical analyses were performed using SAS software (SAS Institute, Cary, NC, version 9.4).

Study population

A total of 316 adult patients who were CMV positive underwent allo-HCT from 1 January 2015 to 31 July 2021. Period 1 comprised 186 patients and the period 2 comprised 130 patients (Figure 1). The baseline characteristics of the population and variables used to calculate CMV risk score are presented in Table 1. Median age was 56.0 years (interquartile range 43.5-64.0), and 52.5% of patients were male. Regarding patients and transplantation modalities, period 1 and period 2 were comparable, except for haploidentical transplantations which were slightly less frequent in period 2. According to the CMV risk score, 156 (49.4%) patients were at low risk and 160 (50.6%) at high risk for CMV infection, and this distribution of patients was balanced in both periods (50% vs 50% in period 1 and 48.5% vs 51.5% in period 2). The median follow-up was 59.5 (95% CI 56.5-64.5) months for period 1 and 17.8 (14.3-20.3) months for period 2.

Figure 1.

Study design.

Figure 1.

Study design.

Close modal
Table 1.

Study population characteristics

Total (n = 316)Period 1 (n = 186)Period 2 (n = 130)P
Age – median (IQR) 56.0 (43.5-64.0) 56.0 (42.0-64.0) 58.5 (47.0-65.0) .23 
< 50 y - n (%) 106 (33.5) 65 (35.0) 41 (31.5)  
50-60 y - n (%) 93 (29.4) 57 (30.7) 36 (27.7)  
> 60 y - n (%) 117 (37.0) 64 (34.4) 53 (40.8)  
Sex - n (%)    .50 
Male 166 (52.5) 103 (55.4) 63 (48.5)  
Female 150 (47.5) 83 (44.6) 67 (51.5)  
Disease - n (%)    .85 
Acute myeloid leukemia 132 (41.8) 74 (39.8) 58 (44.6)  
Myelodysplastic syndrome 58 (18.4) 34 (18.3) 24 (18.5)  
Acute lymphoblastic leukemia 48 (15.2) 27 (14.5) 21 (16.2)  
Lymphoma 24 (7.6) 16 (8.6) 8 (6.2)  
Primary/secondary myelofibrosis 22 (7.0) 14 (7.5) 8 (6.2)  
Others 32 (10.1) 21 (11.3) 11 (8.5)  
Second transplant - n (%) 21 (6.6) 16 (8.6) 5 (3.8) .095 
Donor type - n (%)    .044 
Matched sibling 66 (20.9) 42 (22.6) 24 (18.5)  
Matched unrelated 155 (49.1) 84 (45.2) 71 (54.6)  
Mismatched unrelated 42 (13.3) 21 (11.3) 21 (16.2)  
Haploidentical 53 (16.8) 39 (21.0) 14 (10.8)  
Donor CMV serostatus - n (%)    .43 
Positive 224 (70.9) 135 (72.6) 89 (68.5)  
Negative 92 (29.1) 51 (27.4) 41 (31.5)  
Stem cell source - n (%)    .066  
Bone marrow 141 (44.6) 92 (49.5) 49 (37.7)  
Peripheral blood stem cell 171 (54.1) 94 (50.5) 77 (59.2)  
Cord blood 4 (1.3) 0 (0) 4 (3.1)  
Conditioning regimen - n (%)    .56 
Myeloablative 147 (46.5) 84 (45.2) 63 (48.5)  
Reduced intensity 169 (53.5) 102 (54.8) 67 (51.5)  
Total body irradiation - n (%) 62 (19.6) 34 (18.3) 28 (21.5) .47 
GVHD prophylaxis - n (%)    .14 
Cyclosporin A + methotrexate 229 (72.5) 129 (69.4) 100 (76.9)  
Cyclosporin A + MMF 87 (27.5) 57 (30.6) 30 (23.1)  
In vivo T-cell depletion - n (%) 219 (69.3) 127 (68.3) 92 (70.8) .64 
Antithymocyte globulin 155 (49.1) 82 (44.1) 73 (56.2)  
Post-transplant cyclophosphamide 64 (20.3) 45 (24.2) 19 (14.6)  
CMV risk score - n (%)    .79 
Low-risk 156 (49.4) 93 (50.0) 63 (48.5)  
High-risk 160 (50.6) 93 (50.0) 67 (51.5)  
Total (n = 316)Period 1 (n = 186)Period 2 (n = 130)P
Age – median (IQR) 56.0 (43.5-64.0) 56.0 (42.0-64.0) 58.5 (47.0-65.0) .23 
< 50 y - n (%) 106 (33.5) 65 (35.0) 41 (31.5)  
50-60 y - n (%) 93 (29.4) 57 (30.7) 36 (27.7)  
> 60 y - n (%) 117 (37.0) 64 (34.4) 53 (40.8)  
Sex - n (%)    .50 
Male 166 (52.5) 103 (55.4) 63 (48.5)  
Female 150 (47.5) 83 (44.6) 67 (51.5)  
Disease - n (%)    .85 
Acute myeloid leukemia 132 (41.8) 74 (39.8) 58 (44.6)  
Myelodysplastic syndrome 58 (18.4) 34 (18.3) 24 (18.5)  
Acute lymphoblastic leukemia 48 (15.2) 27 (14.5) 21 (16.2)  
Lymphoma 24 (7.6) 16 (8.6) 8 (6.2)  
Primary/secondary myelofibrosis 22 (7.0) 14 (7.5) 8 (6.2)  
Others 32 (10.1) 21 (11.3) 11 (8.5)  
Second transplant - n (%) 21 (6.6) 16 (8.6) 5 (3.8) .095 
Donor type - n (%)    .044 
Matched sibling 66 (20.9) 42 (22.6) 24 (18.5)  
Matched unrelated 155 (49.1) 84 (45.2) 71 (54.6)  
Mismatched unrelated 42 (13.3) 21 (11.3) 21 (16.2)  
Haploidentical 53 (16.8) 39 (21.0) 14 (10.8)  
Donor CMV serostatus - n (%)    .43 
Positive 224 (70.9) 135 (72.6) 89 (68.5)  
Negative 92 (29.1) 51 (27.4) 41 (31.5)  
Stem cell source - n (%)    .066  
Bone marrow 141 (44.6) 92 (49.5) 49 (37.7)  
Peripheral blood stem cell 171 (54.1) 94 (50.5) 77 (59.2)  
Cord blood 4 (1.3) 0 (0) 4 (3.1)  
Conditioning regimen - n (%)    .56 
Myeloablative 147 (46.5) 84 (45.2) 63 (48.5)  
Reduced intensity 169 (53.5) 102 (54.8) 67 (51.5)  
Total body irradiation - n (%) 62 (19.6) 34 (18.3) 28 (21.5) .47 
GVHD prophylaxis - n (%)    .14 
Cyclosporin A + methotrexate 229 (72.5) 129 (69.4) 100 (76.9)  
Cyclosporin A + MMF 87 (27.5) 57 (30.6) 30 (23.1)  
In vivo T-cell depletion - n (%) 219 (69.3) 127 (68.3) 92 (70.8) .64 
Antithymocyte globulin 155 (49.1) 82 (44.1) 73 (56.2)  
Post-transplant cyclophosphamide 64 (20.3) 45 (24.2) 19 (14.6)  
CMV risk score - n (%)    .79 
Low-risk 156 (49.4) 93 (50.0) 63 (48.5)  
High-risk 160 (50.6) 93 (50.0) 67 (51.5)  

