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
. .

Originally observed as a “peculiar interstitial pneumonia” seen in recipients of allogeneic hematopoietic stem cell transplantation (HSCT) and a major cause of death post-transplantation, human cytomegalovirus (CMV) infections have hindered the success of HSCT since the inception of the technique in the 1970s.1  Despite advances in antiviral therapy, improved understanding of host immunity to CMV and the development of superior CMV diagnostic methods during the past 50 years, CMV infection continues to be one of the most common and life-threatening infectious complications post-HSCT and is strongly associated with overall higher nonrelapse mortality and lower overall survival (OS).2,3  As the use of human leukocyte antigen (HLA) -haploidentical hematopoietic transplantation with post-transplant cyclophosphamide (HaploCy) as the backbone of graft-versus-host disease (GVHD) prophylaxis has expanded worldwide and now accounts for 16 percent of all HSCT,4-6  several reports from individual transplant centers have suggested that HaploCy is associated with increased incidence of CMV infection as well as an earlier onset of CMV in the post-transplant course relative to patients receiving HLA-matched HSCT.7-10  No large data set to date has examined the association between HaploCy and CMV infection. Moreover, it is unclear as to whether the donor source (HLA-haplo) or the PTCy-based GVHD prophylaxis is an increased risk factor for CMV infection.

In the current report, Dr. Scott Goldsmith and colleagues and the Center for International Blood and Marrow Transplant Research (CIBMTR) have tried to determine whether haploidentical donor source and/or PTCy-based prophylaxis increase the risk of CMV infection after HSCT by comparing outcomes to a control cohort of HLA-matched sibling donor transplant recipients treated with calcineurin-based GVHD prophylaxis. Data were reported to the CIBMTR between 2012 and 2017 for patients with acute myeloid leukemia, acute lymphoblastic leukemia, and myelodysplastic syndrome (MDS) who received HaploCy (n = 757), HLA-matched related sibling (Sib) with PTCy (SibCy; n = 403), and HLA-matched related siblings with calcineurin inhibitor–based GVHD prophylaxis (SibCNI, n = 1,605). CMV infection was defined as blood stream only (DNAemia) and/or tissue-invasive (end-organ) disease. The term “CMV reactivation” was not used in the study. Three major analyses were performed in this retrospective registry study.

First, the authors examined the incidence of CMV DNAemia and CMV disease at transplant day 180 in the three transplant cohorts (HaploCy, SibCy, and SibCNI). The incidence of CMV DNAemia was significantly higher by day 180 in patients receiving PTCy (HaploCy, 42% [99% CI, 37-46] and SibCy, 37% [99% CI, 31-43]) compared to patients receiving SibCNI (23% [99% CI, 20-26], p<.001). The median time to CMV reactivation was faster in recipients of PTCy (HaploCy, 38 days [range, 2-176 days], SibCy, 32 days [range, 5-136 days]) compared to SibCNI (42 days [range, 4-176 days], p<.001). The incidence of CMV organ disease was low for all patients; the cumulative incidence of CMV by day 100 was 2.8 percent (HaploCy), 3.4 percent (SibCy), and 1.4 percent (SibCNI). The incidence increased modestly by day 180, but remained nonsignificant between cohorts (p=.115).

In the second analysis — the “CMV serostatus analysis” — the three transplant cohorts (above) were divided further into three subpopulations based on donor/recipient (D/R) CMV serostatus and classified as follows: CMV-seropositive recipient (R+), CMV seropositive donor (D+/R), and CMV seronegative recipient and donor (D/R). The principal outcome of interest was CMV infection (DNAemia and/or organ disease). As expected, the incidence of CMV was highest for CMV seropositive recipients (R+); the day 100 cumulative incidence of CMV DNAemia among R+ patients was 51 percent (99% CI, 46-57) for HaploCy, 48 percent (99% CI, 41-56) for SibCy, and 29 percent for SibCNI. Donor CMV seropositivity (D+/R) was also associated with increased incidence of CMV DNAemia at day 100 (HaploCy, 21% [99% CI, 8-37]; SibCy, 12% [99% CI, 2-30]; and SibCNI, 11% [99% CI, 6-18]). The day 100 cumulative incidence of CMV DNAemia among D/R patients was low: 2.4 percent (99% CI, 0.2-7.1) for HaploCy, 2.6 percent (99% CI, 0-9.2) for SibCy, and 1.3 percent (99% CI, 0.2-3.4) for SibCNI. In multivariate analysis only CMV serostatus influenced CMV infection. The R+ patients (R+ HaploCy, R+ SibCy, and R+ SibCNI) had significantly higher risk of CMV infection as compared to the reference cohort of seronegative Sib HSCT with conventional GVHD prophylaxis (D/R SibCNI) with hazard ratios (HRs) of 50 (99% CI, 14-181), 48 (99% CI, 13-177), and 24 (99% CI, 7-82) among R+ HaploCy, R+ SibCy, and R+ SibCNI, respectively (p<.0001 for all comparisons). Significantly, among R+ subgroups, the use of PTCy for GVHD prophylaxis doubled the risk for CMV infections, and donor source (Haplo or matched sibling donor) did not impact the risk (R+ HaploCy vs. R+ SibCNI, HR 2.1 [99% CI, 1.4-3.1]; p<.0001; R+ SibCy vs. R+ SibCNI, HR 2.0 [99% CI, 1.3-3.1]; p=.0001).