CMV, cytomegalovirus; GVHD, graft versus host disease; IQR, interquartile range; MMF, mycophenolate mofetil.

Excluding cord blood.

CsCMVi and CMV disease

Cumulative incidences of csCMVi and CMV disease based on the risk of CMV infection and period are shown in Figure 2 and Table 2. The median delay between allo-HCT and the first csCMVi was 50 days (range: 12-526). In patients at high risk with systematic use of letermovir in period 2, csCMVi incidence was lower than in period 1 (P < .001) by week 14 after transplantation: 10.5% (95% CI 4.6-19.2) vs 51.6% (41.0-61.3), and beyond week 14 with lower incidences persisting by week 24: 16.9% (8.9-27.0) vs 52.7% (42.0-62.3), and by 1 year after transplantation: 28.2% (17.3-40.2) vs 53.8% (43.0-63.3), respectively. CMV disease was not significantly different (P = .69) between the 2 periods: 0% vs 3.2% (95% CI 0.9-8.4) by week 14, 1.5% (0.1-7.2) vs 4.3% (1.4-9.9) by week 24, and 3.3% (0.6-10.3) vs 4.3% (1.4-9.9) by 1 year after transplantation.

Figure 2.

Cumulative incidence of csCMVi according to CMV infection risk and period.

Figure 2.

Cumulative incidence of csCMVi according to CMV infection risk and period.

Close modal
Table 2.

Cumulative incidences (95% CI) of csCMVi and secondary outcomes (CMV disease, OS, PFS, CIR, NRM) based on the risk of CMV infection and period

Low-risk (n = 156)High-risk (n = 160)
Period 1n = 93Period 2n = 63PPeriod 1n = 93Period 2n = 67P
Letermovir (until wk 14) 0 (0%) 18/63 (28.6%)  0 (0%) 67 (100%)  
csCMVi   .003   < .001 
14-wk csCMVi 29.0% (20.2-38.5) 7.9% (2.9-16.3)  51.6% (41.0-61.3) 10.5% (4.6-19.2)  
24-wk csCMVi 33.3% (23.9-43.0) 11.2% (4.9-20.5)  52.7% (42.0-62.3) 16.9% (8.9-27.0)  
1-y csCMVi 34.4% (24.9-44.1) 12.9% (6.0-22.6)  53.8% (43.0-63.3) 28.2% (17.3-40.2)  
CMV disease   .85   .69 
14-wk CMV disease 0% 0%  3.2% (0.9-8.4) 0%  
24-wk CMV disease 1.1% (0.1-5.3) 1.7% (0.1-8.0)  4.3% (1.4-9.9) 1.5% (0.1-7.2)  
1-y CMV disease 2.2% (0.4-6.9) 1.7% (0.1-8.0)  4.3% (1.4-9.9) 3.3% (0.6-10.3)  
OS   .56   .08 
14-wk OS 92.5% (84.9-96.3) 90.5% (80.0-95.6)  87.1% (78.4-92.5) 89.6% (79.3-94.9)  
24-wk OS 88.2% (79.7-93.3) 85.6% (74.1-92.2)  80.6% (71.1-87.3) 89.6% (79.3-94.9)  
1-y OS 78.5% (68.7-85.6) 74.9% (61.8-84.1)  70.9% (60.5-79.0) 83.4% (71.0-90.8)  
PFS   .52   .06 
14-wk PFS 89.2% (80.9-94.1) 84.1% (72.5-91.1)  82.8% (73.5-89.1) 88.1% (77.5-93.8)  
24-wk PFS 80.6% (71.1-87.3) 76.1% (63.6-84.9)  76.3% (66.3-83.7) 86.6% (75.8-92.8)  
1-y PFS 69.9% (59.5-78.1) 65.3% (51.8-75.9)  62.3% (51.6-71.2) 77.6% (65.0-86.2)  
CIR   .60   .14 
14-wk CIR 4.3% (1.4-9.9) 6.4% (2.0-14.3)  4.3% (1.4-9.9) 1.5% (0.1-7.1)  
24-wk CIR 11.8% (6.3-19.3) 14.4% (7.0-24.2)  7.5% (3.3-14.1) 3.0% (0.6-9.3)  
1-y CIR 18.3% (11.2-26.8) 21.6% (12.1-32.8)  17.3% (10.4-25.7) 7.9% (2.9-16.2)  
NRM   .80   .31 
14-wk NRM 6.5% (2.6-12.7) 9.5% (3.8-18.3)  12.9% (7.0-20.6) 10.5% (4.6-19.2)  
24-wk NRM 7.5% (3.3-14.1) 9.5% (3.8-18.3)  16.1% (9.5-24.3) 10.5% (4.6-19.2)  
1-y NRM 11.8% (6.3-19.3) 13.1% (6.1-23.0)  20.4% (12.9-29.2) 14.5% (7.0-24.6)  
Late csCMVi >14 wk   .52   .20 
24-wk late csCMVi 6.5% (2.6-12.7) 3.4% (0.6-10.4)  9.7% (4.7-16.8) 7.9% (2.9-16.3)  
1-y late csCMVi 9.7% (4.7-16.7) 6.8% (2.1-15.1)  11.8% (6.2-19.3) 21.1% (11.5-32.7)  
Low-risk (n = 156)High-risk (n = 160)
Period 1n = 93Period 2n = 63PPeriod 1n = 93Period 2n = 67P
Letermovir (until wk 14) 0 (0%) 18/63 (28.6%)  0 (0%) 67 (100%)  
csCMVi   .003   < .001 
14-wk csCMVi 29.0% (20.2-38.5) 7.9% (2.9-16.3)  51.6% (41.0-61.3) 10.5% (4.6-19.2)  
24-wk csCMVi 33.3% (23.9-43.0) 11.2% (4.9-20.5)  52.7% (42.0-62.3) 16.9% (8.9-27.0)  
1-y csCMVi 34.4% (24.9-44.1) 12.9% (6.0-22.6)  53.8% (43.0-63.3) 28.2% (17.3-40.2)  
CMV disease   .85   .69 
14-wk CMV disease 0% 0%  3.2% (0.9-8.4) 0%  
24-wk CMV disease 1.1% (0.1-5.3) 1.7% (0.1-8.0)  4.3% (1.4-9.9) 1.5% (0.1-7.2)  
1-y CMV disease 2.2% (0.4-6.9) 1.7% (0.1-8.0)  4.3% (1.4-9.9) 3.3% (0.6-10.3)  
OS   .56   .08 
14-wk OS 92.5% (84.9-96.3) 90.5% (80.0-95.6)  87.1% (78.4-92.5) 89.6% (79.3-94.9)  
24-wk OS 88.2% (79.7-93.3) 85.6% (74.1-92.2)  80.6% (71.1-87.3) 89.6% (79.3-94.9)  
1-y OS 78.5% (68.7-85.6) 74.9% (61.8-84.1)  70.9% (60.5-79.0) 83.4% (71.0-90.8)  
PFS   .52   .06 
14-wk PFS 89.2% (80.9-94.1) 84.1% (72.5-91.1)  82.8% (73.5-89.1) 88.1% (77.5-93.8)  
24-wk PFS 80.6% (71.1-87.3) 76.1% (63.6-84.9)  76.3% (66.3-83.7) 86.6% (75.8-92.8)  
1-y PFS 69.9% (59.5-78.1) 65.3% (51.8-75.9)  62.3% (51.6-71.2) 77.6% (65.0-86.2)  
CIR   .60   .14 
14-wk CIR 4.3% (1.4-9.9) 6.4% (2.0-14.3)  4.3% (1.4-9.9) 1.5% (0.1-7.1)  
24-wk CIR 11.8% (6.3-19.3) 14.4% (7.0-24.2)  7.5% (3.3-14.1) 3.0% (0.6-9.3)  
1-y CIR 18.3% (11.2-26.8) 21.6% (12.1-32.8)  17.3% (10.4-25.7) 7.9% (2.9-16.2)  
NRM   .80   .31 
14-wk NRM 6.5% (2.6-12.7) 9.5% (3.8-18.3)  12.9% (7.0-20.6) 10.5% (4.6-19.2)  
24-wk NRM 7.5% (3.3-14.1) 9.5% (3.8-18.3)  16.1% (9.5-24.3) 10.5% (4.6-19.2)  
1-y NRM 11.8% (6.3-19.3) 13.1% (6.1-23.0)  20.4% (12.9-29.2) 14.5% (7.0-24.6)  
Late csCMVi >14 wk   .52   .20 
24-wk late csCMVi 6.5% (2.6-12.7) 3.4% (0.6-10.4)  9.7% (4.7-16.8) 7.9% (2.9-16.3)  
1-y late csCMVi 9.7% (4.7-16.7) 6.8% (2.1-15.1)  11.8% (6.2-19.3) 21.1% (11.5-32.7)  

In patients at low risk, no patients received letermovir in period 1 and 18 of 63 patients (28.6%) received letermovir in period 2. Despite this sparing use of letermovir during period 2, the incidence of csCMVi was also significantly lower than that in period 1 (P = .003) by week 14: 7.9% (95% CI 2.9-16.3) vs 29.0% (20.2-38.5), by week 24: 11.2% (4.9-20.5) vs 33.3% (23.9-43.0), and by 1 year after transplantation: 12.9% (6.0-22.6) vs 34.4% (24.9-44.1), respectively. CMV disease was not significantly different (P = .85) between the 2 periods: 0% vs 0% by week 14, 1.7% (95% CI 0.1-8.0) vs 1.1% (0.1-5.3) by week 24, and 1.7% (0.1-8.0) vs 2.2% (0.4-6.9) by 1 year posttransplant.