The third analysis examined the effect of CMV DNAemia and CMV serostatus on transplant related outcomes that included OS, disease-free survival, cumulative incidences of relapse, non-relapse mortality (NRM), and GVHD by two years. Univariate analysis revealed that donor or recipient positive CMV serostatus was associated with inferior NRM at 100 days, one year, and two years. In multivariate analysis, CMV seropositive recipients (R+) in the HaploCy cohort had significantly higher risk of NRM and inferior OS compared to R+ SibCNI (HR, 1.4 [99% CI, 1.1-1.7], p=.0001). Additionally, the R+ HaploCy cohort had significantly inferior OS compared to D/R HaploCy cohort (HR, 1.6 [99% CI, 1.1-2.5]; p<.004).

In univariate analysis, there was no difference in the development of grade 2 to 4 acute GVHD among the three transplant cohorts or according to CMV serostatus. The incidence of chronic GVHD at six months was similar across the cohorts and CMV serostatus. The incidence of chronic GVHD at two years was lower in R+ HaploCy (HR, 0.68 [99% CI, 0.47-0.98]; p=.0064) and R+ SibCy (HR, 0.56 [99% CI, 0.39-0.81], p=.0001) compared to D/R SibCNI. However, only the R+ SibCy cohort had a significantly lower risk of chronic GVHD compared to R+ SibCNI. In patients without CMV DNAemia, HaploCy was associated with lower incidence of chronic GVHD (HR, 0.60 [99% CI, 0.42-0.85], p=.0002), and a non-significant trend was seen in patients with SibCy. Among patients with HaploCy, there was a higher incidence of chronic GVHD in those who developed CMV DNAemia as compared to those without CMV DNAemia (HR, 1.6 [99% CI, 1.0-2.3]; p=.006). The incidence of chronic GVHD for HaploCy patients who developed CMV was similar to the SibCNI cohort. Finally, CMV serostatus did not influence the incidence of relapse at two years in any of the transplant cohorts.

In summary, Dr. Goldsmith and colleagues on behalf of the CIBMTR have provided a comprehensive analysis of CMV-associated disease in HSCT allograft recipients in the posttransplant cyclophosphamide era. In this large patient cohort (n=2,765) the use of PTCy was associated with a significantly increased risk of CMV infection, regardless of whether the allograft was from haploidentical or a matched sibling donor. The patients with the highest risk for CMV infection were CMV seropositive recipients (R+), and this high-risk group had inferior NRM and OS. PTCy was associated with lower incidence of chronic GVHD as compared to conventional CNI-based GVHD prophylaxis. However, the development of CMV infection diminished the benefit of chronic GVHD protection among PTCy recipients to that of SibCNI reference cohort. Based on this analysis, CMV serostatus should be incorporated into risk stratification for PTCy-based GVHD prophylaxis, especially since the role of PTCy is rapidly expanding into HLA-identical transplants. CMV seropositive recipients and patients with a seropositive donors should be considered high-risk for CMV infection when a PTCy-based GVHD prophylaxis strategy is planned. Vigilant monitoring for CMV and anti-CMV antiviral prophylaxis should be considered. Of note, all patients in the current analysis were treated between 2012 and 2017, before the introduction of letermovir.11  Future analysis should investigate the role of letermovir prophylaxis in CMV-seropositive recipients and patients with CMV-seropositive donors receiving PTCy.

Dr. Feeney and Dr. O’Dwyer indicated no relevant conflicts of interest.

1.
Neiman
P
,
Wasserman
PB
,
Wentworth
BB
, et al
.
Interstitial pneumonia and cytomegalovirus infection as complications of human marrow transplantation
.
Transplantation
.
1973
;
15
(
5
):
478
485
.
2.
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
.
3.
Schmidt-Hieber
M
,
Labopin
M
,
Beelen
D
, et al
.
CMV serostatus still has an important prognostic impact in de novo acute leukemia patients after allogeneic stem cell transplantation: a report from the Acute Leukemia Working Party of EBMT
.
Blood
.
2013
;
122
(
19
):
3359
3364
.
4.
Luznik
L
,
O’Donnell
PV
,
Symons
HJ
, et al
.
HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide
.
Biol Blood Marrow Transplant
.
2008
;
14
(
6
):
641
650
.
5.
Passweg
JR
,
Baldomero
H
,
Chabannon
C
, et al
.
The EBMT activity survey on hematopoietic-cell transplantation and cellular therapy 2018: CAR-T's come into focus
.
Bone Marrow Transplant
.
2020
;
55
(
8
):
1604
1613
.
6.
D’Souza
A
,
Fretham
C
,
Lee
SJ
, et al
.
Current use of and trends in hematopoietic cell transplantation in the United States
.
Biol Blood Marrow Transplant
.
2020
;
26
(
8
):
e177
e182
.
7.
Goldsmith
SR
,
Slade
M
,
DiPersio
JF
, et al
.
Cytomegalovirus viremia, disease, and impact on relapse in T-cell replete peripheral blood haploidentical hematopoietic cell transplantation with post-transplant cyclophosphamide
.
Haematologica
.
2016
;
101
(
11
):
e465
e468
.
8.
Ramlal
R
,
Sasaki
K
,
Lozano Cerrada
S
, et al
.
Viral reactivation in haploidentical transplants using post-transplantation cyclophosphamide – a single institution experience
.
Transplant and Cellular Therapy
.
2016
;
22
(
3_supplement
):
S375
-
S376
.
9.
Shmueli
E
,
Or
R
,
Shapira
MY
, et al
.
High rate of cytomegalovirus drug resistance among patients receiving preemptive antiviral treatment after haploidentical stem cell transplantation
.
J Infect Dis
.
2014
;
209
(
4
):
557
561
.
10.
Della Chiesa
M
,
Moretta
L
,
Muccio
L
, et al
.
Haploidentical haematopoietic stem cell transplantation: Role of NK cells and effect of cytomegalovirus infections
.
Curr Top Microbiol Immunol
.
2016
;
395
:
209
224
.
11.
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
.