Figure 3 shows the efficacy of letermovir used in period 2 compared to period 1 for preventing csCMVi by week 14, according to subgroups defined by baseline characteristics. All subgroups benefited from letermovir use (nonsignificant heterogeneity) with hazard ratio (HR) consistently < 0.50. The effect of letermovir was more pronounced in patients at high risk: HR = 0.15 (95% CI 0.07-0.32), P < .001 than in patients at low risk: HR = 0.25 (0.10-0.65), P = .005. The most important and significant differences were observed in females: HR = 0.07 (95% CI 0.02-0.28), P < .001 and in patients with CMV-negative donor: HR = 0.07 (0.02-0.29), P < .001.

Figure 3.

Letermovir efficacy in preventing csCMVi by week 14 according to subgroups in period 2 compared to period 1.

Figure 3.

Letermovir efficacy in preventing csCMVi by week 14 according to subgroups in period 2 compared to period 1.

Close modal

Sparing use of letermovir in patients at low risk during period 2

As specified, letermovir was not systematically used in patients at low risk during period 2 (n = 63). On one hand, 18 patients (28.6%) eventually received letermovir because of the use of corticosteroids before week 14, with a median delay of 28 days (range: 16-50) from transplantation before starting letermovir. The reason for initiation of corticosteroids was primarily acute GVHD (aGVHD), except for 2 patients (engraftment syndrome and immune-mediated hemolytic anemia). None of the 18 patients experienced csCMVi. On the other hand, among patients at low risk who did not receive letermovir (n = 45), 5 patients (11.1%) experienced csCMVi by week 14 and were successfully treated with a single course of preemptive therapy without CMV disease. In addition, 23 patients (51.1%) experienced transient positive CMV DNA without csCMVi while 17 (37.8%) never had detectable CMV viral load by week 14. The median time of first detectable CMV DNA was 42 days (range: 21-77) and the median quantifiable CMV viral load was 2.72 IU/mL (range 2.21-3.49). No CMV DNA was found ≥3.5 log10 IU/mL. Figure 4 shows the CMV viral loads of patients at low risk without csCMVi and who did not receive letermovir.

Figure 4.

CMV viral loads for patients at low-risk without csCMVi and who did not receive letermovir in period 2. Two patients are depicted and the number of patients without detectable CMV viral load is indicated at each week.

Figure 4.

CMV viral loads for patients at low-risk without csCMVi and who did not receive letermovir in period 2. Two patients are depicted and the number of patients without detectable CMV viral load is indicated at each week.

Close modal

Secondary endpoints

OS, PFS, CIR, and NRM based on the risk of CMV infection and period are shown in Figure 5 and Table 2. In patients at high risk, higher OS in period 2 than in period 1 was found but did not reach a statistically significant level (1-year OS after transplantation: 83.4% [95% CI 71.0-90.8] in period 2 vs 70.9% [60.5-79.0] in period 1, P = .08). A similar result was found for PFS comparing period 2 to period 1 (1-year after transplantation: 77.6% [95% CI 65.0-86.2] vs 62.3% [51.6-71.2] respectively, P = .06). The CIR did not significantly differ across the 2 periods (P = .14) nor did NRM (P = .31). In patients at low risk, no significative differences were observed between the 2 periods for OS (P = .56), PFS (P = .52), CIR (P = .60), and NRM (P = .80).

Figure 5.

Kaplan-Meier analysis according to CMV infection risk and period. (A) OS, (B) PFS, (C) CIR, and (D) NRM.

Figure 5.

Kaplan-Meier analysis according to CMV infection risk and period. (A) OS, (B) PFS, (C) CIR, and (D) NRM.

Close modal

Exploratory end point: late csCMVi (>week 14)

A landmark analysis was performed from week 14 to explore the incidence of CMV infection after the discontinuation of letermovir, whether or not the patient had a prior CMV infection before week 14. In patients at high risk, there was no significant difference in the incidence of late csCMVi between period 2 and period 1 (1 year after transplantation: 21.1% [95% CI 11.5-32.7] in period 2 vs 11.8% [6.2-19.3] in period 1, P = .20). Similarly, among patients at low risk, the incidences of late csCMVi between the 2 periods were not significantly different between period 2 and period 1 (1 year after transplantation: 6.8% [95% CI 2.1-15.1] vs 9.7% [4.7-16.7], P = .52). The cumulative incidences of late csCMVi based on the risk of CMV infection and period are shown in Figure 6. Overall, 36 patients (13 low-risk and 23 high-risk) experienced late csCMVi in both periods. Of the 36 patients, 17 patients (47.2%) had experienced prior csCMVi before week 14, and 23 patients (63.9%) had received corticosteroids after week 14. Reasons of corticosteroids were late-onset aGVHD (n = 10), corticosteroids-refractory aGVHD (n = 5), GVHD after donor lymphocyte infusion (n = 4), overlap syndrome (n = 2), cryptogenic organizing pneumonia (n = 1), and late capillary leak syndrome (n = 1). Only 6 patients (16.7%) had neither prior csCMVi before week 14 nor late corticosteroid use.

Figure 6.

Cumulative incidences of late csCMVi (>week 14) according to CMV infection risk and period.

Figure 6.

Cumulative incidences of late csCMVi (>week 14) according to CMV infection risk and period.

Close modal

The randomized pivotal trial comparing letermovir with placebo was consistent for the prevention of csCMVi in patients at high and low risk of CMV infection; consequently, the approval was for all patients who were CMV positive and was not based on individual risk of CMV infection.5 It should be noted that in the prophylaxis of fungal infections, voriconazole or posaconazole are labeled in only select patients at high-risk based on randomized phase 3 studies, which however did not consider the individual risk of fungal infection.21,22 Moreover, many studies have shown that the risk of CMV infection depends on transplantation modalities and the characteristics of the donor–recipient pair.12-16 It is therefore reasonable to assume that letermovir would not have the same benefit in all patients. Most of these studies explored the risk factors of CMV infection independently to distinguish between high-risk and low-risk patients. Because risk factors do not have the same strong association with CMV infection, we had previously developed a CMV risk score considering 6 weighted variables at the time of transplantation.17 Using this more sophisticated tool to measure the risk of csCMVi and having a dynamic strategy in patients at low risk receiving corticosteroids, we have shown that sparse use of letermovir in patients at low risk could be achieved resulting in low csCMVi incidence without impairing clinical outcomes as CMV disease, OS, or NRM. Indeed, with 28.6% of the patients at low risk eventually receiving letermovir in period 2, the low incidence of csCMVi by week 14 (7.9%) was similar to that of the letermovir phase 3 study (7.7%) or real-life studies in which letermovir was used in all patients.23-26 The decrease in csCMVi in period 2 compared with that in period 1 was thus explained by the use of letermovir in patients receiving corticosteroids, almost all for aGVHD, which is known to be a major factor in CMV infection.27-30 At the same time, we found an important beneficial of letermovir in patients at high risk in csCMVi prevention with an improved and nearly statistically significant OS through 1 year after transplantation. It seems thus reasonable to assume that a larger number of patients would have allowed for reaching statistical significance. We assume that by focusing on patients at high risk in larger, ideally randomized studies, an additional OS benefit of letermovir could be demonstrated.

Compared with other antiviral drugs tested unsuccessfully in the prophylaxis of CMV infection after allo-HCT, letermovir acts through a unique mechanism of action by inhibiting the viral terminase complex through its binding to pUL56.31-33 Moreover, letermovir is highly specific for CMV, without activity against other herpesviruses or adenovirus.34 This specificity is probably one of the main reasons for its successful use in allo-HCT, in which the side effects often restrict the use of antivirals.35,36 But letermovir, as a P-glycoprotein and an organic anion transporting polypeptide 1B1/3 transporter substrate, a moderate inhibitor of CYP3A, and an inducer of CYP2C9/19, has multiple drug interactions, which might prove a significant point in patients who are polymedicated. The most relevant interactions in the allo-HCT setting are the immunosuppressive drugs (cyclosporine, tacrolimus, and sirolimus) and voriconazole.37,38 Although letermovir dosage reduction (480-240 mg/d) is mandatory if used with cyclosporine and therapeutic monitoring should be carried out for key drugs, the tolerance of letermovir and other coadministered drugs could be challenged, especially in a real-world setting in which patients present potentially more comorbidities. In this context, the benefit-risk balance may be adversely affected in patients at low risk of CMV infection, highlighting the importance of assessing whether letermovir is really needed in this category of patient.

By comparison to HIV-positive patients, the term blips has been used to describe transient episodes of detectable CMV DNA in the blood.10 There has been a long-standing debate about the point at which preventive treatment should be initiated.39 In our study, we systematically used 3.5 log10 IU/mL as the threshold, based on previous studies and recommendations.3,18-20 We showed that a number of patients at low risk not receiving letermovir in period 2 had transient positive CMV viral load but the OS was not affected. In the retrospective study by Green et al7, CMV viral load >250 IU/mL was associated with increased early death (<day 60), but the impact was considerably attenuated after day 60, suggesting that patients in this study with early positive CMV viral load could have been very distinct. Moreover, CMV replication without csCMVi is not necessarily a negative event, and decreased exposure to CMV antigen in patients receiving letermovir has been shown to delay CMV-specific T-cells reconstitution.9,40,41 This delay in the reconstitution of CMV-specific immunity has already been suspected in letermovir phase 3 study with the observation of an increase in csCMVi after the discontinuation of letermovir. One option being considered to address this issue is to extend the letermovir exposure period to 200 days after transplantation (NCT03930615) in patients at high risk for CMV infection. Probably, letermovir at this time may be useful in patients who are still heavily immunocompromised, especially those suffering from GVHD.42 Methods have been developed to quantify specific immune recovery, the main ones being CMV quantiferon and enzyme-linked immunospot, but these approaches are difficult to implement routinely in all centers and standardization with uniform thresholds are still lacking.43-45 

To investigate late CMV infection, we performed a landmark analysis from week 14, which allows the analysis of csCMVi occurring after week 14, even if the patient was previously infected. We reported late csCMVi in both risk groups, independent of letermovir use by week 14. Thus, delayed CMV-specific immunity because of letermovir may not be the exclusive cause of late csCMVi. We considered other risk factors in 36 patients with late csCMVi from both risk groups and both time periods. Interestingly, we observed that nearly all these patients had received corticosteroids after week 14 or had experienced a previous csCMVi before week 14. A role for prolonged letermovir exposure after week 14 or even resumption in case of late GVHD, for instance after a donor lymphocyte infusion, could be identified in this specific case of patients with persistent severe immunodeficiency or as a secondary prophylaxis.46 Admittedly, we have only investigated this possibility in an exploratory manner and this assumption would require a specific study. But, overall, a dynamic strategy that adapts to each patient's changing risk could make letermovir more cost-effective. 47,48 

Some limitations should be noted. The main one is that patients were not randomized between the routine strategy with letermovir and the risk-based strategy, but using time periods (period 1 and period 2) was an acceptable alternative method because transplantation modalities have not radically changed over time. A single-center study was another limitation: the difficulty of doing a multicenter study being variable in the management of CMV infection between centers. Finally, haploidentical transplants were not included in our CMV risk score and consistently received letermovir in P2, but this strategy seems appropriate given that it has been described as associated with a high risk of CMV infection.49 

In conclusion, we showed that a risk-based strategy for the use of letermovir in recipients of allo-HCT who are CMV-positive proves an effective strategy that maintains the strong efficacy of letermovir in patients at high risk but allows some patients at low risk not to use letermovir. A randomized study between routine letermovir use and a risk-based letermovir use strategy would provide a high level of evidence for these findings.

The authors thank Matthieu Penet for his help in identifying outpatients receiving antiviral treatment. They also thank Rachel Tipton for the English proofreading of this manuscript.

This study was not endorsed by a research grant.

Contribution: M. Sourisseau, I.Y.-A. and D.B. designed the study; P.C., M. Srour, A.C., V.C., and L.M. recruited the patients and applied the risk-based letermovir use strategy; E.F., S.A., F.V., and K.F. managed patients for CMV infection; E.K.A. reported viral quantitative data; N.S. reported pharmacological data; M.K. collected clinical data; H.B. performed statistical analysis; M.S. Sourisseau and D.B. wrote the initial draft of the manuscript; and all authors read and approved the final manuscript.

Conflict-of-interest disclosure: I.Y.-A. and D.B. received an honorarium from Merck & Co, unrelated to this article. E.K.A. received support from Merck & Co to attend HIV conferences. The remaining authors declare no competing financial interests.

Correspondence: David Beauvais, UAM allogreffe de cellules souches hématopoïétiques, CHU Lille, 2 avenue Oscar Lambret, F-59037 Lille Cedex, France; e-mail: david.beauvais@chru-lille.fr.

1.
Ljungman
P
,
Brandan
R
.
Factors influencing cytomegalovirus seropositivity in stem cell transplant patients and donors
.
Haematologica
.
2007
. ;
92
(
8
):
1139
-
1142
.
2.
Ljungman
P
,
Boeckh
M
,
Hirsch
HH
, et al
.
Definitions of cytomegalovirus infection and disease in transplant patients for use in clinical trials
.
Clin Infect Dis
.
2017
. ;
64
(
1
):
87
-
91
.
3.
Emery
V
,
Zuckerman
M
,
Jackson
G
, et al
.
Management of cytomegalovirus infection in haemopoietic stem cell transplantation
.
Br J Haematol
.
2013
. ;
162
(
1
):
25
-
39
.
4.
Ljungman
P
,
de la Camara
R
,
Robin
C
, et al
.
Guidelines for the management of cytomegalovirus infection in patients with haematological malignancies and after stem cell transplantation from the 2017 European Conference on Infections in Leukaemia (ECIL 7)
.
Lancet Infect Dis
.
2019
. ;
19
(
8
):
e260
-
e272
.
5.
Marty
FM
,
Ljungman
P
,
Chemaly
RF
, et al
.
Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation
.
N Engl J Med
.
2017
. ;
377
(
25
):
2433
-
2444
.
6.
Chen
K
,
Cheng
MP
,
Hammond
SP
,
Einsele
H
,
Marty
FM
.
Antiviral prophylaxis for cytomegalovirus infection in allogeneic hematopoietic cell transplantation
.
Blood Adv
.
2018
. ;
2
(
16
):
2159
-
2175
.
7.
Green
ML
,
Leisenring
W
,
Xie
H
, et al
.
Cytomegalovirus viral load and mortality after haemopoietic stem cell transplantation in the era of pre-emptive therapy: a retrospective cohort study
.
Lancet Haematol
.
2016
. ;
3
(
3
):
e119
-
127
.
8.
Teira
P
,
Battiwalla
M
,
Ramanathan
M
, et al
.
Early cytomegalovirus reactivation remains associated with increased transplant-related mortality in the current era: a CIBMTR analysis
.
Blood
.
2016
. ;
127
(
20
):
2427
-
2438
.
9.
Schleiss
MR
.
Letermovir and HCT: too much of a good thing?
.
Blood
.
2021
. ;
138
(
1
):
1
-
2
.
10.
Hill
JA
,
Mayer
BT
,
Xie
H
, et al
.
Kinetics of double-stranded DNA viremia after allogeneic hematopoietic cell transplantation
.
Clin Infect Dis
.
2018
. ;
66
(
3
):
368
-
375
.
11.
Kotton
CN
,
Kumar
D
,
Caliendo
AM
, et al
.
The Third International Consensus guidelines on the management of cytomegalovirus in solid-organ transplantation
.
Transplantation
.
2018
. ;
102
(
6
):
900
-
931
.
12.
Ljungman
P
,
Brand
R
,
Hoek
J
, et al
.
Donor cytomegalovirus status influences the outcome of allogeneic stem cell transplant: a study by the European group for blood and marrow transplantation
.
Clin Infect Dis
.
2014
. ;
59
(
4
):
473
-
481
.
13.
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
. ;
15
(
6
):
694
-
703
.
14.
Meijer
E
,
Dekker
AW
,
Verdonck
LF
.
Influence of antithymocyte globulin dose on outcome in cytomegalovirus-seropositive recipients of partially T cell-depleted stem cell grafts from matched-unrelated donors
.
Br J Haematol
.
2003
. ;
121
(
3
):
473
-
476
.
15.
Kalpoe
JS
,
van der Heiden
PLJ
,
Vaessen
N
,
Claas
ECJ
,
Barge
RM
,
Kroes
ACM
.
Comparable incidence and severity of cytomegalovirus infections following T cell-depleted allogeneic stem cell transplantation preceded by reduced intensity or myeloablative conditioning
.
Bone Marrow Transplant
.
2007
. ;
40
(
2
):
137
-
143
.
16.
Junghanss
C
,
Storb
R
,
Maris
MB
, et al
.
Impact of unrelated donor status on the incidence and outcome of cytomegalovirus infections after non-myeloablative allogeneic stem cell transplantation
.
Br J Haematol
.
2003
. ;
123
(
4
):
662
-
670
.
17.
Beauvais
D
,
Drumez
E
,
Blaise
D
, et al
.
Scoring system for clinically significant CMV infection in seropositive recipients following allogenic hematopoietic cell transplant: an SFGM-TC study
.
Bone Marrow Transplant
.
2021
. ;
56
(
6
):
1305
-
1315
.
18.
Gerna
G
,
Lilleri
D
,
Caldera
D
,
Furione
M
,
Zenone Bragotti
L
,
Alessandrino
EP
.
Validation of a DNAemia cutoff for preemptive therapy of cytomegalovirus infection in adult hematopoietic stem cell transplant recipients
.
Bone Marrow Transplant
.
2008
. ;
41
(
10
):
873
-
879
.
19.
Solano
C
,
Muñoz-Cobo
B
,
Giménez
E
, et al
.
Pre-emptive antiviral therapy for active CMV infection in adult allo-SCT patients guided by plasma CMV DNAemia quantitation using a real-time PCR assay: clinical experience at a single center
.
Bone Marrow Transplant
.
2013
. ;
48
(
7
):
1010
-
1012
.
20.
Brissot
E
,
Alsuliman
T
,
Beauvais
D
, et al
.
[Antiviral prophylaxis for CMV, HSV/VZV and HBV in allogeneic hematopoietic cell transplantation in adult patients: guidelines from the Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC)]
.
Bull Cancer
.
2019
. ;
107
(
1S
):
S1
-
S6
.
21.
Marks
DI
,
Pagliuca
A
,
Kibbler
CC
, et al
.
Voriconazole versus itraconazole for antifungal prophylaxis following allogeneic haematopoietic stem-cell transplantation
.
Br J Haematol
.
2011
. ;
155
(
3
):
318
-
327
.
22.
Ullmann
AJ
,
Lipton
JH
,
Vesole
DH
, et al
.
Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease
.
N Engl J Med
.
2007
. ;
356
(
4
):
335
-
347
.
23.
Anderson
A
,
Raja
M
,
Vazquez
N
,
Morris
M
,
Komanduri
K
,
Camargo
J
.
Clinical ‘real-world’ experience with letermovir for prevention of cytomegalovirus infection in allogeneic hematopoietic cell transplant recipients
.
Clin Transplant
.
2020
. ;
34
(
7
):
e13866
.
24.
Lin
A
,
Maloy
M
,
Su
Y
, et al
.
Letermovir for primary and secondary cytomegalovirus prevention in allogeneic hematopoietic cell transplant recipients: real-world experience
.
Transpl Infect Dis
.
2019
. ;
21
(
6
):
e13187
.
25.
Martino
M
,
Pitino
A
,
Gori
M
, et al
.
Letermovir prophylaxis for cytomegalovirus infection in allogeneic stem cell transplantation: a real-world experience
.
Front Oncol
.
2021
. ;
11
:
740079
.
26.
Beauvais
D
,
Robin
C
,
Thiebaut
A
, et al
.
Effective letermovir prophylaxis of CMV infection post allogeneic hematopoietic cell transplantation: results from the French temporary authorization of use compassionate program
.
J Clin Virol
.
2022
. ;
148
:
105106
.
27.
Ljungman
P
,
Perez-Bercoff
L
,
Jonsson
J
, et al
.
Risk factors for the development of cytomegalovirus disease after allogeneic stem cell transplantation
.
Haematologica
.
2006
. ;
91
(
1
):
78
-
83
.
28.
Cantoni
N
,
Hirsch
HH
,
Khanna
N
, et al
.
Evidence for a bidirectional relationship between cytomegalovirus replication and acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2010
. ;
16
(
9
):
1309
-
1314
.
29.
Cohen
L
,
Yeshurun
M
,
Shpilberg
O
,
Ram
R
.
Risk factors and prognostic scale for cytomegalovirus (CMV) infection in CMV-seropositive patients after allogeneic hematopoietic cell transplantation
.
Transpl Infect Dis
.
2015
. ;
17
(
4
):
510
-
517
.
30.
Wolfe
D
,
Zhao
Q
,
Siegel
E
, et al
.
Letermovir prophylaxis and cytomegalovirus reactivation in adult hematopoietic cell transplant recipients with and without acute graft versus host disease
.
Cancers
.
2021
. ;
13
(
21
):
5572
.
31.
Marty
FM
,
Ljungman
P
,
Papanicolaou
GA
, et al
.
Maribavir prophylaxis for prevention of cytomegalovirus disease in recipients of allogeneic stem-cell transplants: a phase 3, double-blind, placebo-controlled, randomised trial
.
Lancet Infect Dis
.
2011
. ;
11
(
4
):
284
-
292
.
32.
Marty
FM
,
Winston
DJ
,
Chemaly
RF
, et al
.
A randomized, double-blind, placebo-controlled phase 3 trial of oral brincidofovir for cytomegalovirus prophylaxis in allogeneic hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2019
. ;
25
(
2
):
369
-
381
.
33.
Goldner
T
,
Hewlett
G
,
Ettischer
N
,
Ruebsamen-Schaeff
H
,
Zimmermann
H
,
Lischka
P
.
The novel anticytomegalovirus compound AIC246 (letermovir) inhibits human cytomegalovirus replication through a specific antiviral mechanism that involves the viral terminase
.
J Virol
.
2011
. ;
85
(
20
):
10884
-
10893
.
34.
Marschall
M
,
Stamminger
T
,
Urban
A
, et al
.
In vitro evaluation of the activities of the novel anticytomegalovirus compound AIC246 (letermovir) against herpesviruses and other human pathogenic viruses
.
Antimicrob Agents Chemother
.
2012
. ;
56
(
2
):
1135
-
1137
.
35.
Venton
G
,
Crocchiolo
R
,
Fürst
S
, et al
.
Risk factors of ganciclovir-related neutropenia after allogeneic stem cell transplantation: a retrospective monocentre study on 547 patients
.
Clin Microbiol Infect
.
2014
. ;
20
(
2
):
160
-
166
.
36.
Foster
GG
,
Grant
MJ
,
Thomas
SM
, et al
.
Treatment with foscarnet after allogeneic hematopoietic cell transplant (allo-HCT) is associated with long-term loss of renal function
.
Biol Blood Marrow Transplant
.
2020
. ;
26
(
9
):
1597
-
1606
.
37.
McCrea
JB
,
Macha
S
,
Adedoyin
A
, et al
.
Pharmacokinetic drug-drug interactions between letermovir and the immunosuppressants cyclosporine, tacrolimus, sirolimus, and mycophenolate mofetil
.
J Clin Pharmacol
.
2019
. ;
59
(
10
):
1331
-
1339
.
38.
Nakashima
T
,
Inamoto
Y
,
Fukushi
Y
, et al
.
Drug interaction between letermovir and voriconazole after allogeneic hematopoietic cell transplantation
.
Int J Hematol
.
2021
. ;
113
(
6
):
872
-
876
.
39.
Huntley
D
,
Talaya
A
,
Giménez
E
, et al
.
Features of cytomegalovirus DNAemia blips in allogeneic hematopoietic stem cell transplant recipients: implications for optimization of preemptive antiviral therapy strategies
.
Biol Blood Marrow Transplant
.
2020
. ;
26
(
5
):
972
-
977
.
40.
Zamora
D
,
Duke
ER
,
Xie
H
, et al
.
Cytomegalovirus-specific T-cell reconstitution following letermovir prophylaxis after hematopoietic cell transplantation
.
Blood
.
2021
. ;
138
(
1
):
34
-
43
.
41.
Lugthart
G
,
van Ostaijen-Ten Dam
MM
,
Jol-van der Zijde
CM
, et al
.
Early cytomegalovirus reactivation leaves a specific and dynamic imprint on the reconstituting T cell compartment long-term after hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2014
. ;
20
(
5
):
655
-
661
.
42.
Liu
LW
,
Yn
A
,
Gao
F
, et al
.
Letermovir discontinuation at D+100 after allogeneic stem cell transplant is associated with increased CMV-related mortality
.
Transplant Cell Ther
.
2022
. ;
S2666-6367
(
22
):
01314
-
01318
.
43.
Paouri
B
,
Soldatou
A
,
Petrakou
E
, et al
.
Quantiferon-cytomegalovirus assay: a potentially useful tool in the evaluation of CMV-specific CD8+ T-cell reconstitution in pediatric hematopoietic stem cell transplant patients
.
Pediatr Transplant
.
2018
. ;
22
(
5
):
e13220
.
44.
Seo
E
,
Choi
ES
,
Kim
JH
, et al
.
Immunologic monitoring of cytomegalovirus (CMV) enzyme-linked immune absorbent spot (ELISPOT) for controlling clinically significant CMV infection in pediatric allogeneic hematopoietic stem cell transplant recipients
.
PLoS One
.
2021
. ;
16
(
2
):
e0246191
.
45.
Ljungman
P
.
Would monitoring CMV immune responses allow improved control of CMV in stem cell transplant patients
.
J Clin Virol
.
2006
. ;
35
(
4
):
493
-
495
.
46.
Robin
C
,
Thiebaut
A
,
Alain
S
, et al
.
Letermovir for secondary prophylaxis of cytomegalovirus infection and disease after allogeneic hematopoietic cell transplantation: results from the French compassionate program
.
Biol Blood Marrow Transplant
.
2020
. ;
26
(
5
):
978
-
984
.
47.
Chan
TS-Y
,
Cheng
SS-Y
,
Chen
W-T
, et al
.
Cost-effectiveness of letermovir as cytomegalovirus prophylaxis in adult recipients of allogeneic hematopoietic stem cell transplantation in Hong Kong
.
J Med Econ
.
2020
. ;
23
(
12
):
1485
-
1492
.
48.
Peffault De Latour
R
,
Chevallier
P
,
Blaise
D
, et al
.
Clinical and economic impact of treated CMV infection in adult CMV-seropositive patients after allogeneic hematopoietic cell transplantation
.
J Med Virol
.
2020
. ;
92
(
12
):
3665
-
3673
.
49.
Goldsmith
SR
,
Abid
MB
,
Auletta
JJ
, et al
.
Posttransplant cyclophosphamide is associated with increased cytomegalovirus infection: a CIBMTR analysis
.
Blood
.
2021
. ;
137
(
23
):
3291
-
3305
.

Author notes

Data are available on request from the corresponding author, David Beauvais (david.beauvais@chru-lille.fr).